1
|
Cai X, Liu L, Xia F, Papadimos TJ, Wang Q. Apelin-13 reverses bupivacaine-induced cardiotoxicity: an experimental study. BRAZILIAN JOURNAL OF ANESTHESIOLOGY (ELSEVIER) 2024; 74:844501. [PMID: 38583586 PMCID: PMC11015498 DOI: 10.1016/j.bjane.2024.844501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
INTRODUCTION Cardiac arrest or arrhythmia caused by bupivacaine may be refractory to treatment. Apelin has been reported to directly increase the frequency of spontaneous activation and the propagation of action potentials, ultimately promoting cardiac contractility. This study aimed to investigate the effects of apelin-13 in reversing cardiac suppression induced by bupivacaine in rats. METHODS A rat model of cardiac suppression was established by a 3-min continuous intravenous infusion of bupivacaine at the rate of 5 mg.kg-1.min-1, and serial doses of apelin-13 (50, 150 and 450 μg.kg-1) were administered to rescue cardiac suppression to identify its dose-response relationship. We used F13A, an inhibitor of Angiotensin Receptor-Like 1 (APJ), and Protein Kinase C (PKC) inhibitor chelerythrine to reverse the effects of apelin-13. Moreover, the protein expressions of PKC, Nav1.5, and APJ in ventricular tissues were measured using Western blotting and immunofluorescence assay. RESULTS Compared to the control rats, the rats subjected to continuous intravenous administration of bupivacaine had impaired hemodynamic stability. Administration of apelin-13, in a dose-dependent manner, significantly improved hemodynamic parameters in rats with bupivacaine-induced cardiac suppression (p < 0.05), and apelin-13 treatment also significantly upregulated the protein expressions of p-PKC and Nav1.5 (p < 0.05), these effects were abrogated by F13A or chelerythrine (p < 0.05). CONCLUSION Exogenous apelin-13, at least in part, activates the PKC signaling pathway through the apelin/APJ system to improve cardiac function in a rat model of bupivacaine-induced cardiac suppression.
Collapse
Affiliation(s)
- Xixi Cai
- The First Affiliated Hospital of Wenzhou Medical University, Department of Anesthesiology, Zhejiang Province, China
| | - Le Liu
- The First Affiliated Hospital of Wenzhou Medical University, Department of Anesthesiology, Zhejiang Province, China
| | - Fangfang Xia
- The First Affiliated Hospital of Wenzhou Medical University, Department of Anesthesiology, Zhejiang Province, China
| | - Thomas J Papadimos
- The Ohio State University Wexner Medical Center, Department of Anesthesiology, Ohio, USA
| | - Quanguang Wang
- The First Affiliated Hospital of Wenzhou Medical University, Department of Anesthesiology, Zhejiang Province, China.
| |
Collapse
|
2
|
Lesage A, Lorenzini M, Burel S, Sarlandie M, Bibault F, Lindskog C, Maloney D, Silva JR, Townsend RR, Nerbonne JM, Marionneau C. Determinants of iFGF13-mediated regulation of myocardial voltage-gated sodium (NaV) channels in mouse. J Gen Physiol 2023; 155:e202213293. [PMID: 37516919 PMCID: PMC10374952 DOI: 10.1085/jgp.202213293] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/14/2023] [Accepted: 06/30/2023] [Indexed: 07/31/2023] Open
Abstract
Posttranslational regulation of cardiac NaV1.5 channels is critical in modulating channel expression and function, yet their regulation by phosphorylation of accessory proteins has gone largely unexplored. Using phosphoproteomic analysis of NaV channel complexes from adult mouse left ventricles, we identified nine phosphorylation sites on intracellular fibroblast growth factor 13 (iFGF13). To explore the potential roles of these phosphosites in regulating cardiac NaV currents, we abolished expression of iFGF13 in neonatal and adult mouse ventricular myocytes and rescued it with wild-type (WT), phosphosilent, or phosphomimetic iFGF13-VY. While the increased rate of closed-state inactivation of NaV channels induced by Fgf13 knockout in adult cardiomyocytes was completely restored by adenoviral-mediated expression of WT iFGF13-VY, only partial rescue was observed in neonatal cardiomyocytes after knockdown. The knockdown of iFGF13 in neonatal ventricular myocytes also shifted the voltage dependence of channel activation toward hyperpolarized potentials, a shift that was not reversed by WT iFGF13-VY expression. Additionally, we found that iFGF13-VY is the predominant isoform in adult ventricular myocytes, whereas both iFGF13-VY and iFGF13-S are expressed comparably in neonatal ventricular myocytes. Similar to WT iFGF13-VY, each of the iFGF13-VY phosphomutants studied restored NaV channel inactivation properties in both models. Lastly, Fgf13 knockout also increased the late Na+ current in adult cardiomyocytes, and this effect was restored with expression of WT and phosphosilent iFGF13-VY. Together, our results demonstrate that iFGF13 is highly phosphorylated and displays differential isoform expression in neonatal and adult ventricular myocytes. While we found no roles for iFGF13 phosphorylation, our results demonstrate differential effects of iFGF13 on neonatal and adult mouse ventricular NaV channels.
Collapse
Affiliation(s)
- Adrien Lesage
- CNRS, INSERM, L’institut du Thorax, Nantes Université, Nantes, France
| | - Maxime Lorenzini
- CNRS, INSERM, L’institut du Thorax, Nantes Université, Nantes, France
| | - Sophie Burel
- CNRS, INSERM, L’institut du Thorax, Nantes Université, Nantes, France
| | - Marine Sarlandie
- CNRS, INSERM, L’institut du Thorax, Nantes Université, Nantes, France
| | - Floriane Bibault
- CNRS, INSERM, L’institut du Thorax, Nantes Université, Nantes, France
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
| | | | - Jonathan R. Silva
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - R. Reid Townsend
- Department of Cell Biology and Physiology, Washington University Medical School, St. Louis, MO, USA
- Department of Medicine, Washington University Medical School, St. Louis, MO, USA
| | - Jeanne M. Nerbonne
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University Medical School, St. Louis, MO, USA
- Department of Developmental Biology, Washington University Medical School, St. Louis, MO, USA
| | - Céline Marionneau
- CNRS, INSERM, L’institut du Thorax, Nantes Université, Nantes, France
| |
Collapse
|
3
|
Lesage A, Lorenzini M, Burel S, Sarlandie M, Bibault F, Maloney D, Silva JR, Reid Townsend R, Nerbonne JM, Marionneau C. FHF2 phosphorylation and regulation of native myocardial Na V 1.5 channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.526475. [PMID: 36778222 PMCID: PMC9915605 DOI: 10.1101/2023.01.31.526475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Phosphorylation of the cardiac Na V 1.5 channel pore-forming subunit is extensive and critical in modulating channel expression and function, yet the regulation of Na V 1.5 by phosphorylation of its accessory proteins remains elusive. Using a phosphoproteomic analysis of Na V channel complexes purified from mouse left ventricles, we identified nine phosphorylation sites on Fibroblast growth factor Homologous Factor 2 (FHF2). To determine the roles of phosphosites in regulating Na V 1.5, we developed two models from neonatal and adult mouse ventricular cardiomyocytes in which FHF2 expression is knockdown and rescued by WT, phosphosilent or phosphomimetic FHF2-VY. While the increased rates of closed-state and open-state inactivation of Na V channels induced by the FHF2 knockdown are completely restored by the FHF2-VY isoform in adult cardiomyocytes, sole a partial rescue is obtained in neonatal cardiomyocytes. The FHF2 knockdown also shifts the voltage-dependence of activation towards hyperpolarized potentials in neonatal cardiomyocytes, which is not rescued by FHF2-VY. Parallel investigations showed that the FHF2-VY isoform is predominant in adult cardiomyocytes, while expression of FHF2-VY and FHF2-A is comparable in neonatal cardiomyocytes. Similar to WT FHF2-VY, however, each FHF2-VY phosphomutant restores the Na V channel inactivation properties in both models, preventing identification of FHF2 phosphosite roles. FHF2 knockdown also increases the late Na + current in adult cardiomyocytes, which is restored similarly by WT and phosphosilent FHF2-VY. Together, our results demonstrate that ventricular FHF2 is highly phosphorylated, implicate differential roles for FHF2 in regulating neonatal and adult mouse ventricular Na V 1.5, and suggest that the regulation of Na V 1.5 by FHF2 phosphorylation is highly complex. eTOC Summary Lesage et al . identify the phosphorylation sites of FHF2 from mouse left ventricular Na V 1.5 channel complexes. While no roles for FHF2 phosphosites could be recognized yet, the findings demonstrate differential FHF2-dependent regulation of neonatal and adult mouse ventricular Na V 1.5 channels.
Collapse
|
4
|
PKC regulation of ion channels: The involvement of PIP 2. J Biol Chem 2022; 298:102035. [PMID: 35588786 PMCID: PMC9198471 DOI: 10.1016/j.jbc.2022.102035] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 11/24/2022] Open
Abstract
Ion channels are integral membrane proteins whose gating has been increasingly shown to depend on the presence of the low-abundance membrane phospholipid, phosphatidylinositol (4,5) bisphosphate. The expression and function of ion channels is tightly regulated via protein phosphorylation by specific kinases, including various PKC isoforms. Several channels have further been shown to be regulated by PKC through altered surface expression, probability of channel opening, shifts in voltage dependence of their activation, or changes in inactivation or desensitization. In this review, we survey the impact of phosphorylation of various ion channels by PKC isoforms and examine the dependence of phosphorylated ion channels on phosphatidylinositol (4,5) bisphosphate as a mechanistic endpoint to control channel gating.
Collapse
|
5
|
Horváth B, Szentandrássy N, Almássy J, Dienes C, Kovács ZM, Nánási PP, Banyasz T. Late Sodium Current of the Heart: Where Do We Stand and Where Are We Going? Pharmaceuticals (Basel) 2022; 15:ph15020231. [PMID: 35215342 PMCID: PMC8879921 DOI: 10.3390/ph15020231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 02/05/2023] Open
Abstract
Late sodium current has long been linked to dysrhythmia and contractile malfunction in the heart. Despite the increasing body of accumulating information on the subject, our understanding of its role in normal or pathologic states is not complete. Even though the role of late sodium current in shaping action potential under physiologic circumstances is debated, it’s unquestioned role in arrhythmogenesis keeps it in the focus of research. Transgenic mouse models and isoform-specific pharmacological tools have proved useful in understanding the mechanism of late sodium current in health and disease. This review will outline the mechanism and function of cardiac late sodium current with special focus on the recent advances of the area.
