1
|
Aldridge JL, Alexander ED, Franklin AA, Frasier CR. Decreased ability to manage increases in reactive oxygen species may underlie susceptibility to arrhythmias in mice lacking Scn1b. Am J Physiol Heart Circ Physiol 2024; 327:H723-H732. [PMID: 39120465 PMCID: PMC11482272 DOI: 10.1152/ajpheart.00265.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Scn1b plays essential roles in the heart, where it encodes β1-subunits that serve as modifiers of gene expression, cell surface channel activity, and cardiac conductivity. Reduced β1 function is linked to electrical instability in various diseases with cardiac manifestations and increased susceptibility to arrhythmias. Recently, we demonstrated that loss of Scn1b in mice leads to compromised mitochondria energetics and reactive oxygen species (ROS) production. In this study, we examined the link between increased ROS and arrhythmia susceptibility in Scn1b-/- mice. In addition, ROS-scavenging capacity can be overwhelmed during prolonged oxidative stress, increasing arrhythmia susceptibility. Therefore, we isolated whole hearts and cardiomyocytes from Scn1b-/- and Scn1b+/+ mice and subjected them to an oxidative challenge with diamide, a glutathione oxidant. Next, we analyzed gene expression and activity of antioxidant enzymes in Scn1b-/- hearts. Cells isolated from Scn1b-/- hearts died faster and displayed higher rates of ROS accumulation preceding cell death compared with those from Scn1b+/+. Furthermore, Scn1b-/- hearts showed higher arrhythmia scores and spent less time free of arrhythmia. Lastly, we found that protein expression and enzymatic activity of glutathione peroxidase is increased in Scn1b-/- hearts compared with wild type. Our results indicate that Scn1b-/- mice have decreased capability to manage ROS during prolonged oxidative stress. ROS accumulation is elevated and appears to overwhelm ROS scavenging through the glutathione system. This imbalance creates the potential for altered cell energetics that may underlie increased susceptibility to arrhythmias or other adverse cardiac outcomes.NEW & NOTEWORTHY Using an oxidative challenge, we demonstrated that isolated cells from Scn1b-/- mice are more susceptible to cell death and surges in reactive oxygen species accumulation. At the whole organ level, they were also more susceptible to the formation of cardiac arrhythmias. This may in part be due to changes to the glutathione antioxidant system.
Collapse
Affiliation(s)
- Jessa L Aldridge
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, United States
| | - Emily Davis Alexander
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, United States
| | - Allison A Franklin
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, United States
| | - Chad R Frasier
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, United States
| |
Collapse
|
2
|
Aldridge JL, Alexander ED, Franklin AA, Harrington E, Al-Ghzawi F, Frasier CR. Sex differences in cardiac mitochondrial respiration and reactive oxygen species production may predispose Scn1a -/+ mice to cardiac arrhythmias and Sudden Unexpected Death in Epilepsy. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 9:100090. [PMID: 39390983 PMCID: PMC11466061 DOI: 10.1016/j.jmccpl.2024.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Dravet Syndrome (DS) is a pediatric-onset epilepsy with an elevated risk of Sudden Unexpected Death in Epilepsy (SUDEP). Most individuals with DS possess mutations in the voltage-gated sodium channel gene Scn1a, expressed in both the brain and heart. Previously, mutations in Scn1a have been linked to arrhythmia. We used a Scn1a -/+ DS mouse model to investigate changes to cardiac mitochondrial function that may underlie arrhythmias and SUDEP. We detected significant alterations in mitochondrial bioenergetics that were sex-specific. Mitochondria from male Scn1a -/+ hearts had deficits in maximal (p = 0.02) and Complex II-linked respiration (p = 0.03). Male Scn1a -/+ mice were also more susceptible to cardiac arrhythmias under increased workload. When isolated cardiomyocytes were subjected to diamide, cardiomyocytes from male Scn1a -/+ hearts were less resistant to thiol oxidation. They had decreased survivability compared to Scn1a +/+ (p = 0.02) despite no whole-heart differences. Lastly, there were no changes in mitochondrial ROS production between DS and wild-type mitochondria at basal conditions, but Scn1a -/+ mitochondria accumulated more ROS during hypoxia/reperfusion. This study determines novel sex-linked differences in mitochondrial and antioxidant function in Scn1a-linked DS. Importantly, we found that male Scn1a -/+ mice are more susceptible to cardiac arrhythmias than female Scn1a -/+ mice. When developing new therapeutics to address SUDEP risk in DS, sex should be considered.
Collapse
Affiliation(s)
- Jessa L. Aldridge
- East Tennessee State University, Quillen College of Medicine, Department of Biomedical Sciences, Johnson City, TN, United States of America
| | - Emily Davis Alexander
- East Tennessee State University, Quillen College of Medicine, Department of Biomedical Sciences, Johnson City, TN, United States of America
| | - Allison A. Franklin
- East Tennessee State University, Quillen College of Medicine, Department of Biomedical Sciences, Johnson City, TN, United States of America
| | - Elizabeth Harrington
- East Tennessee State University, Quillen College of Medicine, Department of Biomedical Sciences, Johnson City, TN, United States of America
| | - Farah Al-Ghzawi
- East Tennessee State University, Quillen College of Medicine, Department of Biomedical Sciences, Johnson City, TN, United States of America
| | - Chad R. Frasier
- East Tennessee State University, Quillen College of Medicine, Department of Biomedical Sciences, Johnson City, TN, United States of America
| |
Collapse
|
3
|
Aldridge JL, Alexander ED, Franklin AA, Frasier CR. Altered cardiac energetics in mice lacking Scn1b. Cardiovasc Res 2024; 120:979-981. [PMID: 38641853 PMCID: PMC11288732 DOI: 10.1093/cvr/cvae087] [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: 12/21/2023] [Revised: 02/21/2024] [Accepted: 02/29/2024] [Indexed: 04/21/2024] Open
Affiliation(s)
- Jessa L Aldridge
- Quillen College of Medicine, Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| | - Emily Davis Alexander
- Quillen College of Medicine, Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| | - Allison A Franklin
- Quillen College of Medicine, Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| | - Chad R Frasier
- Quillen College of Medicine, Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
4
|
Williams ZJ, Payne LB, Wu X, Gourdie RG. New focus on cardiac voltage-gated sodium channel β1 and β1B: Novel targets for treating and understanding arrhythmias? Heart Rhythm 2024:S1547-5271(24)02742-5. [PMID: 38908461 DOI: 10.1016/j.hrthm.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Voltage-gated sodium channels (VGSCs) are transmembrane protein complexes that are vital to the generation and propagation of action potentials in nerve and muscle fibers. The canonical VGSC is generally conceived as a heterotrimeric complex formed by 2 classes of membrane-spanning subunit: an α-subunit (pore forming) and 2 β-subunits (non-pore forming). NaV1.5 is the main sodium channel α-subunit of mammalian ventricle, with lower amounts of other α-subunits, including NaV1.6, being present. There are 4 β-subunits (β1-β4) encoded by 4 genes (SCN1B-SCN4B), each of which is expressed in cardiac tissues. Recent studies suggest that in addition to assignments in channel gating and trafficking, products of Scn1b may have novel roles in conduction of action potential in the heart and intracellular signaling. This includes evidence that the β-subunit extracellular amino-terminal domain facilitates adhesive interactions in intercalated discs and that its carboxyl-terminal region is a substrate for a regulated intramembrane proteolysis (RIP) signaling pathway, with a carboxyl-terminal peptide generated by β1 RIP trafficked to the nucleus and altering transcription of various genes, including NaV1.5. In addition to β1, the Scn1b gene encodes for an alternative splice variant, β1B, which contains an identical extracellular adhesion domain to β1 but has a unique carboxyl-terminus. Although β1B is generally understood to be a secreted variant, evidence indicates that when co-expressed with NaV1.5, it is maintained at the cell membrane, suggesting potential unique roles for this understudied protein. In this review, we focus on what is known of the 2 β-subunit variants encoded by Scn1b in heart, with particular focus on recent findings and the questions raised by this new information. We also explore data that indicate β1 and β1B may be attractive targets for novel antiarrhythmic therapeutics.
