1
|
Masjoan Juncos JX, Nadeem F, Shakil S, El-Husari M, Zafar I, Louch WE, Halade GV, Zaky A, Ahmad A, Ahmad S. Myocardial SERCA2 Protects Against Cardiac Damage and Dysfunction Caused by Inhaled Bromine. J Pharmacol Exp Ther 2024; 390:146-158. [PMID: 38772719 PMCID: PMC11192580 DOI: 10.1124/jpet.123.002084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/23/2024] Open
Abstract
Myocardial sarcoendoplasmic reticulum calcium ATPase 2 (SERCA2) activity is critical for heart function. We have demonstrated that inhaled halogen (chlorine or bromine) gases inactivate SERCA2, impair calcium homeostasis, increase proteolysis, and damage the myocardium ultimately leading to cardiac dysfunction. To further elucidate the mechanistic role of SERCA2 in halogen-induced myocardial damage, we used bromine-exposed cardiac-specific SERCA2 knockout (KO) mice [tamoxifen-administered SERCA2 (flox/flox) Tg (αMHC-MerCreMer) mice] and compared them to the oil-administered controls. We performed echocardiography and hemodynamic analysis to investigate cardiac function 24 hours after bromine (600 ppm for 30 minutes) exposure and measured cardiac injury markers in plasma and proteolytic activity in cardiac tissue and performed electron microscopy of the left ventricle (LV). Cardiac-specific SERCA2 knockout mice demonstrated enhanced toxicity to bromine. Bromine exposure increased ultrastructural damage, perturbed LV shape geometry, and demonstrated acutely increased phosphorylation of phospholamban in the KO mice. Bromine-exposed KO mice revealed significantly enhanced mean arterial pressure and sphericity index and decreased LV end diastolic diameter and LV end systolic pressure when compared with the bromine-exposed control FF mice. Strain analysis showed loss of synchronicity, evidenced by an irregular endocardial shape in systole and irregular vector orientation of contractile motion across different segments of the LV in KO mice, both at baseline and after bromine exposure. These studies underscore the critical role of myocardial SERCA2 in preserving cardiac ultrastructure and function during toxic halogen gas exposures. SIGNIFICANCE STATEMENT: Due to their increased industrial production and transportation, halogens such as chlorine and bromine pose an enhanced risk of exposure to the public. Our studies have demonstrated that inhalation of these halogens leads to the inactivation of cardiopulmonary SERCA2 and results in calcium overload. Using cardiac-specific SERCA2 KO mice, these studies further validated the role of SERCA2 in bromine-induced myocardial injury. These studies highlight the increased susceptibility of individuals with pathological loss of cardiac SERCA2 to the effects of bromine.
Collapse
Affiliation(s)
- Juan Xavier Masjoan Juncos
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama (J.X.M.J., F.N., S.S., M.E.-H., I.Z, A.Z., A.A., S.A.); Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway (W.E.L.); and Division of Cardiovascular Sciences, University of South Florida, Tampa, Florida (G.V.H.)
| | - Fahad Nadeem
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama (J.X.M.J., F.N., S.S., M.E.-H., I.Z, A.Z., A.A., S.A.); Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway (W.E.L.); and Division of Cardiovascular Sciences, University of South Florida, Tampa, Florida (G.V.H.)
| | - Shazia Shakil
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama (J.X.M.J., F.N., S.S., M.E.-H., I.Z, A.Z., A.A., S.A.); Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway (W.E.L.); and Division of Cardiovascular Sciences, University of South Florida, Tampa, Florida (G.V.H.)
| | - Malik El-Husari
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama (J.X.M.J., F.N., S.S., M.E.-H., I.Z, A.Z., A.A., S.A.); Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway (W.E.L.); and Division of Cardiovascular Sciences, University of South Florida, Tampa, Florida (G.V.H.)
| | - Iram Zafar
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama (J.X.M.J., F.N., S.S., M.E.-H., I.Z, A.Z., A.A., S.A.); Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway (W.E.L.); and Division of Cardiovascular Sciences, University of South Florida, Tampa, Florida (G.V.H.)
| | - William E Louch
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama (J.X.M.J., F.N., S.S., M.E.-H., I.Z, A.Z., A.A., S.A.); Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway (W.E.L.); and Division of Cardiovascular Sciences, University of South Florida, Tampa, Florida (G.V.H.)
