51
|
Bragança B, Nogueira-Marques S, Ferreirinha F, Fontes-Sousa AP, Correia-de-Sá P. The Ionotropic P2X4 Receptor has Unique Properties in the Heart by Mediating the Negative Chronotropic Effect of ATP While Increasing the Ventricular Inotropy. Front Pharmacol 2019; 10:1103. [PMID: 31611793 PMCID: PMC6769074 DOI: 10.3389/fphar.2019.01103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/28/2019] [Indexed: 01/09/2023] Open
Abstract
Background: Mounting evidence indicate that reducing the sinoatrial node (SAN) activity may be a useful therapeutic strategy to control of heart failure. Purines, like ATP and its metabolite adenosine, consistently reduce the SAN spontaneous activity leading to negative cardiac chronotropy, with variable effects on the force of myocardial contraction (inotropy). Apart from adenosine A1 receptors, the human SAN expresses high levels of ATP-sensitive ionotropic P2X4 receptors (P2X4R), yet their cardiac role is unexplored. Methods: Here, we investigated the activity of P2 purinoceptors on isolated spontaneously beating atria (chronotropy) and on 2 Hz-paced right ventricular (RV, inotropy) strips from Wistar rats. Results: ATP (pEC 50 = 4.05) and its stable analogue ATPγS (pEC 50 = 4.69) concentration-dependently reduced atrial chronotropy. Inhibition of ATP breakdown into adenosine by NTPDases with POM-1 failed to modify ATP-induced negative chronotropy. The effect of ATP on atrial rate was attenuated by a broad-spectrum P2 antagonist, PPADS, as well as by 5-BDBD, which selectively blocks the P2X4R subtype; however, no effect was observed upon blocking the A1 receptor with DPCPX. The P2X4R positive allosteric modulator, ivermectin, increased the negative chronotropic response of ATP. Likewise, CTP, a P2X agonist that does not generate adenosine, replicated the P2X4R-mediated negative chronotropism of ATP. Inhibition of the Na+/Ca2+ exchanger (NCX) with KB-R7943 and ORM-10103, but not blockage of the HCN channel with ZD7288, mimicked the effect of the P2X4R blocker, 5-BDBD. In paced RV strips, ATP caused a mild negative inotropic effect, which magnitude was 2 to 3-fold increased by 5-BDBD and KB-R7943. Immunofluorescence confocal microscopy studies confirm that cardiomyocytes of the rat SAN and RV co-express P2X4R and NCX1 proteins. Conclusions: Data suggest that activation of ATP-sensitive P2X4R slows down heart rate by reducing the SAN activity while increasing the magnitude of ventricular contractions. The mechanism underlying the dual effect of ATP in the heart may involve inhibition of intracellular Ca2+-extrusion by bolstering NCX function in the reverse mode. Thus, targeting the P2X4R activation may create novel well-tolerated heart-rate lowering drugs with potential benefits in patients with deteriorated ventricular function.
Collapse
Affiliation(s)
- Bruno Bragança
- Laboratório de Farmacologia e Neurobiologia, Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal.,Hospital Pedro Hispano, ULS Matosinhos, Matosinhos, Portugal
| | - Sílvia Nogueira-Marques
- Laboratório de Farmacologia e Neurobiologia, Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Fátima Ferreirinha
- Laboratório de Farmacologia e Neurobiologia, Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana Patrícia Fontes-Sousa
- Laboratório de Farmacologia e Neurobiologia, Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
52
|
Hu RM, Tester DJ, Li R, Sun T, Peterson BZ, Ackerman MJ, Makielski JC, Tan BH. Mexiletine rescues a mixed biophysical phenotype of the cardiac sodium channel arising from the SCN5A mutation, N406K, found in LQT3 patients. Channels (Austin) 2019; 12:176-186. [PMID: 29983085 PMCID: PMC6104686 DOI: 10.1080/19336950.2018.1475794] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction: Individual mutations in the SCN5A-encoding cardiac sodium channel α-subunit usually cause a single cardiac arrhythmia disorder, some cause mixed biophysical or clinical phenotypes. Here we report an infant, female patient harboring a N406K mutation in SCN5A with a marked and mixed biophysical phenotype and assess pathogenic mechanisms. Methods and Results: A patient suffered from recurrent seizures during sleep and torsades de pointes with a QTc of 530 ms. Mutational analysis identified a N406K mutation in SCN5A. The mutation was engineered by site-directed mutagenesis and heterologously expressed in HEK293 cells. After 48 hours incubation with and without mexiletine, macroscopic voltage-gated sodium current (INa) was measured with standard whole-cell patch clamp techniques. SCN5A-N406K elicited both a significantly decreased peak INa and a significantly increased late INa compared to wide-type (WT) channels. Furthermore, mexiletine both restored the decreased peak INa of the mutant channel and inhibited the increased late INa of the mutant channel. Conclusion: SCN5A-N406K channel displays both “gain-of-function” in late INa and “loss-of-function” in peak INa density contributing to a mixed biophysical phenotype. Moreover, our finding may provide the first example that mexiletine exerts a dual rescue of both “gain-of-function” and “loss-of-function” of the mutant sodium channel.
Collapse
Affiliation(s)
- Rou-Mu Hu
- a Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital , Capital Medical University , Beijing , China.,b Division of Cardiovascular Medicine, Department of Medicine , University of Wisconsin , Madison , Wisconsin , USA
| | - David J Tester
- c Departments of Medicine, Pediatrics, and Molecular Pharmacology and Experimental Therapeutics , Mayo Clinic , Rochester , MN , USA
| | - Ryan Li
- d Departments of Pediatrics, and Cellular & Molecular Physiology , Pennsylvania State University College of Medicine , Hershey , PA , USA
| | - Tianyu Sun
- d Departments of Pediatrics, and Cellular & Molecular Physiology , Pennsylvania State University College of Medicine , Hershey , PA , USA
| | - Blaise Z Peterson
- d Departments of Pediatrics, and Cellular & Molecular Physiology , Pennsylvania State University College of Medicine , Hershey , PA , USA
| | - Michael J Ackerman
- c Departments of Medicine, Pediatrics, and Molecular Pharmacology and Experimental Therapeutics , Mayo Clinic , Rochester , MN , USA
| | - Jonathan C Makielski
- b Division of Cardiovascular Medicine, Department of Medicine , University of Wisconsin , Madison , Wisconsin , USA
| | - Bi-Hua Tan
- b Division of Cardiovascular Medicine, Department of Medicine , University of Wisconsin , Madison , Wisconsin , USA.,d Departments of Pediatrics, and Cellular & Molecular Physiology , Pennsylvania State University College of Medicine , Hershey , PA , USA
| |
Collapse
|
53
|
Pereira PJS, Pugsley MK, Troncy E, Tan W, Pouliot M, Harper C, Prefontaine A, Easter A, Wallis R, Miraucourt L, Huang H, Accardi MV, Boulay E, Maghezzi MS, Authier S. Incidence of spontaneous arrhythmias in freely moving healthy untreated Sprague-Dawley rats. J Pharmacol Toxicol Methods 2019; 99:106589. [PMID: 31154034 DOI: 10.1016/j.vascn.2019.106589] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/07/2019] [Accepted: 05/25/2019] [Indexed: 12/18/2022]
Abstract
Spontaneous arrhythmia characterization in healthy rats can support interpretation when studying novel therapies. Male (n = 55) and female (n = 40) Sprague-Dawley rats with telemetry transmitters for a derivation II ECG. Arrhythmias were assessed from continuous ECG monitoring over a period of 24-48 h, and data analyzed using an automated detection algorithm with 100% manual over-read. While a total of 1825 spontaneous ventricular premature beats (VPB) were identified, only 7 rats (or 7.4%) did not present with any over the recording period. Spontaneous episode(s) of ventricular tachycardia (VT) were noted in males (27%) and females (3%). The incidence of VPB was significantly higher (p < 0.01) during the night time (7 pm-7 am) compared to daytime, while males presented with significantly (p < 0.001) more VPB than females. Most VPB were observed as single ectopic beats, followed by salvos (2 or 3 consecutive VPBs), and VT (i.e. 4 consecutive VPBs). Most VPBs were single premature ventricular contractions (PVCs) (57%), while the remaining were escape complexes (43%). Spontaneous premature junctional complexes (PJC) were also observed and were significantly more frequent during the night, and in males. Lastly, 596 episodes of spontaneous 2nd-degree atrioventricular (AV) block were identified and were significantly more frequent during the day time in males. Most 2nd-degree AV block episodes were Mobitz type I (57%), with a significantly (p < 0.05) higher incidence in males. This work emphasizes the importance of obtaining sufficient baseline data when undertaking arrhythmia analysis in safety study and provides a better understanding of both sex- and time- dependent effects of spontaneous arrhythmias in rats.
Collapse
Affiliation(s)
| | | | - Eric Troncy
- Faculty of Veterinary Medicine, Université de Montréal, St-Hyacinthe, QC, Canada
| | - Wendy Tan
- Faculty of Veterinary Medicine, Université de Montréal, St-Hyacinthe, QC, Canada
| | | | | | | | - Alison Easter
- Praxis Precision Medicines, Cambridge, MA, United States of America
| | - Rob Wallis
- Safety Pharmacology Consultant, London, United Kingdom
| | | | | | | | - Emmanuel Boulay
- Citoxlab, Laval, QC, Canada; Faculty of Veterinary Medicine, Université de Montréal, St-Hyacinthe, QC, Canada
| | | | - Simon Authier
- Citoxlab, Laval, QC, Canada; Faculty of Veterinary Medicine, Université de Montréal, St-Hyacinthe, QC, Canada.
| |
Collapse
|
54
|
Del Canto I, Santamaría L, Genovés P, Such-Miquel L, Arias-Mutis O, Zarzoso M, Soler C, Parra G, Tormos Á, Alberola A, Such L, Chorro FJ. Effects of the Inhibition of Late Sodium Current by GS967 on Stretch-Induced Changes in Cardiac Electrophysiology. Cardiovasc Drugs Ther 2019; 32:413-425. [PMID: 30173392 DOI: 10.1007/s10557-018-6822-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE Mechanical stretch increases sodium and calcium entry into myocytes and activates the late sodium current. GS967, a triazolopyridine derivative, is a sodium channel blocker with preferential effects on the late sodium current. The present study evaluates whether GS967 inhibits or modulates the arrhythmogenic electrophysiological effects of myocardial stretch. METHODS Atrial and ventricular refractoriness and ventricular fibrillation modifications induced by acute stretch were studied in Langendorff-perfused rabbit hearts (n = 28) using epicardial multiple electrodes and high-resolution mapping techniques under control conditions and during the perfusion of GS967 at different concentrations (0.03, 0.1, and 0.3 μM). RESULTS On comparing ventricular refractoriness, conduction velocity and wavelength obtained before stretch had no significant changes under each GS967 concentration while atrial refractoriness increased under GS967 0.3 μM. Under GS967, the stretch-induced changes were attenuated, and no significant differences were observed between before and during stretch. GS967 0.3 μM diminished the normal stretch-induced changes resulting in longer (less shortened) atrial refractoriness (138 ± 26 ms vs 95 ± 9 ms; p < 0.01), ventricular refractoriness (155 ± 18 ms vs 124 ± 16 ms; p < 0.01) and increments in spectral concentration (23 ± 5% vs 17 ± 2%; p < 0.01), the fifth percentile of ventricular activation intervals (46 ± 8 ms vs 31 ± 3 ms; p < 0.05), and wavelength of ventricular fibrillation (2.5 ±0.5 cm vs 1.7 ± 0.3 cm; p < 0.05) during stretch. The stretch-induced increments in dominant frequency during ventricular fibrillation (control = 38%, 0.03 μM = 33%, 0.1 μM = 33%, 0.3 μM = 14%; p < 0.01) and the stretch-induced increments in arrhythmia complexity index (control = 62%, 0.03μM = 41%, 0.1 μM = 32%, 0.3 μM = 16%; p < 0.05) progressively decreased on increasing the GS967 concentration. CONCLUSIONS GS967 attenuates stretch-induced changes in cardiac electrophysiology.