Collapse
Affiliation(s)
- Zachary J Williams
- Fralin Biomedical Research Institute, Virginia Polytechnic University, Roanoke, Virginia
| | - Laura Beth Payne
- Fralin Biomedical Research Institute, Virginia Polytechnic University, Roanoke, Virginia
| | - Xiaobo Wu
- Fralin Biomedical Research Institute, Virginia Polytechnic University, Roanoke, Virginia
| | - Robert G Gourdie
- Fralin Biomedical Research Institute, Virginia Polytechnic University, Roanoke, Virginia; School of Medicine, Virgina Polytechnic University, Roanoke, Virginia; Department of Biomedical Engineering and Mechanics, Virginia Polytechnic University, Blacksburg, Virginia.
| |
Collapse
|
5
|
Moreno-Manuel AI, Macías Á, Cruz FM, Gutiérrez LK, Martínez F, González-Guerra A, Martínez Carrascoso I, Bermúdez-Jimenez FJ, Sánchez-Pérez P, Vera-Pedrosa ML, Ruiz-Robles JM, Bernal JA, Jalife J. The Kir2.1E299V mutation increases atrial fibrillation vulnerability while protecting the ventricles against arrhythmias in a mouse model of short QT syndrome type 3. Cardiovasc Res 2024; 120:490-505. [PMID: 38261726 PMCID: PMC11060485 DOI: 10.1093/cvr/cvae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/24/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024] Open
Abstract
AIMS Short QT syndrome type 3 (SQTS3) is a rare arrhythmogenic disease caused by gain-of-function mutations in KCNJ2, the gene coding the inward rectifier potassium channel Kir2.1. We used a multidisciplinary approach and investigated arrhythmogenic mechanisms in an in-vivo model of de-novo mutation Kir2.1E299V identified in a patient presenting an extremely abbreviated QT interval and paroxysmal atrial fibrillation. METHODS AND RESULTS We used intravenous adeno-associated virus-mediated gene transfer to generate mouse models, and confirmed cardiac-specific expression of Kir2.1WT or Kir2.1E299V. On ECG, the Kir2.1E299V mouse recapitulated the QT interval shortening and the atrial-specific arrhythmia of the patient. The PR interval was also significantly shorter in Kir2.1E299V mice. Patch-clamping showed extremely abbreviated action potentials in both atrial and ventricular Kir2.1E299V cardiomyocytes due to a lack of inward-going rectification and increased IK1 at voltages positive to -80 mV. Relative to Kir2.1WT, atrial Kir2.1E299V cardiomyocytes had a significantly reduced slope conductance at voltages negative to -80 mV. After confirming a higher proportion of heterotetrameric Kir2.x channels containing Kir2.2 subunits in the atria, in-silico 3D simulations predicted an atrial-specific impairment of polyamine block and reduced pore diameter in the Kir2.1E299V-Kir2.2WT channel. In ventricular cardiomyocytes, the mutation increased excitability by shifting INa activation and inactivation in the hyperpolarizing direction, which protected the ventricle against arrhythmia. Moreover, Purkinje myocytes from Kir2.1E299V mice manifested substantially higher INa density than Kir2.1WT, explaining the abbreviation in the PR interval. CONCLUSION The first in-vivo mouse model of cardiac-specific SQTS3 recapitulates the electrophysiological phenotype of a patient with the Kir2.1E299V mutation. Kir2.1E299V eliminates rectification in both cardiac chambers but protects against ventricular arrhythmias by increasing excitability in both Purkinje-fiber network and ventricles. Consequently, the predominant arrhythmias are supraventricular likely due to the lack of inward rectification and atrial-specific reduced pore diameter of the Kir2.1E299V-Kir2.2WT heterotetramer.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Action Potentials
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/physiopathology
- Arrhythmias, Cardiac/metabolism
- Atrial Fibrillation/genetics
- Atrial Fibrillation/physiopathology
- Atrial Fibrillation/metabolism
- Disease Models, Animal
- Genetic Predisposition to Disease
- Heart Rate/genetics
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Mice, Inbred C57BL
- Mice, Transgenic
- Mutation
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Phenotype
- Potassium Channels, Inwardly Rectifying/genetics
- Potassium Channels, Inwardly Rectifying/metabolism
Collapse
Affiliation(s)
- Ana I Moreno-Manuel
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Álvaro Macías
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Francisco M Cruz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Lilian K Gutiérrez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Fernando Martínez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Andrés González-Guerra
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Isabel Martínez Carrascoso
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Francisco José Bermúdez-Jimenez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Department of Cardiology, Hospital Universitario Virgen de las Nieves, 18014 Granada, Spain
| | - Patricia Sánchez-Pérez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | | | - Juan Manuel Ruiz-Robles
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Juan A Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Departments of Internal Medicine and Molecular and Integrative Physiology, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 4810, USA
| |
Collapse
|
6
|
Sanchez-Sandoval AL, Hernández-Plata E, Gomora JC. Voltage-gated sodium channels: from roles and mechanisms in the metastatic cell behavior to clinical potential as therapeutic targets. Front Pharmacol 2023; 14:1206136. [PMID: 37456756 PMCID: PMC10348687 DOI: 10.3389/fphar.2023.1206136] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
During the second half of the last century, the prevalent knowledge recognized the voltage-gated sodium channels (VGSCs) as the proteins responsible for the generation and propagation of action potentials in excitable cells. However, over the last 25 years, new non-canonical roles of VGSCs in cancer hallmarks have been uncovered. Their dysregulated expression and activity have been associated with aggressive features and cancer progression towards metastatic stages, suggesting the potential use of VGSCs as cancer markers and prognostic factors. Recent work has elicited essential information about the signalling pathways modulated by these channels: coupling membrane activity to transcriptional regulation pathways, intracellular and extracellular pH regulation, invadopodia maturation, and proteolytic activity. In a promising scenario, the inhibition of VGSCs with FDA-approved drugs as well as with new synthetic compounds, reduces cancer cell invasion in vitro and cancer progression in vivo. The purpose of this review is to present an update regarding recent advances and ongoing efforts to have a better understanding of molecular and cellular mechanisms on the involvement of both pore-forming α and auxiliary β subunits of VGSCs in the metastatic processes, with the aim at proposing VGSCs as new oncological markers and targets for anticancer treatments.
Collapse
Affiliation(s)
- Ana Laura Sanchez-Sandoval
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Medicina Genómica, Hospital General de México “Dr Eduardo Liceaga”, Mexico City, Mexico
| | - Everardo Hernández-Plata
- Consejo Nacional de Humanidades, Ciencias y Tecnologías and Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
7
|
Salvage SC, Jeevaratnam K, Huang CL, Jackson AP. Cardiac sodium channel complexes and arrhythmia: structural and functional roles of the β1 and β3 subunits. J Physiol 2023; 601:923-940. [PMID: 36354758 PMCID: PMC10953345 DOI: 10.1113/jp283085] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/04/2022] [Indexed: 11/12/2022] Open
Abstract
In cardiac myocytes, the voltage-gated sodium channel NaV 1.5 opens in response to membrane depolarisation and initiates the action potential. The NaV 1.5 channel is typically associated with regulatory β-subunits that modify gating and trafficking behaviour. These β-subunits contain a single extracellular immunoglobulin (Ig) domain, a single transmembrane α-helix and an intracellular region. Here we focus on the role of the β1 and β3 subunits in regulating NaV 1.5. We catalogue β1 and β3 domain specific mutations that have been associated with inherited cardiac arrhythmia, including Brugada syndrome, long QT syndrome, atrial fibrillation and sudden death. We discuss how new structural insights into these proteins raises new questions about physiological function.