| | - Ganesh V Halade
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama (J.X.M.J., F.N., S.S., M.E.-H., I.Z, A.Z., A.A., S.A.); Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway (W.E.L.); and Division of Cardiovascular Sciences, University of South Florida, Tampa, Florida (G.V.H.)
| | - Ahmed Zaky
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama (J.X.M.J., F.N., S.S., M.E.-H., I.Z, A.Z., A.A., S.A.); Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway (W.E.L.); and Division of Cardiovascular Sciences, University of South Florida, Tampa, Florida (G.V.H.)
| | - Aftab Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama (J.X.M.J., F.N., S.S., M.E.-H., I.Z, A.Z., A.A., S.A.); Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway (W.E.L.); and Division of Cardiovascular Sciences, University of South Florida, Tampa, Florida (G.V.H.)
| | - Shama Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama (J.X.M.J., F.N., S.S., M.E.-H., I.Z, A.Z., A.A., S.A.); Institute for Experimental Medical Research, Oslo University Hospital and KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway (W.E.L.); and Division of Cardiovascular Sciences, University of South Florida, Tampa, Florida (G.V.H.)
| |
Collapse
|
2
|
Badura K, Buławska D, Dąbek B, Witkowska A, Lisińska W, Radzioch E, Skwira S, Młynarska E, Rysz J, Franczyk B. Primary Electrical Heart Disease-Principles of Pathophysiology and Genetics. Int J Mol Sci 2024; 25:1826. [PMID: 38339103 PMCID: PMC10855675 DOI: 10.3390/ijms25031826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/27/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
Primary electrical heart diseases, often considered channelopathies, are inherited genetic abnormalities of cardiomyocyte electrical behavior carrying the risk of malignant arrhythmias leading to sudden cardiac death (SCD). Approximately 54% of sudden, unexpected deaths in individuals under the age of 35 do not exhibit signs of structural heart disease during autopsy, suggesting the potential significance of channelopathies in this group of age. Channelopathies constitute a highly heterogenous group comprising various diseases such as long QT syndrome (LQTS), short QT syndrome (SQTS), idiopathic ventricular fibrillation (IVF), Brugada syndrome (BrS), catecholaminergic polymorphic ventricular tachycardia (CPVT), and early repolarization syndromes (ERS). Although new advances in the diagnostic process of channelopathies have been made, the link between a disease and sudden cardiac death remains not fully explained. Evolving data in electrophysiology and genetic testing suggest previously described diseases as complex with multiple underlying genes and a high variety of factors associated with SCD in channelopathies. This review summarizes available, well-established information about channelopathy pathogenesis, genetic basics, and molecular aspects relative to principles of the pathophysiology of arrhythmia. In addition, general information about diagnostic approaches and management is presented. Analyzing principles of channelopathies and their underlying causes improves the understanding of genetic and molecular basics that may assist general research and improve SCD prevention.
Collapse
Affiliation(s)
- Krzysztof Badura
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Dominika Buławska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Bartłomiej Dąbek
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Alicja Witkowska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Wiktoria Lisińska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Ewa Radzioch
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Sylwia Skwira
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland (S.S.)
| |
Collapse
|
3
|
Chakraborty P, Aggarwal AK, Nair MKK, Massé S, Riazi S, Nanthakumar K. Restoration of calcium release synchrony: A novel target for heart failure and ventricular arrhythmia. Heart Rhythm 2023; 20:1773-1781. [PMID: 37678492 DOI: 10.1016/j.hrthm.2023.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/13/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023]
Abstract
Myocardial calcium (Ca2+) signaling plays a crucial role in contractile function and membrane electrophysiology. An abnormal myocardial Ca2+ transient is linked to heart failure and ventricular arrhythmias. At the subcellular level, the synchronous release of Ca2+ sparks from sarcoplasmic Ca2+ release units determines the configuration and amplitude of the global Ca2+ transient. This narrative review evaluates the role of aberrant Ca2+ release synchrony in the pathophysiology of cardiomyopathies and ventricular arrhythmias. The potential therapeutic benefits of restoration of Ca2+ release synchrony in heart failure and ventricular arrhythmias are also discussed.
