1
|
Gallego-Delgado M, Cámara-Checa A, Rubio-Alarcón M, Heredero-Jung D, de la Fuente-Blanco L, Rapún J, Plata-Izquierdo B, Pérez-Martín S, Cebrián J, Moreno de Redrojo L, García-Berrocal B, Delpón E, Sánchez PL, Villacorta E, Caballero R. Variable Penetrance and Expressivity of a Rare Pore Loss-of-Function Mutation (p.L889V) of Nav1.5 Channels in Three Spanish Families. Int J Mol Sci 2024; 25:4686. [PMID: 38731905 PMCID: PMC11083067 DOI: 10.3390/ijms25094686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 05/13/2024] Open
Abstract
A novel rare mutation in the pore region of Nav1.5 channels (p.L889V) has been found in three unrelated Spanish families that produces quite diverse phenotypic manifestations (Brugada syndrome, conduction disease, dilated cardiomyopathy, sinus node dysfunction, etc.) with variable penetrance among families. We clinically characterized the carriers and recorded the Na+ current (INa) generated by p.L889V and native (WT) Nav1.5 channels, alone or in combination, to obtain further insight into the genotypic-phenotypic relationships in patients carrying SCN5A mutations and in the molecular determinants of the Nav1.5 channel function. The variant produced a strong dominant negative effect (DNE) since the peak INa generated by p.L889V channels expressed in Chinese hamster ovary cells, either alone (-69.4 ± 9.0 pA/pF) or in combination with WT (-62.2 ± 14.6 pA/pF), was significantly (n ≥ 17, p < 0.05) reduced compared to that generated by WT channels alone (-199.1 ± 44.1 pA/pF). The mutation shifted the voltage dependence of channel activation and inactivation to depolarized potentials, did not modify the density of the late component of INa, slightly decreased the peak window current, accelerated the recovery from fast and slow inactivation, and slowed the induction kinetics of slow inactivation, decreasing the fraction of channels entering this inactivated state. The membrane expression of p.L889V channels was low, and in silico molecular experiments demonstrated profound alterations in the disposition of the pore region of the mutated channels. Despite the mutation producing a marked DNE and reduction in the INa and being located in a critical domain of the channel, its penetrance and expressivity are quite variable among the carriers. Our results reinforce the argument that the incomplete penetrance and phenotypic variability of SCN5A loss-of-function mutations are the result of a combination of multiple factors, making it difficult to predict their expressivity in the carriers despite the combination of clinical, genetic, and functional studies.
Collapse
Affiliation(s)
- María Gallego-Delgado
- Department of Cardiology, CSUR Cardiopatías Familiares, Institute of Biomedical Research of Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), 37007 Salamaca, Spain; (M.G.-D.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain (M.R.-A.); (J.R.); (J.C.); (R.C.)
| | - Anabel Cámara-Checa
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain (M.R.-A.); (J.R.); (J.C.); (R.C.)
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, 28040 Madrid, Spain
| | - Marcos Rubio-Alarcón
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain (M.R.-A.); (J.R.); (J.C.); (R.C.)
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, 28040 Madrid, Spain
| | - David Heredero-Jung
- Department of Biochemistry, CSUR Cardiopatías Familiares, Institute of Biomedical Research of Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), 37007 Salamaca, Spain
| | - Laura de la Fuente-Blanco
- Department of Cardiology, CSUR Cardiopatías Familiares, Institute of Biomedical Research of Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), 37007 Salamaca, Spain; (M.G.-D.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain (M.R.-A.); (J.R.); (J.C.); (R.C.)
| | - Josu Rapún
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain (M.R.-A.); (J.R.); (J.C.); (R.C.)
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, 28040 Madrid, Spain
| | - Beatriz Plata-Izquierdo
- Department of Pediatrics, Institute of Biomedical Research of Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Gerencia Regional de Salud de Castilla y Leon (SACYL), CIBERCV, 37007 Salamaca, Spain;
| | - Sara Pérez-Martín
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain (M.R.-A.); (J.R.); (J.C.); (R.C.)
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, 28040 Madrid, Spain
| | - Jorge Cebrián
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain (M.R.-A.); (J.R.); (J.C.); (R.C.)
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, 28040 Madrid, Spain
| | - Lucía Moreno de Redrojo
- Department of Cardiology, CSUR Cardiopatías Familiares, Institute of Biomedical Research of Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), 37007 Salamaca, Spain; (M.G.-D.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain (M.R.-A.); (J.R.); (J.C.); (R.C.)
| | - Belén García-Berrocal
- Department of Biochemistry, CSUR Cardiopatías Familiares, Institute of Biomedical Research of Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), 37007 Salamaca, Spain
| | - Eva Delpón
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain (M.R.-A.); (J.R.); (J.C.); (R.C.)
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, 28040 Madrid, Spain
| | - Pedro L. Sánchez
- Department of Cardiology, CSUR Cardiopatías Familiares, Institute of Biomedical Research of Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), 37007 Salamaca, Spain; (M.G.-D.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain (M.R.-A.); (J.R.); (J.C.); (R.C.)
| | - Eduardo Villacorta
- Department of Cardiology, CSUR Cardiopatías Familiares, Institute of Biomedical Research of Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), 37007 Salamaca, Spain; (M.G.-D.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain (M.R.-A.); (J.R.); (J.C.); (R.C.)
| | - Ricardo Caballero
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain (M.R.-A.); (J.R.); (J.C.); (R.C.)
- Department of Pharmacology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, 28040 Madrid, Spain
| |
Collapse
|
2
|
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: 2] [Impact Index Per Article: 2.0] [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
|
3
|
Jameson MB, Ríos-Pérez EB, Liu F, Eichel CA, Robertson GA. Pairwise biosynthesis of ion channels stabilizes excitability and mitigates arrhythmias. Proc Natl Acad Sci U S A 2023; 120:e2305295120. [PMID: 37816059 PMCID: PMC10589643 DOI: 10.1073/pnas.2305295120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/14/2023] [Indexed: 10/12/2023] Open
Abstract
Coordinated expression of ion channels is crucial for cardiac rhythms, neural signaling, and cell cycle progression. Perturbation of this balance results in many disorders including cardiac arrhythmias. Prior work revealed association of mRNAs encoding cardiac NaV1.5 (SCN5A) and hERG1 (KCNH2), but the functional significance of this association was not established. Here, we provide a more comprehensive picture of KCNH2, SCN5A, CACNA1C, and KCNQ1 transcripts collectively copurifying with nascent hERG1, NaV1.5, CaV1.2, or KCNQ1 channel proteins. Single-molecule fluorescence in situ hybridization (smFISH) combined with immunofluorescence reveals that the channel proteins are synthesized predominantly as heterotypic pairs from discrete molecules of mRNA, not as larger cotranslational complexes. Puromycin disrupted colocalization of mRNA with its encoded protein, as expected, but remarkably also pairwise mRNA association, suggesting that transcript association relies on intact translational machinery or the presence of the nascent protein. Targeted depletion of KCHN2 by specific shRNA resulted in concomitant reduction of all associated mRNAs, with a corresponding reduction in the encoded channel currents. This co-knockdown effect, originally described for KCNH2 and SCN5A, thus appears to be a general phenomenon among transcripts encoding functionally related proteins. In multielectrode array recordings, proarrhythmic behavior arose when IKr was reduced by the selective blocker dofetilide at IC50 concentrations, but not when equivalent reductions were mediated by shRNA, suggesting that co-knockdown mitigates proarrhythmic behavior expected from the selective reduction of a single channel species. We propose that coordinated, cotranslational association of functionally related ion channel mRNAs confers electrical stability by co-regulating complementary ion channels in macromolecular complexes.