Collapse
Affiliation(s)
| | | | - Christopher L.‐H. Huang
- Department of BiochemistryUniversity of CambridgeCambridgeUK
- Department of PhysiologyDevelopment and NeuroscienceUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
8
|
Inácio JM, Nunes MM, Almeida M, Cristo F, Anjos R, Belo JA. Gene-Edited Human-Induced Pluripotent Stem Cell Lines to Elucidate DAND5 Function throughout Cardiac Differentiation. Cells 2023; 12:520. [PMID: 36831187 PMCID: PMC9954670 DOI: 10.3390/cells12040520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
(1) Background: The contribution of gene-specific variants for congenital heart disease, one of the most common congenital disabilities, is still far from our complete understanding. Here, we applied a disease model using human-induced pluripotent stem cells (hiPSCs) to evaluate the function of DAND5 on human cardiomyocyte (CM) differentiation and proliferation. (2) Methods: Taking advantage of our DAND5 patient-derived iPSC line, we used CRISPR-Cas9 gene-editing to generate a set of isogenic hiPSCs (DAND5-corrected and DAND5 full-mutant). The hiPSCs were differentiated into CMs, and RT-qPCR and immunofluorescence profiled the expression of cardiac markers. Cardiomyocyte proliferation was analysed by flow cytometry. Furthermore, we used a multi-electrode array (MEA) to study the functional electrophysiology of DAND5 hiPSC-CMs. (3) Results: The results indicated that hiPSC-CM proliferation is affected by DAND5 levels. Cardiomyocytes derived from a DAND5 full-mutant hiPSC line are more proliferative when compared with gene-corrected hiPSC-CMs. Moreover, parallel cardiac differentiations showed a differential cardiac gene expression profile, with upregulated cardiac progenitor markers in DAND5-KO hiPSC-CMs. Microelectrode array (MEA) measurements demonstrated that DAND5-KO hiPSC-CMs showed prolonged field potential duration and increased spontaneous beating rates. In addition, conduction velocity is reduced in the monolayers of hiPSC-CMs with full-mutant genotype. (4) Conclusions: The absence of DAND5 sustains the proliferation of hiPSC-CMs, which alters their electrophysiological maturation properties. These results using DAND5 hiPSC-CMs consolidate the findings of the in vitro and in vivo mouse models, now in a translational perspective. Altogether, the data will help elucidate the molecular mechanism underlying this human heart disease and potentiates new therapies for treating adult CHD.
Collapse
Affiliation(s)
- José M. Inácio
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
| | - Mafalda M. Nunes
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
| | - Micael Almeida
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
| | - Fernando Cristo
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
| | - Rui Anjos
- Hospital de Santa Cruz, Centro Hospitalar Lisboa Ocidental, 1449-005 Lisboa, Portugal
| | - José A. Belo
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
| |
Collapse
|
9
|
Zhu Z, Bolt E, Newmaster K, Osei-Bonsu W, Cohen S, Cuddapah VA, Gupta S, Paudel S, Samanta D, Dang LT, Carney PR, Naik S. SCN1B Genetic Variants: A Review of the Spectrum of Clinical Phenotypes and a Report of Early Myoclonic Encephalopathy. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1507. [PMID: 36291443 PMCID: PMC9600564 DOI: 10.3390/children9101507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022]
Abstract
Background: Pathogenic variants in SCN1B, the gene encoding voltage-gated sodium channel b1/b1B subunits are associated with a spectrum of epileptic disorders. This study describes a child with early myoclonic encephalopathy and a compound heterozygous variant in the SCN1B gene (p.Arg85Cys and c.3G>C/p.Met1), along with the child’s clinical response to anti-seizure medications (ASMs) and the ketogenic diet. We reviewed the current clinical literature pertinent to SCN1B-related epilepsy. Methods: We described the evaluation and management of a patient with SCN1B-related developmental and epileptic encephalopathy (DEE). We used the Medline and Pubmed databases to review the various neurological manifestations associated with SCN1B genetic variants, and summarize the functional studies performed on SCN1B variants. Results: We identified 20 families and six individuals (including the index case described herein) reported to have SCN1B-related epilepsy. Individuals with monoallelic pathogenic variants in SCN1B often present with genetic epilepsy with febrile seizures plus (GEFS+), while those with biallelic pathogenic variants may present with developmental and epileptic encephalopathy (DEE). Individuals with DEE present with seizures of various semiologies (commonly myoclonic seizures) and status epilepticus at early infancy and are treated with various antiseizure medications. In our index case, adjunctive fenfluramine was started at 8 months of age at 0.2 mg/kg/day with gradual incremental increases to the final dose of 0.7 mg/kg/day over 5 weeks. Fenfluramine was effective in the treatment of seizures, resulting in a 50% reduction in myoclonic seizures, status epilepticus, and generalized tonic-clonic seizures, as well as a 70−90% reduction in focal seizures, with no significant adverse effects. Following the initiation of fenfluramine at eight months of age, there was also a 50% reduction in the rate of hospitalizations. Conclusions: SCN1B pathogenic variants cause epilepsy and neurodevelopmental impairment with variable expressivity and incomplete penetrance. The severity of disease is associated with the zygosity of the pathogenic variants. Biallelic variants in SCN1B can result in early myoclonic encephalopathy, and adjunctive treatment with fenfluramine may be an effective treatment for SCN1B-related DEE. Further research on the efficacy and safety of using newer ASMs, such as fenfluramine in patients under the age of 2 years is needed.
Collapse
Affiliation(s)
- Zahra Zhu
- College of Medicine, Penn State University, Hershey, PA 17033, USA
| | - Elizabeth Bolt
- College of Medicine, Penn State University, Hershey, PA 17033, USA
| | - Kyra Newmaster
- College of Medicine, Penn State University, Hershey, PA 17033, USA
| | - Wendy Osei-Bonsu
- College of Medicine, Penn State University, Hershey, PA 17033, USA
| | - Stacey Cohen
- Epilepsy Neurogenetics Initiative, Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Vishnu Anand Cuddapah
- Epilepsy Neurogenetics Initiative, Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Siddharth Gupta
- Kennedy Krieger Institute, Department of Neurology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sita Paudel
- Department of Pediatrics and Neurology, Penn State Health Milton Hershey Medical Center, Hershey, PA 17033, USA
| | - Debopam Samanta
- Division of Pediatric Neurology, Arkansas Children’s Hospital, Little Rock, AR 72202, USA
| | - Louis T. Dang
- Department of Pediatrics, Division of Pediatric Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Paul R. Carney
- Pediatric Neurology Division, University of Missouri Health Care, Columbia, MO 65212, USA
| | - Sunil Naik
- Department of Pediatrics and Neurology, Penn State Health Milton Hershey Medical Center, Hershey, PA 17033, USA
| |
Collapse
|
10
|
Hodges SL, Bouza AA, Isom LL. Therapeutic Potential of Targeting Regulated Intramembrane Proteolysis Mechanisms of Voltage-Gated Ion Channel Subunits and Cell Adhesion Molecules. Pharmacol Rev 2022; 74:1028-1048. [PMID: 36113879 PMCID: PMC9553118 DOI: 10.1124/pharmrev.121.000340] [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: 11/08/2021] [Accepted: 05/13/2022] [Indexed: 10/03/2023] Open
Abstract
Several integral membrane proteins undergo regulated intramembrane proteolysis (RIP), a tightly controlled process through which cells transmit information across and between intracellular compartments. RIP generates biologically active peptides by a series of proteolytic cleavage events carried out by two primary groups of enzymes: sheddases and intramembrane-cleaving proteases (iCLiPs). Following RIP, fragments of both pore-forming and non-pore-forming ion channel subunits, as well as immunoglobulin super family (IgSF) members, have been shown to translocate to the nucleus to function in transcriptional regulation. As an example, the voltage-gated sodium channel β1 subunit, which is also an IgSF-cell adhesion molecule (CAM), is a substrate for RIP. β1 RIP results in generation of a soluble intracellular domain, which can regulate gene expression in the nucleus. In this review, we discuss the proposed RIP mechanisms of voltage-gated sodium, potassium, and calcium channel subunits as well as the roles of their generated proteolytic products in the nucleus. We also discuss other RIP substrates that are cleaved by similar sheddases and iCLiPs, such as IgSF macromolecules, including CAMs, whose proteolytically generated fragments function in the nucleus. Importantly, dysfunctional RIP mechanisms are linked to human disease. Thus, we will also review how understanding RIP events and subsequent signaling processes involving ion channel subunits and IgSF proteins may lead to the discovery of novel therapeutic targets. SIGNIFICANCE STATEMENT: Several ion channel subunits and immunoglobulin superfamily molecules have been identified as substrates of regulated intramembrane proteolysis (RIP). This signal transduction mechanism, which generates polypeptide fragments that translocate to the nucleus, is an important regulator of gene transcription. RIP may impact diseases of excitability, including epilepsy, cardiac arrhythmia, and sudden death syndromes. A thorough understanding of the role of RIP in gene regulation is critical as it may reveal novel therapeutic strategies for the treatment of previously intractable diseases.