Collapse
Affiliation(s)
- Praloy Chakraborty
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Heart Rhythm Institute, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - Arjun K Aggarwal
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Madhav Krishna Kumar Nair
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Stéphane Massé
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Sheila Riazi
- Malignant Hyperthermia Investigation Unit, Department of Anesthesia and Pain Management, University Health Network, Toronto, Ontario, Canada
| | - Kumaraswamy Nanthakumar
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
4
|
Jin Q, Greenstein JL, Winslow RL. Estimating the probability of early afterdepolarizations and predicting arrhythmic risk associated with long QT syndrome type 1 mutations. Biophys J 2023; 122:4042-4056. [PMID: 37705243 PMCID: PMC10598291 DOI: 10.1016/j.bpj.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/29/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023] Open
Abstract
Early afterdepolarizations (EADs) are action potential (AP) repolarization abnormalities that can trigger lethal arrhythmias. Simulations using biophysically detailed cardiac myocyte models can reveal how model parameters influence the probability of these cellular arrhythmias; however, such analyses can pose a huge computational burden. We have previously developed a highly simplified approach in which logistic regression models (LRMs) map parameters of complex cell models to the probability of ectopic beats. Here, we extend this approach to predict the probability of EADs (P(EAD)) as a mechanistic metric of arrhythmic risk. We use the LRM to investigate how changes in parameters of the slow-activating delayed rectifier current (IKs) affect P(EAD) for 17 different long QT syndrome type 1 (LQTS1) mutations. In this LQTS1 clinical arrhythmic risk prediction task, we compared P(EAD) for these 17 mutations with two other recently published model-based arrhythmia risk metrics (AP morphology metric across populations of myocyte models and transmural repolarization prolongation based on a one-dimensional [1D] tissue-level model). These model-based risk metrics yield similar prediction performance; however, each fails to stratify clinical risk for a significant number of the 17 studied LQTS1 mutations. Nevertheless, an interpretable ensemble model using multivariate linear regression built by combining all of these model-based risk metrics successfully predicts the clinical risk of 17 mutations. These results illustrate the potential of computational approaches in arrhythmia risk prediction.
Collapse
Affiliation(s)
- Qingchu Jin
- Department of Biomedical Engineering and Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Joseph L Greenstein
- Department of Biomedical Engineering and Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Raimond L Winslow
- Department of Biomedical Engineering and Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
5
|
You T, Xie Y, Luo C, Zhang K, Zhang H. Mechanistic insights into spontaneous transition from cellular alternans to ventricular fibrillation. Physiol Rep 2023; 11:e15619. [PMID: 36863774 PMCID: PMC9981424 DOI: 10.14814/phy2.15619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/04/2023] [Accepted: 02/05/2023] [Indexed: 03/04/2023] Open
Abstract
T-wave alternans (TWA) has been used for predicting the risk of malignant cardiac arrhythmias and sudden cardiac death (SCD) in multiple clinical settings; however, possible mechanism(s) underlying the spontaneous transition from cellular alternans reflected by TWA to arrhythmias in impaired repolarization remains unclear. The healthy guinea pig ventricular myocytes under E-4031 blocking IKr (0.1 μM, N = 12; 0.3 μM, N = 10; 1 μM, N = 10) were evaluated using whole-cell patch-clamp. The electrophysiological properties of isolated perfused guinea pig hearts under E-4031 (0.1 μM, N = 5; 0.3 μM, N = 5; 1 μM, N = 5) were evaluated using dual- optical mapping. The amplitude/threshold/restitution curves of action potential duration (APD) alternans and potential mechanism(s) underlying the spontaneous transition of cellular alternans to ventricular fibrillation (VF) were examined. There were longer APD80 and increased amplitude and threshold of APD alternans in E-4031 group compared with baseline group, which was reflected by more pronounced arrhythmogenesis at the tissue level, and were associated with steep restitution curves of the APD and the conduction velocity (CV). Conduction of AP alternans augmented tissue's functional spatiotemporal heterogeneity of regional AP/Ca alternans, as well as the AP/Ca dispersion, leading to localized uni-directional conduction block that spontaneous facilitated the formation of reentrant excitation waves without the need for additional premature stimulus. Our results provide a possible mechanism for the spontaneous transition from cardiac electrical alternans in cellular action potentials and intercellular conduction without the involvement of premature excitations, and explain the increased susceptibility to ventricular arrhythmias in impaired repolarization. In this study, we implemented voltage-clamp and dual-optical mapping approaches to investigate the underlying mechanism(s) for the arrhythmogenesis of cardiac alternans in the guinea pig heart at cellular and tissue levels. Our results demonstrated a spontaneous development of reentry from cellular alternans, arising from a combined actions of restitution properties of action potential duration, conduction velocity of excitation wave and interplay between alternants of action potential and the intracellular Ca handling. We believe this study provides new insights into underlying the mechanism, by which cellular cardiac alternans spontaneously evolves into cardiac arrhythmias.