Collapse
Affiliation(s)
- Balázs Horváth
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Norbert Szentandrássy
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
- Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| | - János Almássy
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Csaba Dienes
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Zsigmond Máté Kovács
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Péter P. Nánási
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
- Department of Dental Physiology and Pharmacology, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamas Banyasz
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
- Correspondence: ; Tel.: +36-(52)-255-575; Fax: +36-(52)-255-116
| |
Collapse
|
6
|
Function and regulation of phosphatase 1 in healthy and diseased heart. Cell Signal 2021; 90:110203. [PMID: 34822978 DOI: 10.1016/j.cellsig.2021.110203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022]
Abstract
Reversible phosphorylation of ion channels and calcium-handling proteins provides precise post-translational regulation of cardiac excitation and contractility. Serine/threonine phosphatases govern dephosphorylation of the majority of cardiac proteins. Accordingly, dysfunction of this regulation contributes to the development and progression of heart failure and atrial fibrillation. On the molecular level, these changes include alterations in the expression level and phosphorylation status of Ca2+ handling and excitation-contraction coupling proteins provoked by dysregulation of phosphatases. The serine/threonine protein phosphatase PP1 is one a major player in the regulation of cardiac excitation-contraction coupling. PP1 essentially impacts on cardiac physiology and pathophysiology via interactions with the cardiac ion channels Cav1.2, NKA, NCX and KCNQ1, sarcoplasmic reticulum-bound Ca2+ handling proteins such as RyR2, SERCA and PLB as well as the contractile proteins MLC2, TnI and MyBP-C. PP1 itself but also PP1-regulatory proteins like inhibitor-1, inhibitor-2 and heat-shock protein 20 are dysregulated in cardiac disease. Therefore, they represent interesting targets to gain more insights in heart pathophysiology and to identify new treatment strategies for patients with heart failure or atrial fibrillation. We describe the genetic and holoenzymatic structure of PP1 and review its role in the heart and cardiac disease. Finally, we highlight the importance of the PP1 regulatory proteins for disease manifestation, provide an overview of genetic models to study the role of PP1 for the development of heart failure and atrial fibrillation and discuss possibilities of pharmacological interventions.
Collapse
|
7
|
Zheng Y, Wan X, Yang D, Ramirez-Navarro A, Liu H, Fu JD, Deschênes I. A Heart Failure-Associated SCN5A Splice Variant Leads to a Reduction in Sodium Current Through Coupled-Gating With the Wild-Type Channel. Front Physiol 2021; 12:661429. [PMID: 33828490 PMCID: PMC8019726 DOI: 10.3389/fphys.2021.661429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Nav1.5, encoded by the gene SCN5A, is the predominant voltage-gated sodium channel expressed in the heart. It initiates the cardiac action potential and thus is crucial for normal heart rhythm and function. Dysfunctions in Nav1.5 have been involved in multiple congenital or acquired cardiac pathological conditions such as Brugada syndrome (BrS), Long QT Syndrome Type 3, and heart failure (HF), all of which can lead to sudden cardiac death (SCD) - one of the leading causes of death worldwide. Our lab has previously reported that Nav1.5 forms dimer channels with coupled gating. We also found that Nav1.5 BrS mutants can exert a dominant-negative (DN) effect and impair the function of wildtype (WT) channels through coupled-gating with the WT. It was previously reported that reduction in cardiac sodium currents (INa), observed in HF, could be due to the increased expression of an SCN5A splice variant - E28D, which results in a truncated sodium channel (Nav1.5-G1642X). In this study, we hypothesized that this SCN5A splice variant leads to INa reduction in HF through biophysical coupling with the WT. We showed that Nav1.5-G1642X is a non-functional channel but can interact with the WT, resulting in a DN effect on the WT channel. We found that both WT and the truncated channel Nav1.5-G1642X traffic at the cell surface, suggesting biophysical coupling. Indeed, we found that the DN effect can be abolished by difopein, an inhibitor of the biophysical coupling. Interestingly, the sodium channel polymorphism H558R, which has beneficial effect in HF patients, could also block the DN effect. In summary, the HF-associated splice variant Nav1.5-G1642X suppresses sodium currents in heart failure patients through a mechanism involving coupled-gating with the wildtype sodium channel.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Xiaoping Wan
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Dandan Yang
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Angelina Ramirez-Navarro
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Haiyan Liu
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Ji-Dong Fu
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Isabelle Deschênes
- Department of Physiology and Cell Biology, Frick Center for Heart Failure and Arrhythmias, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
8
|
Lorenzini M, Burel S, Lesage A, Wagner E, Charrière C, Chevillard PM, Evrard B, Maloney D, Ruff KM, Pappu RV, Wagner S, Nerbonne JM, Silva JR, Townsend RR, Maier LS, Marionneau C. Proteomic and functional mapping of cardiac NaV1.5 channel phosphorylation sites. J Gen Physiol 2021; 153:211660. [PMID: 33410863 PMCID: PMC7797897 DOI: 10.1085/jgp.202012646] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 10/23/2020] [Accepted: 12/03/2020] [Indexed: 12/16/2022] Open
Abstract
Phosphorylation of the voltage-gated Na+ (NaV) channel NaV1.5 regulates cardiac excitability, yet the phosphorylation sites regulating its function and the underlying mechanisms remain largely unknown. Using a systematic, quantitative phosphoproteomic approach, we analyzed NaV1.5 channel complexes purified from nonfailing and failing mouse left ventricles, and we identified 42 phosphorylation sites on NaV1.5. Most sites are clustered, and three of these clusters are highly phosphorylated. Analyses of phosphosilent and phosphomimetic NaV1.5 mutants revealed the roles of three phosphosites in regulating NaV1.5 channel expression and gating. The phosphorylated serines S664 and S667 regulate the voltage dependence of channel activation in a cumulative manner, whereas the nearby S671, the phosphorylation of which is increased in failing hearts, regulates cell surface NaV1.5 expression and peak Na+ current. No additional roles could be assigned to the other clusters of phosphosites. Taken together, our results demonstrate that ventricular NaV1.5 is highly phosphorylated and that the phosphorylation-dependent regulation of NaV1.5 channels is highly complex, site specific, and dynamic.
Collapse
Affiliation(s)
- Maxime Lorenzini
- Université de Nantes, Centre national de la recherche scientifique, Institut National de la Santé et de la Recherche Médicale, l'Institut du thorax, Nantes, France
| | - Sophie Burel
- Université de Nantes, Centre national de la recherche scientifique, Institut National de la Santé et de la Recherche Médicale, l'Institut du thorax, Nantes, France
| | - Adrien Lesage
- Université de Nantes, Centre national de la recherche scientifique, Institut National de la Santé et de la Recherche Médicale, l'Institut du thorax, Nantes, France
| | - Emily Wagner
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO
| | - Camille Charrière
- Université de Nantes, Centre national de la recherche scientifique, Institut National de la Santé et de la Recherche Médicale, l'Institut du thorax, Nantes, France
| | - Pierre-Marie Chevillard
- Université de Nantes, Centre national de la recherche scientifique, Institut National de la Santé et de la Recherche Médicale, l'Institut du thorax, Nantes, France
| | - Bérangère Evrard
- Université de Nantes, Centre national de la recherche scientifique, Institut National de la Santé et de la Recherche Médicale, l'Institut du thorax, Nantes, France
| | - Dan Maloney
- Bioinformatics Solutions Inc., Waterloo, Ontario, Canada
| | - Kiersten M Ruff
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO
| | - Rohit V Pappu
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO
| | - Stefan Wagner
- Department of Internal Medicine II, University Heart Center, University Hospital Regensburg, Regensburg, Germany
| | - Jeanne M Nerbonne
- Department of Developmental Biology, Washington University Medical School, St. Louis, MO.,Department of Medicine, Washington University Medical School, St. Louis, MO
| | - Jonathan R Silva
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO
| | - R Reid Townsend
- Department of Medicine, Washington University Medical School, St. Louis, MO.,Department of Cell Biology and Physiology, Washington University Medical School, St. Louis, MO
| | - Lars S Maier
- Department of Internal Medicine II, University Heart Center, University Hospital Regensburg, Regensburg, Germany
| | - Céline Marionneau
- Université de Nantes, Centre national de la recherche scientifique, Institut National de la Santé et de la Recherche Médicale, l'Institut du thorax, Nantes, France
| |
Collapse
|
9
|
Brewer KR, Kuenze G, Vanoye CG, George AL, Meiler J, Sanders CR. Structures Illuminate Cardiac Ion Channel Functions in Health and in Long QT Syndrome. Front Pharmacol 2020; 11:550. [PMID: 32431610 PMCID: PMC7212895 DOI: 10.3389/fphar.2020.00550] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
The cardiac action potential is critical to the production of a synchronized heartbeat. This electrical impulse is governed by the intricate activity of cardiac ion channels, among them the cardiac voltage-gated potassium (Kv) channels KCNQ1 and hERG as well as the voltage-gated sodium (Nav) channel encoded by SCN5A. Each channel performs a highly distinct function, despite sharing a common topology and structural components. These three channels are also the primary proteins mutated in congenital long QT syndrome (LQTS), a genetic condition that predisposes to cardiac arrhythmia and sudden cardiac death due to impaired repolarization of the action potential and has a particular proclivity for reentrant ventricular arrhythmias. Recent cryo-electron microscopy structures of human KCNQ1 and hERG, along with the rat homolog of SCN5A and other mammalian sodium channels, provide atomic-level insight into the structure and function of these proteins that advance our understanding of their distinct functions in the cardiac action potential, as well as the molecular basis of LQTS. In this review, the gating, regulation, LQTS mechanisms, and pharmacological properties of KCNQ1, hERG, and SCN5A are discussed in light of these recent structural findings.
