1
|
Boileve A, Romito O, Hof T, Levallois A, Brard L, d'Hers S, Fouchet A, Simard C, Guinamard R, Brette F, Sallé L. EPAC1 and 2 inhibit K + currents via PLC/PKC and NOS/PKG pathways in rat ventricular cardiomyocytes. Am J Physiol Cell Physiol 2024; 327:C557-C570. [PMID: 38985989 DOI: 10.1152/ajpcell.00582.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/29/2024] [Accepted: 06/19/2024] [Indexed: 07/12/2024]
Abstract
The exchange protein directly activated by cAMP (EPAC) has been implicated in cardiac proarrhythmic signaling pathways including spontaneous diastolic Ca2+ leak from sarcoplasmic reticulum and increased action potential duration (APD) in isolated ventricular cardiomyocytes. The action potential (AP) lengthening following acute EPAC activation is mainly due to a decrease of repolarizing steady-state K+ current (IKSS) but the mechanisms involved remain unknown. This study aimed to assess the role of EPAC1 and EPAC2 in the decrease of IKSS and to investigate the underlying signaling pathways. AP and K+ currents were recorded with the whole cell configuration of the patch-clamp technique in freshly isolated rat ventricular myocytes. EPAC1 and EPAC2 were pharmacologically activated with 8-(4-chlorophenylthio)-2'-O-methyl-cAMP acetoxymethyl ester (8-CPTAM, 10 µmol/L) and inhibited with R-Ce3F4 and ESI-05, respectively. Inhibition of EPAC1 and EPAC2 significantly decreased the effect of 8-CPTAM on APD and IKSS showing that both EPAC isoforms are involved in these effects. Unexpectedly, calmodulin-dependent protein kinase II (CaMKII) inhibition by AIP or KN-93, and Ca2+ chelation by intracellular BAPTA, did not impact the response to 8-CPTAM. However, inhibition of PLC/PKC and nitric oxide synthase (NOS)/PKG pathways partially prevents the 8-CPTAM-dependent decrease of IKSS. Finally, the cumulative inhibition of PKC and PKG blocked the 8-CPTAM effect, suggesting that these two actors work along parallel pathways to regulate IKSS upon EPAC activation. On the basis of such findings, we propose that EPAC1 and EPAC2 are involved in APD lengthening by inhibiting a K+ current via both PLC/PKC and NOS/PKG pathways. This may have pathological implications since EPAC is upregulated in diseases such as cardiac hypertrophy.NEW & NOTEWORHTY Exchange protein directly activated by cAMP (EPAC) proteins modulate ventricular electrophysiology at the cellular level. Both EPAC1 and EPAC2 isoforms participate in this effect. Mechanistically, PLC/PKC and nitric oxide synthase (NO)/PKG pathways are involved in regulating K+ repolarizing current whereas the well-known downstream effector of EPAC, calmodulin-dependent protein kinase II (CaMKII), does not participate. This may have pathological implications since EPAC is upregulated in diseases such as cardiac hypertrophy. Thus, EPAC inhibition may be a new approach to prevent arrhythmias under pathological conditions.
Collapse
Affiliation(s)
- Arthur Boileve
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Olivier Romito
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Thomas Hof
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Aurélia Levallois
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Laura Brard
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Sarah d'Hers
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Alexandre Fouchet
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Christophe Simard
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Romain Guinamard
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Fabien Brette
- PhyMedExp, INSERM U1046, CNRS 9412, Université de Montpellier, Montpellier, France
| | - Laurent Sallé
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| |
Collapse
|
2
|
Mazhar F, Bartolucci C, Regazzoni F, Paci M, Dedè L, Quarteroni A, Corsi C, Severi S. A detailed mathematical model of the human atrial cardiomyocyte: integration of electrophysiology and cardiomechanics. J Physiol 2024; 602:4543-4583. [PMID: 37641426 DOI: 10.1113/jp283974] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Mechano-electric regulations (MER) play an important role in the maintenance of cardiac performance. Mechano-calcium and mechano-electric feedback (MCF and MEF) pathways adjust the cardiomyocyte contractile force according to mechanical perturbations and affects electro-mechanical coupling. MER integrates all these regulations in one unit resulting in a complex phenomenon. Computational modelling is a useful tool to accelerate the mechanistic understanding of complex experimental phenomena. We have developed a novel model that integrates the MER loop for human atrial cardiomyocytes with proper consideration of feedforward and feedback pathways. The model couples a modified version of the action potential (AP) Koivumäki model with the contraction model by Quarteroni group. The model simulates iso-sarcometric and isometric twitches and the feedback effects on AP and Ca2+-handling. The model showed a biphasic response of Ca2+ transient (CaT) peak to increasing pacing rates and highlights the possible mechanisms involved. The model has shown a shift of the threshold for AP and CaT alternans from 4.6 to 4 Hz under post-operative atrial fibrillation, induced by depressed SERCA activity. The alternans incidence was dependent on a chain of mechanisms including RyRs availability time, MCF coupling, CaMKII phosphorylation, and the stretch levels. As a result, the model predicted a 10% slowdown of conduction velocity for a 20% stretch, suggesting a role of stretch in creation of substrate formation for atrial fibrillation. Overall, we conclude that the developed model provides a physiological CaT followed by a physiological twitch. This model can open pathways for the future studies of human atrial electromechanics. KEY POINTS: With the availability of human atrial cellular data, interest in atrial-specific model integration has been enhanced. We have developed a detailed mathematical model of human atrial cardiomyocytes including the mechano-electric regulatory loop. The model has gone through calibration and evaluation phases against a wide collection of available human in-vitro data. The usefulness of the model for analysing clinical problems has been preliminaryly tested by simulating the increased incidence of Ca2+ transient and action potential alternans at high rates in post-operative atrial fibrillation condition. The model determines the possible role of mechano-electric feedback in alternans incidence, which can increase vulnerability to atrial arrhythmias by varying stretch levels. We found that our physiologically accurate description of Ca2+ handling can reproduce many experimental phenomena and can help to gain insights into the underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Fazeelat Mazhar
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | - Chiara Bartolucci
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | | | - Michelangelo Paci
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Luca Dedè
- MOX - Dipartimento di Matematica, Politecnico di Milano, Milan, Italy
| | - Alfio Quarteroni
- MOX - Dipartimento di Matematica, Politecnico di Milano, Milan, Italy
- Mathematics Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Cristiana Corsi
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | - Stefano Severi
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| |
Collapse
|
3
|
Ni H, Morotti S, Zhang X, Dobrev D, Grandi E. Integrative human atrial modelling unravels interactive protein kinase A and Ca2+/calmodulin-dependent protein kinase II signalling as key determinants of atrial arrhythmogenesis. Cardiovasc Res 2023; 119:2294-2311. [PMID: 37523735 PMCID: PMC11318383 DOI: 10.1093/cvr/cvad118] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/18/2023] [Accepted: 06/05/2023] [Indexed: 08/02/2023] Open
Abstract
AIMS Atrial fibrillation (AF), the most prevalent clinical arrhythmia, is associated with atrial remodelling manifesting as acute and chronic alterations in expression, function, and regulation of atrial electrophysiological and Ca2+-handling processes. These AF-induced modifications crosstalk and propagate across spatial scales creating a complex pathophysiological network, which renders AF resistant to existing pharmacotherapies that predominantly target transmembrane ion channels. Developing innovative therapeutic strategies requires a systems approach to disentangle quantitatively the pro-arrhythmic contributions of individual AF-induced alterations. METHODS AND RESULTS Here, we built a novel computational framework for simulating electrophysiology and Ca2+-handling in human atrial cardiomyocytes and tissues, and their regulation by key upstream signalling pathways [i.e. protein kinase A (PKA), and Ca2+/calmodulin-dependent protein kinase II (CaMKII)] involved in AF-pathogenesis. Populations of atrial cardiomyocyte models were constructed to determine the influence of subcellular ionic processes, signalling components, and regulatory networks on atrial arrhythmogenesis. Our results reveal a novel synergistic crosstalk between PKA and CaMKII that promotes atrial cardiomyocyte electrical instability and arrhythmogenic triggered activity. Simulations of heterogeneous tissue demonstrate that this cellular triggered activity is further amplified by CaMKII- and PKA-dependent alterations of tissue properties, further exacerbating atrial arrhythmogenesis. CONCLUSIONS Our analysis reveals potential mechanisms by which the stress-associated adaptive changes turn into maladaptive pro-arrhythmic triggers at the cellular and tissue levels and identifies potential anti-AF targets. Collectively, our integrative approach is powerful and instrumental to assemble and reconcile existing knowledge into a systems network for identifying novel anti-AF targets and innovative approaches moving beyond the traditional ion channel-based strategy.
Collapse
Affiliation(s)
- Haibo Ni
- Department of Pharmacology, University of California Davis,
451 Health Sciences Drive, Davis, CA 95616, USA
| | - Stefano Morotti
- Department of Pharmacology, University of California Davis,
451 Health Sciences Drive, Davis, CA 95616, USA
| | - Xianwei Zhang
- Department of Pharmacology, University of California Davis,
451 Health Sciences Drive, Davis, CA 95616, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, Faculty of Medicine, University
Duisburg-Essen, Essen, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and
Université de Montréal, Montréal, Canada
- Department of Molecular Physiology and Biophysics, Baylor College of
Medicine, Houston, TX, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis,
451 Health Sciences Drive, Davis, CA 95616, USA
| |
Collapse
|
4
|
Reyes Gaido OE, Nkashama LJ, Schole KL, Wang Q, Umapathi P, Mesubi OO, Konstantinidis K, Luczak ED, Anderson ME. CaMKII as a Therapeutic Target in Cardiovascular Disease. Annu Rev Pharmacol Toxicol 2023; 63:249-272. [PMID: 35973713 PMCID: PMC11019858 DOI: 10.1146/annurev-pharmtox-051421-111814] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
CaMKII (the multifunctional Ca2+ and calmodulin-dependent protein kinase II) is a highly validated signal for promoting a variety of common diseases, particularly in the cardiovascular system. Despite substantial amounts of convincing preclinical data, CaMKII inhibitors have yet to emerge in clinical practice. Therapeutic inhibition is challenged by the diversity of CaMKII isoforms and splice variants and by physiological CaMKII activity that contributes to learning and memory. Thus, uncoupling the harmful and beneficial aspects of CaMKII will be paramount to developing effective therapies. In the last decade, several targeting strategies have emerged, including small molecules, peptides, and nucleotides, which hold promise in discriminating pathological from physiological CaMKII activity. Here we review the cellular and molecular biology of CaMKII, discuss its role in physiological and pathological signaling, and consider new findings and approaches for developing CaMKII therapeutics.