Collapse
Affiliation(s)
- Samantha L Hodges
- Departments of Pharmacology (S.L.H., A.A.B., L.L.I.), Neurology (L.L.I.), and Molecular & Integrative Physiology (L.L.I.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Alexandra A Bouza
- Departments of Pharmacology (S.L.H., A.A.B., L.L.I.), Neurology (L.L.I.), and Molecular & Integrative Physiology (L.L.I.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Lori L Isom
- Departments of Pharmacology (S.L.H., A.A.B., L.L.I.), Neurology (L.L.I.), and Molecular & Integrative Physiology (L.L.I.), University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
11
|
Cervantes DO, Pizzo E, Ketkar H, Parambath SP, Tang S, Cianflone E, Cannata A, Vinukonda G, Jain S, Jacobson JT, Rota M. Scn1b expression in the adult mouse heart modulates Na + influx in myocytes and reveals a mechanistic link between Na + entry and diastolic function. Am J Physiol Heart Circ Physiol 2022; 322:H975-H993. [PMID: 35394857 PMCID: PMC9076421 DOI: 10.1152/ajpheart.00465.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 11/22/2022]
Abstract
Voltage-gated sodium channels (VGSCs) are macromolecular assemblies composed of a number of proteins regulating channel conductance and properties. VGSCs generate Na+ current (INa) in myocytes and play fundamental roles in excitability and impulse conduction in the heart. Moreover, VGSCs condition mechanical properties of the myocardium, a process that appears to involve the late component of INa. Variants in the gene SCN1B, encoding the VGSC β1- and β1B-subunits, result in inherited neurological disorders and cardiac arrhythmias. But the precise contributions of β1/β1B-subunits and VGSC integrity to the overall function of the adult heart remain to be clarified. For this purpose, adult mice with cardiac-restricted, inducible deletion of Scn1b (conditional knockout, cKO) were studied. Myocytes from cKO mice had increased densities of fast (+20%)- and slow (+140%)-inactivating components of INa, with respect to control cells. By echocardiography and invasive hemodynamics, systolic function was preserved in cKO mice, but diastolic properties and ventricular compliance were compromised, with respect to control animals. Importantly, inhibition of late INa with GS967 normalized left ventricular filling pattern and isovolumic relaxation time in cKO mice. At the cellular level, cKO myocytes presented delayed kinetics of Ca2+ transients and cell mechanics, defects that were corrected by inhibition of INa. Collectively, these results document that VGSC β1/β1B-subunits modulate electrical and mechanical function of the heart by regulating, at least in part, Na+ influx in cardiomyocytes.NEW & NOTEWORTHY We have investigated the consequences of deletion of Scn1b, the gene encoding voltage-gated sodium channel β1-subunits, on myocyte and cardiac function. Our findings support the notion that Scn1b expression controls properties of Na+ influx and Ca2+ cycling in cardiomyocytes affecting the modality of cell contraction and relaxation. These effects at the cellular level condition electrical recovery and diastolic function in vivo, substantiating the multifunctional role of β1-subunits in the physiology of the heart.
Collapse
Affiliation(s)
| | - Emanuele Pizzo
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Harshada Ketkar
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, New York
| | - Sreema P Parambath
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, New York
| | - Samantha Tang
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, New York
| | - Eleonora Cianflone
- Department of Physiology, New York Medical College, Valhalla, New York
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Antonio Cannata
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | | | - Sudhir Jain
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, New York
| | - Jason T Jacobson
- Department of Physiology, New York Medical College, Valhalla, New York
- Department of Cardiology, Westchester Medical Center, Valhalla, New York
| | - Marcello Rota
- Department of Physiology, New York Medical College, Valhalla, New York
| |
Collapse
|
12
|
Ramos-Mondragon R, Edokobi N, Hodges SL, Wang S, Bouza AA, Canugovi C, Scheuing C, Juratli L, Abel WR, Noujaim SF, Madamanchi NR, Runge MS, Lopez-Santiago LF, Isom LL. Neonatal Scn1b-null mice have sinoatrial node dysfunction, altered atrial structure, and atrial fibrillation. JCI Insight 2022; 7:152050. [PMID: 35603785 PMCID: PMC9220823 DOI: 10.1172/jci.insight.152050] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
Loss-of-function (LOF) variants in SCN1B, encoding the voltage-gated sodium channel β1/β1B subunits, are linked to neurological and cardiovascular diseases. Scn1b-null mice have spontaneous seizures and ventricular arrhythmias and die by approximately 21 days after birth. β1/β1B Subunits play critical roles in regulating the excitability of ventricular cardiomyocytes and maintaining ventricular rhythmicity. However, whether they also regulate atrial excitability is unknown. We used neonatal Scn1b-null mice to model the effects of SCN1B LOF on atrial physiology in pediatric patients. Scn1b deletion resulted in altered expression of genes associated with atrial dysfunction. Scn1b-null hearts had a significant accumulation of atrial collagen, increased susceptibility to pacing induced atrial fibrillation (AF), sinoatrial node (SAN) dysfunction, and increased numbers of cholinergic neurons in ganglia that innervate the SAN. Atropine reduced the incidence of AF in null animals. Action potential duration was prolonged in null atrial myocytes, with increased late sodium current density and reduced L-type calcium current density. Scn1b LOF results in altered atrial structure and AF, demonstrating the critical role played by Scn1b in atrial physiology during early postnatal mouse development. Our results suggest that SCN1B LOF variants may significantly impact the developing pediatric heart.
Collapse
Affiliation(s)
| | | | | | | | | | - Chandrika Canugovi
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | | | - Sami F. Noujaim
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Nageswara R. Madamanchi
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Marschall S. Runge
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Lori L. Isom
- Department of Pharmacology and
- Department of Neurology and
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
13
|
O' Brien S, Holmes AP, Johnson DM, Kabir SN, O' Shea C, O' Reilly M, Avezzu A, Reyat JS, Hall AW, Apicella C, Ellinor PT, Niederer S, Tucker NR, Fabritz L, Kirchhof P, Pavlovic D. Increased atrial effectiveness of flecainide conferred by altered biophysical properties of sodium channels. J Mol Cell Cardiol 2022; 166:23-35. [PMID: 35114252 PMCID: PMC7616974 DOI: 10.1016/j.yjmcc.2022.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/13/2022] [Accepted: 01/26/2022] [Indexed: 11/25/2022]
Abstract
Atrial fibrillation (AF) affects over 1% of the population and is a leading cause of stroke and heart failure in the elderly. A feared side effect of sodium channel blocker therapy, ventricular pro-arrhythmia, appears to be relatively rare in patients with AF. The biophysical reasons for this relative safety of sodium blockers are not known. Our data demonstrates intrinsic differences between atrial and ventricular cardiac voltage-gated sodium currents (INa), leading to reduced maximum upstroke velocity of action potential and slower conduction, in left atria compared to ventricle. Reduced atrial INa is only detected at physiological membrane potentials and is driven by alterations in sodium channel biophysical properties and not by NaV1.5 protein expression. Flecainide displayed greater inhibition of atrial INa, greater reduction of maximum upstroke velocity of action potential, and slowed conduction in atrial cells and tissue. Our work highlights differences in biophysical properties of sodium channels in left atria and ventricles and their response to flecainide. These differences can explain the relative safety of sodium channel blocker therapy in patients with atrial fibrillation.