Collapse
Affiliation(s)
- Margaret B. Jameson
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI53705
| | - Erick B. Ríos-Pérez
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI53705
| | - Fang Liu
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI53705
| | - Catherine A. Eichel
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI53705
| | - Gail A. Robertson
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI53705
| |
Collapse
|
4
|
Selimi Z, Rougier JS, Abriel H, Kucera JP. A detailed analysis of single-channel Na v 1.5 recordings does not reveal any cooperative gating. J Physiol 2023; 601:3847-3868. [PMID: 37470338 DOI: 10.1113/jp284861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/23/2023] [Indexed: 07/21/2023] Open
Abstract
Cardiac voltage-gated sodium (Na+ ) channels (Nav 1.5) are crucial for myocardial electrical excitation. Recent studies based on single-channel recordings have suggested that Na+ channels interact functionally and exhibit coupled gating. However, the analysis of such recordings frequently relies on manual interventions, which can lead to bias. Here, we developed an automated pipeline to de-trend and idealize single-channel currents, and assessed possible functional interactions in cell-attached patch clamp experiments in HEK293 cells expressing human Nav 1.5 channels as well as in adult mouse and rabbit ventricular cardiomyocytes. Our pipeline involved de-trending individual sweeps by linear optimization using a library of predefined functions, followed by digital filtering and baseline offset. Subsequently, the processed sweeps were idealized based on the idea that the ensemble average of the idealized current identified by thresholds between current levels reconstructs at best the ensemble average current from the de-trended sweeps. This reconstruction was achieved by non-linear optimization. To ascertain functional interactions, we examined the distribution of the numbers of open channels at every time point during the activation protocol and compared it to the distribution expected for independent channels. We also examined whether the channels tended to synchronize their openings and closings. However, we did not uncover any solid evidence of such interactions in our recordings. Rather, our results indicate that wild-type Nav 1.5 channels are independent entities or exhibit only very weak functional interactions that are probably irrelevant under physiological conditions. Nevertheless, our unbiased analysis will be important for further studies examining whether auxiliary proteins potentiate functional Na+ channel interactions. KEY POINTS: Nav 1.5 channels are critical for cardiac excitation. They are part of macromolecular interacting complexes, and it was previously suggested that two neighbouring channels may functionally interact and exhibit coupled gating. Manual interventions when processing single-channel recordings can lead to bias and inaccurate data interpretation. We developed an automated pipeline to de-trend and idealize single-channel currents and assessed possible functional interactions between Nav 1.5 channels in HEK293 cells and cardiomyocytes during activation protocols using the cell-attached patch clamp technique. In recordings consisting of up to 1000 sweeps from the same patch, our analysis did not reveal any evidence of functional interactions or coupled gating between wild-type Nav 1.5 channels. Our unbiased analysis may be useful in further studies examining how Na+ channel interactions are affected by mutations and auxiliary proteins.
Collapse
Affiliation(s)
- Zoja Selimi
- Department of Physiology, University of Bern, Bern, Switzerland
| | | | - Hugues Abriel
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Jan P Kucera
- Department of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Zheng Y, Deschênes I. Protein 14-3-3 Influences the Response of the Cardiac Sodium Channel Na v1.5 to Antiarrhythmic Drugs. J Pharmacol Exp Ther 2023; 384:417-428. [PMID: 36460339 PMCID: PMC9976794 DOI: 10.1124/jpet.122.001407] [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: 08/11/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
The cardiac sodium channel Nav1.5 is a key contributor to the cardiac action potential, and dysregulations in Nav1.5 can lead to cardiac arrhythmias. Nav1.5 is a target of numerous antiarrhythmic drugs (AADs). Previous studies identified the protein 14-3-3 as a regulator of Nav1.5 biophysical coupling. Inhibition of 14-3-3 can remove the Nav1.5 functional coupling and has been shown to inhibit the dominant-negative effect of Brugada syndrome mutations. However, it is unknown whether the coupling regulation is involved with AADs' modulation of Nav1.5. Indeed, AADs could reveal important structural and functional information about Nav1.5 coupling. Here, we investigated the modulation of Nav1.5 by four classic AADs, quinidine, lidocaine, mexiletine, and flecainide, in the presence of 14-3-3 inhibition. The experiments were carried out by high-throughput patch-clamp experiments in an HEK293 Nav1.5 stable cell line. We found that 14-3-3 inhibition can enhance acute block by quinidine, whereas the block by other drugs was not affected. We also saw changes in the use- and dose-dependency of quinidine, lidocaine, and mexiletine when inhibiting 14-3-3. Inhibiting 14-3-3 also shifted the channel activation toward hyperpolarized voltages in the presence of the four drugs studied and slowed the recovery of inactivation in the presence of quinidine. Our results demonstrated that the protein 14-3-3 and Nav1.5 coupling could impact the effects of AADs. Therefore, 14-3-3 and Nav1.5 coupling are new mechanisms to consider in the development of drugs targeting Nav1.5. SIGNIFICANCE STATEMENT: The cardiac sodium channel Nav1.5 is a target of commonly used antiarrhythmic drugs, and Nav1.5 function is regulated by the protein 14-3-3. The present study demonstrated that the regulation of Nav1.5 by 14-3-3 influences Nav1.5's response to antiarrhythmic drugs. This study provides detailed information about how 14-3-3 differentially regulated Nav1.5 functions under the influence of different drug subtypes. These findings will guide future molecular studies investigating Nav1.5 and antiarrhythmic drugs outcomes.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio (Y.Z., I.D.) and Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio (Y.Z.)
| | - Isabelle Deschênes
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio (Y.Z., I.D.) and Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio (Y.Z.)
| |
Collapse
|
6
|
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
|
7
|
Marchal GA, Remme CA. Subcellular diversity of Nav1.5 in cardiomyocytes: distinct functions, mechanisms and targets. J Physiol 2023; 601:941-960. [PMID: 36469003 DOI: 10.1113/jp283086] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/24/2022] [Indexed: 12/11/2022] Open
Abstract
In cardiomyocytes, the rapid depolarisation of the membrane potential is mediated by the α-subunit of the cardiac voltage-gated Na+ channel (NaV 1.5), encoded by the gene SCN5A. This ion channel allows positively charged Na+ ions to enter the cardiomyocyte, resulting in the fast upstroke of the action potential and is therefore crucial for cardiac excitability and electrical propagation. This essential role is underscored by the fact that dysfunctional NaV 1.5 is associated with high risk for arrhythmias and sudden cardiac death. However, development of therapeutic interventions regulating NaV 1.5 has been limited due to the complexity of NaV 1.5 structure and function and its diverse roles within the cardiomyocyte. In particular, research from the last decade has provided us with increased knowledge on the subcellular distribution of NaV 1.5 as well as the proteins which it interacts with in distinct cardiomyocyte microdomains. We here review these insights, detailing the potential role of NaV 1.5 within subcellular domains as well as its dysfunction in the setting of arrhythmia disorders. We furthermore provide an overview of current knowledge on the pathways involved in (microdomain-specific) trafficking of NaV 1.5, and their potential as novel targets. Unravelling the complexity of NaV 1.5 (dys)function may ultimately facilitate the development of therapeutic strategies aimed at preventing lethal arrhythmias. This is not only of importance for pathophysiological conditions where sodium current is specifically decreased within certain subcellular regions, such as in arrhythmogenic cardiomyopathy and Duchenne muscular dystrophy, but also for other acquired and inherited disorders associated with NaV 1.5.