Collapse
Affiliation(s)
- Kathryn R. Brewer
- Center for Structural Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, United States
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
| | - Georg Kuenze
- Center for Structural Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, United States
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Carlos G. Vanoye
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Alfred L. George
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, United States
- Department of Chemistry, Vanderbilt University, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, United States
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - Charles R. Sanders
- Center for Structural Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, United States
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
10
|
Zaitsev AV, Warren M. "Heart Oddity": Intrinsically Reduced Excitability in the Right Ventricle Requires Compensation by Regionally Specific Stress Kinase Function. Front Physiol 2020; 11:86. [PMID: 32132931 PMCID: PMC7040197 DOI: 10.3389/fphys.2020.00086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/27/2020] [Indexed: 11/13/2022] Open
Abstract
The traditional view of ventricular excitation and conduction is an all-or-nothing response mediated by a regenerative activation of the inward sodium channel, which gives rise to an essentially constant conduction velocity (CV). However, whereas there is no obvious biological need to tune-up ventricular conduction, the principal molecular components determining CV, such as sodium channels, inward-rectifier potassium channels, and gap junctional channels, are known targets of the “stress” protein kinases PKA and calcium/calmodulin dependent protein kinase II (CaMKII), and are thus regulatable by signal pathways converging on these kinases. In this mini-review we will expose deficiencies and controversies in our current understanding of how ventricular conduction is regulated by stress kinases, with a special focus on the chamber-specific dimension in this regulation. In particular, we will highlight an odd property of cardiac physiology: uniform CV in ventricles requires co-existence of mutually opposing gradients in cardiac excitability and stress kinase function. While the biological advantage of this peculiar feature remains obscure, it is important to recognize the clinical implications of this phenomenon pertinent to inherited or acquired conduction diseases and therapeutic interventions modulating activity of PKA or CaMKII.
Collapse
Affiliation(s)
- Alexey V Zaitsev
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Mark Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
11
|
El Refaey M, Musa H, Murphy NP, Lubbers ER, Skaf M, Han M, Cavus O, Koenig SN, Wallace MJ, Gratz D, Bradley E, Alsina KM, Wehrens XHT, Hund TJ, Mohler PJ. Protein Phosphatase 2A Regulates Cardiac Na + Channels. Circ Res 2019; 124:737-746. [PMID: 30602331 DOI: 10.1161/circresaha.118.314350] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
RATIONALE Voltage-gated Na+ channel ( INa) function is critical for normal cardiac excitability. However, the Na+ channel late component ( INa,L) is directly associated with potentially fatal forms of congenital and acquired human arrhythmia. CaMKII (Ca2+/calmodulin-dependent kinase II) enhances INa,L in response to increased adrenergic tone. However, the pathways that negatively regulate the CaMKII/Nav1.5 axis are unknown and essential for the design of new therapies to regulate the pathogenic INa,L. OBJECTIVE To define phosphatase pathways that regulate INa,L in vivo. METHODS AND RESULTS A mouse model lacking a key regulatory subunit (B56α) of the PP (protein phosphatase) 2A holoenzyme displayed aberrant action potentials after adrenergic stimulation. Unbiased computational modeling of B56α KO (knockout) mouse myocyte action potentials revealed an unexpected role of PP2A in INa,L regulation that was confirmed by direct INa,L recordings from B56α KO myocytes. Further, B56α KO myocytes display decreased sensitivity to isoproterenol-induced induction of arrhythmogenic INa,L, and reduced CaMKII-dependent phosphorylation of Nav1.5. At the molecular level, PP2A/B56α complex was found to localize and coimmunoprecipitate with the primary cardiac Nav channel, Nav1.5. CONCLUSIONS PP2A regulates Nav1.5 activity in mouse cardiomyocytes. This regulation is critical for pathogenic Nav1.5 late current and requires PP2A-B56α. Our study supports B56α as a novel target for the treatment of arrhythmia.
Collapse
Affiliation(s)
- Mona El Refaey
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Physiology and Cell Biology, Ohio State University, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., P.J.M.)
| | - Hassan Musa
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Physiology and Cell Biology, Ohio State University, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., P.J.M.)
| | - Nathaniel P Murphy
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Physiology and Cell Biology, Ohio State University, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., P.J.M.)
| | - Ellen R Lubbers
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Physiology and Cell Biology, Ohio State University, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., P.J.M.)
| | - Michel Skaf
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Physiology and Cell Biology, Ohio State University, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., P.J.M.)
| | - Mei Han
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Physiology and Cell Biology, Ohio State University, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., P.J.M.)
| | - Omer Cavus
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Physiology and Cell Biology, Ohio State University, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., P.J.M.)
| | - Sara N Koenig
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Physiology and Cell Biology, Ohio State University, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., P.J.M.)
| | - Michael J Wallace
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Physiology and Cell Biology, Ohio State University, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., P.J.M.)
| | - Daniel Gratz
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Biomedical Engineering, Ohio State University College of Engineering, Columbus (D.G., T.J.H.)
| | - Elisa Bradley
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Internal Medicine, Ohio State University College of Medicine, Columbus (E.B., T.J.H., P.J.M.)
| | - Katherina M Alsina
- Department of Molecular Physiology and Biophysics (K.M.A.), Baylor College of Medicine, Houston, TX.,Division of Cardiology, Department of Medicine (K.M.A.), Baylor College of Medicine, Houston, TX.,Division of Cardiology, Department of Pediatrics (K.M.A.), Baylor College of Medicine, Houston, TX
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (X.H.T.W.)
| | - Thomas J Hund
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Internal Medicine, Ohio State University College of Medicine, Columbus (E.B., T.J.H., P.J.M.).,Department of Biomedical Engineering, Ohio State University College of Engineering, Columbus (D.G., T.J.H.)
| | - Peter J Mohler
- From the Ohio State University College of Medicine and Wexner Medical Center, The Frick Center for Heart Failure and Arrhythmia, The Dorothy M. Davis Heart and Lung Research Institute, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., D.G., E.B., T.J.H., P.J.M.).,Department of Internal Medicine, Ohio State University College of Medicine, Columbus (E.B., T.J.H., P.J.M.).,Department of Physiology and Cell Biology, Ohio State University, Columbus (M.E.R., H.M., N.P.M., E.R.L., M.S., M.H., O.C., S.N.K., M.J.W., P.J.M.)
| |
Collapse
|
12
|
Heathfield LJ, Martin LJ, Ramesar R. Massively parallel sequencing in sudden unexpected death in infants: A case report in South Africa. FORENSIC SCIENCE INTERNATIONAL GENETICS SUPPLEMENT SERIES 2019. [DOI: 10.1016/j.fsigss.2019.10.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
13
|
A fundamental evaluation of the electrical properties and function of cardiac transverse tubules. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118502. [PMID: 31269418 DOI: 10.1016/j.bbamcr.2019.06.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 06/07/2019] [Accepted: 06/28/2019] [Indexed: 11/20/2022]
Abstract
This work discusses active and passive electrical properties of transverse (T-)tubules in ventricular cardiomyocytes to understand the physiological roles of T-tubules. T-tubules are invaginations of the lateral membrane that provide a large surface for calcium-handling proteins to facilitate sarcomere shortening. Higher heart rates correlate with higher T-tubular densities in mammalian ventricular cardiomyocytes. We assess ion dynamics in T-tubules and the effects of sodium current in T-tubules on the extracellular potential, which leads to a partial reduction of the sodium current in deep segments of a T-tubule. We moreover reflect on the impact of T-tubules on macroscopic conduction velocity, integrating fundamental principles of action potential propagation and conduction. We also theoretically assess how the conduction velocity is affected by different T-tubular sodium current densities. Lastly, we critically assess literature on ion channel expression to determine whether action potentials can be initiated in T-tubules.
Collapse
|
14
|
Zaitsev AV, Torres NS, Cawley KM, Sabry AD, Warren JS, Warren M. Conduction in the right and left ventricle is differentially regulated by protein kinases and phosphatases: implications for arrhythmogenesis. Am J Physiol Heart Circ Physiol 2019; 316:H1507-H1527. [PMID: 30875259 PMCID: PMC6620685 DOI: 10.1152/ajpheart.00660.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/19/2019] [Accepted: 03/07/2019] [Indexed: 12/19/2022]
Abstract
The "stress" kinases cAMP-dependent protein kinase (PKA) and calcium/calmodulin-dependent protein kinase II (CaMKII), phosphorylate the Na+ channel Nav1.5 subunit to regulate its function. However, how the channel regulation translates to ventricular conduction is poorly understood. We hypothesized that the stress kinases positively and differentially regulate conduction in the right (RV) and the left (LV) ventricles. We applied the CaMKII blocker KN93 (2.75 μM), PKA blocker H89 (10 μM), and broad-acting phosphatase blocker calyculin (30 nM) in rabbit hearts paced at a cycle length (CL) of 150-8,000 ms. We used optical mapping to determine the distribution of local conduction delays (inverse of conduction velocity). Control hearts exhibited constant and uniform conduction at all tested CLs. Calyculin (15-min perfusion) accelerated conduction, with greater effect in the RV (by 15.3%) than in the LV (by 4.1%; P < 0.05). In contrast, both KN93 and H89 slowed down conduction in a chamber-, time-, and CL-dependent manner, with the strongest effect in the RV outflow tract (RVOT). Combined KN93 and H89 synergistically promoted conduction slowing in the RV (KN93: 24.7%; H89: 29.9%; and KN93 + H89: 114.2%; P = 0.0016) but not the LV. The progressive depression of RV conduction led to conduction block and reentrant arrhythmias. Protein expression levels of both the CaMKII-δ isoform and the PKA catalytic subunit were higher in the RVOT than in the apical LV (P < 0.05). Thus normal RV conduction requires a proper balance between kinase and phosphatase activity. Dysregulation of this balance due to pharmacological interventions or disease is potentially proarrhythmic. NEW & NOTEWORTHY We show that uniform ventricular conduction requires a precise physiological balance of the activities of calcium/calmodulin-dependent protein kinase II (CaMKII), PKA, and phosphatases, which involves region-specific expression of CaMKII and PKA. Inhibiting CaMKII and/or PKA activity elicits nonuniform conduction depression, with the right ventricle becoming vulnerable to the development of conduction disturbances and ventricular fibrillation/ventricular tachycardia.