Collapse
Affiliation(s)
- Oscar E Reyes Gaido
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
| | | | - Kate L Schole
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
| | - Qinchuan Wang
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
| | - Priya Umapathi
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
| | - Olurotimi O Mesubi
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
| | - Klitos Konstantinidis
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
| | - Elizabeth D Luczak
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
| | - Mark E Anderson
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
- Departments of Physiology and Genetic Medicine and Program in Cellular and Molecular Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Takla M, Edling CE, Zhang K, Saadeh K, Tse G, Salvage SC, Huang CL, Jeevaratnam K. Transcriptional profiles of genes related to electrophysiological function in Scn5a +/- murine hearts. Physiol Rep 2021; 9:e15043. [PMID: 34617689 PMCID: PMC8495800 DOI: 10.14814/phy2.15043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 11/24/2022] Open
Abstract
The Scn5a gene encodes the major pore-forming Nav 1.5 (α) subunit, of the voltage-gated Na+ channel in cardiomyocytes. The key role of Nav 1.5 in action potential initiation and propagation in both atria and ventricles predisposes organisms lacking Scn5a or carrying Scn5a mutations to cardiac arrhythmogenesis. Loss-of-function Nav 1.5 genetic abnormalities account for many cases of the human arrhythmic disorder Brugada syndrome (BrS) and related conduction disorders. A murine model with a heterozygous Scn5a deletion recapitulates many electrophysiological phenotypes of BrS. This study examines the relationships between its Scn5a+/- genotype, resulting transcriptional changes, and the consequent phenotypic presentations of BrS. Of 62 selected protein-coding genes related to cardiomyocyte electrophysiological or homeostatic function, concentrations of mRNA transcribed from 15 differed significantly from wild type (WT). Despite halving apparent ventricular Scn5a transcription heterozygous deletion did not significantly downregulate its atrial expression, raising possibilities of atria-specific feedback mechanisms. Most of the remaining 14 genes whose expression differed significantly between WT and Scn5a+/- animals involved Ca2+ homeostasis specifically in atrial tissue, with no overlap with any ventricular changes. All statistically significant changes in expression were upregulations in the atria and downregulations in the ventricles. This investigation demonstrates the value of future experiments exploring for and clarifying links between transcriptional control of Scn5a and of genes whose protein products coordinate Ca2+ regulation and examining their possible roles in BrS.
Collapse
Affiliation(s)
- Michael Takla
- Faculty of Health and Medical ScienceUniversity of SurreyGuildfordUK
- Christ’s CollegeUniversity of CambridgeCambridgeUK
| | | | - Kevin Zhang
- Faculty of Health and Medical ScienceUniversity of SurreyGuildfordUK
- School of MedicineImperial College LondonLondonUK
| | - Khalil Saadeh
- Faculty of Health and Medical ScienceUniversity of SurreyGuildfordUK
- Clinical SchoolUniversity of CambridgeCambridgeUK
| | - Gary Tse
- Faculty of Health and Medical ScienceUniversity of SurreyGuildfordUK
- Second Hospital of Tianjin Medical UniversityTianjinChina
| | | | - Christopher L.‐H. Huang
- Faculty of Health and Medical ScienceUniversity of SurreyGuildfordUK
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | | |
Collapse
|
6
|
Implications of SGLT Inhibition on Redox Signalling in Atrial Fibrillation. Int J Mol Sci 2021; 22:ijms22115937. [PMID: 34073033 PMCID: PMC8198069 DOI: 10.3390/ijms22115937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained (atrial) arrhythmia, a considerable global health burden and often associated with heart failure. Perturbations of redox signalling in cardiomyocytes provide a cellular substrate for the manifestation and maintenance of atrial arrhythmias. Several clinical trials have shown that treatment with sodium-glucose linked transporter inhibitors (SGLTi) improves mortality and hospitalisation in heart failure patients independent of the presence of diabetes. Post hoc analysis of the DECLARE-TIMI 58 trial showed a 19% reduction in AF in patients with diabetes mellitus (hazard ratio, 0.81 (95% confidence interval: 0.68-0.95), n = 17.160) upon treatment with SGLTi, regardless of pre-existing AF or heart failure and independent from blood pressure or renal function. Accordingly, ongoing experimental work suggests that SGLTi not only positively impact heart failure but also counteract cellular ROS production in cardiomyocytes, thereby potentially altering atrial remodelling and reducing AF burden. In this article, we review recent studies investigating the effect of SGLTi on cellular processes closely interlinked with redox balance and their potential effects on the onset and progression of AF. Despite promising insight into SGLTi effect on Ca2+ cycling, Na+ balance, inflammatory and fibrotic signalling, mitochondrial function and energy balance and their potential effect on AF, the data are not yet conclusive and the importance of individual pathways for human AF remains to be established. Lastly, an overview of clinical studies investigating SGLTi in the context of AF is provided.
Collapse
|
7
|
Wang W, Shen W, Zhang S, Luo G, Wang K, Xu Y, Zhang H. The Role of CaMKII Overexpression and Oxidation in Atrial Fibrillation-A Simulation Study. Front Physiol 2021; 11:607809. [PMID: 33391023 PMCID: PMC7775483 DOI: 10.3389/fphys.2020.607809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/19/2020] [Indexed: 11/13/2022] Open
Abstract
This simulation study aims to investigate how the Calcium/calmodulin-dependent protein kinase II (CaMKII) overexpression and oxidation would influence the cardiac electrophysiological behavior and its arrhythmogenic mechanism in atria. A new-built CaMKII oxidation module and a refitted CaMKII overexpression module were integrated into a mouse atrial cell model for analyzing cardiac electrophysiological variations in action potential (AP) characteristics and intracellular Ca2+ cycling under different conditions. Simulation results showed that CaMKII overexpression significantly increased the phosphorylation level of its downstream target proteins, resulting in prolonged AP and smaller calcium transient amplitude, and impaired the Ca2+ cycling stability. These effects were exacerbated by extra reactive oxygen species, which oxidized CaMKII and led to continuous high CaMKII activation in both systolic and diastolic phases. Intracellular Ca2+ depletion and sustained delayed afterdepolarizations (DADs) were observed under co-existing CaMKII overexpression and oxidation, which could be effectively reversed by clamping the phosphorylation level of ryanodine receptor (RyR). We also found that the stability of RyR release highly depended on a delicate balance between the level of RyR phosphorylation and sarcoplasmic reticulum Ca2+ concentration, which was closely related to the genesis of DADs. We concluded that the CaMKII overexpression and oxidation have a synergistic role in increasing the activity of CaMKII, and the unstable RyR may be the key downstream target in the CaMKII arrhythmogenic mechanism. Our simulation provides detailed mechanistic insights into the arrhythmogenic effect of CaMKII overexpression and oxidation, which suggests CaMKII as a promising target in the therapy of atrial fibrillation.
Collapse
Affiliation(s)
- Wei Wang
- Shenzhen Key Laboratory of Visual Object Detection and Recognition, Harbin Institute of Technology, Shenzhen, China.,Peng Cheng Lab, Shenzhen, China
| | - Weijian Shen
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
| | - Shanzhuo Zhang
- Department of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Gongning Luo
- Department of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Kuanquan Wang
- Department of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yong Xu
- Shenzhen Key Laboratory of Visual Object Detection and Recognition, Harbin Institute of Technology, Shenzhen, China
| | - Henggui Zhang
- Peng Cheng Lab, Shenzhen, China.,Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
8
|
Tinaquero D, Crespo-García T, Utrilla RG, Nieto-Marín P, González-Guerra A, Rubio-Alarcón M, Cámara-Checa A, Dago M, Matamoros M, Pérez-Hernández M, Tamargo M, Cebrián J, Jalife J, Tamargo J, Bernal JA, Caballero R, Delpón E. The p.P888L SAP97 polymorphism increases the transient outward current (I to,f) and abbreviates the action potential duration and the QT interval. Sci Rep 2020; 10:10707. [PMID: 32612162 PMCID: PMC7329876 DOI: 10.1038/s41598-020-67109-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 06/01/2020] [Indexed: 11/09/2022] Open
Abstract
Synapse-Associated Protein 97 (SAP97) is an anchoring protein that in cardiomyocytes targets to the membrane and regulates Na+ and K+ channels. Here we compared the electrophysiological effects of native (WT) and p.P888L SAP97, a common polymorphism. Currents were recorded in cardiomyocytes from mice trans-expressing human WT or p.P888L SAP97 and in Chinese hamster ovary (CHO)-transfected cells. The duration of the action potentials and the QT interval were significantly shorter in p.P888L-SAP97 than in WT-SAP97 mice. Compared to WT, p.P888L SAP97 significantly increased the charge of the Ca-independent transient outward (Ito,f) current in cardiomyocytes and the charge crossing Kv4.3 channels in CHO cells by slowing Kv4.3 inactivation kinetics. Silencing or inhibiting Ca/calmodulin kinase II (CaMKII) abolished the p.P888L-induced Kv4.3 charge increase, which was also precluded in channels (p.S550A Kv4.3) in which the CaMKII-phosphorylation is prevented. Computational protein-protein docking predicted that p.P888L SAP97 is more likely to form a complex with CaMKII than WT. The Na+ current and the current generated by Kv1.5 channels increased similarly in WT-SAP97 and p.P888L-SAP97 cardiomyocytes, while the inward rectifier current increased in WT-SAP97 but not in p.P888L-SAP97 cardiomyocytes. The p.P888L SAP97 polymorphism increases the Ito,f, a CaMKII-dependent effect that may increase the risk of arrhythmias.