Collapse
Affiliation(s)
- Sian O' Brien
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK
| | - Andrew P Holmes
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK; School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
| | - Daniel M Johnson
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK; School of Life, Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, UK
| | - S Nashitha Kabir
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK
| | - Christopher O' Shea
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK
| | - Molly O' Reilly
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK
| | - Adelisa Avezzu
- School of Biomedical Engineering & Imaging Sciences, Kings' College London, London, UK
| | - Jasmeet S Reyat
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK
| | - Amelia W Hall
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02129, USA; Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Clara Apicella
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK
| | - Patrick T Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02129, USA; Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Steven Niederer
- School of Biomedical Engineering & Imaging Sciences, Kings' College London, London, UK
| | - Nathan R Tucker
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02129, USA; Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Masonic Medical Research Institute, Utica, NY, 13501, USA
| | - Larissa Fabritz
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK; University Center of Cardiovascular Science, University Heart and Vascular Center UKE, Hamburg, Germany; Department of Cardiology, University Heart and Vascular Center UKE, Hamburg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Paulus Kirchhof
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK; Department of Cardiology, University Heart and Vascular Center UKE, Hamburg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Davor Pavlovic
- Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK.
| |
Collapse
|
14
|
Rubio-Alarcón M, Cámara-Checa A, Dago M, Crespo-García T, Nieto-Marín P, Marín M, Merino JL, Toquero J, Salguero-Bodes R, Tamargo J, Cebrián J, Delpón E, Caballero R. Zfhx3 Transcription Factor Represses the Expression of SCN5A Gene and Decreases Sodium Current Density (I Na). Int J Mol Sci 2021; 22:13031. [PMID: 34884836 PMCID: PMC8657907 DOI: 10.3390/ijms222313031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 02/02/2023] Open
Abstract
The ZFHX3 and SCN5A genes encode the zinc finger homeobox 3 (Zfhx3) transcription factor (TF) and the human cardiac Na+ channel (Nav1.5), respectively. The effects of Zfhx3 on the expression of the Nav1.5 channel, and in cardiac excitability, are currently unknown. Additionally, we identified three Zfhx3 variants in probands diagnosed with familial atrial fibrillation (p.M1260T) and Brugada Syndrome (p.V949I and p.Q2564R). Here, we analyzed the effects of native (WT) and mutated Zfhx3 on Na+ current (INa) recorded in HL-1 cardiomyocytes. ZFHX3 mRNA can be detected in human atrial and ventricular samples. In HL-1 cardiomyocytes, transfection of Zfhx3 strongly reduced peak INa density, while the silencing of endogenous expression augmented it (from -65.9 ± 8.9 to -104.6 ± 10.8 pA/pF; n ≥ 8, p < 0.05). Zfhx3 significantly reduced the transcriptional activity of human SCN5A, PITX2, TBX5, and NKX25 minimal promoters. Consequently, the mRNA and/or protein expression levels of Nav1.5 and Tbx5 were diminished (n ≥ 6, p < 0.05). Zfhx3 also increased the expression of Nedd4-2 ubiquitin-protein ligase, enhancing Nav1.5 proteasomal degradation. p.V949I, p.M1260T, and p.Q2564R Zfhx3 produced similar effects on INa density and time- and voltage-dependent properties in WT. WT Zfhx3 inhibits INa as a result of a direct repressor effect on the SCN5A promoter, the modulation of Tbx5 increasing on the INa, and the increased expression of Nedd4-2. We propose that this TF participates in the control of cardiac excitability in human adult cardiac tissue.
Collapse
Affiliation(s)
- Marcos Rubio-Alarcón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - Anabel Cámara-Checa
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - María Dago
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - Teresa Crespo-García
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - Paloma Nieto-Marín
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - María Marín
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - José Luis Merino
- Department of Cardiology, Hospital Universitario La Paz, Instituto de Investigación Sanitaria la Paz, CIBERCV, 28046 Madrid, Spain;
| | - Jorge Toquero
- Department of Cardiology, Hospital Universitario Puerta de Hierro, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, CIBERCV, Majadahonda, 28222 Madrid, Spain;
| | - Rafael Salguero-Bodes
- Department of Cardiology, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre, CIBERCV, 28041 Madrid, Spain;
| | - Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - Jorge Cebrián
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Gregorio Marañón, CIBERCV, 28040 Madrid, Spain; (M.R.-A.); (A.C.-C.);; (T.C.-G.); (P.N.-M.); (M.M.); (J.T.); (E.D.); (R.C.)
| |
Collapse
|
15
|
Chen L, He Y, Wang X, Ge J, Li H. Ventricular voltage-gated ion channels: Detection, characteristics, mechanisms, and drug safety evaluation. Clin Transl Med 2021; 11:e530. [PMID: 34709746 PMCID: PMC8516344 DOI: 10.1002/ctm2.530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac voltage-gated ion channels (VGICs) play critical roles in mediating cardiac electrophysiological signals, such as action potentials, to maintain normal heart excitability and contraction. Inherited or acquired alterations in the structure, expression, or function of VGICs, as well as VGIC-related side effects of pharmaceutical drug delivery can result in abnormal cellular electrophysiological processes that induce life-threatening cardiac arrhythmias or even sudden cardiac death. Hence, to reduce possible heart-related risks, VGICs must be acknowledged as important targets in drug discovery and safety studies related to cardiac disease. In this review, we first summarize the development and application of electrophysiological techniques that are employed in cardiac VGIC studies alone or in combination with other techniques such as cryoelectron microscopy, optical imaging and optogenetics. Subsequently, we describe the characteristics, structure, mechanisms, and functions of various well-studied VGICs in ventricular myocytes and analyze their roles in and contributions to both physiological cardiac excitability and inherited cardiac diseases. Finally, we address the implications of the structure and function of ventricular VGICs for drug safety evaluation. In summary, multidisciplinary studies on VGICs help researchers discover potential targets of VGICs and novel VGICs in heart, enrich their knowledge of the properties and functions, determine the operation mechanisms of pathological VGICs, and introduce groundbreaking trends in drug therapy strategies, and drug safety evaluation.
Collapse
Affiliation(s)
- Lulan Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| | - Yue He
- Department of CardiologyShanghai Xuhui District Central Hospital & Zhongshan‐xuhui HospitalShanghaiChina
| | - Xiangdong Wang
- Institute of Clinical Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| | - Hua Li
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
16
|
Blandin CE, Gravez BJ, Hatem SN, Balse E. Remodeling of Ion Channel Trafficking and Cardiac Arrhythmias. Cells 2021; 10:cells10092417. [PMID: 34572065 PMCID: PMC8468138 DOI: 10.3390/cells10092417] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 01/08/2023] Open
Abstract
Both inherited and acquired cardiac arrhythmias are often associated with the abnormal functional expression of ion channels at the cellular level. The complex machinery that continuously traffics, anchors, organizes, and recycles ion channels at the plasma membrane of a cardiomyocyte appears to be a major source of channel dysfunction during cardiac arrhythmias. This has been well established with the discovery of mutations in the genes encoding several ion channels and ion channel partners during inherited cardiac arrhythmias. Fibrosis, altered myocyte contacts, and post-transcriptional protein changes are common factors that disorganize normal channel trafficking during acquired cardiac arrhythmias. Channel availability, described notably for hERG and KV1.5 channels, could be another potent arrhythmogenic mechanism. From this molecular knowledge on cardiac arrhythmias will emerge novel antiarrhythmic strategies.
Collapse
Affiliation(s)
- Camille E. Blandin
- INSERM, Unité de Recherche sur les Maladies Cardiovasculaires, le Métabolisme et la Nutrition—UNITE 1166, Sorbonne Université, EQUIPE 3, F-75013 Paris, France; (C.E.B.); (B.J.G.); (S.N.H.)
| | - Basile J. Gravez
- INSERM, Unité de Recherche sur les Maladies Cardiovasculaires, le Métabolisme et la Nutrition—UNITE 1166, Sorbonne Université, EQUIPE 3, F-75013 Paris, France; (C.E.B.); (B.J.G.); (S.N.H.)
| | - Stéphane N. Hatem
- INSERM, Unité de Recherche sur les Maladies Cardiovasculaires, le Métabolisme et la Nutrition—UNITE 1166, Sorbonne Université, EQUIPE 3, F-75013 Paris, France; (C.E.B.); (B.J.G.); (S.N.H.)
- ICAN—Institute of Cardiometabolism and Nutrition, Institute of Cardiology, Pitié-Salpêtrière Hospital, Sorbonne University, F-75013 Paris, France
| | - Elise Balse
- INSERM, Unité de Recherche sur les Maladies Cardiovasculaires, le Métabolisme et la Nutrition—UNITE 1166, Sorbonne Université, EQUIPE 3, F-75013 Paris, France; (C.E.B.); (B.J.G.); (S.N.H.)