Collapse
Affiliation(s)
- Irene Del Canto
- CIBER CV. Carlos III Health Institute, Madrid, Spain.,Department of Electronics, Universitat Politècnica de València, Valencia, Spain
| | - Laura Santamaría
- Department of Physiology, Valencia University - Estudi General, Valencia, Spain
| | | | - Luis Such-Miquel
- CIBER CV. Carlos III Health Institute, Madrid, Spain.,Department of Physiotherapy, Valencia University - Estudi General, Valencia, Spain
| | | | - Manuel Zarzoso
- Department of Physiotherapy, Valencia University - Estudi General, Valencia, Spain
| | - Carlos Soler
- Department of Physiology, Valencia University - Estudi General, Valencia, Spain
| | - Germán Parra
- Department of Physiology, Valencia University - Estudi General, Valencia, Spain
| | - Álvaro Tormos
- CIBER CV. Carlos III Health Institute, Madrid, Spain.,Department of Electronics, Universitat Politècnica de València, Valencia, Spain
| | - Antonio Alberola
- CIBER CV. Carlos III Health Institute, Madrid, Spain.,Department of Physiology, Valencia University - Estudi General, Valencia, Spain
| | - Luis Such
- CIBER CV. Carlos III Health Institute, Madrid, Spain.,Department of Physiology, Valencia University - Estudi General, Valencia, Spain
| | - Francisco J Chorro
- CIBER CV. Carlos III Health Institute, Madrid, Spain. .,Service of Cardiology, Valencia University Clinic Hospital, INCLIVA, Valencia, Spain. .,Department of Medicine, Valencia University - Estudi General, Valencia, Spain. .,Servicio de Cardiología, Hospital Clínico Universitario, Avda. Blasco Ibañez 17, 46010, Valencia, Spain.
| |
Collapse
|
55
|
Mlynarova J, Trentin-Sonoda M, Gaisler da Silva F, Major JL, Salih M, Carneiro-Ramos MS, Tuana BS. SLMAP3 isoform modulates cardiac gene expression and function. PLoS One 2019; 14:e0214669. [PMID: 30934005 PMCID: PMC6443179 DOI: 10.1371/journal.pone.0214669] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 03/18/2019] [Indexed: 12/23/2022] Open
Abstract
The sarcolemmal membrane associated proteins (SLMAPs) belong to the super family of tail anchored membrane proteins which serve diverse roles in biology including cell growth, protein trafficking and ion channel regulation. Mutations in human SLMAP have been linked to Brugada syndrome with putative deficits in trafficking of the sodium channel (Nav1.5) to the cell membrane resulting in aberrant electrical activity and heart function. Three main SLMAP isoforms (SLMAP1 (35 kDa), SLMAP2 (45 kDa), and SLMAP3 (91 kDa)) are expressed in myocardium but their precise role remains to be defined. Here we generated transgenic (Tg) mice with cardiac-specific expression of the SLMAP3 isoform during postnatal development which present with a significant decrease (20%) in fractional shortening and (11%) in cardiac output at 5 weeks of age. There was a lack of any notable cardiac remodeling (hypertrophy, fibrosis or fetal gene activation) in Tg hearts but the electrocardiogram indicated a significant increase (14%) in the PR interval and a decrease (43%) in the R amplitude. Western blot analysis indicated a selective and significant decrease (55%) in protein levels of Nav1.5 while 45% drop in its transcript levels were detectable by qRT-PCR. Significant decreases in the protein and transcript levels of the calcium transport system of the sarcoplasmic reticulum (SERCA2a/PLN) were also evident in Tg hearts. These data reveal a novel role for SLMAP3 in the selective regulation of important ion transport proteins at the level of gene expression and suggest that it may be a unique target in cardiovascular function and disease.
Collapse
Affiliation(s)
- Jana Mlynarova
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Mayra Trentin-Sonoda
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Fernanda Gaisler da Silva
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Jennifer L. Major
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Maysoon Salih
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | | | - Balwant S. Tuana
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- * E-mail:
| |
Collapse
|
56
|
Coates MD, Vrana KE, Ruiz-Velasco V. The influence of voltage-gated sodium channels on human gastrointestinal nociception. Neurogastroenterol Motil 2019; 31:e13460. [PMID: 30216585 DOI: 10.1111/nmo.13460] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/01/2018] [Accepted: 08/07/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Abdominal pain is a frequent and persistent problem in the most common gastrointestinal disorders, including irritable bowel syndrome and inflammatory bowel disease. Pain adversely impacts quality of life, incurs significant healthcare expenditures, and remains a challenging issue to manage with few safe therapeutic options currently available. It is imperative that new methods are developed for identifying and treating this symptom. A variety of peripherally active neuroendocrine signaling elements have the capability to influence gastrointestinal pain perception. A large and growing body of evidence suggests that voltage-gated sodium channels (VGSCs) play a critical role in the development and modulation of nociceptive signaling associated with the gut. Several VGSC isoforms demonstrate significant promise as potential targets for improved diagnosis and treatment of gut-based disorders associated with hyper- and hyposensitivity to abdominal pain. PURPOSE In this article, we critically review key investigations that have evaluated the potential role that VGSCs play in visceral nociception and discuss recent advances related to this topic. Specifically, we discuss the following: (a) what is known about the structure and basic function of VGSCs, (b) the role that each VGSC plays in gut nociception, particularly as it relates to human physiology, and (c) potential diagnostic and therapeutic uses of VGSCs to manage disorders associated with chronic abdominal pain.
Collapse
Affiliation(s)
- Matthew D Coates
- Division of Gastroenterology & Hepatology, Department of Medicine, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Kent E Vrana
- Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Victor Ruiz-Velasco
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
57
|
Rivaud MR, Baartscheer A, Verkerk AO, Beekman L, Rajamani S, Belardinelli L, Bezzina CR, Remme CA. Enhanced late sodium current underlies pro-arrhythmic intracellular sodium and calcium dysregulation in murine sodium channelopathy. Int J Cardiol 2018; 263:54-62. [PMID: 29754923 DOI: 10.1016/j.ijcard.2018.03.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/23/2018] [Accepted: 03/09/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Long QT syndrome mutations in the SCN5A gene are associated with an enhanced late sodium current (INa,L) which may lead to pro-arrhythmic action potential prolongation and intracellular calcium dysregulation. We here investigated the dynamic relation between INa,L, intracellular sodium ([Na+]i) and calcium ([Ca2+]i) homeostasis and pro-arrhythmic events in the setting of a SCN5A mutation. METHODS AND RESULTS Wild-type (WT) and Scn5a1798insD/+ (MUT) mice (age 3-5 months) carrying the murine homolog of the SCN5A-1795insD mutation on two distinct genetic backgrounds (FVB/N and 129P2) were studied. [Na+]i, [Ca2+]i and Ca2+ transient amplitude were significantly increased in 129P2-MUT myocytes as compared to WT, but not in FVB/N-MUT. Accordingly, INa,L wassignificantly more enhanced in 129P2-MUT than in FVB/N-MUT myocytes, consistent with a dose-dependent correlation. Quantitative RT-PCR analysis revealed intrinsic differences in mRNA expression levels of the sodium/potassium pump, the sodium/hydrogen exchanger, and sodium‑calcium exchanger between the two mouse strains. The rate of increase in [Na+]i, [Ca2+]i and Ca2+ transient amplitude following the application of the Na+/K+-ATPase inhibitor ouabain was significantly greater in 129P2-MUT than in 129P2-WT myocytes and was normalized by the INa,L inhibitor ranolazine. Furthermore, ranolazine decreased the incidence of pro-arrhythmic calcium after-transients elicited in 129P2-MUT myocytes. CONCLUSIONS In this study we established a causal link between the magnitude of INa,L, extent of Na+ and Ca2+ dysregulation, and incidence of pro-arrhythmic events in murine Scn5a1798insD/+ myocytes. Furthermore, our findings provide mechanistic insight into the anti-arrhythmic potential of pharmacological inhibition of INa,L in patients with LQT3 syndrome.
Collapse
Affiliation(s)
- Mathilde R Rivaud
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, Meibergdreef 15, 1105AZ Amsterdam, The Netherlands
| | - Antonius Baartscheer
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, Meibergdreef 15, 1105AZ Amsterdam, The Netherlands
| | - Arie O Verkerk
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, Meibergdreef 15, 1105AZ Amsterdam, The Netherlands; Department of Medical Biology, Academic Medical Center, Meibergdreef 15, 1105AZ Amsterdam, The Netherlands
| | - Leander Beekman
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, Meibergdreef 15, 1105AZ Amsterdam, The Netherlands
| | | | | | - Connie R Bezzina
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, Meibergdreef 15, 1105AZ Amsterdam, The Netherlands
| | - Carol Ann Remme
- Heart Center, Department of Clinical and Experimental Cardiology, Academic Medical Center, Meibergdreef 15, 1105AZ Amsterdam, The Netherlands.
| |
Collapse
|
58
|
Wang W, Zhang S, Ni H, Garratt CJ, Boyett MR, Hancox JC, Zhang H. Mechanistic insight into spontaneous transition from cellular alternans to arrhythmia-A simulation study. PLoS Comput Biol 2018; 14:e1006594. [PMID: 30500818 PMCID: PMC6291170 DOI: 10.1371/journal.pcbi.1006594] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 12/12/2018] [Accepted: 10/23/2018] [Indexed: 02/01/2023] Open
Abstract
Cardiac electrical alternans (CEA), manifested as T-wave alternans in ECG, is a clinical biomarker for predicting cardiac arrhythmias and sudden death. However, the mechanism underlying the spontaneous transition from CEA to arrhythmias remains incompletely elucidated. In this study, multiscale rabbit ventricular models were used to study the transition and a potential role of INa in perpetuating such a transition. It was shown CEA evolved into either concordant or discordant action potential (AP) conduction alternans in a homogeneous one-dimensional tissue model, depending on tissue AP duration and conduction velocity (CV) restitution properties. Discordant alternans was able to cause conduction failure in the model, which was promoted by impaired sodium channel with either a reduced or increased channel current. In a two-dimensional homogeneous tissue model, a combined effect of rate- and curvature-dependent CV broke-up alternating wavefronts at localised points, facilitating a spontaneous transition from CEA to re-entry. Tissue inhomogeneity or anisotropy further promoted break-up of re-entry, leading to multiple wavelets. Similar observations have also been seen in human atrial cellular and tissue models. In conclusion, our results identify a mechanism by which CEA spontaneously evolves into re-entry without a requirement for premature ventricular complexes or pre-existing tissue heterogeneities, and demonstrated the important pro-arrhythmic role of impaired sodium channel activity. These findings are model-independent and have potential human relevance.
Collapse
Affiliation(s)
- Wei Wang
- Biological Physics Group, School of Physics & Astronomy, The University of Manchester, Manchester, United Kingdom
| | - Shanzhuo Zhang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Haibo Ni
- Biological Physics Group, School of Physics & Astronomy, The University of Manchester, Manchester, United Kingdom
| | - Clifford J. Garratt
- Manchester Heart Centre, Manchester Royal Infirmary, Manchester, United Kingdom
| | - Mark R. Boyett
- Manchester Heart Centre, Manchester Royal Infirmary, Manchester, United Kingdom
| | - Jules C. Hancox
- Biological Physics Group, School of Physics & Astronomy, The University of Manchester, Manchester, United Kingdom
- School of Physiology, Pharmacology and Neuroscience, and Cardiovascular Research Laboratories, School of Medical Sciences, University of Bristol, Bristol, United Kingdom
| | - Henggui Zhang
- Biological Physics Group, School of Physics & Astronomy, The University of Manchester, Manchester, United Kingdom
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
- Space Institute of Southern China, Shenzhen, China
| |
Collapse
|
59
|
Lou J, Wu H, Wang L, Zhao L, Li X, Kang Y, Wen K, Yin Y. Taurine-magnesium coordination compound, a potential anti-arrhythmic complex, improves aconitine-induced arrhythmias through regulation of multiple ion channels. Toxicol Appl Pharmacol 2018; 356:182-190. [PMID: 30125596 DOI: 10.1016/j.taap.2018.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/03/2018] [Accepted: 08/14/2018] [Indexed: 12/14/2022]
Abstract
Taurine-magnesium coordination compound (TMCC) exhibits antiarrhythmic effects in cesium-chloride-and ouabain-induced arrhythmias; however, the mechanism underlying these effects on arrhythmia remains poorly understood. Here, we investigated the effects of TMCC on aconitine-induced arrhythmia in vivo and the electrophysiological effects of this compound in rat ventricular myocytes in vitro. Aconitine was used to induce arrhythmias in rats, and the dosages required to produce ventricular premature contraction (VPC), ventricular tachycardia (VT), ventricular fibrillation (VF), and cardiac arrest (CA) were recorded. Additionally, the sodium current (INa) and L-type calcium current (ICa,L) were analyzed in normal and aconitine-treated ventricular myocytes using whole-cell patch-clamp recording. In vivo, intravenous administration of TMCC produced marked antiarrhythmic effects, as indicated by the increased dose of aconitine required to induce VPC, VT, VF, and CA. Moreover, this effect was abolished by administration of sodium channel opener veratridine and calcium channel agonist Bay K8644. In vitro, TMCC inhibited aconitine-induced increases in INa and ICa,L. These results revealed that TMCC inhibited aconitine-induced arrhythmias through effects on INa and ICa,L.