Collapse
Affiliation(s)
- David Tinaquero
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | - Teresa Crespo-García
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | - Raquel G Utrilla
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | - Paloma Nieto-Marín
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | | | - Marcos Rubio-Alarcón
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | - Anabel Cámara-Checa
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | - María Dago
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | - Marcos Matamoros
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | - Marta Pérez-Hernández
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | - María Tamargo
- Cardiology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | - Jorge Cebrián
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | - José Jalife
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Department of Internal Medicine/Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Juan Tamargo
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | | | - Ricardo Caballero
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain.
| | - Eva Delpón
- Department of Pharmacology and Toxicology. School of Medicine. Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón. CIBERCV, Madrid, Spain
| | | |
Collapse
|
9
|
Tracy E, Rowe G, LeBlanc AJ. Cardiac tissue remodeling in healthy aging: the road to pathology. Am J Physiol Cell Physiol 2020; 319:C166-C182. [PMID: 32432929 DOI: 10.1152/ajpcell.00021.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review aims to highlight the normal physiological remodeling that occurs in healthy aging hearts, including changes that occur in contractility, conduction, valve function, large and small coronary vessels, and the extracellular matrix. These "normal" age-related changes serve as the foundation that supports decreased plasticity and limited ability for tissue remodeling during pathophysiological states such as myocardial ischemia and heart failure. This review will identify populations at greater risk for poor tissue remodeling in advanced age along with present and future therapeutic strategies that may ameliorate dysfunctional tissue remodeling in aging hearts.
Collapse
Affiliation(s)
- Evan Tracy
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| | - Gabrielle Rowe
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| | - Amanda J LeBlanc
- Department of Physiology, Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| |
Collapse
|
10
|
Takla M, Huang CLH, Jeevaratnam K. The cardiac CaMKII-Na v1.5 relationship: From physiology to pathology. J Mol Cell Cardiol 2020; 139:190-200. [PMID: 31958466 DOI: 10.1016/j.yjmcc.2019.12.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/20/2019] [Accepted: 12/30/2019] [Indexed: 12/19/2022]
Abstract
The SCN5A gene encodes Nav1.5, which, as the cardiac voltage-gated Na+ channel's pore-forming α subunit, is crucial for the initiation and propagation of atrial and ventricular action potentials. The arrhythmogenic propensity of inherited SCN5A mutations implicates the Na+ channel in determining cardiomyocyte excitability under normal conditions. Cytosolic kinases have long been known to alter the kinetic profile of Nav1.5 inactivation via phosphorylation of specific residues. Recent substantiation of both the role of calmodulin-dependent kinase II (CaMKII) in modulating the properties of the Nav1.5 inactivation gate and the significant rise in oxidation-dependent autonomous CaMKII activity in structural heart disease has raised the possibility of a novel pathway for acquired arrhythmias - the CaMKII-Nav1.5 relationship. The aim of this review is to: (1) outline the relationship's translation from physiological adaptation to pathological vicious circle; and (2) discuss the relative merits of each of its components as pharmacological targets.
Collapse
Affiliation(s)
- Michael Takla
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, United Kingdom
| | - Christopher L-H Huang
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, United Kingdom; Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, United Kingdom; Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom.
| |
Collapse
|
11
|
The KN-93 Molecule Inhibits Calcium/Calmodulin-Dependent Protein Kinase II (CaMKII) Activity by Binding to Ca 2+/CaM. J Mol Biol 2019; 431:1440-1459. [PMID: 30753871 DOI: 10.1016/j.jmb.2019.02.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/14/2018] [Accepted: 02/04/2019] [Indexed: 12/22/2022]
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) is a multifunctional serine/threonine protein kinase that transmits calcium signals in various cellular processes. CaMKII is activated by calcium-bound calmodulin (Ca2+/CaM) through a direct binding mechanism involving a regulatory C-terminal α-helix in CaMKII. The Ca2+/CaM binding triggers transphosphorylation of critical threonine residues proximal to the CaM-binding site leading to the autoactivated state of CaMKII. The demonstration of its critical roles in pathophysiological processes has elevated CaMKII to a key target in the management of numerous diseases. The molecule KN-93 is the most widely used inhibitor for studying the cellular and in vivo functions of CaMKII. It is widely believed that KN-93 binds directly to CaMKII, thus preventing kinase activation by competing with Ca2+/CaM. Herein, we employed surface plasmon resonance, NMR, and isothermal titration calorimetry to characterize this presumed interaction. Our results revealed that KN-93 binds directly to Ca2+/CaM and not to CaMKII. This binding would disrupt the ability of Ca2+/CaM to interact with CaMKII, effectively inhibiting CaMKII activation. Our findings also indicated that KN-93 can specifically compete with a CaMKIIδ-derived peptide for binding to Ca2+/CaM. As indicated by the surface plasmon resonance and isothermal titration calorimetry data, apparently at least two KN-93 molecules can bind to Ca2+/CaM. Our findings provide new insight into how in vitro and in vivo data obtained with KN-93 should be interpreted. They further suggest that other Ca2+/CaM-dependent, non-CaMKII activities should be considered in KN-93-based mechanism-of-action studies and drug discovery efforts.
Collapse
|
12
|
Yoo S, Aistrup G, Shiferaw Y, Ng J, Mohler PJ, Hund TJ, Waugh T, Browne S, Gussak G, Gilani M, Knight BP, Passman R, Goldberger JJ, Wasserstrom JA, Arora R. Oxidative stress creates a unique, CaMKII-mediated substrate for atrial fibrillation in heart failure. JCI Insight 2018; 3:120728. [PMID: 30385719 DOI: 10.1172/jci.insight.120728] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 09/27/2018] [Indexed: 12/31/2022] Open
Abstract
The precise mechanisms by which oxidative stress (OS) causes atrial fibrillation (AF) are not known. Since AF frequently originates in the posterior left atrium (PLA), we hypothesized that OS, via calmodulin-dependent protein kinase II (CaMKII) signaling, creates a fertile substrate in the PLA for triggered activity and reentry. In a canine heart failure (HF) model, OS generation and oxidized-CaMKII-induced (Ox-CaMKII-induced) RyR2 and Nav1.5 signaling were increased preferentially in the PLA (compared with left atrial appendage). Triggered Ca2+ waves (TCWs) in HF PLA myocytes were particularly sensitive to acute ROS inhibition. Computational modeling confirmed a direct relationship between OS/CaMKII signaling and TCW generation. CaMKII phosphorylated Nav1.5 (CaMKII-p-Nav1.5 [S571]) was located preferentially at the intercalated disc (ID), being nearly absent at the lateral membrane. Furthermore, a decrease in ankyrin-G (AnkG) in HF led to patchy dropout of CaMKII-p-Nav1.5 at the ID, causing its distribution to become spatially heterogeneous; this corresponded to preferential slowing and inhomogeneity of conduction noted in the HF PLA. Computational modeling illustrated how conduction slowing (e.g., due to increase in CaMKII-p-Nav1.5) interacts with fibrosis to cause reentry in the PLA. We conclude that OS via CaMKII leads to substrate for triggered activity and reentry in HF PLA by mechanisms independent of but complementary to fibrosis.
Collapse
Affiliation(s)
- Shin Yoo
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Gary Aistrup
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yohannes Shiferaw
- Department of Physics, California State University, Northridge, California, USA
| | - Jason Ng
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Peter J Mohler
- Dorothy M. Davis Heart and Lung Research Institute, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Thomas J Hund
- Dorothy M. Davis Heart and Lung Research Institute, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Trent Waugh
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Suzanne Browne
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Georg Gussak
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mehul Gilani
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bradley P Knight
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rod Passman
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jeffrey J Goldberger
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - J Andrew Wasserstrom
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rishi Arora
- Feinberg Cardiovascular Research and Renal Institute, Northwestern University-Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
13
|
Fan X, Yu Y, Lan H, Ou X, Yang L, Li T, Cao J, Zeng X, Li M. Ca2+/Calmodulin-Dependent Protein Kinase II (CaMKII) Increases Small-Conductance Ca2+-Activated K+ Current in Patients with Chronic Atrial Fibrillation. Med Sci Monit 2018; 24:3011-3023. [PMID: 29737974 PMCID: PMC5963316 DOI: 10.12659/msm.909684] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Increased small-conductance Ca2+-activated K+ current (SK), abnormal intracellular Ca2+ handling, and enhanced expression and activity of Ca2+/calmodulin-dependent protein kinase II (CaMKII) have been found in clinical and/or experimental models of atrial fibrillation (AF), but the cumulative effect of these phenomena and their mechanisms in AF are still unclear. This study aimed to test the hypothesis that CaMKII increases SK current in human chronic AF. Material/Methods Right atrial appendage tissues from patients with either sinus rhythm (SR) or AF and neonatal rat atrial myocytes were used. Patch clamp, qRT-PCR, and Western blotting techniques were used to perform the study. Results Compared to SR, the apamin-sensitive SK current (IKAS) was significantly increased, but the mRNA and protein levels of SK1, SK2, and SK3 were significantly decreased. In AF, the steady-state Ca2+ response curve of IKAS was shifted leftward and the [Ca2+]i level was significantly increased. CaMKII inhibitors (KN-93 or autocamtide-2-related inhibitory peptide (AIP)) reduced the IKAS in both AF and SR. The inhibitory effect of KN-93 or AIP on IKAS was greater in AF than in SR. The expression levels of calmodulin, CaMKII, and autophosphorylated CaMKII at Thr287 (but not at Thr286) were significantly increased in AF. Furthermore, KN-93 inhibited the expression of (Thr287)p-CaMKII and SK2 in neonatal rat atrial myocytes. Conclusions SK current is increased via the enhanced activation of CaMKII in patients with AF. This finding may explain the difference between SK current and channels expression in AF, and thus may provide a therapeutic target for AF.
Collapse
Affiliation(s)
- Xuehui Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Yiyan Yu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Huan Lan
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Xianhong Ou
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Lijie Yang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Jiming Cao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Xiaorong Zeng
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| | - Miaoling Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China (mainland)
| |
Collapse
|
14
|
Grandi E, Dobrev D. Non-ion channel therapeutics for heart failure and atrial fibrillation: Are CaMKII inhibitors ready for clinical use? J Mol Cell Cardiol 2017; 121:300-303. [PMID: 29079077 DOI: 10.1016/j.yjmcc.2017.10.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/22/2017] [Indexed: 01/17/2023]
Abstract
The Ca2+-calmodulin dependent protein kinase II (CaMKII) is an established central mediator of electrophysiological and contractile responses to cardiac stress, and its hyper-activation in cardiac diseases has been linked to heart failure (HF) and arrhythmia. Here we summarize the evidence supporting the role of CaMKII as a critical nodal point for therapeutic intervention against HF and atrial and ventricular tachyarrhythmias. Targeting of CaMKII in heart with inhibitors possessing appropriate selectivity might represent a novel therapeutic approach for HF and arrhythmias.
Collapse
Affiliation(s)
- Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, CA, USA.