Collapse
Affiliation(s)
- Gerard A Marchal
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands.,National Institute of Optics, National Research Council (CNR-INO), Sesto Fiorentino, Florence, Italy
| | - Carol Ann Remme
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Yu G, Chakrabarti S, Tischenko M, Chen AL, Wang Z, Cho H, French BA, Naga Prasad SV, Chen Q, Wang QK. Gene therapy targeting protein trafficking regulator MOG1 in mouse models of Brugada syndrome, arrhythmias, and mild cardiomyopathy. Sci Transl Med 2022; 14:eabf3136. [PMID: 35675436 DOI: 10.1126/scitranslmed.abf3136] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Brugada syndrome (BrS) is a fatal arrhythmia that causes an estimated 4% of all sudden death in high-incidence areas. SCN5A encodes cardiac sodium channel NaV1.5 and causes 25 to 30% of BrS cases. Here, we report generation of a knock-in (KI) mouse model of BrS (Scn5aG1746R/+). Heterozygous KI mice recapitulated some of the clinical features of BrS, including an ST segment abnormality (a prominent J wave) on electrocardiograms and development of spontaneous ventricular tachyarrhythmias (VTs), seizures, and sudden death. VTs were caused by shortened cardiac action potential duration and late phase 3 early afterdepolarizations associated with reduced sodium current density (INa) and increased Kcnd3 and Cacna1c expression. We developed a gene therapy using adeno-associated virus serotype 9 (AAV9) vector-mediated MOG1 delivery for up-regulation of MOG1, a chaperone that binds to NaV1.5 and traffics it to the cell surface. MOG1 was chosen for gene therapy because the large size of the SCN5A coding sequence (6048 base pairs) exceeds the packaging capacity of AAV vectors. AAV9-MOG1 gene therapy increased cell surface expression of NaV1.5 and ventricular INa, reversed up-regulation of Kcnd3 and Cacna1c expression, normalized cardiac action potential abnormalities, abolished J waves, and blocked VT in Scn5aG1746R/+ mice. Gene therapy also rescued the phenotypes of cardiac arrhythmias and contractile dysfunction in heterozygous humanized KI mice with SCN5A mutation p.D1275N. Using a small chaperone protein may have broad implications for targeting disease-causing genes exceeding the size capacity of AAV vectors.
Collapse
Affiliation(s)
- Gang Yu
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan 430074, P. R. China.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Susmita Chakrabarti
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Miroslava Tischenko
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Ai-Lan Chen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Department of Cardiology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 511436, P. R. China
| | - Zhijie Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan 430074, P. R. China.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Hyosuk Cho
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Brent A French
- Department of Biomedical Engineering, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Sathyamangla V Naga Prasad
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Qiuyun Chen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Qing K Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.,Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan 430074, P. R. China.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
9
|
O'Neill MJ, Muhammad A, Li B, Wada Y, Hall L, Solus JF, Short L, Roden DM, Glazer AM. Dominant negative effects of SCN5A missense variants. Genet Med 2022; 24:1238-1248. [PMID: 35305865 PMCID: PMC9262418 DOI: 10.1016/j.gim.2022.02.010] [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: 10/12/2021] [Revised: 02/15/2022] [Accepted: 02/15/2022] [Indexed: 10/18/2022] Open
Abstract
PURPOSE Up to 30% of patients with Brugada syndrome (BrS) carry loss-of-function (LoF) variants in the cardiac sodium channel gene SCN5A encoding for the protein NaV1.5. Recent studies suggested that NaV1.5 can dimerize, and some variants exert dominant negative effects. In this study, we sought to explore the generality of missense variant NaV1.5 dominant negative effects and their clinical severity. METHODS We identified 35 LoF variants (<10% of wild type [WT] peak current) and 15 partial LoF variants (10%-50% of WT peak current) that we assessed for dominant negative effects. SCN5A variants were studied in HEK293T cells, alone or in heterozygous coexpression with WT SCN5A using automated patch clamp. To assess the clinical risk, we compared the prevalence of dominant negative vs putative haploinsufficient (frameshift, splice, or nonsense) variants in a BrS consortium and the Genome Aggregation Database population database. RESULTS In heterozygous expression with WT, 32 of 35 LoF and 6 of 15 partial LoF variants showed reduction to <75% of WT-alone peak current, showing a dominant negative effect. Individuals with dominant negative LoF variants had an elevated disease burden compared with the individuals with putative haploinsufficient variants (2.7-fold enrichment in BrS cases, P = .019). CONCLUSION Most SCN5A missense LoF variants exert a dominant negative effect. This class of variant confers an especially high burden of BrS.
Collapse
Affiliation(s)
- Matthew J O'Neill
- Vanderbilt University School of Medicine, Medical Scientist Training Program, Vanderbilt University, Nashville, TN
| | - Ayesha Muhammad
- Vanderbilt University School of Medicine, Medical Scientist Training Program, Vanderbilt University, Nashville, TN
| | - Bian Li
- Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Vanderbilt University Medical Center, Nashville, TN
| | - Yuko Wada
- Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Vanderbilt University Medical Center, Nashville, TN
| | - Lynn Hall
- Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Vanderbilt University Medical Center, Nashville, TN
| | - Joseph F Solus
- Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Vanderbilt University Medical Center, Nashville, TN
| | - Laura Short
- Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Vanderbilt University Medical Center, Nashville, TN
| | - Dan M Roden
- Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Vanderbilt University Medical Center, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Andrew M Glazer
- Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Vanderbilt University Medical Center, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
10
|
Horváth B, Szentandrássy N, Almássy J, Dienes C, Kovács ZM, Nánási PP, Banyasz T. Late Sodium Current of the Heart: Where Do We Stand and Where Are We Going? Pharmaceuticals (Basel) 2022; 15:ph15020231. [PMID: 35215342 PMCID: PMC8879921 DOI: 10.3390/ph15020231] [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: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 02/05/2023] Open
Abstract
Late sodium current has long been linked to dysrhythmia and contractile malfunction in the heart. Despite the increasing body of accumulating information on the subject, our understanding of its role in normal or pathologic states is not complete. Even though the role of late sodium current in shaping action potential under physiologic circumstances is debated, it’s unquestioned role in arrhythmogenesis keeps it in the focus of research. Transgenic mouse models and isoform-specific pharmacological tools have proved useful in understanding the mechanism of late sodium current in health and disease. This review will outline the mechanism and function of cardiac late sodium current with special focus on the recent advances of the area.
Collapse
Affiliation(s)
- Balázs Horváth
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Norbert Szentandrássy
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
- Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| | - János Almássy
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Csaba Dienes
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Zsigmond Máté Kovács
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Péter P. Nánási
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
- Department of Dental Physiology and Pharmacology, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamas Banyasz
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
- Correspondence: ; Tel.: +36-(52)-255-575; Fax: +36-(52)-255-116
| |
Collapse
|
11
|
Daimi H, Lozano-Velasco E, Aranega A, Franco D. Genomic and Non-Genomic Regulatory Mechanisms of the Cardiac Sodium Channel in Cardiac Arrhythmias. Int J Mol Sci 2022; 23:1381. [PMID: 35163304 PMCID: PMC8835759 DOI: 10.3390/ijms23031381] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 12/19/2022] Open
Abstract
Nav1.5 is the predominant cardiac sodium channel subtype, encoded by the SCN5A gene, which is involved in the initiation and conduction of action potentials throughout the heart. Along its biosynthesis process, Nav1.5 undergoes strict genomic and non-genomic regulatory and quality control steps that allow only newly synthesized channels to reach their final membrane destination and carry out their electrophysiological role. These regulatory pathways are ensured by distinct interacting proteins that accompany the nascent Nav1.5 protein along with different subcellular organelles. Defects on a large number of these pathways have a tremendous impact on Nav1.5 functionality and are thus intimately linked to cardiac arrhythmias. In the present review, we provide current state-of-the-art information on the molecular events that regulate SCN5A/Nav1.5 and the cardiac channelopathies associated with defects in these pathways.