Collapse
Affiliation(s)
- Jianshi Lou
- Department of Pharmacology, Tianjin Medical University, Tianjin, PR China
| | - Hong Wu
- Department of Pharmacology, Tianjin Medical University, Tianjin, PR China; Mudanjiang Medical University, Mudanjiang, PR China
| | - Lingfang Wang
- Institute of Translational Medicine, Nanchang University, Nanchang, PR China
| | - Lin Zhao
- International College, Tianjin Medical University, Tianjin, PR China
| | - Xin Li
- Department of Pharmacology, Tianjin Medical University, Tianjin, PR China
| | - Yi Kang
- Department of Pharmacology, Tianjin Medical University, Tianjin, PR China
| | - Ke Wen
- Department of Pharmacology, Tianjin Medical University, Tianjin, PR China
| | - Yongqiang Yin
- Department of Pharmacology, Tianjin Medical University, Tianjin, PR China.
| |
Collapse
|
60
|
Arvanitis DA, Vafiadaki E, Johnson DM, Kranias EG, Sanoudou D. The Histidine-Rich Calcium Binding Protein in Regulation of Cardiac Rhythmicity. Front Physiol 2018; 9:1379. [PMID: 30319456 PMCID: PMC6171002 DOI: 10.3389/fphys.2018.01379] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 09/11/2018] [Indexed: 12/16/2022] Open
Abstract
Sudden unexpected cardiac death (SCD) accounts for up to half of all-cause mortality of heart failure patients. Standardized cardiology tools such as electrocardiography, cardiac imaging, electrophysiological and serum biomarkers cannot accurately predict which patients are at risk of life-threatening arrhythmic episodes. Recently, a common variant of the histidine-rich calcium binding protein (HRC), the Ser96Ala, was identified as a potent biomarker of malignant arrhythmia triggering in these patients. HRC has been shown to be involved in the regulation of cardiac sarcoplasmic reticulum (SR) Ca2+ cycling, by binding and storing Ca2+ in the SR, as well as interacting with the SR Ca2+ uptake and release complexes. The underlying mechanisms, elucidated by studies at the molecular, biochemical, cellular and intact animal levels, indicate that transversion of Ser96 to Ala results in abolishment of an HRC phosphorylation site by Fam20C kinase and dysregulation of SR Ca2+ cycling. This is mediated through aberrant SR Ca2+ release by the ryanodine receptor (RyR2) quaternary complex, due to the impaired HRC/triadin interaction, and depressed SR Ca2+ uptake by the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA2) pump, due to the impaired HRC/SERCA2 interaction. Pharmacological intervention with KN-93, an inhibitor of Ca2+/calmodulin-dependent protein kinase II (CaMKII), in the HRC Ser96Ala mouse model, reduced the occurrence of malignant cardiac arrhythmias. Herein, we summarize the current evidence on the pivotal role of HRC in the regulation of cardiac rhythmicity and the importance of HRC Ser96Ala as a genetic modifier for arrhythmias in the setting of heart failure.
Collapse
Affiliation(s)
- Demetrios A Arvanitis
- Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Elizabeth Vafiadaki
- Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Daniel M Johnson
- Department of Cardiothoracic Surgery, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Evangelia G Kranias
- Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece.,Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Despina Sanoudou
- Molecular Biology Division, Biomedical Research Foundation, Academy of Athens, Athens, Greece.,Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
61
|
Agwa AJ, Blomster LV, Craik DJ, King GF, Schroeder CI. Efficient Enzymatic Ligation of Inhibitor Cystine Knot Spider Venom Peptides: Using Sortase A To Form Double-Knottins That Probe Voltage-Gated Sodium Channel NaV1.7. Bioconjug Chem 2018; 29:3309-3319. [DOI: 10.1021/acs.bioconjchem.8b00505] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Akello J. Agwa
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Linda V. Blomster
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - David J. Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Christina I. Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
62
|
Human iPSC-Derived Cardiomyocytes for Investigation of Disease Mechanisms and Therapeutic Strategies in Inherited Arrhythmia Syndromes: Strengths and Limitations. Cardiovasc Drugs Ther 2018; 31:325-344. [PMID: 28721524 PMCID: PMC5550530 DOI: 10.1007/s10557-017-6735-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During the last two decades, significant progress has been made in the identification of genetic defects underlying inherited arrhythmia syndromes, which has provided some clinical benefit through elucidation of gene-specific arrhythmia triggers and treatment. However, for most arrhythmia syndromes, clinical management is hindered by insufficient knowledge of the functional consequences of the mutation in question, the pro-arrhythmic mechanisms involved, and hence the most optimal treatment strategy. Moreover, disease expressivity and sensitivity to therapeutic interventions often varies between mutations and/or patients, underlining the need for more individualized strategies. The development of the induced pluripotent stem cell (iPSC) technology now provides the opportunity for generating iPSC-derived cardiomyocytes (CMs) from human material (hiPSC-CMs), enabling patient- and/or mutation-specific investigations. These hiPSC-CMs may furthermore be employed for identification and assessment of novel therapeutic strategies for arrhythmia syndromes. However, due to their relative immaturity, hiPSC-CMs also display a number of essential differences as compared to adult human CMs, and hence there are certain limitations in their use. We here review the electrophysiological characteristics of hiPSC-CMs, their use for investigating inherited arrhythmia syndromes, and their applicability for identification and assessment of (novel) anti-arrhythmic treatment strategies.
Collapse
|
63
|
Josifovska S, Vazharova R, Balabanski L, Malinov M, Kaneva A, Panov S, Hadjidekova S, Toncheva D. Double heterozygosity of novel variants found in patients with severe clinical phenotype of cardiovascular disorders. BIOTECHNOL BIOTEC EQ 2018. [DOI: 10.1080/13102818.2018.1433064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Slavica Josifovska
- Laboratory of Molecular Biology, Faculty of Natural Sciences and Mathematics, “Ss.Cyril and Methodius” University, Skopje, Macedonia
- Genome Centre, GARH “Malinov”, Sofia, Bulgaria
- Department of Medical Genetics, Medical University of Sofia, Sofia, Bulgaria
| | - Radoslava Vazharova
- Genome Centre, GARH “Malinov”, Sofia, Bulgaria
- Department of Biology, Medical Genetics and Microbiology, Faculty of Medicine, Sofia University “St. Kliment Ohridski,” Sofia, Bulgaria
| | | | | | - Anna Kaneva
- Department of Pediatric Cardiology, National Heart Hospital, Sofia, Bulgaria
| | - Sasho Panov
- Laboratory of Molecular Biology, Faculty of Natural Sciences and Mathematics, “Ss.Cyril and Methodius” University, Skopje, Macedonia
| | - Savina Hadjidekova
- Department of Medical Genetics, Medical University of Sofia, Sofia, Bulgaria
| | - Draga Toncheva
- Department of Medical Genetics, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
64
|
Haverinen J, Hassinen M, Korajoki H, Vornanen M. Cardiac voltage-gated sodium channel expression and electrophysiological characterization of the sodium current in the zebrafish (Danio rerio) ventricle. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 138:59-68. [PMID: 29655910 DOI: 10.1016/j.pbiomolbio.2018.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/22/2018] [Accepted: 04/11/2018] [Indexed: 12/11/2022]
Abstract
Na+ channel α-subunit composition of the zebrafish heart and electrophysiological properties of Na+ current (INa) of zebrafish ventricular myocytes were examined. Eight Na+ channel α-subunits were expressed in both atrium and ventricle of the zebrafish heart. Nav1.5Lb, an orthologue to the human Nav1.5, was clearly the predominant isoform in both chambers representing 65.2 ± 4.1% and 83.1 ± 2.1% of all Na+ channel transcripts in atrium and ventricle, respectively. Nav1.4b, an orthologue to human Nav1.4, formed 34.1 ± 4.1 and 16.2 ± 2.0% of the Na+ channel transcripts in atrium and ventricle, respectively. The density of INa and the rate of action potential upstroke in zebrafish ventricular myocytes at 28 °C were similar to those of human ventricles at the comparable temperature. Na+ channel isoforms and the main electrophysiological characteristics of the INa are largely similar in zebrafish and human hearts indicating evolutionary conservation of Na+ channel composition and function. The zebrafish INa differs from the human cardiac INa in terms of higher tetrodotoxin sensitivity (IC50-value = 5.3 ± 0.1 nM) and slower inactivation kinetics. The zebrafish INa was inhibited with tricaine (MS-222) with an IC50-value of 1.2 ± 0.18 mM (336 mg l-1), suggesting some care in the use of MS-222 as an anesthetic.
Collapse
Affiliation(s)
- Jaakko Haverinen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Finland
| | - Minna Hassinen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Finland
| | - Hanna Korajoki
- Department of Environmental and Biological Sciences, University of Eastern Finland, Finland
| | - Matti Vornanen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Finland.
| |
Collapse
|
65
|
Abstract
INTRODUCTION AND OBJECTIVES The importance of sodium channels for the normal electrical activity of the heart is emphasized by the fact that mutations (inherited or de novo) in genes that encode for these channels or their associated proteins cause arrhythmogenic syndromes such as the Brugada syndrome and the long QT syndrome (LQTS). The aim of this study is to conduct a review of the literature on the mutations in the sodium channel complex responsible for heart disease and the implications of a close relationship between genetics and the clinical aspects of the main cardiac channelopathies, namely at the level of diagnosis, risk stratification, prognosis, screening of family members and treatment. METHODS The online Pubmed® database was used to search for articles published in this field in indexed journals. The MeSH database was used to define the following query: "Mutation [Mesh] AND Sodium Channels [Mesh] AND Heart Diseases [Mesh]", and articles published in the last 15 years, written in English or Portuguese and referring to research in human beings were included. CONCLUSIONS In the past few years, significant advances have been made to clarify the genetic and molecular basis of these syndromes. A greater understanding of the underlying pathophysiological mechanisms showed the importance of the relationship between genotype and phenotype and led to progress in the clinical approach to these patients. However, it is still necessary to improve diagnostic capacity, optimize risk stratification, and develop new specific treatments according to the genotype-phenotype binomial.
Collapse
|
66
|
Sinus Bradycardia in Carriers of the SCN5A-1795insD Mutation: Unraveling the Mechanism through Computer Simulations. Int J Mol Sci 2018; 19:ijms19020634. [PMID: 29473904 PMCID: PMC5855856 DOI: 10.3390/ijms19020634] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/13/2018] [Accepted: 02/19/2018] [Indexed: 11/25/2022] Open
Abstract
The SCN5A gene encodes the pore-forming α-subunit of the ion channel that carries the cardiac fast sodium current (INa). The 1795insD mutation in SCN5A causes sinus bradycardia, with a mean heart rate of 70 beats/min in mutation carriers vs. 77 beats/min in non-carriers from the same family (lowest heart rate 41 vs. 47 beats/min). To unravel the underlying mechanism, we incorporated the mutation-induced changes in INa into a recently developed comprehensive computational model of a single human sinoatrial node cell (Fabbri–Severi model). The 1795insD mutation reduced the beating rate of the model cell from 74 to 69 beats/min (from 49 to 43 beats/min in the simulated presence of 20 nmol/L acetylcholine). The mutation-induced persistent INa per se resulted in a substantial increase in beating rate. This gain-of-function effect was almost completely counteracted by the loss-of-function effect of the reduction in INa conductance. The further loss-of-function effect of the shifts in steady-state activation and inactivation resulted in an overall loss-of-function effect of the 1795insD mutation. We conclude that the experimentally identified mutation-induced changes in INa can explain the clinically observed sinus bradycardia. Furthermore, we conclude that the Fabbri–Severi model may prove a useful tool in understanding cardiac pacemaker activity in humans.