Collapse
Affiliation(s)
- Alexey V Zaitsev
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah , Salt Lake City, Utah
- Department of Bioengineering, University of Utah , Salt Lake City, Utah
| | - Natalia S Torres
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah , Salt Lake City, Utah
| | - Keiko M Cawley
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah , Salt Lake City, Utah
| | - Amira D Sabry
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah , Salt Lake City, Utah
| | - Junco S Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah , Salt Lake City, Utah
- Department of Internal Medicine, School of Medicine, University of Utah , Salt Lake City, Utah
| | - Mark Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah , Salt Lake City, Utah
- Department of Bioengineering, University of Utah , Salt Lake City, Utah
| |
Collapse
|
15
|
Iqbal SM, Lemmens‐Gruber R. Phosphorylation of cardiac voltage-gated sodium channel: Potential players with multiple dimensions. Acta Physiol (Oxf) 2019; 225:e13210. [PMID: 30362642 PMCID: PMC6590314 DOI: 10.1111/apha.13210] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 10/14/2018] [Accepted: 10/14/2018] [Indexed: 12/11/2022]
Abstract
Cardiomyocytes are highly coordinated cells with multiple proteins organized in micro domains. Minor changes or interference in subcellular proteins can cause major disturbances in physiology. The cardiac sodium channel (NaV1.5) is an important determinant of correct electrical activity in cardiomyocytes which are localized at intercalated discs, T‐tubules and lateral membranes in the form of a macromolecular complex with multiple interacting protein partners. The channel is tightly regulated by post‐translational modifications for smooth conduction and propagation of action potentials. Among regulatory mechanisms, phosphorylation is an enzymatic and reversible process which modulates NaV1.5 channel function by attaching phosphate groups to serine, threonine or tyrosine residues. Phosphorylation of NaV1.5 is implicated in both normal physiological and pathological processes and is carried out by multiple kinases. In this review, we discuss and summarize recent literature about the (a) structure of NaV1.5 channel, (b) formation and subcellular localization of NaV1.5 channel macromolecular complex, (c) post‐translational phosphorylation and regulation of NaV1.5 channel, and (d) how these phosphorylation events of NaV1.5 channel alter the biophysical properties and affect the channel during disease status. We expect, by reviewing these aspects will greatly improve our understanding of NaV1.5 channel biology, physiology and pathology, which will also provide an insight into the mechanism of arrythmogenesis at molecular level.
Collapse
Affiliation(s)
- Shahid M. Iqbal
- Department of Pharmacology and Toxicology University of Vienna Vienna Austria
- Drugs Regulatory Authority of Pakistan Telecom Foundation (TF) Complex Islamabad Pakistan
| | - Rosa Lemmens‐Gruber
- Department of Pharmacology and Toxicology University of Vienna Vienna Austria
| |
Collapse
|
16
|
Marionneau C, Abriel H. Cardiac Sodium Current Under Sympathetic Control Protein Phosphatase 2A Regulates Cardiac Na+ Channels. Circ Res 2019; 124:674-676. [PMID: 30817259 DOI: 10.1161/circresaha.119.314680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
| | - Hugues Abriel
- Ion Channels and Channelopathies Laboratory, Institute for Biochemistry and Molecular Medicine, University of Bern, Switzerland (H.A.)
| |
Collapse
|
17
|
Abstract
Fast opening and closing of voltage-gated sodium channels are crucial for proper propagation of the action potential through excitable tissues. Unlike potassium channels, sodium channel α-subunits are believed to form functional monomers. Yet, an increasing body of literature shows inconsistency with the traditional idea of a single α-subunit functioning as a monomer. Here we demonstrate that sodium channel α-subunits not only physically interact with each other but they actually assemble, function and gate as a dimer. We identify the region involved in the dimerization and demonstrate that 14-3-3 protein mediates the coupled gating. Importantly we show conservation of this mechanism among mammalian sodium channels. Our study not only shifts conventional paradigms in regard to sodium channel assembly, structure, and function but importantly this discovery of the mechanism involved in channel dimerization and biophysical coupling could open the door to new approaches and targets to treat and/or prevent sodium channelopathies. Voltage-gated sodium channels are expressed in excitable tissues and mutations have been linked to cardiac arrhythmias and channelopathies. Here the authors show that the sodium channel α-subunits interact to form a dimer and gate as dimer and that this functional dimerisation is conserved.
Collapse
|
18
|
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
|
19
|
Veeraraghavan R, Györke S, Radwański PB. Neuronal sodium channels: emerging components of the nano-machinery of cardiac calcium cycling. J Physiol 2017; 595:3823-3834. [PMID: 28195313 DOI: 10.1113/jp273058] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/05/2016] [Indexed: 01/07/2023] Open
Abstract
Excitation-contraction coupling is the bridge between cardiac electrical activation and mechanical contraction. It is driven by the influx of Ca2+ across the sarcolemma triggering Ca2+ release from the sarcoplasmic reticulum (SR) - a process termed Ca2+ -induced Ca2+ release (CICR) - followed by re-sequestration of Ca2+ into the SR. The Na+ /Ca2+ exchanger inextricably couples the cycling of Ca2+ and Na+ in cardiac myocytes. Thus, influx of Na+ via voltage-gated Na+ channels (NaV ) has emerged as an important regulator of CICR both in health and in disease. Recent insights into the subcellular distribution of cardiac and neuronal NaV isoforms and their ultrastructural milieu have important implications for the roles of these channels in mediating Ca2+ -driven arrhythmias. This review will discuss functional insights into the role of neuronal NaV isoforms vis-à-vis cardiac NaV s in triggering such arrhythmias and their potential as therapeutic targets in the context of the aforementioned structural observations.
Collapse
Affiliation(s)
- Rengasayee Veeraraghavan
- Virginia Tech Carilion Research Institute, and Center for Heart and Regenerative Medicine, Virginia Polytechnic University, Roanoke, VA, USA
| | - Sándor Györke
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, Ohio State University Wexner Medical Center, 473 West 12th Avenue, Room 510, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, College of Medicine, Ohio State University, Columbus, OH, USA
| | - Przemysław B Radwański
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, Ohio State University Wexner Medical Center, 473 West 12th Avenue, Room 510, Columbus, OH, 43210, USA.,Department of Physiology and Cell Biology, College of Medicine, Ohio State University, Columbus, OH, USA.,Division of Pharmacy Practice and Science, College of Pharmacy, Ohio State University, Columbus, OH, USA
| |
Collapse
|
20
|
Warren M, Sciuto KJ, Taylor TG, Garg V, Torres NS, Shibayama J, Spitzer KW, Zaitsev AV. Blockade of CaMKII depresses conduction preferentially in the right ventricular outflow tract and promotes ischemic ventricular fibrillation in the rabbit heart. Am J Physiol Heart Circ Physiol 2017; 312:H752-H767. [PMID: 28130334 DOI: 10.1152/ajpheart.00347.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 11/22/2022]
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) regulates the principle ion channels mediating cardiac excitability and conduction, but how this regulation translates to the normal and ischemic heart remains unknown. Diverging results on CaMKII regulation of Na+ channels further prevent predicting how CaMKII activity regulates excitability and conduction in the intact heart. To address this deficiency, we tested the effects of the CaMKII blocker KN93 (1 and 2.75 μM) and its inactive analog KN92 (2.75 μM) on conduction and excitability in the left (LV) and right (RV) ventricles of rabbit hearts during normal perfusion and global ischemia. We used optical mapping to determine local conduction delays and the optical action potential (OAP) upstroke velocity (dV/dtmax). At baseline, local conduction delays were similar between RV and LV, whereas the OAP dV/dtmax was lower in RV than in LV. At 2.75 μM, KN93 heterogeneously slowed conduction and reduced dV/dtmax, with the largest effect in the RV outflow tract (RVOT). This effect was further exacerbated by ischemia, leading to recurrent conduction block in the RVOT and early ventricular fibrillation (at 6.7 ± 0.9 vs. 18.2 ± 0.8 min of ischemia in control, P < 0.0001). Neither KN92 nor 1 μM KN93 depressed OAP dV/dtmax or conduction. Rabbit cardiomyocytes isolated from RVOT exhibited a significantly lower dV/dtmax than those isolated from the LV. KN93 (2.75 μM) significantly reduced dV/dtmax in cells from both locations. This led to frequency-dependent intermittent activation failure occurring predominantly in RVOT cells. Thus CaMKII blockade exacerbates intrinsically lower excitability in the RVOT, which is proarrhythmic during ischemia.NEW & NOTEWORTHY We show that calcium/calmodulin-dependent protein kinase II (CaMKII) blockade exacerbates intrinsically lower excitability in the right ventricular outflow tract, which causes highly nonuniform chamber-specific slowing of conduction and facilitates ventricular fibrillation during ischemia. Constitutive CaMKII activity is necessary for uniform and safe ventricular conduction, and CaMKII block is potentially proarrhythmic.
Collapse
Affiliation(s)
- Mark Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah
| | - Katie J Sciuto
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah
| | - Tyson G Taylor
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah
| | - Vivek Garg
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah
| | - Natalia S Torres
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah
| | - Junko Shibayama
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah
| | - Kenneth W Spitzer
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah
| | - Alexey V Zaitsev
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah
| |
Collapse
|
21
|
Abstract
Voltage-gated sodium channels (VGSCs) are critical determinants of excitability. The properties of VGSCs are thought to be tightly controlled. However, VGSCs are also subjected to extensive modifications. Multiple posttranslational modifications that covalently modify VGSCs in neurons and muscle have been identified. These include, but are not limited to, phosphorylation, ubiquitination, palmitoylation, nitrosylation, glycosylation, and SUMOylation. Posttranslational modifications of VGSCs can have profound impact on cellular excitability, contributing to normal and abnormal physiology. Despite four decades of research, the complexity of VGSC modulation is still being determined. While some modifications have similar effects on the various VGSC isoforms, others have isoform-specific interactions. In addition, while much has been learned about how individual modifications can impact VGSC function, there is still more to be learned about how different modifications can interact. Here we review what is known about VGSC posttranslational modifications with a focus on the breadth and complexity of the regulatory mechanisms that impact VGSC properties.