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
15
|
Ellinwood N, Dobrev D, Morotti S, Grandi E. Revealing kinetics and state-dependent binding properties of I Kur-targeting drugs that maximize atrial fibrillation selectivity. CHAOS (WOODBURY, N.Y.) 2017; 27:093918. [PMID: 28964116 PMCID: PMC5573366 DOI: 10.1063/1.5000226] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 07/06/2017] [Indexed: 06/07/2023]
Abstract
The KV1.5 potassium channel, which underlies the ultra-rapid delayed-rectifier current (IKur) and is predominantly expressed in atria vs. ventricles, has emerged as a promising target to treat atrial fibrillation (AF). However, while numerous KV1.5-selective compounds have been screened, characterized, and tested in various animal models of AF, evidence of antiarrhythmic efficacy in humans is still lacking. Moreover, current guidelines for pre-clinical assessment of candidate drugs heavily rely on steady-state concentration-response curves or IC50 values, which can overlook adverse cardiotoxic effects. We sought to investigate the effects of kinetics and state-dependent binding of IKur-targeting drugs on atrial electrophysiology in silico and reveal the ideal properties of IKur blockers that maximize anti-AF efficacy and minimize pro-arrhythmic risk. To this aim, we developed a new Markov model of IKur that describes KV1.5 gating based on experimental voltage-clamp data in atrial myocytes from patient right-atrial samples in normal sinus rhythm. We extended the IKur formulation to account for state-specificity and kinetics of KV1.5-drug interactions and incorporated it into our human atrial cell model. We simulated 1- and 3-Hz pacing protocols in drug-free conditions and with a [drug] equal to the IC50 value. The effects of binding and unbinding kinetics were determined by examining permutations of the forward (kon) and reverse (koff) binding rates to the closed, open, and inactivated states of the KV1.5 channel. We identified a subset of ideal drugs exhibiting anti-AF electrophysiological parameter changes at fast pacing rates (effective refractory period prolongation), while having little effect on normal sinus rhythm (limited action potential prolongation). Our results highlight that accurately accounting for channel interactions with drugs, including kinetics and state-dependent binding, is critical for developing safer and more effective pharmacological anti-AF options.
Collapse
Affiliation(s)
- Nicholas Ellinwood
- Department of Pharmacology, University of California Davis, Davis, California 95616, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Stefano Morotti
- Department of Pharmacology, University of California Davis, Davis, California 95616, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, California 95616, USA
| |
Collapse
|
16
|
Groen C, Bähring R. Modulation of human Kv4.3/KChIP2 channel inactivation kinetics by cytoplasmic Ca 2. Pflugers Arch 2017; 469:1457-1470. [PMID: 28735419 DOI: 10.1007/s00424-017-2039-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/11/2017] [Accepted: 07/13/2017] [Indexed: 10/19/2022]
Abstract
The transient outward current (I to) in the human heart is mediated by Kv4.3 channels complexed with Kv channel interacting protein (KChIP) 2, a cytoplasmic Ca2+-binding EF-hand protein known to modulate Kv4.3 inactivation gating upon heterologous co-expression. We studied Kv4.3 channels co-expressed with wild-type (wt) or EF-hand-mutated (ΔEF) KChIP2 in human embryonic kidney (HEK) 293 cells. Co-expression took place in the absence or presence of BAPTA-AM, and macroscopic currents were recorded in the whole-cell patch-clamp configuration with different free Ca2+ concentrations in the patch-pipette. Our data indicate that Ca2+ is not necessary for Kv4.3/KChIP2 complex formation. The Kv4.3/KChIP2-mediated current decay was faster and the recovery of Kv4.3/KChIP2 channels from inactivation slower with 50 μM Ca2+ than with BAPTA (nominal Ca2+-free) in the patch-pipette. The apparent Ca2+-mediated slowing of recovery kinetics was still observed when EF-hand 4 of KChIP2 was mutated (ΔEF4) but not when EF-hand 2 (ΔEF2) was mutated, and turned into a Ca2+-mediated acceleration of recovery kinetics when EF-hand 3 (ΔEF3) was mutated. In the presence of the Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitor KN-93 cytoplasmic Ca2+ (50 μM) induced an acceleration of Kv4.3/KChIP2 recovery kinetics, which was still observed when EF-hand 2 was mutated (ΔEF2) but not when EF-hand 3 (ΔEF3) or EF-hand 4 (ΔEF4) was mutated. Our results support the notion that binding of Ca2+ to KChIP2 EF-hands can acutely modulate Kv4.3/KChIP2 channel inactivation gating, but the Ca2+-dependent gating modulation depends on CaMKII action. Our findings speak for an acute modulation of I to kinetics and frequency-dependent I to availability in cardiomyocytes under conditions with elevated Ca2+ levels and CaMKII activity.
Collapse
Affiliation(s)
- Christiane Groen
- Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Robert Bähring
- Institut für Zelluläre und Integrative Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| |
Collapse
|
17
|
Neef S, Mann C, Zwenger A, Dybkova N, Maier LS. Reduction of SR Ca2+ leak and arrhythmogenic cellular correlates by SMP-114, a novel CaMKII inhibitor with oral bioavailability. Basic Res Cardiol 2017; 112:45. [DOI: 10.1007/s00395-017-0637-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/09/2017] [Indexed: 01/13/2023]
|
18
|
Greiser M. Calcium signalling silencing in atrial fibrillation. J Physiol 2017; 595:4009-4017. [PMID: 28332202 DOI: 10.1113/jp273045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/05/2017] [Indexed: 01/19/2023] Open
Abstract
Subcellular calcium signalling silencing is a novel and distinct cellular and molecular adaptive response to rapid cardiac activation. Calcium signalling silencing develops during short-term sustained rapid atrial activation as seen clinically during paroxysmal atrial fibrillation (AF). It is the first 'anti-arrhythmic' adaptive response in the setting of AF and appears to counteract the maladaptive changes that lead to intracellular Ca2+ signalling instability and Ca2+ -based arrhythmogenicity. Calcium signalling silencing results in a failed propagation of the [Ca2+ ]i signal to the myocyte centre both in patients with AF and in a rabbit model. This adaptive mechanism leads to a substantial reduction in the expression levels of calcium release channels (ryanodine receptors, RyR2) in the sarcoplasmic reticulum, and the frequency of Ca2+ sparks and arrhythmogenic Ca2+ waves remains low. Less Ca2+ release per [Ca2+ ]i transient, increased fast Ca2+ buffering strength, shortened action potentials and reduced L-type Ca2+ current contribute to a substantial reduction of intracellular [Na+ ]. These features of Ca2+ signalling silencing are distinct and in contrast to the changes attributed to Ca2+ -based arrhythmogenicity. Some features of Ca2+ signalling silencing prevail in human AF suggesting that the Ca2+ signalling 'phenotype' in AF is a sum of Ca2+ stabilizing (Ca2+ signalling silencing) and Ca2+ destabilizing (arrhythmogenic unstable Ca2+ signalling) factors. Calcium signalling silencing is a part of the mechanisms that contribute to the natural progression of AF and may limit the role of Ca2+ -based arrhythmogenicity after the onset of AF.
Collapse
Affiliation(s)
- Maura Greiser
- Center for Biomedical Engineering and Technology and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Heijman J, Ghezelbash S, Wehrens XHT, Dobrev D. Serine/Threonine Phosphatases in Atrial Fibrillation. J Mol Cell Cardiol 2017; 103:110-120. [PMID: 28077320 DOI: 10.1016/j.yjmcc.2016.12.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/15/2016] [Accepted: 12/20/2016] [Indexed: 12/19/2022]
Abstract
Serine/threonine protein phosphatases control dephosphorylation of numerous cardiac proteins, including a variety of ion channels and calcium-handling proteins, thereby providing precise post-translational regulation of cardiac electrophysiology and function. Accordingly, dysfunction of this regulation can contribute to the initiation, maintenance and progression of cardiac arrhythmias. Atrial fibrillation (AF) is the most common heart rhythm disorder and is characterized by electrical, autonomic, calcium-handling, contractile, and structural remodeling, which include, among other things, changes in the phosphorylation status of a wide range of proteins. Here, we review AF-associated alterations in the phosphorylation of atrial ion channels, calcium-handling and contractile proteins, and their role in AF-pathophysiology. We highlight the mechanisms controlling the phosphorylation of these proteins and focus on the role of altered dephosphorylation via local type-1, type-2A and type-2B phosphatases (PP1, PP2A, and PP2B, also known as calcineurin, respectively). Finally, we discuss the challenges for phosphatase research, potential therapeutic significance of altered phosphatase-mediated protein dephosphorylation in AF, as well as future directions.
Collapse
Affiliation(s)
- Jordi Heijman
- Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Shokoufeh Ghezelbash
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics, Department of Medicine (Cardiology), Pediatrics, Baylor College of Medicine, Houston, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
20
|
Role of CaMKII and PKA in Early Afterdepolarization of Human Ventricular Myocardium Cell: A Computational Model Study. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2016; 2016:4576313. [PMID: 28053652 PMCID: PMC5178856 DOI: 10.1155/2016/4576313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 10/31/2016] [Indexed: 11/30/2022]
Abstract
Early afterdepolarization (EAD) plays an important role in arrhythmogenesis. Many experimental studies have reported that Ca2+/calmodulin-dependent protein kinase II (CaMKII) and β-adrenergic signaling pathway are two important regulators. In this study, we developed a modified computational model of human ventricular myocyte to investigate the combined role of CaMKII and β-adrenergic signaling pathway on the occurrence of EADs. Our simulation results showed that (1) CaMKII overexpression facilitates EADs through the prolongation of late sodium current's (INaL) deactivation progress; (2) the combined effect of CaMKII overexpression and activation of β-adrenergic signaling pathway further increases the risk of EADs, where EADs could occur at shorter cycle length (2000 ms versus 4000 ms) and lower rapid delayed rectifier K+ current (IKr) blockage (77% versus 85%). In summary, this study computationally demonstrated the combined role of CaMKII and β-adrenergic signaling pathway on the occurrence of EADs, which could be useful for searching for therapy strategies to treat EADs related arrhythmogenesis.