Collapse
|
67
|
Fonseca DJ, Vaz da Silva MJ. Cardiac channelopathies: The role of sodium channel mutations. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2018. [DOI: 10.1016/j.repce.2017.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
68
|
Jackman CP, Ganapathi AM, Asfour H, Qian Y, Allen BW, Li Y, Bursac N. Engineered cardiac tissue patch maintains structural and electrical properties after epicardial implantation. Biomaterials 2018; 159:48-58. [PMID: 29309993 DOI: 10.1016/j.biomaterials.2018.01.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/17/2017] [Accepted: 01/01/2018] [Indexed: 12/22/2022]
Abstract
Functional cardiac tissue engineering holds promise as a candidate therapy for myocardial infarction and heart failure. Generation of "strong-contracting and fast-conducting" cardiac tissue patches capable of electromechanical coupling with host myocardium could allow efficient improvement of heart function without increased arrhythmogenic risks. Towards that goal, we engineered highly functional 1 cm × 1 cm cardiac tissue patches made of neonatal rat ventricular cells which after 2 weeks of culture exhibited force of contraction of 18.0 ± 1.4 mN, conduction velocity (CV) of 32.3 ± 1.8 cm/s, and sustained chronic activation when paced at rates as high as 8.7 ± 0.8 Hz. Patches transduced with genetically-encoded calcium indicator (GCaMP6) were implanted onto adult rat ventricles and after 4-6 weeks assessed for action potential conduction and electrical integration by two-camera optical mapping of GCaMP6-reported Ca2+ transients in the patch and RH237-reported action potentials in the recipient heart. Of the 13 implanted patches, 11 (85%) engrafted, maintained structural integrity, and conducted action potentials with average CVs and Ca2+ transient durations comparable to those before implantation. Despite preserved graft electrical properties, no anterograde or retrograde conduction could be induced between the patch and host cardiomyocytes, indicating lack of electrical integration. Electrical properties of the underlying myocardium were not changed by the engrafted patch. From immunostaining analyses, implanted patches were highly vascularized and expressed abundant electromechanical junctions, but remained separated from the epicardium by a non-myocyte layer. In summary, our studies demonstrate generation of highly functional cardiac tissue patches that can robustly engraft on the epicardial surface, vascularize, and maintain electrical function, but do not couple with host tissue. The lack of graft-host electrical integration is therefore a critical obstacle to development of efficient tissue engineering therapies for heart repair.
Collapse
Affiliation(s)
| | - Asvin M Ganapathi
- Duke University Medical Center, Department of General Surgery, Durham, NC, USA
| | - Huda Asfour
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Ying Qian
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Brian W Allen
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Yanzhen Li
- Duke University, Department of Biomedical Engineering, Durham, NC, USA
| | - Nenad Bursac
- Duke University, Department of Biomedical Engineering, Durham, NC, USA.
| |
Collapse
|
69
|
Abstract
Voltage-gated sodium channels are protein complexes comprised of one pore forming α subunit and two, non-pore forming, β subunits. The voltage-gated sodium channel β subunits were originally identified to function as auxiliary subunits, which modulate the gating, kinetics, and localization of the ion channel pore. Since that time, the five β subunits have been shown to play crucial roles as multifunctional signaling molecules involved in cell adhesion, cell migration, neuronal pathfinding, fasciculation, and neurite outgrowth. Here, we provide an overview of the evidence implicating the β subunits in their conducting and non-conducting roles. Mutations in the β subunit genes (SCN1B-SCN4B) have been linked to a variety of diseases. These include cancer, epilepsy, cardiac arrhythmias, sudden infant death syndrome/sudden unexpected death in epilepsy, neuropathic pain, and multiple neurodegenerative disorders. β subunits thus provide novel therapeutic targets for future drug discovery.
Collapse
Affiliation(s)
- Alexandra A Bouza
- Department of Pharmacology, University of Michigan Medical School, 2200 MSRBIII, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-5632, USA
| | - Lori L Isom
- Department of Pharmacology, University of Michigan Medical School, 2301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-5632, USA.
| |
Collapse
|
70
|
Burel S, Coyan FC, Lorenzini M, Meyer MR, Lichti CF, Brown JH, Loussouarn G, Charpentier F, Nerbonne JM, Townsend RR, Maier LS, Marionneau C. C-terminal phosphorylation of Na V1.5 impairs FGF13-dependent regulation of channel inactivation. J Biol Chem 2017; 292:17431-17448. [PMID: 28882890 PMCID: PMC5655519 DOI: 10.1074/jbc.m117.787788] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 08/23/2017] [Indexed: 11/06/2022] Open
Abstract
Voltage-gated Na+ (NaV) channels are key regulators of myocardial excitability, and Ca2+/calmodulin-dependent protein kinase II (CaMKII)-dependent alterations in NaV1.5 channel inactivation are emerging as a critical determinant of arrhythmias in heart failure. However, the global native phosphorylation pattern of NaV1.5 subunits associated with these arrhythmogenic disorders and the associated channel regulatory defects remain unknown. Here, we undertook phosphoproteomic analyses to identify and quantify in situ the phosphorylation sites in the NaV1.5 proteins purified from adult WT and failing CaMKIIδc-overexpressing (CaMKIIδc-Tg) mouse ventricles. Of 19 native NaV1.5 phosphorylation sites identified, two C-terminal phosphoserines at positions 1938 and 1989 showed increased phosphorylation in the CaMKIIδc-Tg compared with the WT ventricles. We then tested the hypothesis that phosphorylation at these two sites impairs fibroblast growth factor 13 (FGF13)-dependent regulation of NaV1.5 channel inactivation. Whole-cell voltage-clamp analyses in HEK293 cells demonstrated that FGF13 increases NaV1.5 channel availability and decreases late Na+ current, two effects that were abrogated with NaV1.5 mutants mimicking phosphorylation at both sites. Additional co-immunoprecipitation experiments revealed that FGF13 potentiates the binding of calmodulin to NaV1.5 and that phosphomimetic mutations at both sites decrease the interaction of FGF13 and, consequently, of calmodulin with NaV1.5. Together, we have identified two novel native phosphorylation sites in the C terminus of NaV1.5 that impair FGF13-dependent regulation of channel inactivation and may contribute to CaMKIIδc-dependent arrhythmogenic disorders in failing hearts.
Collapse
Affiliation(s)
- Sophie Burel
- From the l'Institut du Thorax, INSERM, CNRS, UNIV Nantes, Nantes 44007, France
| | - Fabien C Coyan
- From the l'Institut du Thorax, INSERM, CNRS, UNIV Nantes, Nantes 44007, France
| | - Maxime Lorenzini
- From the l'Institut du Thorax, INSERM, CNRS, UNIV Nantes, Nantes 44007, France
| | | | - Cheryl F Lichti
- the Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Joan H Brown
- the Department of Pharmacology, University of California at San Diego, La Jolla, California 92093-0636, and
| | - Gildas Loussouarn
- From the l'Institut du Thorax, INSERM, CNRS, UNIV Nantes, Nantes 44007, France
| | - Flavien Charpentier
- From the l'Institut du Thorax, INSERM, CNRS, UNIV Nantes, Nantes 44007, France
| | | | - R Reid Townsend
- Internal Medicine, and
- Cell Biology and Physiology, Washington University Medical School, St. Louis, Missouri 63110
| | - Lars S Maier
- the Department of Internal Medicine II, University Heart Center, University Hospital Regensburg, D-93042 Regensburg, Germany
| | - Céline Marionneau
- From the l'Institut du Thorax, INSERM, CNRS, UNIV Nantes, Nantes 44007, France,
| |
Collapse
|
71
|
Abnormal sodium channel mRNA splicing in hypertrophic cardiomyopathy. Int J Cardiol 2017; 249:282-286. [PMID: 28916354 DOI: 10.1016/j.ijcard.2017.08.071] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 08/03/2017] [Accepted: 08/29/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND Our previous studies showed that in ischemic and nonischemic heart failure (HF), the voltage-gated cardiac Na+ channel α subunit (SCN5A) mRNA is abnormally spliced to produce two truncated transcript variants (E28C and D) that activate the unfolded protein response (UPR). We tested whether SCN5A post-transcriptional regulation was abnormal in hypertrophic cardiomyopathy (HCM). MATERIAL AND METHODS Human heart tissue was obtained from HCM patients. The changes in relative abundances of SCN5A, its variants, splicing factors RBM25 and LUC7A, and PERK, a major effector of the UPR, were analyzed by real time RT-PCR and the expression changes were confirmed by Western Blot. RESULTS We found reduced full-length transcript, increased SCN5A truncation variants and activation of UPR in HCM when compared to control hearts. In these patients, real time RT-PCR revealed that HCM patients had decreased SCN5A mRNA to 27.8±4.07% of control (P<0.01) and an increased abundance of E28C and E28D (3.4±0.3 and 2.8±0.3-fold, respectively, P<0.05). PERK mRNA increased 8.2±3.1 fold (P<0.01) in HCM patients. Western blot confirmed a significant increase of PERK. CONCLUSIONS These data suggested that the full-length SCN5A was reduced in patients with HCM. This reduction was accompanied by abnormal SCN5A pre-mRNA splicing and UPR activation. These changes may contribute to the arrhythmic risk in HCM.
Collapse
|
72
|
Vanninen SUM, Nikus K, Aalto-Setälä K. Electrocardiogram changes and atrial arrhythmias in individuals carrying sodium channel SCN5A D1275N mutation. Ann Med 2017; 49:496-503. [PMID: 28294644 DOI: 10.1080/07853890.2017.1307515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION The cardiac sodium channel SCN5A regulates atrioventricular and ventricular depolarization as well as cardiac conduction. Patients with cardiac electrical abnormalities have an increased risk of sudden cardiac death (SCD) and cardio-embolic stroke. Optimal management of cardiac disease includes the understanding of association between the causative mutations and the clinical phenotype. A 12-lead electrocardiogram (ECG) is an easy and inexpensive tool for finding risk patients. MATERIALS AND METHODS A blood sample for DNA extraction was obtained in a Finnish family with 43 members; systematic 12-lead ECG analysis was performed in 13 of the family members carrying an SCN5A D1275N mutation. Conduction defects and supraventricular arrhythmias, including atrial fibrillation/flutter, atrioventricular nodal re-entry tachycardia (AVNRT) and junctional rhythm were searched for. RESULTS Five (38%) mutation carriers had fascicular or bundle branch block, 10 had atrial arrhythmias; no ventricular arrhythmias were found. Notching of the R- and S waves - including initial QRS fragmentation - and prolonged S-wave upstroke were present in all the affected family members. Notably, four (31%) affected family members had a stroke before the age of 31 and two experienced premature death. CONCLUSIONS A 12-lead ECG can be used to predict arrhythmias in SCN5A D1275N mutation carriers. Key messages The 12-lead ECG may reveal cardiac abnormalities even before clinical symptoms occur. Specific ECG findings - initial QRS fragmentation, prolonged S-wave upstroke as well as supraventricular arrhythmias - were frequently encountered in all SCN5A D1257N mutation carriers. ECG follow-up is recommended for all SCN5A D1275N mutation carriers.