Collapse
Affiliation(s)
- Zifan Pei
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA
| | - Yanling Pan
- Medical Neuroscience Graduate Program, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA
| | - Theodore R Cummins
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA. .,Medical Neuroscience Graduate Program, Indiana University - Purdue University Indianapolis, Indianapolis, IN, USA.
| |
Collapse
|
22
|
Aromolaran AS, Chahine M, Boutjdir M. Regulation of Cardiac Voltage-Gated Sodium Channel by Kinases: Roles of Protein Kinases A and C. Handb Exp Pharmacol 2017; 246:161-184. [PMID: 29032483 DOI: 10.1007/164_2017_53] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In the heart, voltage-gated sodium (Nav) channel (Nav1.5) is defined by its pore-forming α-subunit and its auxiliary β-subunits, both of which are important for its critical contribution to the initiation and maintenance of the cardiac action potential (AP) that underlie normal heart rhythm. The physiological relevance of Nav1.5 is further marked by the fact that inherited or congenital mutations in Nav1.5 channel gene SCN5A lead to altered functional expression (including expression, trafficking, and current density), and are generally manifested in the form of distinct cardiac arrhythmic events, epilepsy, neuropathic pain, migraine, and neuromuscular disorders. However, despite significant advances in defining the pathophysiology of Nav1.5, the molecular mechanisms that underlie its regulation and contribution to cardiac disorders are poorly understood. It is rapidly becoming evident that the functional expression (localization, trafficking and gating) of Nav1.5 may be under modulation by post-translational modifications that are associated with phosphorylation. We review here the molecular basis of cardiac Na channel regulation by kinases (PKA and PKC) and the resulting functional consequences. Specifically, we discuss: (1) recent literature on the structural, molecular, and functional properties of cardiac Nav1.5 channels; (2) how these properties may be altered by phosphorylation in disease states underlain by congenital mutations in Nav1.5 channel and/or subunits such as long QT and Brugada syndromes. Our expectation is that understanding the roles of these distinct and complex phosphorylation processes on the functional expression of Nav1.5 is likely to provide crucial mechanistic insights into Na channel associated arrhythmogenic events and will facilitate the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Ademuyiwa S Aromolaran
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, USA
- Departments of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Mohamed Chahine
- CERVO Brain Research Center, Institut Universitaire en Santé Mentale de Québec, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, USA.
- Departments of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY, USA.
- Department of Medicine, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
23
|
Radwański PB, Ho HT, Veeraraghavan R, Brunello L, Liu B, Belevych AE, Unudurthi SD, Makara MA, Priori SG, Volpe P, Armoundas AA, Dillmann WH, Knollmann BC, Mohler PJ, Hund TJ, Györke S. Neuronal Na + Channels Are Integral Components of Pro-arrhythmic Na +/Ca 2+ Signaling Nanodomain That Promotes Cardiac Arrhythmias During β-adrenergic Stimulation. JACC Basic Transl Sci 2016; 1:251-266. [PMID: 27747307 PMCID: PMC5065245 DOI: 10.1016/j.jacbts.2016.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Cardiac arrhythmias are a leading cause of death in the US. Vast majority of these arrhythmias including catecholaminergic polymorphic ventricular tachycardia (CPVT) are associated with increased levels of circulating catecholamines and involve abnormal impulse formation secondary to aberrant Ca2+ and Na+ handling. However, the mechanistic link between β-AR stimulation and the subcellular/molecular arrhythmogenic trigger(s) remains elusive. METHODS AND RESULTS We performed functional and structural studies to assess Ca2+ and Na+ signaling in ventricular myocyte as well as surface electrocardiograms in mouse models of cardiac calsequestrin (CASQ2)-associated CPVT. We demonstrate that a subpopulation of Na+ channels (neuronal Na+ channels; nNav) that colocalize with RyR2 and Na+/Ca2+ exchanger (NCX) are a part of the β-AR-mediated arrhythmogenic process. Specifically, augmented Na+ entry via nNav in the settings of genetic defects within the RyR2 complex and enhanced sarcoplasmic reticulum (SR) Ca2+-ATPase (SERCA)-mediated SR Ca2+ refill is both an essential and a necessary factor for the arrhythmogenesis. Furthermore, we show that augmentation of Na+ entry involves β-AR-mediated activation of CAMKII subsequently leading to nNav augmentation. Importantly, selective pharmacological inhibition as well as silencing of Nav1.6 inhibit myocyte arrhythmic potential and prevent arrhythmias in vivo. CONCLUSION These data suggest that the arrhythmogenic alteration in Na+/Ca2+ handling evidenced ruing β-AR stimulation results, at least in part, from enhanced Na+ influx through nNav. Therefore, selective inhibition of these channels and Nav1.6 in particular can serve as a potential antiarrhythmic therapy.
Collapse
Affiliation(s)
- Przemysław B Radwański
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA ; Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, US ; Division of Pharmacy Practice and Sciences, College of Pharmacy, The Ohio State University, Columbus, OH, US
| | - Hsiang-Ting Ho
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA ; Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, US
| | - Rengasayee Veeraraghavan
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, Virginia Polytechnic University, Roanoke, VA, USA
| | - Lucia Brunello
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA ; Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, US
| | - Bin Liu
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA ; Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, US
| | - Andriy E Belevych
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA ; Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, US
| | - Sathya D Unudurthi
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA ; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Michael A Makara
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA ; Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, US
| | - Silvia G Priori
- Division of Cardiology and Molecular Cardiology, Maugeri Foundation-University of Pavia, Pavia, Italy
| | - Pompeo Volpe
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Antonis A Armoundas
- Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, MA, USA
| | - Wolfgang H Dillmann
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Bjorn C Knollmann
- Division of Clinical Pharmacology, Vanderbilt University Medical School, Nashville, TN, USA
| | - Peter J Mohler
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA ; Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, US
| | - Thomas J Hund
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA ; Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, Virginia Polytechnic University, Roanoke, VA, USA
| | - Sándor Györke
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA ; Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, US
| |
Collapse
|
24
|
Unudurthi SD, Hund TJ. Late sodium current dysregulation as a causal factor in arrhythmia. Expert Rev Cardiovasc Ther 2016; 14:545-7. [PMID: 26886049 DOI: 10.1586/14779072.2016.1155451] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Sathya D Unudurthi
- a The Dorothy M. Davis Heart and Lung Research Institute.,b Department of Biomedical Engineering , College of Engineering, The Ohio State University , Columbus , OH , USA
| | - Thomas J Hund
- a The Dorothy M. Davis Heart and Lung Research Institute.,c Department of Biomedical Engineering , College of Engineering.,d Department of Internal Medicine , The Ohio State University.,e Wexner Medical Center, The Ohio State University , Columbus , OH , USA
| |
Collapse
|
25
|
Weber S, Meyer-Roxlau S, Wagner M, Dobrev D, El-Armouche A. Counteracting Protein Kinase Activity in the Heart: The Multiple Roles of Protein Phosphatases. Front Pharmacol 2015; 6:270. [PMID: 26617522 PMCID: PMC4643138 DOI: 10.3389/fphar.2015.00270] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/28/2015] [Indexed: 12/19/2022] Open
Abstract
Decades of cardiovascular research have shown that variable and flexible levels of protein phosphorylation are necessary to maintain cardiac function. A delicate balance between phosphorylated and dephosphorylated states of proteins is guaranteed by a complex interplay of protein kinases (PKs) and phosphatases. Serine/threonine phosphatases, in particular members of the protein phosphatase (PP) family govern dephosphorylation of the majority of these cardiac proteins. Recent findings have however shown that PPs do not only dephosphorylate previously phosphorylated proteins as a passive control mechanism but are capable to actively control PK activity via different direct and indirect signaling pathways. These control mechanisms can take place on (epi-)genetic, (post-)transcriptional, and (post-)translational levels. In addition PPs themselves are targets of a plethora of proteinaceous interaction partner regulating their endogenous activity, thus adding another level of complexity and feedback control toward this system. Finally, novel approaches are underway to achieve spatiotemporal pharmacologic control of PPs which in turn can be used to fine-tune misleaded PK activity in heart disease. Taken together, this review comprehensively summarizes the major aspects of PP-mediated PK regulation and discusses the subsequent consequences of deregulated PP activity for cardiovascular diseases in depth.
Collapse
Affiliation(s)
- Silvio Weber
- Department of Pharmacology and Toxicology, Dresden University of Technology , Dresden, Germany
| | - Stefanie Meyer-Roxlau
- Department of Pharmacology and Toxicology, Dresden University of Technology , Dresden, Germany
| | - Michael Wagner
- Department of Pharmacology and Toxicology, Dresden University of Technology , Dresden, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, Faculty of Medicine, West German Heart and Vascular Center , Essen, Germany
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Dresden University of Technology , Dresden, Germany
| |
Collapse
|
26
|
Onwuli DO, Beltran-Alvarez P. An update on transcriptional and post-translational regulation of brain voltage-gated sodium channels. Amino Acids 2015; 48:641-651. [PMID: 26503606 PMCID: PMC4752963 DOI: 10.1007/s00726-015-2122-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 11/29/2022]
Abstract
Voltage-gated sodium channels are essential proteins in brain physiology, as they generate the sodium currents that initiate neuronal action potentials. Voltage-gated sodium channels expression, localisation and function are regulated by a range of transcriptional and post-translational mechanisms. Here, we review our understanding of regulation of brain voltage-gated sodium channels, in particular SCN1A (NaV1.1), SCN2A (NaV1.2), SCN3A (NaV1.3) and SCN8A (NaV1.6), by transcription factors, by alternative splicing, and by post-translational modifications. Our focus is strongly centred on recent research lines, and newly generated knowledge.