- Correspondence:
| |
Collapse
|
17
|
Zhu W, Wang W, Angsutararux P, Mellor RL, Isom LL, Nerbonne JM, Silva JR. Modulation of the effects of class Ib antiarrhythmics on cardiac NaV1.5-encoded channels by accessory NaVβ subunits. JCI Insight 2021; 6:e143092. [PMID: 34156986 PMCID: PMC8410097 DOI: 10.1172/jci.insight.143092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 06/17/2021] [Indexed: 01/28/2023] Open
Abstract
Native myocardial voltage-gated sodium (NaV) channels function in macromolecular complexes comprising a pore-forming (α) subunit and multiple accessory proteins. Here, we investigated the impact of accessory NaVβ1 and NaVβ3 subunits on the functional effects of 2 well-known class Ib antiarrhythmics, lidocaine and ranolazine, on the predominant NaV channel α subunit, NaV1.5, expressed in the mammalian heart. We showed that both drugs stabilized the activated conformation of the voltage sensor of domain-III (DIII-VSD) in NaV1.5. In the presence of NaVβ1, the effect of lidocaine on the DIII-VSD was enhanced, whereas the effect of ranolazine was abolished. Mutating the main class Ib drug-binding site, F1760, affected but did not abolish the modulation of drug block by NaVβ1/β3. Recordings from adult mouse ventricular myocytes demonstrated that loss of Scn1b (NaVβ1) differentially affected the potencies of lidocaine and ranolazine. In vivo experiments revealed distinct ECG responses to i.p. injection of ranolazine or lidocaine in WT and Scn1b-null animals, suggesting that NaVβ1 modulated drug responses at the whole-heart level. In the human heart, we found that SCN1B transcript expression was 3 times higher in the atria than ventricles, differences that could, in combination with inherited or acquired cardiovascular disease, dramatically affect patient response to class Ib antiarrhythmic therapies.
Collapse
Affiliation(s)
- Wandi Zhu
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Wei Wang
- Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Paweorn Angsutararux
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Rebecca L Mellor
- Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Lori L Isom
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jeanne M Nerbonne
- Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.,Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Jonathan R Silva
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
18
|
Mantegazza M, Cestèle S, Catterall WA. Sodium channelopathies of skeletal muscle and brain. Physiol Rev 2021; 101:1633-1689. [PMID: 33769100 DOI: 10.1152/physrev.00025.2020] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels initiate action potentials in nerve, skeletal muscle, and other electrically excitable cells. Mutations in them cause a wide range of diseases. These channelopathy mutations affect every aspect of sodium channel function, including voltage sensing, voltage-dependent activation, ion conductance, fast and slow inactivation, and both biosynthesis and assembly. Mutations that cause different forms of periodic paralysis in skeletal muscle were discovered first and have provided a template for understanding structure, function, and pathophysiology at the molecular level. More recent work has revealed multiple sodium channelopathies in the brain. Here we review the well-characterized genetics and pathophysiology of the periodic paralyses of skeletal muscle and then use this information as a foundation for advancing our understanding of mutations in the structurally homologous α-subunits of brain sodium channels that cause epilepsy, migraine, autism, and related comorbidities. We include studies based on molecular and structural biology, cell biology and physiology, pharmacology, and mouse genetics. Our review reveals unexpected connections among these different types of sodium channelopathies.
Collapse
Affiliation(s)
- Massimo Mantegazza
- Université Cote d'Azur, Valbonne-Sophia Antipolis, France.,CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne-Sophia Antipolis, France.,INSERM, Valbonne-Sophia Antipolis, France
| | - Sandrine Cestèle
- Université Cote d'Azur, Valbonne-Sophia Antipolis, France.,CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne-Sophia Antipolis, France
| | | |
Collapse
|
19
|
Li W, Stauske M, Luo X, Wagner S, Vollrath M, Mehnert CS, Schubert M, Cyganek L, Chen S, Hasheminasab SM, Wulf G, El-Armouche A, Maier LS, Hasenfuss G, Guan K. Disease Phenotypes and Mechanisms of iPSC-Derived Cardiomyocytes From Brugada Syndrome Patients With a Loss-of-Function SCN5A Mutation. Front Cell Dev Biol 2020; 8:592893. [PMID: 33195263 PMCID: PMC7642519 DOI: 10.3389/fcell.2020.592893] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/01/2020] [Indexed: 12/19/2022] Open
Abstract
Brugada syndrome (BrS) is one of the major causes of sudden cardiac death in young people, while the underlying mechanisms are not completely understood. Here, we investigated the pathophysiological phenotypes and mechanisms using induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) from two BrS patients (BrS-CMs) carrying a heterozygous SCN5A mutation p.S1812X. Compared to CMs derived from healthy controls (Ctrl-CMs), BrS-CMs displayed a 50% reduction of INa density, a 69.5% reduction of NaV1.5 expression, and the impaired localization of NaV1.5 and connexin 43 (Cx43) at the cell surface. BrS-CMs exhibited reduced action potential (AP) upstroke velocity and conduction slowing. The Ito in BrS-CMs was significantly augmented, and the ICaL window current probability was increased. Our data indicate that the electrophysiological mechanisms underlying arrhythmia in BrS-CMs may involve both depolarization and repolarization disorders. Cilostazol and milrinone showed dramatic inhibitions of Ito in BrS-CMs and alleviated the arrhythmic activity, suggesting their therapeutic potential for BrS patients.
Collapse
Affiliation(s)
- Wener Li
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Michael Stauske
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Xiaojing Luo
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Stefan Wagner
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,Department of Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Meike Vollrath
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Carola S Mehnert
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Mario Schubert
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Lukas Cyganek
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Simin Chen
- German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Sayed-Mohammad Hasheminasab
- Department of Dermatology, Venereology and Allergy, Charité - Universitätsmedizin Berlin, Berlin, Germany.,CCU Translational Radiation Oncology, German Cancer Consortium Core-Center Heidelberg, National Center for Tumor Diseases, Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gerald Wulf
- Department of Hematology and Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Ali El-Armouche
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Lars S Maier
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,Clinic for Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| |
Collapse
|
20
|
Salvage SC, Huang CLH, Jackson AP. Cell-Adhesion Properties of β-Subunits in the Regulation of Cardiomyocyte Sodium Channels. Biomolecules 2020; 10:biom10070989. [PMID: 32630316 PMCID: PMC7407995 DOI: 10.3390/biom10070989] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/25/2020] [Accepted: 06/27/2020] [Indexed: 12/17/2022] Open
Abstract
Voltage-gated sodium (Nav) channels drive the rising phase of the action potential, essential for electrical signalling in nerves and muscles. The Nav channel α-subunit contains the ion-selective pore. In the cardiomyocyte, Nav1.5 is the main Nav channel α-subunit isoform, with a smaller expression of neuronal Nav channels. Four distinct regulatory β-subunits (β1–4) bind to the Nav channel α-subunits. Previous work has emphasised the β-subunits as direct Nav channel gating modulators. However, there is now increasing appreciation of additional roles played by these subunits. In this review, we focus on β-subunits as homophilic and heterophilic cell-adhesion molecules and the implications for cardiomyocyte function. Based on recent cryogenic electron microscopy (cryo-EM) data, we suggest that the β-subunits interact with Nav1.5 in a different way from their binding to other Nav channel isoforms. We believe this feature may facilitate trans-cell-adhesion between β1-associated Nav1.5 subunits on the intercalated disc and promote ephaptic conduction between cardiomyocytes.
Collapse
Affiliation(s)
- Samantha C. Salvage
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK;
- Correspondence: (S.C.S.); (A.P.J.); Tel.: +44-1223-765950 (S.C.S.); +44-1223-765951 (A.P.J.)
| | - Christopher L.-H. Huang
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK;
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Antony P. Jackson
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK;
- Correspondence: (S.C.S.); (A.P.J.); Tel.: +44-1223-765950 (S.C.S.); +44-1223-765951 (A.P.J.)
| |
Collapse
|
21
|
Glass WG, Duncan AL, Biggin PC. Computational Investigation of Voltage-Gated Sodium Channel β3 Subunit Dynamics. Front Mol Biosci 2020; 7:40. [PMID: 32266288 PMCID: PMC7103644 DOI: 10.3389/fmolb.2020.00040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/19/2020] [Indexed: 01/23/2023] Open
Abstract
Voltage-gated sodium (Na v ) channels form the basis for the initiation of the action potential in excitable cells by allowing sodium ions to pass through the cell membrane. The Na v channel α subunit is known to function both with and without associated β subunits. There is increasing evidence that these β subunits have multiple roles that include not only influencing the voltage-dependent gating but also the ability to alter the spatial distribution of the pore-forming α subunit. Recent structural data has shown possible ways in which β1 subunits may interact with the α subunit. However, the position of the β1 subunit would not be compatible with a previous trimer structure of the β3 subunit. Furthermore, little is currently known about the dynamic behavior of the β subunits both as individual monomers and as higher order oligomers. Here, we use multiscale molecular dynamics simulations to assess the dynamics of the β3, and the closely related, β1 subunit. These findings reveal the spatio-temporal dynamics of β subunits and should provide a useful framework for interpreting future low-resolution experiments such as atomic force microscopy.