Collapse
Affiliation(s)
| | - Kjell Nikus
- a Heart Center, Tampere University Hospital , Tampere , Finland.,b Faculty of Medicine and Life Sciences , University of Tampere , Tampere , Finland
| | - Katriina Aalto-Setälä
- a Heart Center, Tampere University Hospital , Tampere , Finland.,b Faculty of Medicine and Life Sciences , University of Tampere , Tampere , Finland.,c BioMediTech, University of Tampere , Tampere , Finland
| |
Collapse
|
73
|
Cardoso FC, Dekan Z, Smith JJ, Deuis JR, Vetter I, Herzig V, Alewood PF, King GF, Lewis RJ. Modulatory features of the novel spider toxin μ-TRTX-Df1a isolated from the venom of the spider Davus fasciatus. Br J Pharmacol 2017; 174:2528-2544. [PMID: 28542706 DOI: 10.1111/bph.13865] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Naturally occurring dysfunction of voltage-gated sodium (NaV ) channels results in complex disorders such as chronic pain, making these channels an attractive target for new therapies. In the pursuit of novel NaV modulators, we investigated spider venoms for new inhibitors of NaV channels. EXPERIMENTAL APPROACH We used high-throughput screens to identify a NaV modulator in venom of the spider Davus fasciatus. Further characterization of this venom peptide was undertaken using fluorescent and electrophysiological assays, molecular modelling and a rodent pain model. KEY RESULTS We identified a potent NaV inhibitor named μ-TRTX-Df1a. This 34-residue peptide fully inhibited responses mediated by NaV 1.7 endogenously expressed in SH-SY5Y cells. Df1a also inhibited voltage-gated calcium (CaV 3) currents but had no activity against the voltage-gated potassium (KV 2) channel. The modelled structure of Df1a, which contains an inhibitor cystine knot motif, is reminiscent of the NaV channel toxin ProTx-I. Electrophysiology revealed that Df1a inhibits all NaV subtypes tested (hNaV 1.1-1.7). Df1a also slowed fast inactivation of NaV 1.1, NaV 1.3 and NaV 1.5 and modified the voltage-dependence of activation and inactivation of most of the NaV subtypes. Df1a preferentially binds to the domain II voltage-sensor and has additional interactions with the voltage sensors domains III and IV, which probably explains its modulatory features. Df1a was analgesic in vivo, reversing the spontaneous pain behaviours induced by the NaV activator OD1. CONCLUSION AND IMPLICATIONS μ-TRTX-Df1a shows potential as a new molecule for the development of drugs to treat pain disorders mediated by voltage-gated ion channels.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Zoltan Dekan
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Jennifer J Smith
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Jennifer R Deuis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia.,School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia.,School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Volker Herzig
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Paul F Alewood
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
74
|
Chadda KR, Jeevaratnam K, Lei M, Huang CLH. Sodium channel biophysics, late sodium current and genetic arrhythmic syndromes. Pflugers Arch 2017; 469:629-641. [PMID: 28265756 PMCID: PMC5438422 DOI: 10.1007/s00424-017-1959-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 02/14/2017] [Indexed: 12/11/2022]
Abstract
Arrhythmias arise from breakdown of orderly action potential (AP) activation, propagation and recovery driven by interactive opening and closing of successive voltage-gated ion channels, in which one or more Na+ current components play critical parts. Early peak, Na+ currents (INa) reflecting channel activation drive the AP upstroke central to cellular activation and its propagation. Sustained late Na+ currents (INa-L) include contributions from a component with a delayed inactivation timecourse influencing AP duration (APD) and refractoriness, potentially causing pro-arrhythmic phenotypes. The magnitude of INa-L can be analysed through overlaps or otherwise in the overall voltage dependences of the steady-state properties and kinetics of activation and inactivation of the Na+ conductance. This was useful in analysing repetitive firing associated with paramyotonia congenita in skeletal muscle. Similarly, genetic cardiac Na+ channel abnormalities increasing INa-L are implicated in triggering phenomena of automaticity, early and delayed afterdepolarisations and arrhythmic substrate. This review illustrates a wide range of situations that may accentuate INa-L. These include (1) overlaps between steady-state activation and inactivation increasing window current, (2) kinetic deficiencies in Na+ channel inactivation leading to bursting phenomena associated with repetitive channel openings and (3) non-equilibrium gating processes causing channel re-opening due to more rapid recoveries from inactivation. All these biophysical possibilities were identified in a selection of abnormal human SCN5A genotypes. The latter presented as a broad range of clinical arrhythmic phenotypes, for which effective therapeutic intervention would require specific identification and targeting of the diverse electrophysiological abnormalities underlying their increased INa-L.
Collapse
Affiliation(s)
- Karan R Chadda
- Faculty of Health and Medical Sciences, University of Surrey, VSM Building, Guildford, GU2 7AL, UK
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, VSM Building, Guildford, GU2 7AL, UK
- School of Medicine, Perdana University-Royal College of Surgeons Ireland, 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK.
- Department of Biochemistry, University of Cambridge, Hopkins Building, Cambridge, CB2 1QW, UK.
| |
Collapse
|
75
|
Avula UMR, Noonavath M, Wan E. Gender Differences in Atrial Fibrillation. GENDER AND THE GENOME 2017. [DOI: 10.1089/gg.2016.0002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Uma Mahesh R. Avula
- Department of Medicine, Division of Cardiology, Columbia University, New York, New York
| | - Meghana Noonavath
- Department of Medicine, Division of Cardiology, Columbia University, New York, New York
| | - Elaine Wan
- Department of Medicine, Division of Cardiology, Columbia University, New York, New York
| |
Collapse
|
76
|
Avula U, Noonavath M, Wan E. Review Article: Gender Differences in Atrial Fibrillation. GENDER AND THE GENOME 2017. [DOI: 10.1177/247028971700100101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
77
|
Du D, Yang H, Ednie AR, Bennett ES. In-Silico Modeling of the Functional Role of Reduced Sialylation in Sodium and Potassium Channel Gating of Mouse Ventricular Myocytes. IEEE J Biomed Health Inform 2017; 22:631-639. [PMID: 28182562 DOI: 10.1109/jbhi.2017.2664579] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cardiac ion channels are highly glycosylated membrane proteins with up to 30% of the protein's mass containing glycans. Heart diseases often accompany individuals with congenital disorders of glycosylation (CDG). However, cardiac dysfunction among CDG patients is not yet fully understood. There is an urgent need to study how aberrant glycosylation impacts cardiac electrical signaling. Our previous works reported that congenitally reduced sialylation achieved through deletion of the sialyltransferase gene, ST3Gal4, leads to altered gating of voltage-gated Na+ and K+ channels ( and , respectively). However, linking the impact of reduced sialylation on ion channel gating to the action potential (AP) is difficult without performing computer experiments. Also, decomposing the sum of K+ currents is difficult because of complex structures and components of channels (e.g., , and ). In this study, we developed in-silico models to describe the functional role of reduced sialylation in both and gating and the AP using in vitro experimental data. Modeling results showed that reduced sialylation changes gating as follows: 1) The steady-state activation voltages of isoforms are shifted to a more depolarized potential. 2) Aberrant K+ currents ( and ) contribute to a prolonged AP duration, and altered Na+ current ( ) contributes to a shortened AP refractory period. This study contributes to a better understanding of the functional role of reduced sialylation in cardiac dysfunction that shows strong potential to provide new pharmaceutical targets for the treatment of CDG-related heart diseases.
Collapse
|
78
|
Ledwitch KV, Roberts AG. Cardiovascular Ion Channel Inhibitor Drug-Drug Interactions with P-glycoprotein. AAPS JOURNAL 2016; 19:409-420. [PMID: 28028729 DOI: 10.1208/s12248-016-0023-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/19/2016] [Indexed: 12/31/2022]
Abstract
P-glycoprotein (Pgp) is an ATP-binding cassette (ABC) transporter that plays a major role in cardiovascular drug disposition by effluxing a chemically and structurally diverse range of cardiovascular therapeutics. Unfortunately, drug-drug interactions (DDIs) with the transporter have become a major roadblock to effective cardiovascular drug administration because they can cause adverse drug reactions (ADRs) or reduce the efficacy of drugs. Cardiovascular ion channel inhibitors are particularly susceptible to DDIs and ADRs with Pgp because they often have low therapeutic indexes and are commonly coadministered with other drugs that are also Pgp substrates. DDIs from cardiovascular ion channel inhibitors with the transporter occur because of inhibition or induction of the transporter and the transporter's tissue and cellular localization. Inhibiting Pgp can increase absorption and reduce excretion of drugs, leading to elevated drug plasma concentrations and drug toxicity. In contrast, inducing Pgp can have the opposite effect by reducing the drug plasma concentration and its efficacy. A number of in vitro and in vivo studies have already demonstrated DDIs from several cardiovascular ion channel inhibitors with human Pgp and its animal analogs, including verapamil, digoxin, and amiodarone. In this review, Pgp-mediated DDIs and their effects on pharmacokinetics for different categories of cardiovascular ion channel inhibitors are discussed. This information is essential for improving pharmacokinetic predictions of cardiovascular therapeutics, for safer cardiovascular drug administration and for mitigating ADRs emanating from Pgp.
Collapse
Affiliation(s)
- Kaitlyn V Ledwitch
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W. Green St., Athens, Georgia, 30602, USA
| | - Arthur G Roberts
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W. Green St., Athens, Georgia, 30602, USA.
| |
Collapse
|
79
|
Xu Q, Patel D, Zhang X, Veenstra RD. Changes in cardiac Nav1.5 expression, function, and acetylation by pan-histone deacetylase inhibitors. Am J Physiol Heart Circ Physiol 2016; 311:H1139-H1149. [PMID: 27638876 DOI: 10.1152/ajpheart.00156.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 08/24/2016] [Indexed: 12/19/2022]
Abstract
Histone deacetylase (HDAC) inhibitors are small molecule anticancer therapeutics that exhibit limiting cardiotoxicities including QT interval prolongation and life-threatening cardiac arrhythmias. Because the molecular mechanisms for HDAC inhibitor-induced cardiotoxicity are poorly understood, we performed whole cell patch voltage-clamp experiments to measure cardiac sodium currents (INa) from wild-type neonatal mouse ventricular or human-induced pluripotent stem cell-derived cardiomyocytes treated with trichostatin A (TSA), vorinostat (VOR), or romidepsin (FK228). All three pan-HDAC inhibitors dose dependently decreased peak INa density and shifted the voltage activation curve 3- to 8-mV positive. Increases in late INa were not observed despite a moderate slowing of the inactivation rate at low activating potentials (<-40 mV). Scn5a mRNA levels were not significantly altered but NaV1.5 protein levels were significantly reduced. Immunoprecipitation with anti-NaV1.5 and Western blotting with anti-acetyl-lysine antibodies indicated that NaV1.5 acetylation is increased in vivo after HDAC inhibition. FK228 inhibited total cardiac HDAC activity with two apparent IC50s of 5 nM and 1.75 μM, consistent with previous findings with TSA and VOR. FK228 also decreased ventricular gap junction conductance (gj), again consistent with previous findings. We conclude that pan-HDAC inhibition reduces cardiac INa density and NaV1.5 protein levels without affecting late INa amplitude and, thus, probably does not contribute to the reported QT interval prolongation and arrhythmias associated with pan-HDAC inhibitor therapies. Conversely, reductions in gj may enhance the occurrence of triggered activity by limiting electrotonic inhibition and, combined with reduced INa, slow myocardial conduction and increase vulnerability to reentrant arrhythmias.
Collapse
Affiliation(s)
- Qin Xu
- Department of Pharmacology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York; and
| | - Dakshesh Patel
- Department of Pharmacology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York; and
| | - Xian Zhang
- Department of Pharmacology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York; and
| | - Richard D Veenstra
- Department of Pharmacology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York; and .,Department of Cell and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York
| |
Collapse
|
80
|
Zhang X, Guo L, Zeng H, White SL, Furniss M, Balasubramanian B, Lis E, Lagrutta A, Sannajust F, Zhao LL, Xi B, Wang X, Davis M, Abassi YA. Multi-parametric assessment of cardiomyocyte excitation-contraction coupling using impedance and field potential recording: A tool for cardiac safety assessment. J Pharmacol Toxicol Methods 2016; 81:201-16. [DOI: 10.1016/j.vascn.2016.06.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/29/2016] [Accepted: 06/04/2016] [Indexed: 11/17/2022]
|
81
|
Winters JJ, Isom LL. Developmental and Regulatory Functions of Na(+) Channel Non-pore-forming β Subunits. CURRENT TOPICS IN MEMBRANES 2016; 78:315-51. [PMID: 27586289 DOI: 10.1016/bs.ctm.2016.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Voltage-gated Na(+) channels (VGSCs) isolated from mammalian neurons are heterotrimeric complexes containing one pore-forming α subunit and two non-pore-forming β subunits. In excitable cells, VGSCs are responsible for the initiation of action potentials. VGSC β subunits are type I topology glycoproteins, containing an extracellular amino-terminal immunoglobulin (Ig) domain with homology to many neural cell adhesion molecules (CAMs), a single transmembrane segment, and an intracellular carboxyl-terminal domain. VGSC β subunits are encoded by a gene family that is distinct from the α subunits. While α subunits are expressed in prokaryotes, β subunit orthologs did not arise until after the emergence of vertebrates. β subunits regulate the cell surface expression, subcellular localization, and gating properties of their associated α subunits. In addition, like many other Ig-CAMs, β subunits are involved in cell migration, neurite outgrowth, and axon pathfinding and may function in these roles in the absence of associated α subunits. In sum, these multifunctional proteins are critical for both channel regulation and central nervous system development.