Collapse
Affiliation(s)
- Donatus O Onwuli
- School of Biological, Biomedical and Environmental Sciences, University of Hull, Hardy Building Cottingham Road, Hull, HU6 7RX, UK
| | - Pedro Beltran-Alvarez
- School of Biological, Biomedical and Environmental Sciences, University of Hull, Hardy Building Cottingham Road, Hull, HU6 7RX, UK.
| |
Collapse
|
27
|
Detta N, Frisso G, Salvatore F. The multi-faceted aspects of the complex cardiac Nav1.5 protein in membrane function and pathophysiology. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015. [PMID: 26209461 DOI: 10.1016/j.bbapap.2015.07.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The aim of this mini-review is to draw together the main concepts and findings that have emerged from recent studies of the cardiac channel protein Nav1.5. This complex protein is encoded by the SCN5A gene that, in its mutated form, is implicated in various diseases, particularly channelopathies, specifically at cardiac tissue level. Here we describe the structural, and functional aspects of Nav1.5 including post-translational modifications in normal conditions, and the main human channelopathies in which this protein may be the cause or trigger. Lastly, we also briefly discuss interacting proteins that are relevant for these channel functions in normal and disease conditions.
Collapse
Affiliation(s)
- Nicola Detta
- CEINGE-Biotecnologie Avanzate s.c.ar.l., Naples, Italy
| | - Giulia Frisso
- CEINGE-Biotecnologie Avanzate s.c.ar.l., Naples, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - Francesco Salvatore
- CEINGE-Biotecnologie Avanzate s.c.ar.l., Naples, Italy; IRCCS-Fondazione SDN, Naples, Italy.
| |
Collapse
|
28
|
Glynn P, Musa H, Wu X, Unudurthi SD, Little S, Qian L, Wright PJ, Radwanski PB, Gyorke S, Mohler PJ, Hund TJ. Voltage-Gated Sodium Channel Phosphorylation at Ser571 Regulates Late Current, Arrhythmia, and Cardiac Function In Vivo. Circulation 2015; 132:567-77. [PMID: 26187182 DOI: 10.1161/circulationaha.114.015218] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 06/12/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Voltage-gated Na(+) channels (Nav) are essential for myocyte membrane excitability and cardiac function. Nav current (INa) is a large-amplitude, short-duration spike generated by rapid channel activation followed immediately by inactivation. However, even under normal conditions, a small late component of INa (INa,L) persists because of incomplete/failed inactivation of a subpopulation of channels. Notably, INa,L is directly linked with both congenital and acquired disease states. The multifunctional Ca(2+)/calmodulin-dependent kinase II (CaMKII) has been identified as an important activator of INa,L in disease. Several potential CaMKII phosphorylation sites have been discovered, including Ser571 in the Nav1.5 DI-DII linker, but the molecular mechanism underlying CaMKII-dependent regulation of INa,L in vivo remains unknown. METHODS AND RESULTS To determine the in vivo role of Ser571, 2 Scn5a knock-in mouse models were generated expressing either: (1) Nav1.5 with a phosphomimetic mutation at Ser571 (S571E), or (2) Nav1.5 with the phosphorylation site ablated (S571A). Electrophysiology studies revealed that Ser571 regulates INa,L but not other channel properties previously linked to CaMKII. Ser571-mediated increases in INa,L promote abnormal repolarization and intracellular Ca(2+) handling and increase susceptibility to arrhythmia at the cellular and animal level. Importantly, Ser571 is required for maladaptive remodeling and arrhythmias in response to pressure overload. CONCLUSIONS Our data provide the first in vivo evidence for the molecular mechanism underlying CaMKII activation of the pathogenic INa,L. Relevant for improved rational design of potential therapies, our findings demonstrate that Ser571-dependent regulation of Nav1.5 specifically tunes INa,L without altering critical physiological components of the current.
Collapse
Affiliation(s)
- Patric Glynn
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Hassan Musa
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Xiangqiong Wu
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Sathya D Unudurthi
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Sean Little
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Lan Qian
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Patrick J Wright
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Przemyslaw B Radwanski
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Sandor Gyorke
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Peter J Mohler
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Thomas J Hund
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.).
| |
Collapse
|
29
|
Abriel H. Genetic background of Brugada syndrome is more complex than what we would like it to be! Cardiovasc Res 2015; 106:351-2. [PMID: 25926434 DOI: 10.1093/cvr/cvv135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Hugues Abriel
- Department of Clinical Research, Ion Channel Research Group, University of Bern, Murtenstrasse 35, Bern 3010, Switzerland
| |
Collapse
|
30
|
Herren AW, Weber DM, Rigor RR, Margulies KB, Phinney BS, Bers DM. CaMKII Phosphorylation of Na(V)1.5: Novel in Vitro Sites Identified by Mass Spectrometry and Reduced S516 Phosphorylation in Human Heart Failure. J Proteome Res 2015; 14:2298-311. [PMID: 25815641 DOI: 10.1021/acs.jproteome.5b00107] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The cardiac voltage-gated sodium channel, Na(V)1.5, drives the upstroke of the cardiac action potential and is a critical determinant of myocyte excitability. Recently, calcium (Ca(2+))/calmodulin(CaM)-dependent protein kinase II (CaMKII) has emerged as a critical regulator of Na(V)1.5 function through phosphorylation of multiple residues including S516, T594, and S571, and these phosphorylation events may be important for the genesis of acquired arrhythmias, which occur in heart failure. However, phosphorylation of full-length human Na(V)1.5 has not been systematically analyzed and Na(V)1.5 phosphorylation in human heart failure is incompletely understood. In the present study, we used label-free mass spectrometry to assess phosphorylation of human Na(V)1.5 purified from HEK293 cells with full coverage of phosphorylatable sites and identified 23 sites that were phosphorylated by CaMKII in vitro. We confirmed phosphorylation of S516 and S571 by LC-MS/MS and found a decrease in S516 phosphorylation in human heart failure, using a novel phospho-specific antibody. This work furthers our understanding of the phosphorylation of Na(V)1.5 by CaMKII under normal and disease conditions, provides novel CaMKII target sites for functional validation, and provides the first phospho-proteomic map of full-length human Na(V)1.5.
Collapse
Affiliation(s)
- Anthony W Herren
- †Department of Pharmacology, University of California Davis, Genome Building 3513, Davis, California 95616, United States
| | - Darren M Weber
- §UC Davis Genome Center, University of California Davis, 451 Health Science Drive, Davis, California 95616, United States
| | - Robert R Rigor
- †Department of Pharmacology, University of California Davis, Genome Building 3513, Davis, California 95616, United States
| | - Kenneth B Margulies
- ∥Perelman School of Medicine, University of Pennsylvania, 415 Curie Boulevard, Philadelphia, Pennsylvania 19104, United States
| | - Brett S Phinney
- §UC Davis Genome Center, University of California Davis, 451 Health Science Drive, Davis, California 95616, United States
| | - Donald M Bers
- †Department of Pharmacology, University of California Davis, Genome Building 3513, Davis, California 95616, United States
| |
Collapse
|
31
|
Ballou LM, Lin RZ, Cohen IS. Control of cardiac repolarization by phosphoinositide 3-kinase signaling to ion channels. Circ Res 2015; 116:127-37. [PMID: 25552692 DOI: 10.1161/circresaha.116.303975] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Upregulation of phosphoinositide 3-kinase (PI3K) signaling is a common alteration in human cancer, and numerous drugs that target this pathway have been developed for cancer treatment. However, recent studies have implicated inhibition of the PI3K signaling pathway as the cause of a drug-induced long-QT syndrome in which alterations in several ion currents contribute to arrhythmogenic drug activity. Surprisingly, some drugs that were thought to induce long-QT syndrome by direct block of the rapid delayed rectifier (IKr) also seem to inhibit PI3K signaling, an effect that may contribute to their arrhythmogenicity. The importance of PI3K in regulating cardiac repolarization is underscored by evidence that QT interval prolongation in diabetes mellitus also may result from changes in multiple currents because of decreased insulin activation of PI3K in the heart. How PI3K signaling regulates ion channels to control the cardiac action potential is poorly understood. Hence, this review summarizes what is known about the effect of PI3K and its downstream effectors, including Akt, on sodium, potassium, and calcium currents in cardiac myocytes. We also refer to some studies in noncardiac cells that provide insight into potential mechanisms of ion channel regulation by this signaling pathway in the heart. Drug development and safety could be improved with a better understanding of the mechanisms by which PI3K regulates cardiac ion channels and the extent to which PI3K inhibition contributes to arrhythmogenic susceptibility.
Collapse
Affiliation(s)
- Lisa M Ballou
- From the Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, NY (L.M.B., R.Z.L., I.S.C.); and the Medical Service, Northport VA Medical Center, NY (R.Z.L.)
| | - Richard Z Lin
- From the Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, NY (L.M.B., R.Z.L., I.S.C.); and the Medical Service, Northport VA Medical Center, NY (R.Z.L.).
| | - Ira S Cohen
- From the Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, NY (L.M.B., R.Z.L., I.S.C.); and the Medical Service, Northport VA Medical Center, NY (R.Z.L.).
| |
Collapse
|
32
|
Regulation of the cardiac Na+ channel NaV1.5 by post-translational modifications. J Mol Cell Cardiol 2015; 82:36-47. [PMID: 25748040 DOI: 10.1016/j.yjmcc.2015.02.013] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 01/28/2015] [Accepted: 02/17/2015] [Indexed: 02/07/2023]
Abstract
The cardiac voltage-gated Na(+) channel, Na(V)1.5, is responsible for the upstroke of the action potential in cardiomyocytes and for efficient propagation of the electrical impulse in the myocardium. Even subtle alterations of Na(V)1.5 function, as caused by mutations in its gene SCN5A, may lead to many different arrhythmic phenotypes in carrier patients. In addition, acquired malfunctions of Na(V)1.5 that are secondary to cardiac disorders such as heart failure and cardiomyopathies, may also play significant roles in arrhythmogenesis. While it is clear that the regulation of Na(V)1.5 protein expression and function tightly depends on genetic mechanisms, recent studies have demonstrated that Na(V)1.5 is the target of various post-translational modifications that are pivotal not only in physiological conditions, but also in disease. In this review, we examine the recent literature demonstrating glycosylation, phosphorylation by Protein Kinases A and C, Ca(2+)/Calmodulin-dependent protein Kinase II, Phosphatidylinositol 3-Kinase, Serum- and Glucocorticoid-inducible Kinases, Fyn and Adenosine Monophosphate-activated Protein Kinase, methylation, acetylation, redox modifications, and ubiquitylation of Na(V)1.5. Modern and sensitive mass spectrometry approaches, applied directly to channel proteins that were purified from native cardiac tissues, have enabled the determination of the precise location of post-translational modification sites, thus providing essential information for understanding the mechanistic details of these regulations. The current challenge is first, to understand the roles of these modifications on the expression and the function of Na(V)1.5, and second, to further identify other chemical modifications. It is postulated that the diversity of phenotypes observed with Na(V)1.5-dependent disorders may partially arise from the complex post-translational modifications of channel protein components.