Collapse
|
21
|
Grandi E, Maleckar MM. Anti-arrhythmic strategies for atrial fibrillation: The role of computational modeling in discovery, development, and optimization. Pharmacol Ther 2016; 168:126-142. [PMID: 27612549 DOI: 10.1016/j.pharmthera.2016.09.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Atrial fibrillation (AF), the most common cardiac arrhythmia, is associated with increased risk of cerebrovascular stroke, and with several other pathologies, including heart failure. Current therapies for AF are targeted at reducing risk of stroke (anticoagulation) and tachycardia-induced cardiomyopathy (rate or rhythm control). Rate control, typically achieved by atrioventricular nodal blocking drugs, is often insufficient to alleviate symptoms. Rhythm control approaches include antiarrhythmic drugs, electrical cardioversion, and ablation strategies. Here, we offer several examples of how computational modeling can provide a quantitative framework for integrating multiscale data to: (a) gain insight into multiscale mechanisms of AF; (b) identify and test pharmacological and electrical therapy and interventions; and (c) support clinical decisions. We review how modeling approaches have evolved and contributed to the research pipeline and preclinical development and discuss future directions and challenges in the field.
Collapse
Affiliation(s)
- Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, USA.
| | | |
Collapse
|
22
|
Yang Z, Wang L, Wang X. Matrine induces the hepatic differentiation of WB-F344 rat hepatic progenitor cells and inhibits Jagged 1/HES1 signaling. Mol Med Rep 2016; 14:3841-7. [DOI: 10.3892/mmr.2016.5668] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 07/13/2016] [Indexed: 11/06/2022] Open
|
23
|
Mason FE, Sossalla S. The Significance of the Late Na+ Current for Arrhythmia Induction and the Therapeutic Antiarrhythmic Potential of Ranolazine. J Cardiovasc Pharmacol Ther 2016; 22:40-50. [DOI: 10.1177/1074248416644989] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The purpose of this article is to review the basis of arrhythmogenesis, the functional and clinical role of the late Na current, and its therapeutic inhibition. Under pathological conditions such as ischemia and heart failure this current is abnormally enhanced and influences cellular electrophysiology as a proarrhythmic substrate in myocardial pathology. Ranolazine the only approved late Na current blocker has been demonstrated to produce antiarrhythmic effects in the atria and the ventricle. We summarize recent experimental and clinical studies of ranolazine and other experimental late Na current blockers and discuss the significance of the available data.
Collapse
Affiliation(s)
- Fleur E. Mason
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany
| | - Samuel Sossalla
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany
- Department of Internal Medicine III (Cardiology and Angiology), University Hospital Schleswig-Holstein, Kiel, Germany
- German Centre for Cardiovascular Research (DZHK), Göttingen & Kiel, Germany
| |
Collapse
|
24
|
Han P, Lloyd T, Chen Z, Xiao Y. Proinflammatory Cytokines Regulate Cementogenic Differentiation of Periodontal Ligament Cells by Wnt/Ca(2+) Signaling Pathway. J Interferon Cytokine Res 2016; 36:328-37. [PMID: 27074616 DOI: 10.1089/jir.2015.0111] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Periodontal inflammation can inhibit cell differentiation of periodontal ligament cells (PDLCs), resulting in decreased bone/cementum regeneration ability. The Wnt signaling pathway, including canonical Wnt/β-catenin signaling and noncanonical Wnt/Ca(2+) signaling, plays essential roles in cell proliferation and differentiation during tooth development. However, little is still known whether noncanonical Wnt/Ca(2+) signaling cascade could regulate cementogenic/osteogenic differentiation capability of PDLCs within an inflammatory environment. Therefore, in this study, human PDLCs (hPDLCs) and their cementogenic differentiation potential were investigated in the presence of cytokines. The data demonstrated that both cytokines interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) inhibited cell proliferation, relative alkaline phosphatase activity, bone/cementum-related gene/protein expression, and canonical Wnt pathway-related gene/protein expression in hPDLCs. Interestingly, both cytokines upregulated the noncanonical Wnt/Ca(2+) signaling-related gene and protein expression in hPDLCs. When the Wnt/Ca(2+) pathway was blocked by Ca(2+)/calmodulin-dependent protein kinase II inhibitor KN93, even in the presence of IL-6 and TNF-α, cementogenesis could be stimulated in hPDLCs. Our data indicate that the Wnt/Ca(2+) pathway plays an inhibitory role on PDLC cementogenic differentiation in inflammatory microenvironments. Therefore, targeting the Wnt/Ca(2+) pathway may provide a novel therapeutic approach to improve periodontal regeneration for periodontal diseases.
Collapse
Affiliation(s)
- Pingping Han
- 1 Institute of Health and Biomedical Innovation, Queensland University of Technology , Brisbane, Australia .,2 Tissue Engineering and Microfluidic Laboratory, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland , Brisbane, Australia
| | - Tain Lloyd
- 3 School of Biomedical Sciences, The University of Queensland , Brisbane, Australia
| | - Zetao Chen
- 1 Institute of Health and Biomedical Innovation, Queensland University of Technology , Brisbane, Australia
| | - Yin Xiao
- 1 Institute of Health and Biomedical Innovation, Queensland University of Technology , Brisbane, Australia
| |
Collapse
|
25
|
Opacic D, van Bragt KA, Nasrallah HM, Schotten U, Verheule S. Atrial metabolism and tissue perfusion as determinants of electrical and structural remodelling in atrial fibrillation. Cardiovasc Res 2016; 109:527-41. [DOI: 10.1093/cvr/cvw007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/12/2016] [Indexed: 12/14/2022] Open
|
26
|
Mesubi OO, Anderson ME. Atrial remodelling in atrial fibrillation: CaMKII as a nodal proarrhythmic signal. Cardiovasc Res 2016; 109:542-57. [PMID: 26762270 DOI: 10.1093/cvr/cvw002] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/05/2016] [Indexed: 01/10/2023] Open
Abstract
CaMKII is a serine-threonine protein kinase that is abundant in myocardium. Emergent evidence suggests that CaMKII may play an important role in promoting atrial fibrillation (AF) by targeting a diverse array of proteins involved in membrane excitability, cell survival, calcium homeostasis, matrix remodelling, inflammation, and metabolism. Furthermore, CaMKII inhibition appears to protect against AF in animal models and correct proarrhythmic, defective intracellular Ca(2+) homeostasis in fibrillating human atrial cells. This review considers current concepts and evidence from animal and human studies on the role of CaMKII in AF.
Collapse
Affiliation(s)
- Olurotimi O Mesubi
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA Department of Medicine, The Johns Hopkins University School of Medicine, 1830 E. Monument Street, Suite 9026, Baltimore, MD 21287, USA
| | - Mark E Anderson
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA Department of Medicine, The Johns Hopkins University School of Medicine, 1830 E. Monument Street, Suite 9026, Baltimore, MD 21287, USA Department of Physiology and the Program in Cellular and Molecular Medicine, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
27
|
Hegyi B, Chen-Izu Y, Jian Z, Shimkunas R, Izu LT, Banyasz T. KN-93 inhibits IKr in mammalian cardiomyocytes. J Mol Cell Cardiol 2015; 89:173-6. [PMID: 26463508 PMCID: PMC4689637 DOI: 10.1016/j.yjmcc.2015.10.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 12/19/2022]
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) inhibitor KN-93 is widely used in multiple fields of cardiac research especially for studying the mechanisms of cardiomyopathy and cardiac arrhythmias. Whereas KN-93 is a potent inhibitor of CaMKII, several off-target effects have also been found in expression cell systems and smooth muscle cells, but there is no information on the KN93 side effects in mammalian ventricular myocytes. In this study we explore the effect of KN-93 on the rapid component of delayed rectifier potassium current (IKr) in the ventricular myocytes from rabbit and guinea pig hearts. Our data indicate that KN-93 exerts direct inhibitory effect on IKr that is not mediated via CaMKII. This off-target effect of KN93 should be taken into account when interpreting the data from using KN93 to investigate the role of CaMKII in cardiac function.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Ye Chen-Izu
- Department of Pharmacology, University of California, Davis, CA, USA; Department of Biomedical Engineering, University of California, Davis, CA USA; Department of Internal Medicine (Cardiology), University of California, Davis, CA USA
| | - Zhong Jian
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Rafael Shimkunas
- Department of Pharmacology, University of California, Davis, CA, USA; Department of Biomedical Engineering, University of California, Davis, CA USA
| | - Leighton T Izu
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Tamas Banyasz
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
28
|
Anderson ME. Oxidant stress promotes disease by activating CaMKII. J Mol Cell Cardiol 2015; 89:160-7. [PMID: 26475411 PMCID: PMC5075238 DOI: 10.1016/j.yjmcc.2015.10.014] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/09/2015] [Accepted: 10/10/2015] [Indexed: 12/31/2022]
Abstract
CaMKII is activated by oxidation of methionine residues residing in the regulatory domain. Oxidized CaMKII (ox-CaMKII) is now thought to participate in cardiovascular and pulmonary diseases and cancer. This invited review summarizes current evidence for the role of ox-CaMKII in disease, considers critical knowledge gaps and suggests new areas for inquiry.
Collapse
Affiliation(s)
- Mark E Anderson
- Johns Hopkins Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, United States.
| |
Collapse
|
29
|
Yang KC, Nerbonne JM. Mechanisms contributing to myocardial potassium channel diversity, regulation and remodeling. Trends Cardiovasc Med 2015; 26:209-18. [PMID: 26391345 DOI: 10.1016/j.tcm.2015.07.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/11/2015] [Accepted: 07/12/2015] [Indexed: 01/19/2023]
Abstract
In the mammalian heart, multiple types of K(+) channels contribute to the control of cardiac electrical and mechanical functioning through the regulation of resting membrane potentials, action potential waveforms and refractoriness. There are similarly vast arrays of K(+) channel pore-forming and accessory subunits that contribute to the generation of functional myocardial K(+) channel diversity. Maladaptive remodeling of K(+) channels associated with cardiac and systemic diseases results in impaired repolarization and increased propensity for arrhythmias. Here, we review the diverse transcriptional, post-transcriptional, post-translational, and epigenetic mechanisms contributing to regulating the expression, distribution, and remodeling of cardiac K(+) channels under physiological and pathological conditions.
Collapse
Affiliation(s)
- Kai-Chien Yang
- Department of Pharmacology, National Taiwan University, Taipei, Taiwan; Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jeanne M Nerbonne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO; Internal Medicine, Washington University School of Medicine, St. Louis, MO; Cardiovascular Division, Washington University School of Medicine, St. Louis, MO.
| |
Collapse
|
30
|
Abstract
Optimal cardiac function depends on proper timing of excitation and contraction in various regions of the heart, as well as on appropriate heart rate. This is accomplished via specialized electrical properties of various components of the system, including the sinoatrial node, atria, atrioventricular node, His-Purkinje system, and ventricles. Here we review the major regionally determined electrical properties of these cardiac regions and present the available data regarding the molecular and ionic bases of regional cardiac function and dysfunction. Understanding these differences is of fundamental importance for the investigation of arrhythmia mechanisms and pharmacotherapy.