Collapse
Affiliation(s)
- J J Winters
- University of Michigan Neuroscience Program, Ann Arbor, MI, United States
| | - L L Isom
- University of Michigan Neuroscience Program, Ann Arbor, MI, United States; University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
82
|
Wang N, Huo R, Cai B, Lu Y, Ye B, Li X, Li F, Xu H. Activation of Wnt/β-catenin signaling by hydrogen peroxide transcriptionally inhibits NaV1.5 expression. Free Radic Biol Med 2016; 96:34-44. [PMID: 27068063 PMCID: PMC4912406 DOI: 10.1016/j.freeradbiomed.2016.04.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 04/03/2016] [Accepted: 04/07/2016] [Indexed: 10/22/2022]
Abstract
Oxidants and canonical Wnt/β-catenin signaling have been shown to decrease cardiac Na(+) channel activity by suppressing NaV1.5 expression. Our aims are to determine if hydrogen peroxide (H2O2), one oxidant of reactive oxygen species (ROS), activates Wnt/β-catenin signaling and promotes β-catenin nuclear activity, leading to suppression of NaV1.5 expression and if this suppression requires the interaction of β-catenin with its nuclear partner, TCF4 (also called TCF7L2) to decrease SCN5a promoter activity. The results demonstrated that H2O2 increased β-catenin, but not TCF4 nuclear localization determined by immunofluorescence without affecting total β-catenin protein level. Furthermore, H2O2 exerted a dose- and time-dependent suppressive effect on NaV1.5 expression. RT-PCR and/or Western blot analyses revealed that overexpressing active form of β-catenin or stabilizing β-catenin by GSK-3β inhibitors, LiCl and Bio, suppressed NaV1.5 expression in HL-1 cells. In contrast, destabilization of β-catenin by a constitutively active GSK-3β mutant (S9A) upregulated NaV1.5 expression. Whole-cell recording showed that LiCl significantly inhibited Na(+) channel activity in these cells. Using immunoprecipitation (IP), we showed that β-catenin interacted with TCF4 indicating that β-catenin as a co-transfactor, regulates NaV1.5 expression through TCF4. Analyses of the SCN5a promoter sequences among different species by using VISTA tools indicated that SCN5a promoter harbors TCF4 binding sites. Chromatin IP assays demonstrated that both β-catenin and TCF4 were recruited in the SCN5a promoter, and regulated its activity. Luciferase promoter assays exhibited that β-catenin inhibited the SCN5a promoter activity at a dose-dependent manner and this inhibition required TCF4. Small interfering (Si) RNA targeting β-catenin significantly increased SCN5a promoter activity, leading to enhanced NaV1.5 expression. As expected, β-catenin SiRNA prevents H2O2 suppressive effects on both SCN5a promoter activity and NaV1.5 expression. Our findings indicate that H2O2 inhibits NaV1.5 expression by activating the Wnt/β-catenin signaling and β-catenin interacts with TCF4 to transcriptionally suppress cardiac NaV1.5 expression.
Collapse
Affiliation(s)
- Ning Wang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Corresponding author at: Haodong Xu, MD, PhD, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave., CHS 13-145E Los Angeles, CA 90095-1732 USA. Tel.: 310-206-3582; fax: 310-267-2658.
| | - Rong Huo
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Corresponding author at: Haodong Xu, MD, PhD, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave., CHS 13-145E Los Angeles, CA 90095-1732 USA. Tel.: 310-206-3582; fax: 310-267-2658.
| | - Benzhi Cai
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Corresponding author at: Haodong Xu, MD, PhD, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave., CHS 13-145E Los Angeles, CA 90095-1732 USA. Tel.: 310-206-3582; fax: 310-267-2658.
| | - Yan Lu
- Division of Cardiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Bo Ye
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Xiang Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Faqian Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Haodong Xu
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Corresponding author at: Haodong Xu, MD, PhD, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave., CHS 13-145E Los Angeles, CA 90095-1732 USA. Tel.: 310-206-3582; fax: 310-267-2658.
| |
Collapse
|
83
|
Liu M, Yang KC, Dudley SC. Cardiac Sodium Channel Mutations: Why so Many Phenotypes? CURRENT TOPICS IN MEMBRANES 2016; 78:513-59. [PMID: 27586294 DOI: 10.1016/bs.ctm.2015.12.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cardiac Na(+) channel (Nav1.5) conducts a depolarizing inward Na(+) current that is responsible for the generation of the upstroke Phase 0 of the action potential. In heart tissue, changes in Na(+) currents can affect conduction velocity and impulse propagation. The cardiac Nav1.5 is also involved in determination of the action potential duration, since some channels may reopen during the plateau phase, generating a persistent or late inward current. Mutations of cardiac Nav1.5 can induce gain or loss of channel function because of an increased late current or a decrease of peak current, respectively. Gain-of-function mutations cause Long QT syndrome type 3 and possibly atrial fibrillation, while loss-of-function channel mutations are associated with a wider variety of phenotypes, such as Brugada syndrome, cardiac conduction disease, dilated cardiomyopathy, and sick sinus node syndrome. The penetrance and phenotypes resulting from Nav1.5 mutations also vary with age, gender, body temperature, circadian rhythm, and between regions of the heart. This phenotypic variability makes it difficult to correlate genotype-phenotype. We propose that mutations are only one contributor to the phenotype and additional modifications on Nav1.5 lead to the phenotypic variability. Possible modifiers include other genetic variations and alterations in the life cycle of Nav1.5 such as gene transcription, RNA processing, translation, posttranslational modifications, trafficking, complex assembly, and degradation. In this chapter, we summarize potential modifiers of cardiac Nav1.5 that could help explain the clinically observed phenotypic variability. Consideration of these modifiers could help improve genotype-phenotype correlations and lead to new therapeutic strategies.
Collapse
Affiliation(s)
- M Liu
- The Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - K-C Yang
- The Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - S C Dudley
- The Warren Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
84
|
Wan E, Abrams J, Weinberg RL, Katchman AN, Bayne J, Zakharov SI, Yang L, Morrow JP, Garan H, Marx SO. Aberrant sodium influx causes cardiomyopathy and atrial fibrillation in mice. J Clin Invest 2015; 126:112-22. [PMID: 26595809 DOI: 10.1172/jci84669] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/15/2015] [Indexed: 01/17/2023] Open
Abstract
Increased sodium influx via incomplete inactivation of the major cardiac sodium channel Na(V)1.5 is correlated with an increased incidence of atrial fibrillation (AF) in humans. Here, we sought to determine whether increased sodium entry is sufficient to cause the structural and electrophysiological perturbations that are required to initiate and sustain AF. We used mice expressing a human Na(V)1.5 variant with a mutation in the anesthetic-binding site (F1759A-Na(V)1.5) and demonstrated that incomplete Na+ channel inactivation is sufficient to drive structural alterations, including atrial and ventricular enlargement, myofibril disarray, fibrosis and mitochondrial injury, and electrophysiological dysfunctions that together lead to spontaneous and prolonged episodes of AF in these mice. Using this model, we determined that the increase in a persistent sodium current causes heterogeneously prolonged action potential duration and rotors, as well as wave and wavelets in the atria, and thereby mimics mechanistic theories that have been proposed for AF in humans. Acute inhibition of the sodium-calcium exchanger, which targets the downstream effects of enhanced sodium entry, markedly reduced the burden of AF and ventricular arrhythmias in this model, suggesting a potential therapeutic approach for AF. Together, our results indicate that these mice will be important for assessing the cellular mechanisms and potential effectiveness of antiarrhythmic therapies.
Collapse
|
85
|
Chaudhari U, Nemade H, Wagh V, Gaspar JA, Ellis JK, Srinivasan SP, Spitkovski D, Nguemo F, Louisse J, Bremer S, Hescheler J, Keun HC, Hengstler JG, Sachinidis A. Identification of genomic biomarkers for anthracycline-induced cardiotoxicity in human iPSC-derived cardiomyocytes: an in vitro repeated exposure toxicity approach for safety assessment. Arch Toxicol 2015; 90:2763-2777. [PMID: 26537877 PMCID: PMC5065579 DOI: 10.1007/s00204-015-1623-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 10/20/2015] [Indexed: 11/25/2022]
Abstract
The currently available techniques for the safety evaluation of candidate drugs are usually cost-intensive and time-consuming and are often insufficient to predict human relevant cardiotoxicity. The purpose of this study was to develop an in vitro repeated exposure toxicity methodology allowing the identification of predictive genomics biomarkers of functional relevance for drug-induced cardiotoxicity in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). The hiPSC-CMs were incubated with 156 nM doxorubicin, which is a well-characterized cardiotoxicant, for 2 or 6 days followed by washout of the test compound and further incubation in compound-free culture medium until day 14 after the onset of exposure. An xCELLigence Real-Time Cell Analyser was used to monitor doxorubicin-induced cytotoxicity while also monitoring functional alterations of cardiomyocytes by counting of the beating frequency of cardiomyocytes. Unlike single exposure, repeated doxorubicin exposure resulted in long-term arrhythmic beating in hiPSC-CMs accompanied by significant cytotoxicity. Global gene expression changes were studied using microarrays and bioinformatics tools. Analysis of the transcriptomic data revealed early expression signatures of genes involved in formation of sarcomeric structures, regulation of ion homeostasis and induction of apoptosis. Eighty-four significantly deregulated genes related to cardiac functions, stress and apoptosis were validated using real-time PCR. The expression of the 84 genes was further studied by real-time PCR in hiPSC-CMs incubated with daunorubicin and mitoxantrone, further anthracycline family members that are also known to induce cardiotoxicity. A panel of 35 genes was deregulated by all three anthracycline family members and can therefore be expected to predict the cardiotoxicity of compounds acting by similar mechanisms as doxorubicin, daunorubicin or mitoxantrone. The identified gene panel can be applied in the safety assessment of novel drug candidates as well as available therapeutics to identify compounds that may cause cardiotoxicity.
Collapse
Affiliation(s)
- Umesh Chaudhari
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - Harshal Nemade
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - Vilas Wagh
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - John Antonydas Gaspar
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - James K Ellis
- Biomolecular Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Sureshkumar Perumal Srinivasan
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - Dimitry Spitkovski
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - Filomain Nguemo
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - Jochem Louisse
- Institute for Health and Consumer Protection, Joint Research Centre, European Commission, Ispra, Italy
| | - Susanne Bremer
- Institute for Health and Consumer Protection, Joint Research Centre, European Commission, Ispra, Italy
| | - Jürgen Hescheler
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany
| | - Hector C Keun
- Biomolecular Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), 44139, Dortmund, Germany
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Str. 39, 50931, Cologne, NRW, Germany.