Collapse
Affiliation(s)
| | | | - Philip C. Biggin
- Structural Bioinformatics and Computational Biochemistry, Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
22
|
Essandoh K, Philippe JM, Jenkins PM, Brody MJ. Palmitoylation: A Fatty Regulator of Myocardial Electrophysiology. Front Physiol 2020; 11:108. [PMID: 32140110 PMCID: PMC7042378 DOI: 10.3389/fphys.2020.00108] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 01/30/2020] [Indexed: 01/02/2023] Open
Abstract
Regulation of cardiac physiology is well known to occur through the action of kinases that reversibly phosphorylate ion channels, calcium handling machinery, and signaling effectors. However, it is becoming increasingly apparent that palmitoylation or S-acylation, the post-translational modification of cysteines with saturated fatty acids, plays instrumental roles in regulating the localization, activity, stability, sorting, and function of numerous proteins, including proteins known to have essential functions in cardiomyocytes. However, the impact of this modification on cardiac physiology requires further investigation. S-acylation is catalyzed by the zDHHC family of S-acyl transferases that localize to intracellular organelle membranes or the sarcolemma. Recent work has begun to uncover functions of S-acylation in the heart, particularly in the regulation of cardiac electrophysiology, including modification of the sodium-calcium exchanger, phospholemman and the cardiac sodium pump, as well as the voltage-gated sodium channel. Elucidating the regulatory functions of zDHHC enzymes in cardiomyocytes and determination of how S-acylation is altered in the diseased heart will shed light on how these modifications participate in cardiac pathogenesis and potentially identify novel targets for the treatment of cardiovascular disease. Indeed, proteins with critical signaling roles in the heart are also S-acylated, including receptors and G-proteins, yet the dynamics and functions of these modifications in myocardial physiology have not been interrogated. Here, we will review what is known about zDHHC enzymes and substrate S-acylation in myocardial physiology and highlight future areas of investigation that will uncover novel functions of S-acylation in cardiac homeostasis and pathophysiology.
Collapse
Affiliation(s)
- Kobina Essandoh
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States
| | - Julie M Philippe
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States
| | - Paul M Jenkins
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States.,Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| | - Matthew J Brody
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
23
|
Biophysical Characterization of Epigallocatechin-3-Gallate Effect on the Cardiac Sodium Channel Na v1.5. Molecules 2020; 25:molecules25040902. [PMID: 32085432 PMCID: PMC7070937 DOI: 10.3390/molecules25040902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/07/2020] [Accepted: 02/15/2020] [Indexed: 12/19/2022] Open
Abstract
Epigallocatechin-3-Gallate (EGCG) has been extensively studied for its protective effect against cardiovascular disorders. This effect has been attributed to its action on multiple molecular pathways and transmembrane proteins, including the cardiac Nav1.5 channels, which are inhibited in a dose-dependent manner. However, the molecular mechanism underlying this effect remains to be unveiled. To this aim, we have characterized the EGCG effect on Nav1.5 using electrophysiology and molecular dynamics (MD) simulations. EGCG superfusion induced a dose-dependent inhibition of Nav1.5 expressed in tsA201 cells, negatively shifted the steady-state inactivation curve, slowed the inactivation kinetics, and delayed the recovery from fast inactivation. However, EGCG had no effect on the voltage-dependence of activation and showed little use-dependent block on Nav1.5. Finally, MD simulations suggested that EGCG does not preferentially stay in the center of the bilayer, but that it spontaneously relocates to the membrane headgroup region. Moreover, no sign of spontaneous crossing from one leaflet to the other was observed, indicating a relatively large free energy barrier associated with EGCG transport across the membrane. These results indicate that EGCG may exert its biophysical effect via access to its binding site through the cell membrane or via a bilayer-mediated mechanism.
Collapse
|
24
|
Aeby A, Sculier C, Bouza AA, Askar B, Lederer D, Schoonjans A, Vander Ghinst M, Ceulemans B, Offord J, Lopez‐Santiago LF, Isom LL. SCN1B-linked early infantile developmental and epileptic encephalopathy. Ann Clin Transl Neurol 2019; 6:2354-2367. [PMID: 31709768 PMCID: PMC6917350 DOI: 10.1002/acn3.50921] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/22/2019] [Accepted: 09/23/2019] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE Patients with Early Infantile Epileptic Encephalopathy (EIEE) 52 have inherited, homozygous variants in the gene SCN1B, encoding the voltage-gated sodium channel (VGSC) β1 and β1B non-pore-forming subunits. METHODS Here, we describe the detailed electroclinical features of a biallelic SCN1B patient with a previously unreported variant, p.Arg85Cys. RESULTS The female proband showed hypotonia from birth, multifocal myoclonus at 2.5 months, then focal seizures and myoclonic status epilepticus (SE) at 3 months, triggered by fever. Auditory brainstem response (ABR) showed bilateral hearing loss. Epilepsy was refractory and the patient had virtually no development. Administration of fenfluramine resulted in a significant reduction in seizure frequency and resolution of SE episodes that persisted after a 2-year follow-up. The patient phenotype is more compatible with early infantile developmental and epileptic encephalopathy (DEE) than with typical Dravet syndrome (DS), as previously diagnosed for other patients with homozygous SCN1B variants. Biochemical and electrophysiological analyses of the SCN1B variant expressed in heterologous cells showed cell surface expression of the mutant β1 subunit, similar to wild-type (WT), but with loss of normal β1-mediated modification of human Nav 1.1-generated sodium current, suggesting that SCN1B-p.Arg85Cys is a loss-of-function (LOF) variant. INTERPRETATION Importantly, a review of the literature in light of our results suggests that the term, early infantile developmental and epileptic encephalopathy, is more appropriate than either EIEE or DS to describe biallelic SCN1B patients.
Collapse
Affiliation(s)
- Alec Aeby
- Pediatric NeurologyQueen Fabiola Children HospitalULBBrusselsBelgium
| | | | - Alexandra A. Bouza
- Department of PharmacologyUniversity of Michigan Medical SchoolAnn ArborMI48109
| | - Brandon Askar
- Department of PharmacologyUniversity of Michigan Medical SchoolAnn ArborMI48109
| | | | | | - Marc Vander Ghinst
- ENT DepartmentULB‐Hôpital ErasmeUniversité libre de Bruxelles (ULB)BrusselsBelgium
- Laboratoire de Cartographie fonctionnelle du CerveauUNI – ULB Neuroscience InstituteUniversité libre de Bruxelles (ULB)BrusselsBelgium
| | | | - James Offord
- Department of PharmacologyUniversity of Michigan Medical SchoolAnn ArborMI48109
| | | | - Lori L. Isom
- Department of PharmacologyUniversity of Michigan Medical SchoolAnn ArborMI48109
| |
Collapse
|
25
|
Maroni M, Körner J, Schüttler J, Winner B, Lampert A, Eberhardt E. β1 and β3 subunits amplify mechanosensitivity of the cardiac voltage-gated sodium channel Nav1.5. Pflugers Arch 2019; 471:1481-1492. [PMID: 31728700 DOI: 10.1007/s00424-019-02324-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 09/25/2019] [Accepted: 10/21/2019] [Indexed: 12/19/2022]
Abstract
In cardiomyocytes, electrical activity is coupled to cellular contraction, thus exposing all proteins expressed in the sarcolemma to mechanical stress. The voltage-gated sodium channel Nav1.5 is the main contributor to the rising phase of the action potential in the heart. There is growing evidence that gating and kinetics of Nav1.5 are modulated by mechanical forces and pathogenic variants that affect mechanosensitivity have been linked to arrhythmias. Recently, the sodium channel β1 subunit has been described to stabilise gating against mechanical stress of Nav1.7 expressed in neurons. Here, we tested the effect of β1 and β3 subunits on mechanosensitivity of the cardiac Nav1.5. β1 amplifies stress-induced shifts of V1/2 of steady-state fast inactivation to hyperpolarised potentials (ΔV1/2: 6.2 mV without and 10.7 mV with β1 co-expression). β3, on the other hand, almost doubles stress-induced speeding of time to sodium current transient peak (Δtime to peak at - 30 mV: 0.19 ms without and 0.37 ms with β3 co-expression). Our findings may indicate that in cardiomyocytes, the interdependence of electrical activity and contraction is used as a means of fine tuning cardiac sodium channel function, allowing quicker but more strongly inactivating sodium currents under conditions of increased mechanical stress. This regulation may help to shorten action potential duration during tachycardia, to prevent re-entry phenomena and thus arrhythmias.