Collapse
Affiliation(s)
- Houria Daimi
- Biochemistry and Molecular Biology Laboratory, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia
| | - Estefanía Lozano-Velasco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (A.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento, 34, 18016 Granada, Spain
| | - Amelia Aranega
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (A.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento, 34, 18016 Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (E.L.-V.); (A.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento, 34, 18016 Granada, Spain
| |
Collapse
|
12
|
Dixon RE, Navedo MF, Binder MD, Santana LF. Mechanisms and Physiological Implications of Cooperative Gating of Ion Channels Clusters. Physiol Rev 2021; 102:1159-1210. [PMID: 34927454 DOI: 10.1152/physrev.00022.2021] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ion channels play a central role in the regulation of nearly every cellular process. Dating back to the classic 1952 Hodgkin-Huxley model of the generation of the action potential, ion channels have always been thought of as independent agents. A myriad of recent experimental findings exploiting advances in electrophysiology, structural biology, and imaging techniques, however, have posed a serious challenge to this long-held axiom as several classes of ion channels appear to open and close in a coordinated, cooperative manner. Ion channel cooperativity ranges from variable-sized oligomeric cooperative gating in voltage-gated, dihydropyridine-sensitive Cav1.2 and Cav1.3 channels to obligatory dimeric assembly and gating of voltage-gated Nav1.5 channels. Potassium channels, transient receptor potential channels, hyperpolarization cyclic nucleotide-activated channels, ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3Rs) have also been shown to gate cooperatively. The implications of cooperative gating of these ion channels range from fine tuning excitation-contraction coupling in muscle cells to regulating cardiac function and vascular tone, to modulation of action potential and conduction velocity in neurons and cardiac cells, and to control of pace-making activity in the heart. In this review, we discuss the mechanisms leading to cooperative gating of ion channels, their physiological consequences and how alterations in cooperative gating of ion channels may induce a range of clinically significant pathologies.
Collapse
Affiliation(s)
- Rose Ellen Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA, United States
| | - Marc D Binder
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| |
Collapse
|
13
|
Fonseca-Barriendos D, Frías-Soria CL, Pérez-Pérez D, Gómez-López R, Borroto Escuela DO, Rocha L. Drug-resistant epilepsy: Drug target hypothesis and beyond the receptors. Epilepsia Open 2021; 7 Suppl 1:S23-S33. [PMID: 34542940 PMCID: PMC9340308 DOI: 10.1002/epi4.12539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/21/2021] [Accepted: 08/27/2021] [Indexed: 12/28/2022] Open
Abstract
Epilepsy is a chronic neurological disorder that affects more than 50 million people worldwide. Despite a recent introduction of antiseizure drugs for the treatment of epileptic seizures, one-third of these patients suffer from drug-resistant epilepsy (DRE). The therapeutic target hypothesis is a cited theory to explain DRE. According to the target hypothesis, the failure to achieve seizure freedom leads to alteration of the structure and/or function of the antiseizure medication (ASM) target. However, this hypothesis fails to explain why patients with DRE do not respond to antiseizure medications of different targets. This review presents different conditions, such as epigenetic mechanisms and protein-protein interactions that may result in alterations of diverse drug targets using different mechanisms. These novel conditions represent new targets to control DRE.
Collapse
Affiliation(s)
| | | | - Daniel Pérez-Pérez
- Plan of Combined Studies in Medicine (PECEM), Faculty of Medicine, UNAM, México City, Mexico
| | - Rosenda Gómez-López
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, México
| | | | - Luisa Rocha
- Pharmacobiology Department, Center for Research and Advanced Studies, México City, México
| |
Collapse
|
14
|
Balla C, Conte E, Selvatici R, Marsano RM, Gerbino A, Farnè M, Blunck R, Vitali F, Armaroli A, Brieda A, Liantonio A, De Luca A, Ferlini A, Rapezzi C, Bertini M, Gualandi F, Imbrici P. Functional Characterization of Two Novel Mutations in SCN5A Associated with Brugada Syndrome Identified in Italian Patients. Int J Mol Sci 2021; 22:ijms22126513. [PMID: 34204499 PMCID: PMC8234720 DOI: 10.3390/ijms22126513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/19/2022] Open
Abstract
Background. Brugada syndrome (BrS) is an autosomal dominantly inherited cardiac disease characterized by “coved type” ST-segment elevation in the right precordial leads, high susceptibility to ventricular arrhythmia and a family history of sudden cardiac death. The SCN5A gene, encoding for the cardiac voltage-gated sodium channel Nav1.5, accounts for ~20–30% of BrS cases and is considered clinically relevant. Methods. Here, we describe the clinical findings of two Italian families affected by BrS and provide the functional characterization of two novel SCN5A mutations, the missense variant Pro1310Leu and the in-frame insertion Gly1687_Ile1688insGlyArg. Results. Despite being clinically different, both patients have a family history of sudden cardiac death and had history of arrhythmic events. The Pro1310Leu mutation significantly reduced peak sodium current density without affecting channel membrane localization. Changes in the gating properties of expressed Pro1310Leu channel likely account for the loss-of-function phenotype. On the other hand, Gly1687_Ile1688insGlyArg channel, identified in a female patient, yielded a nearly undetectable sodium current. Following mexiletine incubation, the Gly1687_Ile1688insGlyArg channel showed detectable, albeit very small, currents and biophysical properties similar to those of the Nav1.5 wild-type channel. Conclusions. Overall, our results suggest that the degree of loss-of-function shown by the two Nav1.5 mutant channels correlates with the aggressive clinical phenotype of the two probands. This genotype-phenotype correlation is fundamental to set out appropriate therapeutical intervention.
Collapse
Affiliation(s)
- Cristina Balla
- Cardiological Center, University of Ferrara, 44121 Ferrara, Italy; (C.B.); (F.V.); (A.B.); (C.R.); (M.B.)
| | - Elena Conte
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (A.L.); (A.D.L.)
| | - Rita Selvatici
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (R.S.); (M.F.); (A.A.); (A.F.)
| | | | - Andrea Gerbino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Marianna Farnè
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (R.S.); (M.F.); (A.A.); (A.F.)
| | - Rikard Blunck
- Department of Physics, Université de Montréal, Montréal, QC H3C 3J7, Canada;
| | - Francesco Vitali
- Cardiological Center, University of Ferrara, 44121 Ferrara, Italy; (C.B.); (F.V.); (A.B.); (C.R.); (M.B.)
| | - Annarita Armaroli
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (R.S.); (M.F.); (A.A.); (A.F.)
| | - Alessandro Brieda
- Cardiological Center, University of Ferrara, 44121 Ferrara, Italy; (C.B.); (F.V.); (A.B.); (C.R.); (M.B.)
| | - Antonella Liantonio
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (A.L.); (A.D.L.)
| | - Annamaria De Luca
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (A.L.); (A.D.L.)