| |
Collapse
|
86
|
Cardoso FC, Dekan Z, Rosengren KJ, Erickson A, Vetter I, Deuis JR, Herzig V, Alewood PF, King GF, Lewis RJ. Identification and Characterization of ProTx-III [μ-TRTX-Tp1a], a New Voltage-Gated Sodium Channel Inhibitor from Venom of the Tarantula Thrixopelma pruriens. Mol Pharmacol 2015; 88:291-303. [PMID: 25979003 DOI: 10.1124/mol.115.098178] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/15/2015] [Indexed: 01/26/2023] Open
Abstract
Spider venoms are a rich source of ion channel modulators with therapeutic potential. Given the analgesic potential of subtype-selective inhibitors of voltage-gated sodium (NaV) channels, we screened spider venoms for inhibitors of human NaV1.7 (hNaV1.7) using a high-throughput fluorescent assay. Here, we describe the discovery of a novel NaV1.7 inhibitor, μ-TRTX-Tp1a (Tp1a), isolated from the venom of the Peruvian green-velvet tarantula Thrixopelma pruriens. Recombinant and synthetic forms of this 33-residue peptide preferentially inhibited hNaV1.7 > hNaV1.6 > hNaV1.2 > hNaV1.1 > hNaV1.3 channels in fluorescent assays. NaV1.7 inhibition was diminished (IC50 11.5 nM) and the association rate decreased for the C-terminal acid form of Tp1a compared with the native amidated form (IC50 2.1 nM), suggesting that the peptide C terminus contributes to its interaction with hNaV1.7. Tp1a had no effect on human voltage-gated calcium channels or nicotinic acetylcholine receptors at 5 μM. Unlike most spider toxins that modulate NaV channels, Tp1a inhibited hNaV1.7 without significantly altering the voltage dependence of activation or inactivation. Tp1a proved to be analgesic by reversing spontaneous pain induced in mice by intraplantar injection in OD1, a scorpion toxin that potentiates hNaV1.7. The structure of Tp1a as determined using NMR spectroscopy revealed a classic inhibitor cystine knot (ICK) motif. The molecular surface of Tp1a presents a hydrophobic patch surrounded by positively charged residues, with subtle differences from other ICK spider toxins that might contribute to its different pharmacological profile. Tp1a may help guide the development of more selective and potent hNaV1.7 inhibitors for treatment of chronic pain.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Institute for Molecular Bioscience (F.C.C., Z.D., I.V., J.R.D., V.H., P.F.A., G.F.K., R.J.L.), School of Biomedical Sciences (K.J.R.), and School of Chemistry and Molecular Biosciences (A.E.), The University of Queensland, Brisbane, Queensland, Australia
| | - Zoltan Dekan
- Institute for Molecular Bioscience (F.C.C., Z.D., I.V., J.R.D., V.H., P.F.A., G.F.K., R.J.L.), School of Biomedical Sciences (K.J.R.), and School of Chemistry and Molecular Biosciences (A.E.), The University of Queensland, Brisbane, Queensland, Australia
| | - K Johan Rosengren
- Institute for Molecular Bioscience (F.C.C., Z.D., I.V., J.R.D., V.H., P.F.A., G.F.K., R.J.L.), School of Biomedical Sciences (K.J.R.), and School of Chemistry and Molecular Biosciences (A.E.), The University of Queensland, Brisbane, Queensland, Australia
| | - Andelain Erickson
- Institute for Molecular Bioscience (F.C.C., Z.D., I.V., J.R.D., V.H., P.F.A., G.F.K., R.J.L.), School of Biomedical Sciences (K.J.R.), and School of Chemistry and Molecular Biosciences (A.E.), The University of Queensland, Brisbane, Queensland, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience (F.C.C., Z.D., I.V., J.R.D., V.H., P.F.A., G.F.K., R.J.L.), School of Biomedical Sciences (K.J.R.), and School of Chemistry and Molecular Biosciences (A.E.), The University of Queensland, Brisbane, Queensland, Australia
| | - Jennifer R Deuis
- Institute for Molecular Bioscience (F.C.C., Z.D., I.V., J.R.D., V.H., P.F.A., G.F.K., R.J.L.), School of Biomedical Sciences (K.J.R.), and School of Chemistry and Molecular Biosciences (A.E.), The University of Queensland, Brisbane, Queensland, Australia
| | - Volker Herzig
- Institute for Molecular Bioscience (F.C.C., Z.D., I.V., J.R.D., V.H., P.F.A., G.F.K., R.J.L.), School of Biomedical Sciences (K.J.R.), and School of Chemistry and Molecular Biosciences (A.E.), The University of Queensland, Brisbane, Queensland, Australia
| | - Paul F Alewood
- Institute for Molecular Bioscience (F.C.C., Z.D., I.V., J.R.D., V.H., P.F.A., G.F.K., R.J.L.), School of Biomedical Sciences (K.J.R.), and School of Chemistry and Molecular Biosciences (A.E.), The University of Queensland, Brisbane, Queensland, Australia
| | - Glenn F King
- Institute for Molecular Bioscience (F.C.C., Z.D., I.V., J.R.D., V.H., P.F.A., G.F.K., R.J.L.), School of Biomedical Sciences (K.J.R.), and School of Chemistry and Molecular Biosciences (A.E.), The University of Queensland, Brisbane, Queensland, Australia
| | - Richard J Lewis
- Institute for Molecular Bioscience (F.C.C., Z.D., I.V., J.R.D., V.H., P.F.A., G.F.K., R.J.L.), School of Biomedical Sciences (K.J.R.), and School of Chemistry and Molecular Biosciences (A.E.), The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
87
|
Zhao Y, Huang Y, Li W, Wang Z, Zhan S, Zhou M, Yao Y, Zeng Z, Hou Y, Chen Q, Tu X, Wang QK, Huang Z. Post-transcriptional regulation of cardiac sodium channel gene SCN5A expression and function by miR-192-5p. Biochim Biophys Acta Mol Basis Dis 2015. [PMID: 26209011 DOI: 10.1016/j.bbadis.2015.07.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The SCN5A gene encodes cardiac sodium channel Nav1.5 and causes lethal ventricular arrhythmias/sudden death and atrial fibrillation (AF) when mutated. MicroRNAs (miRNAs) are important post-transcriptional regulators of gene expression, and involved in the pathogenesis of many diseases. However, little is known about the regulation of SCN5A by miRNAs. Here we reveal a novel post-transcriptional regulatory mechanism for expression and function of SCN5A/Nav1.5 via miR-192-5p. Bioinformatic analysis revealed that the 3'-UTR of human and rhesus SCN5A, but not elephant, pig, rabbit, mouse, and rat SCN5A, contained a target binding site for miR-192-5p and dual luciferase reporter assays showed that the site was critical for down-regulation of human SCN5A. With Western blot assays and electrophysiological studies, we demonstrated that miR-192-5p significantly reduced expression of SCN5A and Nav1.5 as well as peak sodium current density INa generated by Nav1.5. Notably, in situ hybridization, immunohistochemistry and real-time qPCR analyses showed that miR-192-5p was up-regulated in tissue samples from AF patients, which was associated with down-regulation of SCN5A/Nav1.5. These results demonstrate an important post-transcriptional role of miR-192-5p in post-transcriptional regulation of Nav1.5, reveal a novel role of miR-192-5p in cardiac physiology and disease, and provide a new target for novel miRNA-based antiarrhythmic therapy for diseases with reduced INa.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, PR China
| | - Yuan Huang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, PR China
| | - Weihua Li
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, PR China
| | - Zhijie Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, PR China
| | - Shaopeng Zhan
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, PR China
| | - Mengchen Zhou
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, PR China
| | - Yufeng Yao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, PR China
| | - Zhipeng Zeng
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, PR China
| | - Yuxi Hou
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, PR China
| | - Qiuyun Chen
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, CCLCM, Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Xin Tu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, PR China
| | - Qing K Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, PR China; Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, CCLCM, Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA.
| | - Zhengrong Huang
- Department of Cardiology, The First Affiliated Hospital of Xiamen University, Xiamen, PR China.
| |
Collapse
|
88
|
Gintant G. Cardiac Sodium Current (Na v1.5). METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2015. [DOI: 10.1002/9783527673643.ch12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
89
|
Daimi H, Lozano-Velasco E, Haj Khelil A, Chibani JBE, Barana A, Amorós I, González de la Fuente M, Caballero R, Aranega A, Franco D. Regulation of SCN5A by microRNAs: miR-219 modulates SCN5A transcript expression and the effects of flecainide intoxication in mice. Heart Rhythm 2015; 12:1333-42. [PMID: 25701775 DOI: 10.1016/j.hrthm.2015.02.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND The human cardiac action potential in atrial and ventricular cells is initiated by a fast-activating, fast-inactivating sodium current generated by the SCN5A/Nav1.5 channel in association with its β1/SCN1B subunit. The role of Nav1.5 in the etiology of many cardiac diseases strongly suggests that proper regulation of cell biology and function of the channel is critical for normal cardiac function. Hence, numerous recent studies have focused on the regulatory mechanisms of Nav1.5 biosynthetic and degradation processes as well as its subcellular localization. OBJECTIVE The purpose of this study was to investigate the role of microRNAs in the Scn5a/Nav1.5 posttranscriptional regulation. METHODS Quantitative polymerase chain reaction, immunohistochemical and electrophysiological measurements of distinct microRNA gain-of-function experiments in cardiomyocytes for the assessment of Scn5a expression. RESULTS Functional studies of HL-1 cardiomyocytes and luciferase assays in fibroblasts demonstrate that Scn5a is directly (miR-98, miR-106, miR-200, and miR-219) and indirectly (miR-125 and miR-153) regulated by multiple microRNAs displaying distinct time-dependent profiles. Cotransfection experiments demonstrated that miR-219 and miR-200 have independent opposite effects on Scn5a expression modulation. Of all the microRNAs studied, only miR-219 increases Scn5a expression levels, leading to altered contraction rhythm of HL-1 cardiomyocytes. Electrophysiological analyses in HL-1 cells revealed that miR-219 increases the sodium current. In vivo administration of miR-219 does not alter normal cardiac rhythm, but abolishes some of the effects of flecainide intoxication in mice, particularly QRS prolongation. CONCLUSION This study demonstrates the involvement of multiple microRNAs in the regulation of Scn5a. Particularly, miR-219 increases Scn5a/Nav1.5 transcript and protein expression. Our data suggest that microRNAs, such as miR-219, constitute a promising therapeutical tool to treat sodium cardiac arrhythmias.
Collapse
Affiliation(s)
- Houria Daimi
- Department of Experimental Biology, University of Jaén, Jáen, Spain; Biochemistry and Molecular Biology Laboratory, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | | | - Amel Haj Khelil
- Biochemistry and Molecular Biology Laboratory, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Jemni B E Chibani
- Biochemistry and Molecular Biology Laboratory, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Adriana Barana
- Department of Pharmacology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Irene Amorós
- Department of Pharmacology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | | | - Ricardo Caballero
- Department of Pharmacology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Amelia Aranega
- Department of Experimental Biology, University of Jaén, Jáen, Spain
| | - Diego Franco
- Department of Experimental Biology, University of Jaén, Jáen, Spain.
| |
Collapse
|
90
|
Sialic acids attached to N- and O-glycans within the Nav1.4 D1S5-S6 linker contribute to channel gating. Biochim Biophys Acta Gen Subj 2014; 1850:307-17. [PMID: 25450184 DOI: 10.1016/j.bbagen.2014.10.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/09/2014] [Accepted: 10/23/2014] [Indexed: 01/02/2023]
Abstract
BACKGROUND Voltage-gated Na+ channels (Nav) are responsible for the initiation and conduction of neuronal and muscle action potentials. Nav gating can be altered by sialic acids attached to channel N-glycans, typically through isoform-specific electrostatic mechanisms. METHODS Using two sets of Chinese Hamster Ovary cell lines with varying abilities to glycosylate glycoproteins, we show for the first time that sialic acids attached to O-glycans and N-glycans within the Nav1.4 D1S5-S6 linker modulate Nav gating. RESULTS All measured steady-state and kinetic parameters were shifted to more depolarized potentials under conditions of essentially no sialylation. When sialylation of only N-glycans or of only O-glycans was prevented, the observed voltage-dependent parameter values were intermediate between those observed under full versus no sialylation. Immunoblot gel shift analyses support the biophysical data. CONCLUSIONS The data indicate that sialic acids attached to both N- and O-glycans residing within the Nav1.4 D1S5-S6 linker modulate channel gating through electrostatic mechanisms, with the relative contribution of sialic acids attached to N- versus O-glycans on channel gating being similar. GENERAL SIGNIFICANCE Protein N- and O-glycosylation can modulate ion channel gating simultaneously. These data also suggest that environmental, metabolic, and/or congenital changes in glycosylation that impact sugar substrate levels, could lead, potentially, to changes in Nav sialylation and gating that would modulate AP waveforms and conduction.
Collapse
|
91
|
Wilde AAM, Lieve KVV. Double trouble, stick to the basics should be the rule! Heart Rhythm 2014; 11:2105-6. [PMID: 25063691 DOI: 10.1016/j.hrthm.2014.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Indexed: 10/25/2022]
Affiliation(s)
- Arthur A M Wilde
- Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands.; Princess Al-Jawhara Albrahim Centre of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Krystien V V Lieve
- Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
92
|
Li HX, Zhou YF, Jiang B, Zhao X, Jiang TB, Li X, Yang XJ, Jiang WP. GATA-4 induces changes in electrophysiological properties of rat mesenchymal stem cells. Biochim Biophys Acta Gen Subj 2014; 1840:2060-9. [DOI: 10.1016/j.bbagen.2014.02.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 02/14/2014] [Accepted: 02/19/2014] [Indexed: 01/12/2023]
|
93
|
Gao G, Brahmanandam V, Raicu M, Gu L, Zhou L, Kasturirangan S, Shah A, Negi SI, Wood MR, Desai AA, Tatooles A, Schwartz A, Dudley SC. Enhanced risk profiling of implanted defibrillator shocks with circulating SCN5A mRNA splicing variants: a pilot trial. J Am Coll Cardiol 2014; 63:2261-9. [PMID: 24703920 DOI: 10.1016/j.jacc.2014.02.588] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/26/2014] [Accepted: 02/04/2014] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The aim of this study was to determine the association of SCN5A cardiac sodium (Na(+)) channel mRNA splice variants in white blood cells (WBCs) with risk of arrhythmias in heart failure (HF). BACKGROUND HF is associated with upregulation of two cardiac SCN5A mRNA splice variants that encode prematurely truncated, nonfunctional Na(+) channels. Because circulating WBCs demonstrate similar SCN5A splicing patterns, we hypothesized that these WBC-derived splice variants might further stratify patients with HF who are at risk for arrhythmias. METHODS Simultaneously obtained myocardial core samples and WBCs were compared for SCN5A variants C (VC) and D (VD). Circulating variant levels were compared among patients with HF, divided into three groups: HF without an implantable cardioverter-defibrillator (ICD), HF with an ICD without appropriate intervention, and HF with an ICD with appropriate intervention. RESULTS Myocardial tissue-derived SCN5A variant expression levels strongly correlated with circulating WBC samples for both VC and VD variants (r = 0.78 and 0.75, respectively). After controlling for covariates, patients with HF who had received an appropriate ICD intervention had higher expression levels of both WBC-derived SCN5A variants compared with patients with HF with ICDs who had not received appropriate ICD intervention (odds ratio, 3.25; 95% CI, 1.64-6.45; p = 0.001). Receiver operating characteristic analysis revealed that circulating SCN5A variant levels were highly associated with the risk for appropriate ICD intervention (area under the curve ≥0.97). CONCLUSIONS Circulating expression levels of SCN5A variants were strongly associated with myocardial tissue levels. Furthermore, circulating variant levels were correlative with arrhythmic risk as measured by ICD events in an HF population within 1 year. (Sodium Channel Splicing in Heart Failure Trial [SOCS-HEFT]; NCT01185587).