Collapse
|
33
|
Nav1.5 channels can reach the plasma membrane through distinct N-glycosylation states. Biochim Biophys Acta Gen Subj 2015; 1850:1215-23. [PMID: 25721215 DOI: 10.1016/j.bbagen.2015.02.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 01/26/2015] [Accepted: 02/16/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Like many voltage-gated sodium channels, the cardiac isoform Nav1.5 is well known as a glycoprotein which necessarily undergoes N-glycosylation processing during its transit to the plasma membrane. In some cardiac disorders, especially the Brugada syndrome (BrS), mutations in Nav1.5 encoding gene lead to intracellular retention and consequently trafficking defect of these proteins. We used two BrS mutants as tools to clarify both Nav1.5 glycosylation states and associated secretory behaviors. METHODS Patch-clamp recordings and surface biotinylation assays of HEK293T cells expressing wild-type (WT) and/or mutant Nav1.5 proteins were performed to assess the impact of mutant co-expression on the membrane activity and localization of WT channels. Enzymatic deglycosylation assays and brefeldin A (BFA) treatments were also employed to further characterize recombinant and native Nav1.5 maturation. RESULTS The present data demonstrate that Nav1.5 channels mainly exist as two differentially glycosylated forms. We reveal that dominant negative effects induced by BrS mutants upon WT channel current result from the abnormal surface expression of the fully-glycosylated forms exclusively. Furthermore, we show that core-glycosylated channels can be found at the surface membrane of BFA-treated or untreated cells, but obviously without generating any sodium current. CONCLUSIONS Our findings provide evidence that native and recombinant Nav1.5 subunits are expressed as two distinct matured forms. Fully-glycosylated state of Nav1.5 seems to determine its functionality whereas core-glycosylated forms might be transported to the plasma membrane through an unconventional Golgi-independent secretory route. GENERAL SIGNIFICANCE This work highlights that N-linked glycosylation processing would be critical for Nav1.5 membrane trafficking and function.
Collapse
|
34
|
Abstract
The major cardiac voltage-gated sodium channel Nav1.5 associates with proteins that regulate its biosynthesis, localization, activity and degradation. Identification of partner proteins is crucial for a better understanding of the channel regulation. Using a yeast two-hybrid screen, we identified dynamitin as a Nav1.5-interacting protein. Dynamitin is part of the microtubule-binding multiprotein complex dynactin. When overexpressed it is a potent inhibitor of dynein/kinesin-mediated transport along the microtubules by disrupting the dynactin complex and dissociating cargoes from microtubules. The use of deletion constructs showed that the C-terminal domain of dynamitin is essential for binding to the first intracellular interdomain of Nav1.5. Co-immunoprecipitation assays confirmed the association between Nav1.5 and dynamitin in mouse heart extracts. Immunostaining experiments showed that dynamitin and Nav1.5 co-localize at intercalated discs of mouse cardiomyocytes. The whole-cell patch-clamp technique was applied to test the functional link between Nav1.5 and dynamitin. Dynamitin overexpression in HEK-293 (human embryonic kidney 293) cells expressing Nav1.5 resulted in a decrease in sodium current density in the membrane with no modification of the channel-gating properties. Biotinylation experiments produced similar information with a reduction in Nav1.5 at the cell surface when dynactin-dependent transport was inhibited. The present study strongly suggests that dynamitin is involved in the regulation of Nav1.5 cell-surface density.
Collapse
|
35
|
Beltran-Alvarez P, Feixas F, Osuna S, Díaz-Hernández R, Brugada R, Pagans S. Interplay between R513 methylation and S516 phosphorylation of the cardiac voltage-gated sodium channel. Amino Acids 2014; 47:429-34. [PMID: 25501501 DOI: 10.1007/s00726-014-1890-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 12/04/2014] [Indexed: 10/24/2022]
Abstract
Arginine methylation is a novel post-translational modification within the voltage-gated ion channel superfamily, including the cardiac sodium channel, NaV1.5. We show that NaV1.5 R513 methylation decreases S516 phosphorylation rate by 4 orders of magnitude, the first evidence of protein kinase A inhibition by arginine methylation. Reciprocally, S516 phosphorylation blocks R513 methylation. NaV1.5 p.G514C, associated to cardiac conduction disease, abrogates R513 methylation, while leaving S516 phosphorylation rate unchanged. This is the first report of methylation-phosphorylation cross-talk of a cardiac ion channel.
Collapse
Affiliation(s)
- Pedro Beltran-Alvarez
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona Dr. Josep Trueta, University of Girona, 17003, Girona, Spain,
| | | | | | | | | | | |
Collapse
|
36
|
Hund TJ, Mohler PJ. Role of CaMKII in cardiac arrhythmias. Trends Cardiovasc Med 2014; 25:392-7. [PMID: 25577293 DOI: 10.1016/j.tcm.2014.12.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/01/2014] [Accepted: 12/01/2014] [Indexed: 11/30/2022]
Abstract
Protein phosphorylation is a central mechanism in vertebrates for the regulation of signaling. With regard to the cardiovascular system, phosphorylation of myocyte targets is critical for the regulation of excitation contraction coupling, metabolism, intracellular calcium regulation, mitochondrial activity, transcriptional regulation, and cytoskeletal dynamics. In fact, pathways that tune protein kinase signaling have been a mainstay for cardiovascular therapies for the past 60 years. The calcium/calmodulin-dependent protein kinase II (CaMKII) is a multifunctional serine/threonine kinase with numerous roles in human physiology. Dysfunction in CaMKII-based signaling has been linked with a host of cardiovascular phenotypes including heart failure and arrhythmia, and CaMKII levels are elevated in human and animal disease models of heart disease. While nearly a decade has been invested in targeting CaMKII for the treatment of heart failure and arrhythmia phenotypes, to date, approaches to target the molecule for antiarrhythmic benefit have been unsuccessful for reasons that are still not entirely clear, although (1) lack of compound specificity and (2) the multitude of downstream targets are likely contributing factors. This review will provide an update on current pathways regulated by CaMKII with the goal of illustrating potential upstream regulatory mechanisms and downstream targets that may be modulated for the prevention of cardiac electrical defects. While the review will cover multiple aspects of CaMKII dysfunction in cardiovascular disease, we have given special attention to the potential of CaMKII-associated late Na(+) current as a novel therapeutic target for cardiac arrhythmia.
Collapse
Affiliation(s)
- Thomas J Hund
- The Dorothy M. Davis Heart & Lung Research Institute, OH; Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus, OH
| | - Peter J Mohler
- The Dorothy M. Davis Heart & Lung Research Institute, OH; Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH; Department of Physiology & Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH.
| |
Collapse
|
37
|
Beltran-Alvarez P, Tarradas A, Chiva C, Pérez-Serra A, Batlle M, Pérez-Villa F, Schulte U, Sabidó E, Brugada R, Pagans S. Identification of N-terminal protein acetylation and arginine methylation of the voltage-gated sodium channel in end-stage heart failure human heart. J Mol Cell Cardiol 2014; 76:126-9. [PMID: 25172307 DOI: 10.1016/j.yjmcc.2014.08.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 07/25/2014] [Accepted: 08/15/2014] [Indexed: 11/30/2022]
Abstract
The α subunit of the cardiac voltage-gated sodium channel, NaV1.5, provides the rapid sodium inward current that initiates cardiomyocyte action potentials. Here, we analyzed for the first time the post-translational modifications of NaV1.5 purified from end-stage heart failure human cardiac tissue. We identified R526 methylation as the major post-translational modification of any NaV1.5 arginine or lysine residue. Unexpectedly, we found that the N terminus of NaV1.5 was: 1) devoid of the initiation methionine, and 2) acetylated at the resulting initial alanine residue. This is the first evidence for N-terminal acetylation in any member of the voltage-gated ion channel superfamily. Our results open the door to explore NaV1.5 N-terminal acetylation and arginine methylation levels as drivers or markers of end-stage heart failure.
Collapse
Affiliation(s)
- Pedro Beltran-Alvarez
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona, 17003 Girona, Spain.
| | - Anna Tarradas
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona, 17003 Girona, Spain; Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain
| | - Cristina Chiva
- Proteomics Unit, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Alexandra Pérez-Serra
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona, 17003 Girona, Spain; Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain
| | - Montserrat Batlle
- Thorax Institute, Cardiology Department, Hospital Clínic, University of Barcelona, Institute of Biomedical Research August Pi i Sunyer, 08036 Barcelona, Spain
| | - Félix Pérez-Villa
- Thorax Institute, Cardiology Department, Hospital Clínic, University of Barcelona, Institute of Biomedical Research August Pi i Sunyer, 08036 Barcelona, Spain
| | - Uwe Schulte
- Logopharm GmbH, 79232 March-Buchheim, Germany
| | - Eduard Sabidó
- Proteomics Unit, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Ramon Brugada
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona, 17003 Girona, Spain; Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain.
| | - Sara Pagans
- Cardiovascular Genetics Center, Institut d'Investigació Biomèdica de Girona, 17003 Girona, Spain; Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain.
| |
Collapse
|
38
|
Jerng HH, Pfaffinger PJ. Modulatory mechanisms and multiple functions of somatodendritic A-type K (+) channel auxiliary subunits. Front Cell Neurosci 2014; 8:82. [PMID: 24723849 PMCID: PMC3973911 DOI: 10.3389/fncel.2014.00082] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/03/2014] [Indexed: 12/13/2022] Open
Abstract
Auxiliary subunits are non-conducting, modulatory components of the multi-protein ion channel complexes that underlie normal neuronal signaling. They interact with the pore-forming α-subunits to modulate surface distribution, ion conductance, and channel gating properties. For the somatodendritic subthreshold A-type potassium (ISA) channel based on Kv4 α-subunits, two types of auxiliary subunits have been extensively studied: Kv channel-interacting proteins (KChIPs) and dipeptidyl peptidase-like proteins (DPLPs). KChIPs are cytoplasmic calcium-binding proteins that interact with intracellular portions of the Kv4 subunits, whereas DPLPs are type II transmembrane proteins that associate with the Kv4 channel core. Both KChIPs and DPLPs genes contain multiple start sites that are used by various neuronal populations to drive the differential expression of functionally distinct N-terminal variants. In turn, these N-terminal variants generate tremendous functional diversity across the nervous system. Here, we focus our review on (1) the molecular mechanism underlying the unique properties of different N-terminal variants, (2) the shaping of native ISA properties by the concerted actions of KChIPs and DPLP variants, and (3) the surprising ways that KChIPs and DPLPs coordinate the activity of multiple channels to fine-tune neuronal excitability. Unlocking the unique contributions of different auxiliary subunit N-terminal variants may provide an important opportunity to develop novel targeted therapeutics to treat numerous neurological disorders.