Collapse
Affiliation(s)
- Tingting You
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases)Institute of Cardiovascular Research, Southwest Medical UniversityLuzhouChina
- Department of NeurosurgeryXinqiao Hospital, Army Medical UniversityChongqingChina
| | - Yulong Xie
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases)Institute of Cardiovascular Research, Southwest Medical UniversityLuzhouChina
| | - Cunjin Luo
- School of Computer Science and Electronic EngineeringUniversity of EssexColchesterUK
| | - Kevin Zhang
- School of MedicineImperial College of LondonLondonUK
| | - Henggui Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases)Institute of Cardiovascular Research, Southwest Medical UniversityLuzhouChina
- Department of Physics and AstronomyUniversity of ManchesterManchesterUK
| |
Collapse
|
6
|
McCoy MD, Ullah A, Lederer WJ, Jafri MS. Understanding Calmodulin Variants Affecting Calcium-Dependent Inactivation of L-Type Calcium Channels through Whole-Cell Simulation of the Cardiac Ventricular Myocyte. Biomolecules 2022; 13:72. [PMID: 36671457 PMCID: PMC9855640 DOI: 10.3390/biom13010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Mutations in the calcium-sensing protein calmodulin (CaM) have been linked to two cardiac arrhythmia diseases, Long QT Syndrome 14 (LQT14) and Catecholaminergic Polymorphic Ventricular Tachycardia Type 4 (CPVT4), with varying degrees of severity. Functional characterization of the CaM mutants most strongly associated with LQT14 show a clear disruption of the calcium-dependent inactivation (CDI) of the L-Type calcium channel (LCC). CPVT4 mutants on the other hand are associated with changes in their affinity to the ryanodine receptor. In clinical studies, some variants have been associated with both CPVT4 and LQT15. This study uses simulations in a model for excitation-contraction coupling in the rat ventricular myocytes to understand how LQT14 variant might give the functional phenotype similar to CPVT4. Changing the CaM-dependent transition rate by a factor of 0.75 corresponding to the D96V variant and by a factor of 0.90 corresponding to the F142L or N98S variants, in a physiologically based stochastic model of the LCC prolonger, the action potential duration changed by a small amount in a cardiac myocyte but did not disrupt CICR at 1, 2, and 4 Hz. Under beta-adrenergic simulation abnormal excitation-contraction coupling was observed above 2 Hz pacing for the mutant CaM. The same conditions applied under beta-adrenergic stimulation led to the rapid onset of arrhythmia in the mutant CaM simulations. Simulations with the LQT14 mutations under the conditions of rapid pacing with beta-adrenergic stimulation drives the cardiac myocyte toward an arrhythmic state known as Ca2+ overload. These simulations provide a mechanistic link to a disease state for LQT14-associated mutations in CaM to yield a CPVT4 phenotype. The results show that small changes to the CaM-regulated inactivation of LCC promote arrhythmia and underscore the significance of CDI in proper heart function.