Collapse
Affiliation(s)
- Daniel C Bartos
- Department of Pharmacology, University of California Davis, Davis, California, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, California, USA
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California Davis, Davis, California, USA
| |
Collapse
|
31
|
|
32
|
Greiser M, Kerfant BG, Williams GS, Voigt N, Harks E, Dibb KM, Giese A, Meszaros J, Verheule S, Ravens U, Allessie MA, Gammie JS, van der Velden J, Lederer WJ, Dobrev D, Schotten U. Tachycardia-induced silencing of subcellular Ca2+ signaling in atrial myocytes. J Clin Invest 2014; 124:4759-72. [PMID: 25329692 PMCID: PMC4347234 DOI: 10.1172/jci70102] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 08/28/2014] [Indexed: 01/06/2023] Open
Abstract
Atrial fibrillation (AF) is characterized by sustained high atrial activation rates and arrhythmogenic cellular Ca2+ signaling instability; however, it is not clear how a high atrial rate and Ca2+ instability may be related. Here, we characterized subcellular Ca2+ signaling after 5 days of high atrial rates in a rabbit model. While some changes were similar to those in persistent AF, we identified a distinct pattern of stabilized subcellular Ca2+ signaling. Ca2+ sparks, arrhythmogenic Ca2+ waves, sarcoplasmic reticulum (SR) Ca2+ leak, and SR Ca2+ content were largely unaltered. Based on computational analysis, these findings were consistent with a higher Ca2+ leak due to PKA-dependent phosphorylation of SR Ca2+ channels (RyR2s), fewer RyR2s, and smaller RyR2 clusters in the SR. We determined that less Ca2+ release per [Ca2+]i transient, increased Ca2+ buffering strength, shortened action potentials, and reduced L-type Ca2+ current contribute to a stunning reduction of intracellular Na+ concentration following rapid atrial pacing. In both patients with AF and in our rabbit model, this silencing led to failed propagation of the [Ca2+]i signal to the myocyte center. We conclude that sustained high atrial rates alone silence Ca2+ signaling and do not produce Ca2+ signaling instability, consistent with an adaptive molecular and cellular response to atrial tachycardia.
Collapse
Affiliation(s)
- Maura Greiser
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Benoît-Gilles Kerfant
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - George S.B. Williams
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Niels Voigt
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Erik Harks
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Katharine M. Dibb
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Anne Giese
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Janos Meszaros
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Sander Verheule
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Ursula Ravens
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Maurits A. Allessie
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - James S. Gammie
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - W. Jonathan Lederer
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Dobromir Dobrev
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| | - Ulrich Schotten
- Department of Physiology, Maastricht University, Maastricht, the Netherlands. Center for Biomedical Engineering and Technology, Laboratory of Molecular Cardiology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA. Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany. Unit of Cardiac Physiology, Manchester Academic Health Sciences Centre, Manchester, United Kingdom. Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany. Division of Cardiac Surgery, University of Maryland Medical Center, Baltimore, Maryland, USA. Laboratory for Physiology, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
33
|
Chae YJ, Choi BH, Choi JS, Hahn SJ. Block of Kv4.3 potassium channel by trifluoperazine independent of CaMKII. Neurosci Lett 2014; 578:159-64. [PMID: 24993295 DOI: 10.1016/j.neulet.2014.06.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/12/2014] [Accepted: 06/23/2014] [Indexed: 11/24/2022]
Abstract
Trifluoperazine, a trifluoro-methyl phenothiazine derivative, is widely used in the management of schizophrenia and related psychotic disorders. We studied the effects of trifluoperazine on Kv4.3 currents expressed in CHO cells using the whole-cell patch-clamp technique. Trifluoperazine blocked Kv4.3 in a concentration-dependent manner with an IC50 value of 8.0±0.4 μM and a Hill coefficient of 2.1±0.1. Trifluoperazine also accelerated the inactivation and activation (time-to-peak) kinetics in a concentration-dependent manner. The effects of trifluoperazine on Kv4.3 were completely reversible after washout. The effects of trifluoperazine were not affected by the pretreatment of KN93, which is another CaMKII inhibitor. In addition, the inclusion of CaMKII inhibitory peptide 281-309 in the pipette solution did not modify the effect of trifluoperazine on Kv4.3. Trifluoperazine shifted the activation curve of Kv4.3 in a hyperpolarizing direction but did not affect the slope factor. The block of Kv4.3 by trifluoperazine was voltage-dependent with a steep increase across the voltage range of channel activation. Voltage dependence was also observed over the full range of activation (δ=0.18). Trifluoperazine slowed the time course for recovery from inactivation of Kv4.3. Our results indicated that trifluoperazine blocked Kv4.3 by preferentially binding to the open state of the channel. This effect was not mediated via the inhibition of CaMKII activity.
Collapse
Affiliation(s)
- Yun Ju Chae
- Department of Physiology, Cell Death and Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea
| | - Bok Hee Choi
- Department of Pharmacology, Institute for Medical Science, Chonbuk National University Medical School, Jeonju, Jeonbuk 561-180, Republic of Korea
| | - Jin-Sung Choi
- College of Pharmacy, Integrated Research Institute of Pharmaceutical, The Catholic University of Korea, 43-1 Yeokgok 2-dong, Wonmi-gu, Bucheon, Gyeonggi-do, Republic of Korea
| | - Sang June Hahn
- Department of Physiology, Cell Death and Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea.
| |
Collapse
|
34
|
Affiliation(s)
- Richard Gordan
- Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School
| | | |
Collapse
|
35
|
Vincent KP, McCulloch AD, Edwards AG. Toward a hierarchy of mechanisms in CaMKII-mediated arrhythmia. Front Pharmacol 2014; 5:110. [PMID: 24994983 PMCID: PMC4062880 DOI: 10.3389/fphar.2014.00110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 04/25/2014] [Indexed: 12/16/2022] Open
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) activity has been shown to contribute to arrhythmogenesis in a remarkably broad range of cardiac pathologies. Several of these involve significant structural and electrophysiologic remodeling, whereas others are due to specific channelopathies, and are not typically associated with arrhythmogenic changes to protein expression or cellular and tissue structure. The ability of CaMKII to contribute to arrhythmia across such a broad range of phenotypes suggests one of two interpretations regarding the role of CaMKII in cardiac arrhythmia: (1) some CaMKII-dependent mechanism is a common driver of arrhythmia irrespective of the specific etiology of the disease, or (2) these different etiologies expose different mechanisms by which CaMKII is capable of promoting arrhythmia. In this review, we dissect the available mechanistic evidence to explore these two possibilities and discuss how the various molecular actions of CaMKII promote arrhythmia in different pathophysiologic contexts.
Collapse
Affiliation(s)
- Kevin P Vincent
- Department of Bioengineering, University of California San Diego La Jolla, CA, USA
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego La Jolla, CA, USA ; Department of Medicine, University of California San Diego La Jolla, CA, USA
| | - Andrew G Edwards
- Department of Bioengineering, University of California San Diego La Jolla, CA, USA ; Institute for Experimental Medicine, Oslo University Hospital Ullevål Oslo, Norway ; Simula Research Laboratory Lysaker, Norway
| |
Collapse
|
36
|
Heijman J, Voigt N, Wehrens XHT, Dobrev D. Calcium dysregulation in atrial fibrillation: the role of CaMKII. Front Pharmacol 2014; 5:30. [PMID: 24624086 PMCID: PMC3940963 DOI: 10.3389/fphar.2014.00030] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 02/15/2014] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most frequently encountered clinical arrhythmia and is associated with increased morbidity and mortality. Ectopic activity and reentry are considered major arrhythmogenic mechanisms contributing to the initiation and maintenance of AF. In addition, AF is self-reinforcing through progressive electrical and structural remodeling which stabilize the arrhythmia and make it more difficult to treat. Recent research has suggested an important role for Ca(2+)-dysregulation in AF. Ca(2+)-handling abnormalities may promote ectopic activity, conduction abnormalities facilitating reentry, and AF-related remodeling. In this review article, we summarize the Ca(2+)-handling derangements occurring in AF and discuss their impact on fundamental arrhythmogenic mechanisms. We focus in particular on the role of the multifunctional Ca(2+)/calmodulin-dependent protein kinase type-II (CaMKII), which acts as a major link between Ca(2+)-dysregulation and arrhythmogenesis. CaMKII expression and activity are increased in AF and promote arrhythmogenesis through phosphorylation of various targets involved in cardiac electrophysiology and excitation-contraction coupling. We discuss the implications for potential novel therapeutic strategies for AF based on CaMKII and Ca(2+)-handling abnormalities.
Collapse
Affiliation(s)
- Jordi Heijman
- Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen Essen, Germany
| | - Niels Voigt
- Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen Essen, Germany
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Departments of Molecular Physiology and Biophysics, and Medicine-Cardiology, Baylor College of Medicine Houston, TX, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen Essen, Germany
| |
Collapse
|
37
|
Mustroph J, Maier LS, Wagner S. CaMKII regulation of cardiac K channels. Front Pharmacol 2014; 5:20. [PMID: 24600393 PMCID: PMC3930912 DOI: 10.3389/fphar.2014.00020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 01/31/2014] [Indexed: 11/23/2022] Open
Abstract
Cardiac K channels are critical determinants of cardiac excitability. In hypertrophied and failing myocardium, alterations in the expression and activity of voltage-gated K channels are frequently observed and contribute to the increased propensity for life-threatening arrhythmias. Thus, understanding the mechanisms of disturbed K channel regulation in heart failure (HF) is of critical importance. Amongst others, Ca/calmodulin-dependent protein kinase II (CaMKII) has been identified as an important regulator of K channel activity. In human HF but also various animal models, increased CaMKII expression and activity has been linked to deteriorated contractile function and arrhythmias. This review will discuss the current knowledge about CaMKII regulation of several K channels, its influence on action potential properties, dispersion of repolarization, and arrhythmias with special focus on HF.