Collapse
Affiliation(s)
- Ge Gao
- Section of Cardiology and the Jesse Brown VAMC, University of Illinois at Chicago, Chicago, Illinois
| | - Vikram Brahmanandam
- Section of Cardiology and the Jesse Brown VAMC, University of Illinois at Chicago, Chicago, Illinois
| | - Mihai Raicu
- Section of Cardiology and the Jesse Brown VAMC, University of Illinois at Chicago, Chicago, Illinois
| | - Lianzhi Gu
- Section of Cardiology and the Jesse Brown VAMC, University of Illinois at Chicago, Chicago, Illinois
| | - Li Zhou
- Section of Cardiology and the Jesse Brown VAMC, University of Illinois at Chicago, Chicago, Illinois
| | - Srinivasan Kasturirangan
- Section of Cardiology and the Jesse Brown VAMC, University of Illinois at Chicago, Chicago, Illinois
| | - Anish Shah
- University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Smita I Negi
- The University of Texas Health Science Center, Houston, Texas
| | - Melissa R Wood
- Section of Cardiology and the Jesse Brown VAMC, University of Illinois at Chicago, Chicago, Illinois
| | - Ankit A Desai
- Section of Cardiology and the Jesse Brown VAMC, University of Illinois at Chicago, Chicago, Illinois; Institute for Personalized Respiratory Medicine and Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Antone Tatooles
- Transplant/Mechanical Assist, Advocate Christ Medical Center, Oak Lawn, Illinois
| | - Alan Schwartz
- Departments of Medical Education and Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - Samuel C Dudley
- Section of Cardiology and the Jesse Brown VAMC, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
94
|
Piceatannol facilitates conduction block and ventricular fibrillation induction in ischemia-reperfused rabbit hearts with pacing-induced heart failure. Int J Cardiol 2014; 171:250-8. [DOI: 10.1016/j.ijcard.2013.12.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/21/2013] [Accepted: 12/14/2013] [Indexed: 11/21/2022]
|
95
|
Calhoun JD, Isom LL. The role of non-pore-forming β subunits in physiology and pathophysiology of voltage-gated sodium channels. Handb Exp Pharmacol 2014; 221:51-89. [PMID: 24737232 DOI: 10.1007/978-3-642-41588-3_4] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Voltage-gated sodium channel β1 and β2 subunits were discovered as auxiliary proteins that co-purify with pore-forming α subunits in brain. The other family members, β1B, β3, and β4, were identified by homology and shown to modulate sodium current in heterologous systems. Work over the past 2 decades, however, has provided strong evidence that these proteins are not simply ancillary ion channel subunits, but are multifunctional signaling proteins in their own right, playing both conducting (channel modulatory) and nonconducting roles in cell signaling. Here, we discuss evidence that sodium channel β subunits not only regulate sodium channel function and localization but also modulate voltage-gated potassium channels. In their nonconducting roles, VGSC β subunits function as immunoglobulin superfamily cell adhesion molecules that modulate brain development by influencing cell proliferation and migration, axon outgrowth, axonal fasciculation, and neuronal pathfinding. Mutations in genes encoding β subunits are linked to paroxysmal diseases including epilepsy, cardiac arrhythmia, and sudden infant death syndrome. Finally, β subunits may be targets for the future development of novel therapeutics.
Collapse
Affiliation(s)
- Jeffrey D Calhoun
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109-5632, USA
| | | |
Collapse
|
96
|
Remme CA, Wilde AAM. Targeting sodium channels in cardiac arrhythmia. Curr Opin Pharmacol 2013; 15:53-60. [PMID: 24721654 DOI: 10.1016/j.coph.2013.11.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 11/26/2013] [Accepted: 11/28/2013] [Indexed: 12/15/2022]
Abstract
Cardiac voltage-gated sodium channels are responsible for proper electrical conduction in the heart. During acquired pathological conditions and inherited sodium channelopathies, altered sodium channel function causes conduction disturbances and ventricular arrhythmias. Although the clinical, genetic and biophysical characteristics of cardiac sodium channel disease have been extensively studied, limited progress has been made in the development of treatment strategies targeting sodium channels. Classical non-selective sodium channel blockers have only limited clinical applicability, while more selective inhibitors of the late sodium current constitute a more promising treatment option. Because of our insufficient understanding of their complexity and subcellular diversity, other specific therapeutic targets for modulating sodium channels remain elusive. The current status and future potential of targeting sodium channels in cardiac arrhythmias are discussed.
Collapse
Affiliation(s)
- Carol Ann Remme
- Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, The Netherlands.
| | - Arthur A M Wilde
- Department of Clinical and Experimental Cardiology, Academic Medical Center, University of Amsterdam, The Netherlands
| |
Collapse
|
97
|
Dolz-Gaitón P, Núñez M, Núñez L, Barana A, Amorós I, Matamoros M, Pérez-Hernández M, González de la Fuente M, Álvarez-López M, Macías-Ruiz R, Tercedor-Sánchez L, Jiménez-Jáimez J, Delpón E, Caballero R, Tamargo J. Functional characterization of a novel frameshift mutation in the C-terminus of the Nav1.5 channel underlying a Brugada syndrome with variable expression in a Spanish family. PLoS One 2013; 8:e81493. [PMID: 24363796 PMCID: PMC3868464 DOI: 10.1371/journal.pone.0081493] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 10/14/2013] [Indexed: 12/19/2022] Open
Abstract
Introduction We functionally analyzed a frameshift mutation in the SCN5A gene encoding cardiac Na+ channels (Nav1.5) found in a proband with repeated episodes of ventricular fibrillation who presented bradycardia and paroxysmal atrial fibrillation. Seven relatives also carry the mutation and showed a Brugada syndrome with an incomplete and variable expression. The mutation (p.D1816VfsX7) resulted in a severe truncation (201 residues) of the Nav1.5 C-terminus. Methods and Results Wild-type (WT) and mutated Nav1.5 channels together with hNavβ1 were expressed in CHO cells and currents were recorded at room temperature using the whole-cell patch-clamp. Expression of p.D1816VfsX7 alone resulted in a marked reduction (≈90%) in peak Na+ current density compared with WT channels. Peak current density generated by p.D1816VfsX7+WT was ≈50% of that generated by WT channels. p.D1816VfsX7 positively shifted activation and inactivation curves, leading to a significant reduction of the window current. The mutation accelerated current activation and reactivation kinetics and increased the fraction of channels developing slow inactivation with prolonged depolarizations. However, late INa was not modified by the mutation. p.D1816VfsX7 produced a marked reduction of channel trafficking toward the membrane that was not restored by decreasing incubation temperature during cell culture or by incubation with 300 μM mexiletine and 5 mM 4-phenylbutirate. Conclusion Despite a severe truncation of the C-terminus, the resulting mutated channels generate currents, albeit with reduced amplitude and altered biophysical properties, confirming the key role of the C-terminal domain in the expression and function of the cardiac Na+ channel.
Collapse
Affiliation(s)
- Pablo Dolz-Gaitón
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Mercedes Núñez
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Lucía Núñez
- Complejo Hospitalario Universitario de A Coruña and Instituto de Ciencias de la Salud, Universidad de A Coruña, A Coruña, Spain
| | - Adriana Barana
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Irene Amorós
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Marcos Matamoros
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Marta Pérez-Hernández
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Marta González de la Fuente
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Miguel Álvarez-López
- Arrhytmias Unit, Cardiology Department, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Rosa Macías-Ruiz
- Arrhytmias Unit, Cardiology Department, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Luis Tercedor-Sánchez
- Arrhytmias Unit, Cardiology Department, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Juan Jiménez-Jáimez
- Arrhytmias Unit, Cardiology Department, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Eva Delpón
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid, Spain
- * E-mail:
| | - Ricardo Caballero
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, School of Medicine, Universidad Complutense, Madrid, Spain
| |
Collapse
|
98
|
Abstract
Control and modulation of electrical signaling is vital to normal physiology, particularly in neurons, cardiac myocytes, and skeletal muscle. The orchestrated activities of variable sets of ion channels and transporters, including voltage-gated ion channels (VGICs), are responsible for initiation, conduction, and termination of the action potential (AP) in excitable cells. Slight changes in VGIC activity can lead to severe pathologies including arrhythmias, epilepsies, and paralyses, while normal excitability depends on the precise tuning of the AP waveform. VGICs are heavily posttranslationally modified, with upward of 30% of the mature channel mass consisting of N- and O-glycans. These glycans are terminated typically by negatively charged sialic acid residues that modulate voltage-dependent channel gating directly. The data indicate that sialic acids alter VGIC activity in isoform-specific manners, dependent in part, on the number/location of channel sialic acids attached to the pore-forming alpha and/or auxiliary subunits that often act through saturating electrostatic mechanisms. Additionally, cell-specific regulation of sialylation can affect VGIC gating distinctly. Thus, channel sialylation is likely regulated through two mechanisms that together contribute to a dynamic spectrum of possible gating motifs: a subunit-specific mechanism and regulated (aberrant) changes in the ability of the cell to glycosylate. Recent studies showed that neuronal and cardiac excitability is modulated through regulated changes in voltage-gated Na(+) channel sialylation, suggesting that both mechanisms of differential VGIC sialylation contribute to electrical signaling in the brain and heart. Together, the data provide insight into an important and novel paradigm involved in the control and modulation of electrical signaling.
Collapse
Affiliation(s)
- Andrew R Ednie
- Programs in Cardiovascular Research and Neuroscience, Department of Molecular Pharmacology & Physiology, College of Medicine, University of South Florida, Tampa, Florida, USA
| | | |
Collapse
|
99
|
Late sodium current inhibition in acquired and inherited ventricular (dys)function and arrhythmias. Cardiovasc Drugs Ther 2013; 27:91-101. [PMID: 23292167 DOI: 10.1007/s10557-012-6433-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The late sodium current has been increasingly recognized for its mechanistic role in various cardiovascular pathologies, including angina pectoris, myocardial ischemia, atrial fibrillation, heart failure and congenital long QT syndrome. Although relatively small in magnitude, the late sodium current (I(NaL)) represents a functionally relevant contributor to cardiomyocyte (electro)physiology. Many aspects of I(NaL) itself are as yet still unresolved, including its distribution and function in different cell types throughout the heart, and its regulation by sodium channel accessory proteins and intracellular signalling pathways. Its complexity is further increased by a close interrelationship with the peak sodium current and other ion currents, hindering the development of inhibitors with selective and specific properties. Thus, increased knowledge of the intricacies of the complex nature of I(NaL) during distinct cardiovascular conditions and its potential as a pharmacological target is essential. Here, we provide an overview of the functional and electrophysiological effects of late sodium current inhibition on the level of the ventricular myocyte, and its potential cardioprotective and anti-arrhythmic efficacy in the setting of acquired and inherited ventricular dysfunction and arrhythmias.
Collapse
|
100
|
SCN5A promoter haplotype affects the therapeutic range for serum flecainide concentration in Asian patients. Pharmacogenet Genomics 2013; 23:349-54. [DOI: 10.1097/fpc.0b013e328361fb8d] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|