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (R.S.); (M.F.); (A.A.); (A.F.)
| | - Claudio Rapezzi
- Cardiological Center, University of Ferrara, 44121 Ferrara, Italy; (C.B.); (F.V.); (A.B.); (C.R.); (M.B.)
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy
| | - Matteo Bertini
- Cardiological Center, University of Ferrara, 44121 Ferrara, Italy; (C.B.); (F.V.); (A.B.); (C.R.); (M.B.)
| | - Francesca Gualandi
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (R.S.); (M.F.); (A.A.); (A.F.)
- Correspondence: (F.G.); (P.I.)
| | - Paola Imbrici
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (E.C.); (A.L.); (A.D.L.)
- Correspondence: (F.G.); (P.I.)
| |
Collapse
|
15
|
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: 47] [Impact Index Per Article: 15.7] [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
|
16
|
Zheng Y, Wan X, Yang D, Ramirez-Navarro A, Liu H, Fu JD, Deschênes I. A Heart Failure-Associated SCN5A Splice Variant Leads to a Reduction in Sodium Current Through Coupled-Gating With the Wild-Type Channel. Front Physiol 2021; 12:661429. [PMID: 33828490 PMCID: PMC8019726 DOI: 10.3389/fphys.2021.661429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Nav1.5, encoded by the gene SCN5A, is the predominant voltage-gated sodium channel expressed in the heart. It initiates the cardiac action potential and thus is crucial for normal heart rhythm and function. Dysfunctions in Nav1.5 have been involved in multiple congenital or acquired cardiac pathological conditions such as Brugada syndrome (BrS), Long QT Syndrome Type 3, and heart failure (HF), all of which can lead to sudden cardiac death (SCD) - one of the leading causes of death worldwide. Our lab has previously reported that Nav1.5 forms dimer channels with coupled gating. We also found that Nav1.5 BrS mutants can exert a dominant-negative (DN) effect and impair the function of wildtype (WT) channels through coupled-gating with the WT. It was previously reported that reduction in cardiac sodium currents (INa), observed in HF, could be due to the increased expression of an SCN5A splice variant - E28D, which results in a truncated sodium channel (Nav1.5-G1642X). In this study, we hypothesized that this SCN5A splice variant leads to INa reduction in HF through biophysical coupling with the WT. We showed that Nav1.5-G1642X is a non-functional channel but can interact with the WT, resulting in a DN effect on the WT channel. We found that both WT and the truncated channel Nav1.5-G1642X traffic at the cell surface, suggesting biophysical coupling. Indeed, we found that the DN effect can be abolished by difopein, an inhibitor of the biophysical coupling. Interestingly, the sodium channel polymorphism H558R, which has beneficial effect in HF patients, could also block the DN effect. In summary, the HF-associated splice variant Nav1.5-G1642X suppresses sodium currents in heart failure patients through a mechanism involving coupled-gating with the wildtype sodium channel.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Xiaoping Wan
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Dandan Yang
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Angelina Ramirez-Navarro
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Haiyan Liu
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Ji-Dong Fu
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Isabelle Deschênes
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
17
|
Nathan S, Gabelli SB, Yoder JB, Srinivasan L, Aldrich RW, Tomaselli GF, Ben-Johny M, Amzel LM. Structural basis of cytoplasmic NaV1.5 and NaV1.4 regulation. J Gen Physiol 2020; 153:211587. [PMID: 33306788 PMCID: PMC7953540 DOI: 10.1085/jgp.202012722] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
Voltage-gated sodium channels (NaVs) are membrane proteins responsible for the rapid upstroke of the action potential in excitable cells. There are nine human voltage-sensitive NaV1 isoforms that, in addition to their sequence differences, differ in tissue distribution and specific function. This review focuses on isoforms NaV1.4 and NaV1.5, which are primarily expressed in skeletal and cardiac muscle cells, respectively. The determination of the structures of several eukaryotic NaVs by single-particle cryo-electron microscopy (cryo-EM) has brought new perspective to the study of the channels. Alignment of the cryo-EM structure of the transmembrane channel pore with x-ray crystallographic structures of the cytoplasmic domains illustrates the complementary nature of the techniques and highlights the intricate cellular mechanisms that modulate these channels. Here, we review structural insights into the cytoplasmic C-terminal regulation of NaV1.4 and NaV1.5 with special attention to Ca2+ sensing by calmodulin, implications for disease, and putative channel dimerization.
Collapse
Affiliation(s)
- Sara Nathan
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sandra B Gabelli
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD.,Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jesse B Yoder
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lakshmi Srinivasan
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Richard W Aldrich
- Department of Neuroscience, University of Texas at Austin, Austin, TX
| | - Gordon F Tomaselli
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Manu Ben-Johny
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY
| | - L Mario Amzel
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
18
|
Hichri E, Selimi Z, Kucera JP. Modeling the Interactions Between Sodium Channels Provides Insight Into the Negative Dominance of Certain Channel Mutations. Front Physiol 2020; 11:589386. [PMID: 33250780 PMCID: PMC7674773 DOI: 10.3389/fphys.2020.589386] [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: 07/30/2020] [Accepted: 10/12/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Nav1.5 cardiac Na+ channel mutations can cause arrhythmogenic syndromes. Some of these mutations exert a dominant negative effect on wild-type channels. Recent studies showed that Na+ channels can dimerize, allowing coupled gating. This leads to the hypothesis that allosteric interactions between Na+ channels modulate their function and that these interactions may contribute to the negative dominance of certain mutations. METHODS To investigate how allosteric interactions affect microscopic and macroscopic channel function, we developed a modeling paradigm in which Markovian models of two channels are combined. Allosteric interactions are incorporated by modifying the free energies of the composite states and/or barriers between states. RESULTS Simulations using two generic 2-state models (C-O, closed-open) revealed that increasing the free energy of the composite states CO/OC leads to coupled gating. Simulations using two 3-state models (closed-open-inactivated) revealed that coupled closings must also involve interactions between further composite states. Using two 6-state cardiac Na+ channel models, we replicated previous experimental results mainly by increasing the energies of the CO/OC states and lowering the energy barriers between the CO/OC and the CO/OO states. The channel model was then modified to simulate a negative dominant mutation (Nav1.5 p.L325R). Simulations of homodimers and heterodimers in the presence and absence of interactions showed that the interactions with the variant channel impair the opening of the wild-type channel and thus contribute to negative dominance. CONCLUSION Our new modeling framework recapitulates qualitatively previous experimental observations and helps identifying possible interaction mechanisms between ion channels.