Collapse
Affiliation(s)
- Julian Mustroph
- Department of Cardiology, University Medical Center Göttingen Göttingen, Germany
| | - Lars S Maier
- Department of Cardiology, University Medical Center Göttingen Göttingen, Germany
| | - Stefan Wagner
- Department of Cardiology, University Medical Center Göttingen Göttingen, Germany
| |
Collapse
|
38
|
Martins RP, Kaur K, Hwang E, Ramirez RJ, Willis BC, Filgueiras-Rama D, Ennis SR, Takemoto Y, Ponce-Balbuena D, Zarzoso M, O'Connell RP, Musa H, Guerrero-Serna G, Avula UMR, Swartz MF, Bhushal S, Deo M, Pandit SV, Berenfeld O, Jalife J. Dominant frequency increase rate predicts transition from paroxysmal to long-term persistent atrial fibrillation. Circulation 2014; 129:1472-82. [PMID: 24463369 DOI: 10.1161/circulationaha.113.004742] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Little is known about the mechanisms underlying the transition from paroxysmal to persistent atrial fibrillation (AF). In an ovine model of long-standing persistent AF we tested the hypothesis that the rate of electric and structural remodeling, assessed by dominant frequency (DF) changes, determines the time at which AF becomes persistent. METHODS AND RESULTS Self-sustained AF was induced by atrial tachypacing. Seven sheep were euthanized 11.5±2.3 days after the transition to persistent AF and without reversal to sinus rhythm; 7 sheep were euthanized after 341.3±16.7 days of long-standing persistent AF. Seven sham-operated animals were in sinus rhythm for 1 year. DF was monitored continuously in each group. Real-time polymerase chain reaction, Western blotting, patch clamping, and histological analyses were used to determine the changes in functional ion channel expression and structural remodeling. Atrial dilatation, mitral valve regurgitation, myocyte hypertrophy, and atrial fibrosis occurred progressively and became statistically significant after the transition to persistent AF, with no evidence for left ventricular dysfunction. DF increased progressively during the paroxysmal-to-persistent AF transition and stabilized when AF became persistent. Importantly, the rate of DF increase correlated strongly with the time to persistent AF. Significant action potential duration abbreviation, secondary to functional ion channel protein expression changes (CaV1.2, NaV1.5, and KV4.2 decrease; Kir2.3 increase), was already present at the transition and persisted for 1 year of follow up. CONCLUSIONS In the sheep model of long-standing persistent AF, the rate of DF increase predicts the time at which AF stabilizes and becomes persistent, reflecting changes in action potential duration and densities of sodium, L-type calcium, and inward rectifier currents.
Collapse
Affiliation(s)
- Raphael P Martins
- Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI (R.P.M., K.K., E.H., R.J.R., B.C.W., D.F.-R., S.R.E., Y.T., D.P.-B., M.Z., R.P.O., H.M., G.G.-S., U.M.R.A., S.V.P., O.B., J.J.); Department of Surgery, University of Rochester, Rochester, NY (M.F.S.); and Department of Engineering, Norfolk State University, Norfolk, VA (S.B., M.D.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Purohit A, Rokita AG, Guan X, Chen B, Koval OM, Voigt N, Neef S, Sowa T, Gao Z, Luczak ED, Stefansdottir H, Behunin AC, Li N, El-Accaoui RN, Yang B, Swaminathan PD, Weiss RM, Wehrens XHT, Song LS, Dobrev D, Maier LS, Anderson ME. Oxidized Ca(2+)/calmodulin-dependent protein kinase II triggers atrial fibrillation. Circulation 2013; 128:1748-57. [PMID: 24030498 DOI: 10.1161/circulationaha.113.003313] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is a growing public health problem without adequate therapies. Angiotensin II and reactive oxygen species are validated risk factors for AF in patients, but the molecular pathways connecting reactive oxygen species and AF are unknown. The Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) has recently emerged as a reactive oxygen species-activated proarrhythmic signal, so we hypothesized that oxidized CaMKIIδ could contribute to AF. METHODS AND RESULTS We found that oxidized CaMKII was increased in atria from AF patients compared with patients in sinus rhythm and from mice infused with angiotensin II compared with mice infused with saline. Angiotensin II-treated mice had increased susceptibility to AF compared with saline-treated wild-type mice, establishing angiotensin II as a risk factor for AF in mice. Knock-in mice lacking critical oxidation sites in CaMKIIδ (MM-VV) and mice with myocardium-restricted transgenic overexpression of methionine sulfoxide reductase A, an enzyme that reduces oxidized CaMKII, were resistant to AF induction after angiotensin II infusion. CONCLUSIONS Our studies suggest that CaMKII is a molecular signal that couples increased reactive oxygen species with AF and that therapeutic strategies to decrease oxidized CaMKII may prevent or reduce AF.
Collapse
Affiliation(s)
- Anil Purohit
- Department of Internal Medicine, Division of Cardiovascular Medicine and Cardiovascular Research Center, Carver College of Medicine (A.P., A.G.R., X.G., B.C., O.M.K., Z.G., E.D.L., H.S., A.C.B., R.N.E.-A., P.D.S., R.M.W., L.-S.S., M.E.A.), Department of Obstetrics and Gynecology (B.Y.), and Department of Molecular Physiology and Biophysics (M.E.A.), University of Iowa, Iowa City; Institute of Pharmacology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany, and Division of Experimental Cardiology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (N.V., D.D.); Cardiology and Pneumology, German Heart Center, University Hospital Goettingen, Goettingen, Germany (S.N., T.S., L.S.M.); and Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX (N.L., X.H.T.W.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Westenbrink BD, Edwards AG, McCulloch AD, Brown JH. The promise of CaMKII inhibition for heart disease: preventing heart failure and arrhythmias. Expert Opin Ther Targets 2013; 17:889-903. [PMID: 23789646 DOI: 10.1517/14728222.2013.809064] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Calcium-calmodulin-dependent protein kinase II (CaMKII) has emerged as a central mediator of cardiac stress responses which may serve several critical roles in the regulation of cardiac rhythm, cardiac contractility and growth. Sustained and excessive activation of CaMKII during cardiac disease has, however, been linked to arrhythmias, and maladaptive cardiac remodeling, eventually leading to heart failure (HF) and sudden cardiac death. AREAS COVERED In the current review, the authors describe the unique structural and biochemical properties of CaMKII and focus on its physiological effects in cardiomyocytes. Furthermore, they provide evidence for a role of CaMKII in cardiac pathologies, including arrhythmogenesis, myocardial ischemia and HF development. The authors conclude by discussing the potential for CaMKII as a target for inhibition in heart disease. EXPERT OPINION CaMKII provides a promising nodal point for intervention that may allow simultaneous prevention of HF progression and development of arrhythmias. For future studies and drug development there is a strong rationale for the development of more specific CaMKII inhibitors. In addition, an improved understanding of the differential roles of CaMKII subtypes is required.
Collapse
Affiliation(s)
- B Daan Westenbrink
- University of California, Department of Pharmacology, San Diego, La Jolla, CA, USA
| | | | | | | |
Collapse
|
41
|
Song YH. A Memory Molecule, Ca(2+)/Calmodulin-Dependent Protein Kinase II and Redox Stress; Key Factors for Arrhythmias in a Diseased Heart. Korean Circ J 2013; 43:145-51. [PMID: 23613689 PMCID: PMC3629238 DOI: 10.4070/kcj.2013.43.3.145] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Arrhythmias can develop in various cardiac diseases, such as ischemic heart disease, cardiomyopathy and congenital heart disease. It can also contribute to the aggravation of heart failure and sudden cardiac death. Redox stress and Ca2+ overload are thought to be the important triggering factors in the generation of arrhythmias in failing myocardium. From recent studies, it appears evident that Ca2+/calmodulin-dependent protein kinase II (CaMKII) plays a central role in the arrhythmogenic processes in heart failure by sensing intracellular Ca2+ and redox stress, affecting individual ion channels and thereby leading to electrical instability in the heart. CaMKII, a multifunctional serine/threonine kinase, is an abundant molecule in the neuron and the heart. It has a specific property as "a memory molecule" such that the binding of calcified calmodulin (Ca2+/CaM) to the regulatory domain on CaMKII initially activates this enzyme. Further, it allows autophosphorylation of T287 or oxidation of M281/282 in the regulatory domain, resulting in sustained activation of CaMKII even after the dissociation of Ca2+/CaM. This review provides the understanding of both the structural and functional properties of CaMKII, the experimental findings of the interactions between CaMKII, redox stress and individual ion channels, and the evidences proving the potential participation of CaMKII and oxidative stress in the diverse arrhythmogenic processes in a diseased heart.
Collapse
Affiliation(s)
- Young-Hwan Song
- Department of Pediatrics, Sanggye Paik Hospital, College of Medicine, Inje University, Seoul, Korea
| |
Collapse
|
42
|
Epac activator critically regulates action potential duration by decreasing potassium current in rat adult ventricle. J Mol Cell Cardiol 2013; 57:96-105. [PMID: 23376036 DOI: 10.1016/j.yjmcc.2013.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 12/21/2012] [Accepted: 01/17/2013] [Indexed: 02/08/2023]
Abstract
Sympathetic stimulation is an important modulator of cardiac function via the classic cAMP-dependent signaling pathway, PKA. Recently, this paradigm has been challenged by the discovery of a family of guanine nucleotide exchange proteins directly activated by cAMP (Epac), acting in parallel to the classic signaling pathway. In cardiac myocytes, Epac activation is known to modulate Ca(2+) cycling yet their actions on cardiac ionic currents remain poorly characterized. This study attempts to address this paucity of information using the patch clamp technique to record action potential (AP) and ionic currents on rat ventricular myocytes. Epac was selectively activated by 8-CPT-AM (acetoxymethyl ester form of 8-CPT). AP amplitude, maximum depolarization rate and resting membrane amplitude were unaltered by 8-CPT-AM, strongly suggesting that Na(+) current and inward rectifier K(+) current are not regulated by Epac. In contrast, AP duration was significantly increased by 8-CPT-AM (prolongation of duration at 50% and 90% of repolarization by 41±10% and 43±8% respectively, n=11). L-type Ca(2+) current density was unaltered by 8-CPT-AM (n=16) so this cannot explain the action potential lengthening. However, the steady state component of K(+) current was significantly inhibited by 8-CPT-AM (-38±6%, n=15), while the transient outward K(+) current was unaffected by 8-CPT-AM. These effects were PKA-independent since they were observed in the presence of PKA inhibitor KT5720. Isoprenaline (100nM) induced a significant prolongation of AP duration, even in the presence of KT5720. This study provides the first evidence that the cAMP-binding protein Epac critically modulates cardiac AP duration by decreasing steady state K(+) current. These observations may be relevant to diseases in which Epac is upregulated, like cardiac hypertrophy.