Collapse
Affiliation(s)
- Henry H. Jerng
- Department of Neuroscience, Baylor College of MedicineHouston, TX, USA
| | | |
Collapse
|
39
|
Grandi E, Herren AW. CaMKII-dependent regulation of cardiac Na(+) homeostasis. Front Pharmacol 2014; 5:41. [PMID: 24653702 PMCID: PMC3948048 DOI: 10.3389/fphar.2014.00041] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 02/21/2014] [Indexed: 01/01/2023] Open
Abstract
Na+ homeostasis is a key regulator of cardiac excitation and contraction. The cardiac voltage-gated Na+ channel, NaV1.5, critically controls cell excitability, and altered channel gating has been implicated in both inherited and acquired arrhythmias. Ca2+/calmodulin-dependent protein kinase II (CaMKII), a serine/threonine kinase important in cardiac physiology and disease, phosphorylates NaV1.5 at multiple sites within the first intracellular linker loop to regulate channel gating. Although CaMKII sites on the channel have been identified (S516, T594, S571), the relative role of each of these phospho-sites in channel gating properties remains unclear, whereby both loss-of-function (reduced availability) and gain-of-function (late Na+ current, INaL) effects have been reported. Our review highlights investigating the complex multi-site phospho-regulation of NaV1.5 gating is crucial to understanding the genesis of acquired arrhythmias in heart failure (HF) and CaMKII activated conditions. In addition, the increased Na+ influx accompanying INaL may also indirectly contribute to arrhythmia by promoting Ca2+ overload. While the precise mechanisms of Na+ loading during HF remain unclear, and quantitative analyses of the contribution of INaL are lacking, disrupted Na+ homeostasis is a consistent feature of HF. Computational and experimental observations suggest that both increased diastolic Na+ influx and action potential prolongation due to systolic INaL contribute to disruption of Ca2+ handling in failing hearts. Furthermore, simulations reveal a synergistic interaction between perturbed Na+ fluxes and CaMKII, and confirm recent experimental findings of an arrhythmogenic feedback loop, whereby CaMKII activation is at once a cause and a consequence of Na+ loading.
Collapse
Affiliation(s)
- Eleonora Grandi
- Department of Pharmacology, University of California at Davis Davis, CA, USA
| | - Anthony W Herren
- Department of Pharmacology, University of California at Davis Davis, CA, USA
| |
Collapse
|
40
|
Lu Z, Jiang YP, Wu CYC, Ballou LM, Liu S, Carpenter ES, Rosen MR, Cohen IS, Lin RZ. Increased persistent sodium current due to decreased PI3K signaling contributes to QT prolongation in the diabetic heart. Diabetes 2013; 62:4257-65. [PMID: 23974924 PMCID: PMC3837031 DOI: 10.2337/db13-0420] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Diabetes is an independent risk factor for sudden cardiac death and ventricular arrhythmia complications of acute coronary syndrome. Prolongation of the QT interval on the electrocardiogram is also a risk factor for arrhythmias and sudden death, and the increased prevalence of QT prolongation is an independent risk factor for cardiovascular death in diabetic patients. The pathophysiological mechanisms responsible for this lethal complication are poorly understood. Diabetes is associated with a reduction in phosphoinositide 3-kinase (PI3K) signaling, which regulates the action potential duration (APD) of individual myocytes and thus the QT interval by altering multiple ion currents, including the persistent sodium current INaP. Here, we report a mechanism for diabetes-induced QT prolongation that involves an increase in INaP caused by defective PI3K signaling. Cardiac myocytes of mice with type 1 or type 2 diabetes exhibited an increase in APD that was reversed by expression of constitutively active PI3K or intracellular infusion of phosphatidylinositol 3,4,5-trisphosphate (PIP3), the second messenger produced by PI3K. The diabetic myocytes also showed an increase in INaP that was reversed by activated PI3K or PIP3. The increases in APD and INaP in myocytes translated into QT interval prolongation for both types of diabetic mice. The long QT interval of type 1 diabetic hearts was shortened by insulin treatment ex vivo, and this effect was blocked by a PI3K inhibitor. Treatment of both types of diabetic mouse hearts with an INaP blocker also shortened the QT interval. These results indicate that downregulation of cardiac PI3K signaling in diabetes prolongs the QT interval at least in part by causing an increase in INaP. This mechanism may explain why the diabetic population has an increased risk of life-threatening arrhythmias.
Collapse
Affiliation(s)
- Zhongju Lu
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
| | - Ya-Ping Jiang
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
| | - Chia-Yen C. Wu
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
| | - Lisa M. Ballou
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
| | - Shengnan Liu
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
| | - Eileen S. Carpenter
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| | - Michael R. Rosen
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
- Department of Pharmacology, Columbia University, New York, New York
| | - Ira S. Cohen
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
- Corresponding author: Ira S. Cohen, , or Richard Z. Lin,
| | - Richard Z. Lin
- Department of Physiology and Biophysics and the Institute for Molecular Cardiology, Stony Brook University, Stony Brook, New York
- Medical Service, Northport VA Medical Center, Northport, New York
- Corresponding author: Ira S. Cohen, , or Richard Z. Lin,
| |
Collapse
|
41
|
Heijman J, Dewenter M, El-Armouche A, Dobrev D. Function and regulation of serine/threonine phosphatases in the healthy and diseased heart. J Mol Cell Cardiol 2013; 64:90-8. [PMID: 24051368 DOI: 10.1016/j.yjmcc.2013.09.006] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 09/03/2013] [Accepted: 09/08/2013] [Indexed: 12/20/2022]
Abstract
Protein phosphorylation is a major control mechanism of a wide range of physiological processes and plays an important role in cardiac pathophysiology. Serine/threonine protein phosphatases control the dephosphorylation of a variety of cardiac proteins, thereby fine-tuning cardiac electrophysiology and function. Specificity of protein phosphatases type-1 and type-2A is achieved by multiprotein complexes that target the catalytic subunits to specific subcellular domains. Here, we describe the composition, regulation and target substrates of serine/threonine phosphatases in the heart. In addition, we provide an overview of pharmacological tools and genetic models to study the role of cardiac phosphatases. Finally, we review the role of protein phosphatases in the diseased heart, particularly in ventricular arrhythmias and atrial fibrillation and discuss their role as potential therapeutic targets.
Collapse
Affiliation(s)
- Jordi Heijman
- Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, 45122 Essen, Germany
| | | | | | | |
Collapse
|
42
|
Herren AW, Bers DM, Grandi E. Post-translational modifications of the cardiac Na channel: contribution of CaMKII-dependent phosphorylation to acquired arrhythmias. Am J Physiol Heart Circ Physiol 2013; 305:H431-45. [PMID: 23771687 DOI: 10.1152/ajpheart.00306.2013] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The voltage-gated Na channel isoform 1.5 (NaV1.5) is the pore forming α-subunit of the voltage-gated cardiac Na channel, which is responsible for the initiation and propagation of cardiac action potentials. Mutations in the SCN5A gene encoding NaV1.5 have been linked to changes in the Na current leading to a variety of arrhythmogenic phenotypes, and alterations in the NaV1.5 expression level, Na current density, and/or gating have been observed in acquired cardiac disorders, including heart failure. The precise mechanisms underlying these abnormalities have not been fully elucidated. However, several recent studies have made it clear that NaV1.5 forms a macromolecular complex with a number of proteins that modulate its expression levels, localization, and gating and is the target of extensive post-translational modifications, which may also influence all these properties. We review here the molecular aspects of cardiac Na channel regulation and their functional consequences. In particular, we focus on the molecular and functional aspects of Na channel phosphorylation by the Ca/calmodulin-dependent protein kinase II, which is hyperactive in heart failure and has been causally linked to cardiac arrhythmia. Understanding the mechanisms of altered NaV1.5 expression and function is crucial for gaining insight into arrhythmogenesis and developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Anthony W Herren
- Department of Pharmacology, University of California Davis, Davis, California
| | | | | |
Collapse
|
43
|
SOTTAS VALENTIN, ROUGIER JEANSÉBASTIEN, JOUSSET FLORIAN, KUCERA JANP, SHESTAK ANNA, MAKAROV LEONIDM, ZAKLYAZMINSKAYA ELENAV, ABRIEL HUGUES. Characterization of 2 Genetic Variants of Nav
1.5-Arginine 689 Found in Patients with Cardiac Arrhythmias. J Cardiovasc Electrophysiol 2013; 24:1037-46. [DOI: 10.1111/jce.12173] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 04/06/2013] [Accepted: 04/10/2013] [Indexed: 12/19/2022]
Affiliation(s)
- VALENTIN SOTTAS
- Department of Clinical Research; University of Bern; Switzerland
| | | | | | - JAN P. KUCERA
- Department of Physiology; University of Bern; Switzerland
| | - ANNA SHESTAK
- Russian Research Centre of Surgery RAMS; Laboratory of Medical Genetics; Moscow Russia
| | - LEONID M. MAKAROV
- Center for Syncope and Arrhythmias in Children and Adolescents FMBA; Moscow Russia
| | | | - HUGUES ABRIEL
- Department of Clinical Research; University of Bern; Switzerland
| |
Collapse
|