Collapse
Affiliation(s)
| | | | - Jan P. Kucera
- Department of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
19
|
Rühlmann AH, Körner J, Hausmann R, Bebrivenski N, Neuhof C, Detro-Dassen S, Hautvast P, Benasolo CA, Meents J, Machtens JP, Schmalzing G, Lampert A. Uncoupling sodium channel dimers restores the phenotype of a pain-linked Na v 1.7 channel mutation. Br J Pharmacol 2020; 177:4481-4496. [PMID: 32663327 PMCID: PMC7484505 DOI: 10.1111/bph.15196] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 06/24/2020] [Accepted: 06/27/2020] [Indexed: 12/11/2022] Open
Abstract
Background and Purpose The voltage‐gated sodium channel Nav1.7 is essential for adequate perception of painful stimuli. Mutations in the encoding gene, SCN9A, cause various pain syndromes in humans. The hNav1.7/A1632E channel mutant causes symptoms of erythromelalgia and paroxysmal extreme pain disorder (PEPD), and its main gating change is a strongly enhanced persistent current. On the basis of recently published 3D structures of voltage‐gated sodium channels, we investigated how the inactivation particle binds to the channel, how this mechanism is altered by the hNav1.7/A1632E mutation, and how dimerization modifies function of the pain‐linked mutation. Experimental Approach We applied atomistic molecular simulations to demonstrate the effect of the mutation on channel fast inactivation. Native PAGE was used to demonstrate channel dimerization, and electrophysiological measurements in HEK cells and Xenopus laevis oocytes were used to analyze the links between functional channel dimerization and impairment of fast inactivation by the hNav1.7/A1632E mutation. Key Results Enhanced persistent current through hNav1.7/A1632E channels was caused by impaired binding of the inactivation particle, which inhibits proper functioning of the recently proposed allosteric fast inactivation mechanism. hNav1.7 channels form dimers and the disease‐associated persistent current through hNav1.7/A1632E channels depends on their functional dimerization status: Expression of the synthetic peptide difopein, a 14‐3‐3 inhibitor known to functionally uncouple dimers, decreased hNav1.7/A1632E channel‐induced persistent currents. Conclusion and Implications Functional uncoupling of mutant hNav1.7/A1632E channel dimers restored their defective allosteric fast inactivation mechanism. Our findings support the concept of sodium channel dimerization and reveal its potential relevance for human pain syndromes.
Collapse
Affiliation(s)
- Annika H Rühlmann
- Institute of Physiology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany
| | - Jannis Körner
- Institute of Physiology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany.,Department of Anaesthesiology, Medical Faculty, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany
| | - Ralf Hausmann
- Institute of Clinical Pharmacology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany
| | - Nikolay Bebrivenski
- Institute of Clinical Pharmacology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany
| | - Christian Neuhof
- Institute of Clinical Pharmacology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany
| | - Silvia Detro-Dassen
- Institute of Clinical Pharmacology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany
| | - Petra Hautvast
- Institute of Physiology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany
| | - Carène A Benasolo
- Institute of Clinical Pharmacology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany.,Forschungszentrum Jülich, Institute of Biological Information Processing (IBI-1), Molekular- und Zellphysiologie, and JARA-HPC, Jülich, Germany
| | - Jannis Meents
- Institute of Physiology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany
| | - Jan-Philipp Machtens
- Institute of Clinical Pharmacology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany.,Forschungszentrum Jülich, Institute of Biological Information Processing (IBI-1), Molekular- und Zellphysiologie, and JARA-HPC, Jülich, Germany
| | - Günther Schmalzing
- Institute of Clinical Pharmacology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany
| | - Angelika Lampert
- Institute of Physiology, Uniklinik RWTH Aachen University, Pauwelsstrasse 30, Aachen, Deutschland, 52074, Germany
| |
Collapse
|
20
|
Inter-Regulation of K v4.3 and Voltage-Gated Sodium Channels Underlies Predisposition to Cardiac and Neuronal Channelopathies. Int J Mol Sci 2020; 21:ijms21145057. [PMID: 32709127 PMCID: PMC7404392 DOI: 10.3390/ijms21145057] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/04/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Genetic variants in voltage-gated sodium channels (Nav) encoded by SCNXA genes, responsible for INa, and Kv4.3 channels encoded by KCND3, responsible for the transient outward current (Ito), contribute to the manifestation of both Brugada syndrome (BrS) and spinocerebellar ataxia (SCA19/22). We examined the hypothesis that Kv4.3 and Nav variants regulate each other’s function, thus modulating INa/Ito balance in cardiomyocytes and INa/I(A) balance in neurons. Methods: Bicistronic and other constructs were used to express WT or variant Nav1.5 and Kv4.3 channels in HEK293 cells. INa and Ito were recorded. Results: SCN5A variants associated with BrS reduced INa, but increased Ito. Moreover, BrS and SCA19/22 KCND3 variants associated with a gain of function of Ito, significantly reduced INa, whereas the SCA19/22 KCND3 variants associated with a loss of function (LOF) of Ito significantly increased INa. Auxiliary subunits Navβ1, MiRP3 and KChIP2 also modulated INa/Ito balance. Co-immunoprecipitation and Duolink studies suggested that the two channels interact within the intracellular compartments and biotinylation showed that LOF SCN5A variants can increase Kv4.3 cell-surface expression. Conclusion: Nav and Kv4.3 channels modulate each other’s function via trafficking and gating mechanisms, which have important implications for improved understanding of these allelic cardiac and neuronal syndromes.
Collapse
|
21
|
Salvage SC, Rees JS, McStea A, Hirsch M, Wang L, Tynan CJ, Reed MW, Irons JR, Butler R, Thompson AJ, Martin-Fernandez ML, Huang CL, Jackson AP. Supramolecular clustering of the cardiac sodium channel Nav1.5 in HEK293F cells, with and without the auxiliary β3-subunit. FASEB J 2020; 34:3537-3553. [PMID: 31950564 PMCID: PMC7079131 DOI: 10.1096/fj.201701473rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 09/30/2019] [Accepted: 10/08/2019] [Indexed: 01/24/2023]
Abstract
Voltage-gated sodium channels comprise an ion-selective α-subunit and one or more associated β-subunits. The β3-subunit (encoded by the SCN3B gene) is an important physiological regulator of the heart-specific sodium channel, Nav1.5. We have previously shown that when expressed alone in HEK293F cells, the full-length β3-subunit forms trimers in the plasma membrane. We extend this result with biochemical assays and use the proximity ligation assay (PLA) to identify oligomeric β3-subunits, not just at the plasma membrane, but throughout the secretory pathway. We then investigate the corresponding clustering properties of the α-subunit and the effects upon these of the β3-subunits. The oligomeric status of the Nav1.5 α-subunit in vivo, with or without the β3-subunit, has not been previously investigated. Using super-resolution fluorescence imaging, we show that under conditions typically used in electrophysiological studies, the Nav1.5 α-subunit assembles on the plasma membrane of HEK293F cells into spatially localized clusters rather than individual and randomly dispersed molecules. Quantitative analysis indicates that the β3-subunit is not required for this clustering but β3 does significantly change the distribution of cluster sizes and nearest-neighbor distances between Nav1.5 α-subunits. However, when assayed by PLA, the β3-subunit increases the number of PLA-positive signals generated by anti-(Nav1.5 α-subunit) antibodies, mainly at the plasma membrane. Since PLA can be sensitive to the orientation of proteins within a cluster, we suggest that the β3-subunit introduces a significant change in the relative alignment of individual Nav1.5 α-subunits, but the clustering itself depends on other factors. We also show that these structural and higher-order changes induced by the β3-subunit do not alter the degree of electrophysiological gating cooperativity between Nav1.5 α-subunits. Our data provide new insights into the role of the β3-subunit and the supramolecular organization of sodium channels, in an important model cell system that is widely used to study Nav channel behavior.