Collapse
|
43
|
Butler T, Paul J, Europe-Finner N, Smith R, Chan EC. Role of serine-threonine phosphoprotein phosphatases in smooth muscle contractility. Am J Physiol Cell Physiol 2013; 304:C485-504. [PMID: 23325405 DOI: 10.1152/ajpcell.00161.2012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The degree of phosphorylation of myosin light chain 20 (MLC20) is a major determinant of force generation in smooth muscle. Myosin phosphatases (MPs) contain protein phosphatase (PP) 1 as catalytic subunits and are the major enzymes that dephosphorylate MLC20. MP regulatory targeting subunit 1 (MYPT1), the main regulatory subunit of MP in all smooth muscles, is a key convergence point of contractile and relaxatory pathways. Combinations of regulatory mechanisms, including isoform splicing, multiple phosphorylation sites, and scaffolding proteins, modulate MYPT1 activity with tissue and agonist specificities to affect contraction and relaxation. Other members of the PP1 family that do not target myosin, as well as PP2A and PP2B, dephosphorylate a range of proteins that affect smooth muscle contraction. This review discusses the role of phosphatases in smooth muscle contractility with a focus on MYPT1 in uterine smooth muscle. Myometrium shares characteristics of vascular and other visceral smooth muscles yet, during healthy pregnancy, undergoes hypertrophy, hyperplasia, quiescence, and labor as physiological processes. Myometrium presents an accessible model for the study of normal and pathological smooth muscle function, and a better understanding of myometrial physiology may allow the development of novel therapeutics for the many disorders of myometrial physiology from preterm labor to dysmenorrhea.
Collapse
Affiliation(s)
- Trent Butler
- Mothers and Babies Research Centre, Faculty of Health, University of Newcastle, Callaghan, NSW 2308, Australia
| | | | | | | | | |
Collapse
|
44
|
Rokita AG, Anderson ME. New therapeutic targets in cardiology: arrhythmias and Ca2+/calmodulin-dependent kinase II (CaMKII). Circulation 2013; 126:2125-39. [PMID: 23091085 DOI: 10.1161/circulationaha.112.124990] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Adam G Rokita
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | |
Collapse
|
45
|
Bers DM, Grandi E. Human atrial fibrillation: insights from computational electrophysiological models. Trends Cardiovasc Med 2012; 21:145-50. [PMID: 22732550 DOI: 10.1016/j.tcm.2012.04.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 04/09/2012] [Accepted: 04/10/2012] [Indexed: 11/16/2022]
Abstract
Computational electrophysiology has proven useful to investigate the mechanisms of cardiac arrhythmias at various spatial scales, from isolated myocytes to the whole heart. This article reviews how mathematical modeling has aided our understanding of human atrial myocyte electrophysiology to study the contribution of structural and electrical remodeling to human atrial fibrillation. Potential new avenues of investigation and model development are suggested.
Collapse
Affiliation(s)
- Donald M Bers
- Department of Pharmacology, University of California at Davis, Davis, CA 95616-8636, USA.
| | | |
Collapse
|
46
|
Swaminathan PD, Purohit A, Hund TJ, Anderson ME. Calmodulin-dependent protein kinase II: linking heart failure and arrhythmias. Circ Res 2012; 110:1661-77. [PMID: 22679140 DOI: 10.1161/circresaha.111.243956] [Citation(s) in RCA: 226] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Understanding relationships between heart failure and arrhythmias, important causes of suffering and sudden death, remains an unmet goal for biomedical researchers and physicians. Evidence assembled over the past decade supports a view that activation of the multifunctional Ca(2+) and calmodulin-dependent protein kinase II (CaMKII) favors myocardial dysfunction and cell membrane electrical instability. CaMKII activation follows increases in intracellular Ca(2+) or oxidation, upstream signals with the capacity to transition CaMKII into a Ca(2+) and calmodulin-independent constitutively active enzyme. Constitutively active CaMKII appears poised to participate in disease pathways by catalyzing the phosphorylation of classes of protein targets important for excitation-contraction coupling and cell survival, including ion channels and Ca(2+) homeostatic proteins, and transcription factors that drive hypertrophic and inflammatory gene expression. This rich diversity of downstream targets helps to explain the potential for CaMKII to simultaneously affect mechanical and electrical properties of heart muscle cells. Proof-of-concept studies from a growing number of investigators show that CaMKII inhibition is beneficial for improving myocardial performance and for reducing arrhythmias. We review the molecular physiology of CaMKII and discuss CaMKII actions at key cellular targets and results of animal models of myocardial hypertrophy, dysfunction, and arrhythmias that suggest CaMKII inhibition may benefit myocardial function while reducing arrhythmias.
Collapse
Affiliation(s)
- Paari Dominic Swaminathan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
47
|
David M, Macías Á, Moreno C, Prieto Á, Martínez-Mármol R, Vicente R, González T, Felipe A, Tamkun MM, Valenzuela C. Protein kinase C (PKC) activity regulates functional effects of Kvβ1.3 subunit on KV1.5 channels: identification of a cardiac Kv1.5 channelosome. J Biol Chem 2012; 287:21416-28. [PMID: 22547057 DOI: 10.1074/jbc.m111.328278] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
K(v)1.5 channels are the primary channels contributing to the ultrarapid outward potassium current (I(Kur)). The regulatory K(v)β1.3 subunit converts K(v)1.5 channels from delayed rectifiers with a modest degree of slow inactivation to channels with both fast and slow inactivation components. Previous studies have shown that inhibition of PKC with calphostin C abolishes the fast inactivation induced by K(v)β1.3. In this study, we investigated the mechanisms underlying this phenomenon using electrophysiological, biochemical, and confocal microscopy approaches. To achieve this, we used HEK293 cells (which lack K(v)β subunits) transiently cotransfected with K(v)1.5+K(v)β1.3 and also rat ventricular and atrial tissue to study native α-β subunit interactions. Immunocytochemistry assays demonstrated that these channel subunits colocalize in control conditions and after calphostin C treatment. Moreover, coimmunoprecipitation studies showed that K(v)1.5 and K(v)β1.3 remain associated after PKC inhibition. After knocking down all PKC isoforms by siRNA or inhibiting PKC with calphostin C, K(v)β1.3-induced fast inactivation at +60 mV was abolished. However, depolarization to +100 mV revealed K(v)β1.3-induced inactivation, indicating that PKC inhibition causes a dramatic positive shift of the inactivation curve. Our results demonstrate that calphostin C-mediated abolishment of fast inactivation is not due to the dissociation of K(v)1.5 and K(v)β1.3. Finally, immunoprecipitation and immunocytochemistry experiments revealed an association between K(v)1.5, K(v)β1.3, the receptor for activated C kinase (RACK1), PKCβI, PKCβII, and PKCθ in HEK293 cells. A very similar K(v)1.5 channelosome was found in rat ventricular tissue but not in atrial tissue.
Collapse
Affiliation(s)
- Miren David
- Instituto de Investigaciones Biomédicas, Madrid Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, C/Arturo Duperier 4, 28029 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Grandi E, Workman AJ, Pandit SV. Altered Excitation-Contraction Coupling in Human Chronic Atrial Fibrillation. J Atr Fibrillation 2012; 4:495. [PMID: 28496736 DOI: 10.4022/jafib.495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 02/10/2012] [Accepted: 03/19/2012] [Indexed: 12/19/2022]
Abstract
This review focuses on the (mal)adaptive processes in atrial excitation-contraction coupling occurring in patients with chronic atrial fibrillation. Cellular remodeling includes shortening of the atrial action potential duration and effective refractory period, depressed intracellular Ca2+ transient, and reduced myocyte contractility. Here we summarize the current knowledge of the ionic bases underlying these changes. Understanding the molecular mechanisms of excitation-contraction-coupling remodeling in the fibrillating human atria is important to identify new potential targets for AF therapy.
Collapse
Affiliation(s)
- Eleonora Grandi
- Department of Pharmacology, University of California at Davis, Davis, CA, USA
| | - Antony J Workman
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Sandeep V Pandit
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
49
|
Xie LH, Shanmugam M, Park JY, Zhao Z, Wen H, Tian B, Periasamy M, Babu GJ. Ablation of sarcolipin results in atrial remodeling. Am J Physiol Cell Physiol 2012; 302:C1762-71. [PMID: 22496245 DOI: 10.1152/ajpcell.00425.2011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sarcolipin (SLN) is a key regulator of sarco(endo)plasmic reticulum (SR) Ca(2+)-ATPase (SERCA), and its expression is altered in diseased atrial myocardium. To determine the precise role of SLN in atrial Ca(2+) homeostasis, we developed a SLN knockout (sln-/-) mouse model and demonstrated that ablation of SLN enhances atrial SERCA pump activity. The present study is designed to determine the long-term effects of enhanced SERCA activity on atrial remodeling in the sln-/- mice. Calcium transient measurements show an increase in atrial SR Ca(2+) load and twitch Ca(2+) transients. Patch-clamping experiments demonstrate activation of the forward mode of sodium/calcium exchanger, increased L-type Ca(2+) channel activity, and prolongation of action potential duration at 90% repolarization in the atrial myocytes of sln-/- mice. Spontaneous Ca(2+) waves, delayed afterdepolarization, and triggered activities are frequent in the atrial myocytes of sln-/- mice. Furthermore, loss of SLN in atria is associated with increased interstitial fibrosis and altered expression of genes encoding collagen and other extracellular matrix proteins. Our results also show that the sln-/- mice are susceptible to atrial arrhythmias upon aging. Together, these findings indicate that ablation of SLN results in increased SERCA activity and SR Ca(2+) load, which, in turn, could cause abnormal intracellular Ca(2+) handling and atrial remodeling.
Collapse
Affiliation(s)
- Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, 07103, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Atrial fibrillation is the most common type of cardiac arrhythmia, and is responsible for substantial morbidity and mortality in the general population. Current treatments have moderate efficacy and considerable risks, especially of pro-arrhythmia, highlighting the need for new therapeutic strategies. In recent years, substantial efforts have been invested in developing novel treatments that target the underlying molecular determinants of atrial fibrillation, and several new compounds are under development. This Review focuses on the mechanistic rationale for the development of new anti-atrial fibrillation drugs, on the molecular and structural motifs that they target and on the results obtained so far in experimental and clinical studies.
Collapse
|