Collapse
Affiliation(s)
- Michele Maroni
- Department of Anaesthesiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany.,Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Jannis Körner
- Institute of Physiology, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.,Department of Anaesthesiology, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany
| | - Jürgen Schüttler
- Department of Anaesthesiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Angelika Lampert
- Institute of Physiology, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany
| | - Esther Eberhardt
- Department of Anaesthesiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany.
| |
Collapse
|
26
|
Wu M, Tran PN, Sheng J, Randolph AL, Wu WW. Drug potency on inhibiting late Na + current is sensitive to gating modifier and current region where drug effects were measured. J Pharmacol Toxicol Methods 2019; 100:106605. [PMID: 31255744 DOI: 10.1016/j.vascn.2019.106605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/18/2019] [Accepted: 06/24/2019] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Cardiac late Na+ current (INaL) contributes to ventricular action potential duration. Pathological increase in INaL is arrhythmogenic, and inhibition of INaL offers protection against ventricular repolarization disturbance. Recently, two INaL datasets generated by different laboratories that assessed current inhibition by a panel of clinical drugs as a part of the Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative were published. The results revealed a surprising degree of data variability despite of the use of a standardized voltage protocol. This study investigated whether remaining procedural differences related to experimental methods and data analysis associated with these datasets can produce differences in INaL pharmacology. METHODS Whole cell voltage clamp recordings were performed on cells expressing NaV1.5 α- and β1-subunits to study: 1) the impact of gating modifiers used to augment INaL (ATX-II vs. veratridine), internal solution composition (with vs. without ATP and GTP), and recording temperature (23 °C vs 37 °C) on stability of INaL measured across the duration of a patch clamp experiment; 2) mechanisms of each gating modifier on Na+ channels; and 3) effects of six drugs (lidocaine, mexiletine, chloroquine, ranolazine, ritonavir, and verapamil) on INaL induced by either gating modifier. RESULTS Stability of INaL is affected by the choice of gating modifier, presence of nucleotides in the internal solution, and recording temperature. ATX-II and veratridine produced different changes in Na+ channel gating, inducing mechanistically distinct INaL. Drug potencies on inhibiting INaL were dependent on the choice of gating modifier and current region where drug effects were measured. DISCUSSION INaL pharmacology can be impacted by all experimental factors examined in this study. The effect of gating modifier and current region used to quantify drug inhibition alone led to 30× difference in half inhibitory concentration (IC50) for ritonavir, demonstrating that substantial difference in drug inhibition can be produced. Drug potencies on inhibiting INaL derived from different patch clamp studies may thus not be generalizable. For INaL pharmacology to be useful for in silico modeling or interpreting drug-induced changes in cardiac action potentials or ECG, standardizing INaL experimental procedures including data analysis methods is necessary to minimize data variability.
Collapse
Affiliation(s)
- Min Wu
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States of America
| | - Phu N Tran
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States of America
| | - Jiansong Sheng
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States of America
| | - Aaron L Randolph
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States of America
| | - Wendy W Wu
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States of America.
| |
Collapse
|
27
|
Nof E, Vysochek L, Meisel E, Burashnikov E, Antzelevitch C, Clatot J, Beinart R, Luria D, Glikson M, Oz S. Mutations in Na V1.5 Reveal Calcium-Calmodulin Regulation of Sodium Channel. Front Physiol 2019; 10:700. [PMID: 31231243 PMCID: PMC6560087 DOI: 10.3389/fphys.2019.00700] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/20/2019] [Indexed: 12/02/2022] Open
Abstract
Mutations in the SCN5A gene, encoding the cardiac voltage-gated sodium channel NaV1.5, are associated with inherited cardiac arrhythmia and conduction disease. Ca2+-dependent mechanisms and the involvement of β-subunit (NaVβ) in NaV1.5 regulation are not fully understood. A patient with severe sinus-bradycardia and cardiac conduction-disease was genetically evaluated and compound heterozygosity in the SCN5A gene was found. Mutations were identified in the cytoplasmic DIII-IV linker (K1493del) and the C-terminus (A1924T) of NaV1.5, both are putative CaM-binding domains. These mutants were functionally studied in human embryonic kidney (HEK) cells and HL-1 cells using whole-cell patch clamp technique. Calmodulin (CaM) interaction and cell-surface expression of heterologously expressed NaV1.5 mutants were studied by pull-down and biotinylation assays. The mutation K1493del rendered NaV1.5 non-conductive. NaV1.5K1493del altered the gating properties of co-expressed functional NaV1.5, in a Ca2+ and NaVβ1-dependent manner. NaV1.5A1924T impaired NaVβ1-dependent gating regulation. Ca2+-dependent CaM-interaction with NaV1.5 was blunted in NaV1.5K1493del. Electrical charge substitution at position 1493 did not affect CaM-interaction and channel functionality. Arrhythmia and conduction-disease -associated mutations revealed Ca2+-dependent gating regulation of NaV1.5 channels. Our results highlight the role of NaV1.5 DIII-IV linker in the CaM-binding complex and channel function, and suggest that the Ca2+-sensing machinery of NaV1.5 involves NaVβ1.
Collapse
Affiliation(s)
- Eyal Nof
- Heart Center, Sheba Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Eshcar Meisel
- Heart Center, Sheba Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elena Burashnikov
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - Charles Antzelevitch
- Lankenau Institute for Medical Research, Wynnewood, PA, United States.,Lankenau Heart Institute, Wynnewood, PA, United States.,Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Jerome Clatot
- Lankenau Institute for Medical Research, Wynnewood, PA, United States
| | - Roy Beinart
- Heart Center, Sheba Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - David Luria
- Heart Center, Sheba Medical Center, Ramat Gan, Israel
| | - Michael Glikson
- Heart Center, Sheba Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shimrit Oz
- Heart Center, Sheba Medical Center, Ramat Gan, Israel
| |
Collapse
|
28
|
Abnormal Scn1b and Fxyd1 gene expression in the pulled-through ganglionic colon may influence functional outcome in patients with Hirschsprung's disease. Pediatr Surg Int 2019; 35:9-14. [PMID: 30386899 DOI: 10.1007/s00383-018-4370-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/18/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE Smooth muscle cells are electrically coupled to ICC and PDGFRα+ cells, to regulate smooth muscle contraction. Recent studies have reported that the voltage-gated sodium channel type 1β (Scn1b), and the chloride channel subunit, Fxyd1, are highly expressed by both ICC and PDGFRα+ cells in the mouse colon. We designed this study to investigate the expression of the Scn1b and Fxyd1 genes in the normal human colon and in HSCR. METHODS HSCR tissue specimens (n = 6) were collected at the time of pull-through surgery, while control samples were obtained at the time of colostomy closure in patients with imperforate anus (n = 6). qRT-PCR analysis was undertaken to quantify Scn1b and Fxyd1 gene expression, and immunolabelling of Scn1b and Fxyd1 proteins were visualized using confocal microscopy. RESULTS qRT-PCR analysis revealed significant downregulation of Scn1b and Fxyd1 genes in both aganglionic and ganglionic HSCR specimens compared to controls (p < 0.05). Confocal microscopy revealed a reduction in Scn1b and Fxyd1 protein expression in both aganglionic and ganglionic HSCR colon compared to controls. CONCLUSION Scn1b and Fxyd1 expression was significantly downregulated in HSCR colon. These results add to mounting evidence suggesting that the pulled-through ganglionic segment of bowel in these patients is abnormal, despite the presence of ganglion cells.
Collapse
|