Collapse
Affiliation(s)
| | | | - Alexandra McStea
- Central Laser FacilityResearch Complex at HarwellScience and Technology Facilities CouncilRutherford Appleton LaboratoryOxfordUK
| | - Michael Hirsch
- Central Laser FacilityResearch Complex at HarwellScience and Technology Facilities CouncilRutherford Appleton LaboratoryOxfordUK
| | - Lin Wang
- Central Laser FacilityResearch Complex at HarwellScience and Technology Facilities CouncilRutherford Appleton LaboratoryOxfordUK
| | - Christopher J. Tynan
- Central Laser FacilityResearch Complex at HarwellScience and Technology Facilities CouncilRutherford Appleton LaboratoryOxfordUK
| | - Matthew W. Reed
- Department of Nuclear PhysicsResearch School of Physics and EngineeringAustralian National UniversityCanberrraACTAustralia
| | | | - Richard Butler
- Wellcome Trust/Cancer Research UK Gurdon InstituteUniversity of CambridgeCambridgeUK
| | | | - Marisa L. Martin-Fernandez
- Central Laser FacilityResearch Complex at HarwellScience and Technology Facilities CouncilRutherford Appleton LaboratoryOxfordUK
| | - Christopher L.‐H. Huang
- Deparment of BiochemistryUniversity of CambridgeCambridgeUK
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
22
|
Wadsworth PA, Folorunso O, Nguyen N, Singh AK, D'Amico D, Powell RT, Brunell D, Allen J, Stephan C, Laezza F. High-throughput screening against protein:protein interaction interfaces reveals anti-cancer therapeutics as potent modulators of the voltage-gated Na + channel complex. Sci Rep 2019; 9:16890. [PMID: 31729429 PMCID: PMC6858373 DOI: 10.1038/s41598-019-53110-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/28/2019] [Indexed: 11/09/2022] Open
Abstract
Multiple voltage-gated Na+ (Nav) channelopathies can be ascribed to subtle changes in the Nav macromolecular complex. Fibroblast growth factor 14 (FGF14) is a functionally relevant component of the Nav1.6 channel complex, a causative link to spinocerebellar ataxia 27 (SCA27) and an emerging risk factor for neuropsychiatric disorders. Yet, how this protein:channel complex is regulated in the cell is still poorly understood. To search for key cellular pathways upstream of the FGF14:Nav1.6 complex, we have developed, miniaturized and optimized an in-cell assay in 384-well plates by stably reconstituting the FGF14:Nav1.6 complex using the split-luciferase complementation assay. We then conducted a high-throughput screening (HTS) of 267 FDA-approved compounds targeting known mediators of cellular signaling. Of the 65 hits initially detected, 24 were excluded based on counter-screening and cellular toxicity. Based on target analysis, potency and dose-response relationships, 5 compounds were subsequently repurchased for validation and confirmed as hits. Among those, the tyrosine kinase inhibitor lestaurtinib was highest ranked, exhibiting submicromolar inhibition of FGF14:Nav1.6 assembly. While providing evidence for a robust in-cell HTS platform that can be adapted to search for any channelopathy-associated regulatory proteins, these results lay the potential groundwork for repurposing cancer drugs for neuropsychopharmacology.
Collapse
Affiliation(s)
- Paul A Wadsworth
- MD/PhD Combined Degree Program and Biochemistry and Molecular Biology Graduate Program, The University of Texas Medical Branch, Galveston, Texas, 77555, USA.,Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Oluwarotimi Folorunso
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Nghi Nguyen
- HTS Screening Core, Center for Translational Cancer Research, Texas A&M Health Science Center: Institute of Biosciences and Technology, Houston, TX, 77030, USA
| | - Aditya K Singh
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Daniela D'Amico
- Neuroscience Graduate Program, The University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Reid T Powell
- HTS Screening Core, Center for Translational Cancer Research, Texas A&M Health Science Center: Institute of Biosciences and Technology, Houston, TX, 77030, USA
| | - David Brunell
- HTS Screening Core, Center for Translational Cancer Research, Texas A&M Health Science Center: Institute of Biosciences and Technology, Houston, TX, 77030, USA
| | - John Allen
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Clifford Stephan
- HTS Screening Core, Center for Translational Cancer Research, Texas A&M Health Science Center: Institute of Biosciences and Technology, Houston, TX, 77030, USA
| | - Fernanda Laezza
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, Texas, 77555, USA.
| |
Collapse
|
23
|
Inglis GAS, Wong JC, Butler KM, Thelin JT, Mistretta OC, Wu X, Lin X, English AW, Escayg A. Mutations in the Scn8a DIIS4 voltage sensor reveal new distinctions among hypomorphic and null Na v 1.6 sodium channels. GENES BRAIN AND BEHAVIOR 2019; 19:e12612. [PMID: 31605437 DOI: 10.1111/gbb.12612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/30/2019] [Accepted: 09/22/2019] [Indexed: 12/14/2022]
Abstract
Mutations in the voltage-gated sodium channel gene SCN8A cause a broad range of human diseases, including epilepsy, intellectual disability, and ataxia. Here we describe three mouse lines on the C57BL/6J background with novel, overlapping mutations in the Scn8a DIIS4 voltage sensor: an in-frame 9 bp deletion (Δ9), an in-frame 3 bp insertion (∇3) and a 35 bp deletion that results in a frameshift and the generation of a null allele (Δ35). Scn8a Δ9/+ and Scn8a ∇3/+ heterozygous mutants display subtle motor deficits, reduced acoustic startle response, and are resistant to induced seizures, suggesting that these mutations reduce activity of the Scn8a channel protein, Nav 1.6. Heterozygous Scn8a Δ35/+ mutants show no alterations in motor function or acoustic startle response, but are resistant to induced seizures. Homozygous mutants from each line exhibit premature lethality and severe motor impairments, ranging from uncoordinated gait with tremor (Δ9 and ∇3) to loss of hindlimb control (Δ35). Scn8a Δ9/Δ9 and Scn8a ∇3/∇3 homozygous mutants also exhibit impaired nerve conduction velocity, while normal nerve conduction was observed in Scn8a Δ35/Δ35 homozygous mice. Our results suggest that hypomorphic mutations that reduce Nav 1.6 activity will likely result in different clinical phenotypes compared to null alleles. These three mouse lines represent a valuable opportunity to examine the phenotypic impacts of hypomorphic and null Scn8a mutations without the confound of strain-specific differences.
Collapse
Affiliation(s)
| | - Jennifer C Wong
- Department of Human Genetics, Emory University, Atlanta, Georgia
| | - Kameryn M Butler
- Department of Human Genetics, Emory University, Atlanta, Georgia
| | | | | | - Xuewen Wu
- Department of Cell Biology, Emory University, Atlanta, Georgia.,Department of Otolaryngology, Emory University School of Medicine, Atlanta, Georgia
| | - Xi Lin
- Department of Cell Biology, Emory University, Atlanta, Georgia.,Department of Otolaryngology, Emory University School of Medicine, Atlanta, Georgia
| | | | - Andrew Escayg
- Department of Human Genetics, Emory University, Atlanta, Georgia
| |
Collapse
|
24
|
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
|
25
|
Begemann A, Acuña MA, Zweier M, Vincent M, Steindl K, Bachmann-Gagescu R, Hackenberg A, Abela L, Plecko B, Kroell-Seger J, Baumer A, Yamakawa K, Inoue Y, Asadollahi R, Sticht H, Zeilhofer HU, Rauch A. Further corroboration of distinct functional features in SCN2A variants causing intellectual disability or epileptic phenotypes. Mol Med 2019; 25:6. [PMID: 30813884 PMCID: PMC6391808 DOI: 10.1186/s10020-019-0073-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/05/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Deleterious variants in the voltage-gated sodium channel type 2 (Nav1.2) lead to a broad spectrum of phenotypes ranging from benign familial neonatal-infantile epilepsy (BFNIE), severe developmental and epileptic encephalopathy (DEE) and intellectual disability (ID) to autism spectrum disorders (ASD). Yet, the underlying mechanisms are still incompletely understood. METHODS To further elucidate the genotype-phenotype correlation of SCN2A variants we investigated the functional effects of six variants representing the phenotypic spectrum by whole-cell patch-clamp studies in transfected HEK293T cells and in-silico structural modeling. RESULTS The two variants p.L1342P and p.E1803G detected in patients with early onset epileptic encephalopathy (EE) showed profound and complex changes in channel gating, whereas the BFNIE variant p.L1563V exhibited only a small gain of channel function. The three variants identified in ID patients without seizures, p.R937C, p.L611Vfs*35 and p.W1716*, did not produce measurable currents. Homology modeling of the missense variants predicted structural impairments consistent with the electrophysiological findings. CONCLUSIONS Our findings support the hypothesis that complete loss-of-function variants lead to ID without seizures, small gain-of-function variants cause BFNIE and EE variants exhibit variable but profound Nav1.2 gating changes. Moreover, structural modeling was able to predict the severity of the variant impact, supporting a potential role of structural modeling as a prognostic tool. Our study on the functional consequences of SCN2A variants causing the distinct phenotypes of EE, BFNIE and ID contributes to the elucidation of mechanisms underlying the broad phenotypic variability reported for SCN2A variants.
Collapse
Affiliation(s)
- Anaïs Begemann
- Institute of Medical Genetics, University of Zurich, 8952, Schlieren, Zurich, Switzerland.,radiz-Rare Disease Initiative Zürich, Clinical Research Priority Program for Rare Diseases, University of Zurich, 8006, Zurich, Switzerland
| | - Mario A Acuña
- radiz-Rare Disease Initiative Zürich, Clinical Research Priority Program for Rare Diseases, University of Zurich, 8006, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, 8057, Zurich, Switzerland
| | - Markus Zweier
- Institute of Medical Genetics, University of Zurich, 8952, Schlieren, Zurich, Switzerland.,radiz-Rare Disease Initiative Zürich, Clinical Research Priority Program for Rare Diseases, University of Zurich, 8006, Zurich, Switzerland
| | - Marie Vincent
- Service de génétique médicale, CHU Nantes, 44093, Nantes, France
| | - Katharina Steindl
- Institute of Medical Genetics, University of Zurich, 8952, Schlieren, Zurich, Switzerland.,radiz-Rare Disease Initiative Zürich, Clinical Research Priority Program for Rare Diseases, University of Zurich, 8006, Zurich, Switzerland
| | | | - Annette Hackenberg
- Division of Child Neurology, University Children's Hospital Zurich, 8032, Zurich, Switzerland
| | - Lucia Abela
- radiz-Rare Disease Initiative Zürich, Clinical Research Priority Program for Rare Diseases, University of Zurich, 8006, Zurich, Switzerland.,Division of Child Neurology, University Children's Hospital Zurich, 8032, Zurich, Switzerland
| | - Barbara Plecko
- radiz-Rare Disease Initiative Zürich, Clinical Research Priority Program for Rare Diseases, University of Zurich, 8006, Zurich, Switzerland.,Division of Child Neurology, University Children's Hospital Zurich, 8032, Zurich, Switzerland.,Division of General Pediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Graz, 8036, Graz, Austria
| | - Judith Kroell-Seger
- Children's department, Swiss Epilepsy Centre, Clinic Lengg, 8008, Zurich, Switzerland
| | - Alessandra Baumer
- Institute of Medical Genetics, University of Zurich, 8952, Schlieren, Zurich, Switzerland
| | - Kazuhiro Yamakawa
- Laboratory for Neurogenetics, RIKEN Center for Brain Science, Wako-shi, Saitama, 351-0198, Japan
| | - Yushi Inoue
- National Epilepsy Center, NHO Shizuoka Institute of Epilepsy and Neurological Disorders, Shizuoka, 420-8688, Japan
| | - Reza Asadollahi
- Institute of Medical Genetics, University of Zurich, 8952, Schlieren, Zurich, Switzerland.,radiz-Rare Disease Initiative Zürich, Clinical Research Priority Program for Rare Diseases, University of Zurich, 8006, Zurich, Switzerland
| | - Heinrich Sticht
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Hanns Ulrich Zeilhofer
- radiz-Rare Disease Initiative Zürich, Clinical Research Priority Program for Rare Diseases, University of Zurich, 8006, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, 8057, Zurich, Switzerland.,Institute of Pharmaceutical Sciences, ETH Zurich, 8093, Zürich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057, Zurich, Switzerland
| | - Anita Rauch
- Institute of Medical Genetics, University of Zurich, 8952, Schlieren, Zurich, Switzerland. .,radiz-Rare Disease Initiative Zürich, Clinical Research Priority Program for Rare Diseases, University of Zurich, 8006, Zurich, Switzerland. .,Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057, Zurich, Switzerland. .,Zurich Center for Integrative Human Physiology, University of Zurich, 8057, Zurich, Switzerland.
| |
Collapse
|
26
|
Wilders R. Cellular Mechanisms of Sinus Node Dysfunction in Carriers of the SCN5A-E161K Mutation and Role of the H558R Polymorphism. Front Physiol 2018; 9:1795. [PMID: 30618807 PMCID: PMC6305593 DOI: 10.3389/fphys.2018.01795] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/29/2018] [Indexed: 12/16/2022] Open
Abstract
Background: Carriers of the E161K mutation in the SCN5A gene, encoding the NaV1.5 pore-forming α-subunit of the ion channel carrying the fast sodium current (INa), show sinus bradycardia and occasional exit block. Voltage clamp experiments in mammalian expression systems revealed a mutation-induced 2.5- to 4-fold reduction in INa peak current density as well as a +19 mV shift and reduced steepness of the steady-state activation curve. The highly common H558R polymorphism in NaV1.5 limits this shift to +13 mV, but also introduces a -10 mV shift in steady-state inactivation. Aim: We assessed the cellular mechanism by which the E161K mutation causes sinus node dysfunction in heterozygous mutation carriers as well as the potential role of the H558R polymorphism. Methods: We incorporated the mutation-induced changes in INa into the Fabbri-Severi model of a single human sinoatrial node cell and the Maleckar et al. human atrial cell model, and carried out simulations under control conditions and over a wide range of acetylcholine levels. Results: In absence of the H558R polymorphism, the E161K mutation increased the basic cycle length of the sinoatrial node cell from 813 to 866 ms. In the simulated presence of 10 and 25 nM acetylcholine, basic cycle length increased from 1027 to 1131 and from 1448 to 1795 ms, respectively. The increase in cycle length was the result of a significant slowing of diastolic depolarization. The mutation-induced reduction in INa window current had reduced the contribution of the mutant component of INa to the net membrane current during diastolic depolarization to effectively zero. Highly similar results were obtained in presence of the H558R polymorphism. Atrial excitability was reduced, both in absence and presence of the H558R polymorphism, as reflected by an increase in threshold stimulus current and a concomitant decrease in capacitive current of the atrial cell. Conclusion: We conclude that the experimentally identified mutation-induced changes in INa can explain the clinically observed sinus bradycardia and potentially the occasional exit block. Furthermore, we conclude that the common H558R polymorphism does not significantly alter the effects of the E161K mutation and can thus not explain the reduced penetrance of the E161K mutation.
Collapse
Affiliation(s)
- Ronald Wilders
- Department of Medical Biology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|