Collapse
Affiliation(s)
- Matthew D. McCoy
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
- Innovation Center for Biomedical Informatics, Department of Oncology, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, USA
| | - Aman Ullah
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
| | - W. Jonathan Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| | - M. Saleet Jafri
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| |
Collapse
|
7
|
Dahlen SA, Bernadyn TF, Dixon AJ, Sun B, Xia J, Owens EA, Osei-Owusu P. Dual loss of regulator of G protein signaling 2 and 5 exacerbates ventricular myocyte arrhythmias and disrupts the fine-tuning of G i/o signaling. J Mol Cell Cardiol 2022; 170:34-46. [PMID: 35661621 DOI: 10.1016/j.yjmcc.2022.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/22/2022] [Accepted: 05/14/2022] [Indexed: 10/18/2022]
Abstract
AIMS Cardiac contractility, essential to maintaining proper cardiac output and circulation, is regulated by G protein-coupled receptor (GPCR) signaling. Previously, the absence of regulator of G protein signaling (RGS) 2 and 5, separately, was shown to cause G protein dysregulation, contributing to modest blood pressure elevation and exaggerated cardiac hypertrophic response to pressure-overload. Whether RGS2 and 5 redundantly control G protein signaling to maintain cardiovascular homeostasis is unknown. Here we examined how the dual absence of RGS2 and 5 (Rgs2/5 dbKO) affects blood pressure and cardiac structure and function. METHODS AND RESULTS We found that Rgs2/5 dbKO mice showed left ventricular dilatation at baseline by echocardiography. Cardiac contractile response to dobutamine stress test was sex-dependently reduced in male Rgs2/5 dbKO relative to WT mice. When subjected to surgery-induced stress, male Rgs2/5 dbKO mice had 75% mortality within 72-96 h after surgery, accompanied by elevated baseline blood pressure and decreased cardiac contractile function. At the cellular level, cardiomyocytes (CM) from Rgs2/5 dbKO mice showed augmented Ca2+ transients and increased incidence of arrhythmia without augmented contractile response to electrical field stimulation (EFS) and activation of β-adrenergic receptors (βAR) with isoproterenol. Dual loss of Rgs2 and 5 suppressed forskolin-induced cAMP production, which was restored by Gi/o inactivation with pertussis toxin that also reduced arrhythmogenesis during EFS or βAR stimulation. Cardiomyocyte NCX and PMCA mRNA expression was unaffected in Rgs2/5 dbKO male mice. However, there was an exaggerated elevation of EFS-induced cytoplasmic Ca2+ in the presence of SERCA blockade with thapsigargin. CONCLUSIONS We conclude that RGS2 and 5 promote normal ventricular rhythm by coordinating their regulatory activity towards Gi/o signaling and facilitating cardiomyocyte calcium handling.
Collapse
Affiliation(s)
- Shelby A Dahlen
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States of America
| | - Tyler F Bernadyn
- Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America
| | - Alethia J Dixon
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States of America
| | - Bo Sun
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States of America
| | - Jingsheng Xia
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States of America
| | - Elizabeth A Owens
- Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America
| | - Patrick Osei-Owusu
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States of America; Department of Pharmacology & Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America.
| |
Collapse
|
8
|
Thyroid hormones regulate cardiac repolarization and QT-interval related gene expression in hiPSC cardiomyocytes. Sci Rep 2022; 12:568. [PMID: 35022468 PMCID: PMC8755773 DOI: 10.1038/s41598-021-04659-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/22/2021] [Indexed: 11/08/2022] Open
Abstract
Prolongation of cardiac repolarization (QT interval) represents a dangerous and potentially life-threatening electrical event affecting the heart. Thyroid hormones (THs) are critical for cardiac development and heart function. However, little is known about THs influence on ventricular repolarization and controversial effects on QT prolongation are reported. Human iPSC-derived cardiomyocytes (hiPSC-CMs) and multielectrode array (MEA) systems were used to investigate the influence of 3,3',5-triiodo-L-Thyronine (T3) and 3,3',5,5'-tetraiodo-L-Thyronine (T4) on corrected Field Potential Duration (FPDc), the in vitro analog of QT interval, and on local extracellular Action Potential Duration (APD). Treatment with high THs doses induces a significant prolongation of both FPDc and APD, with the strongest increase reached after 24 h exposure. Preincubation with reverse T3 (rT3), a specific antagonist for nuclear TH receptor binding, significantly reduces T3 effects on FPDc, suggesting a TRs-mediated transcriptional mechanism. RNA-seq analysis showed significant deregulation in genes involved in cardiac repolarization pathways, including several QT-interval related genes. In conclusion, long-time administration of high THs doses induces FPDc prolongation in hiPSC-CMs probably through the modulation of genes linked to QT-interval regulation. These results open the way to investigate new potential diagnostic biomarkers and specific targeted therapies for cardiac repolarization dysfunctions.
Collapse
|
9
|
Baracaldo-Santamaría D, Llinás-Caballero K, Corso-Ramirez JM, Restrepo CM, Dominguez-Dominguez CA, Fonseca-Mendoza DJ, Calderon-Ospina CA. Genetic and Molecular Aspects of Drug-Induced QT Interval Prolongation. Int J Mol Sci 2021; 22:8090. [PMID: 34360853 PMCID: PMC8347245 DOI: 10.3390/ijms22158090] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 12/22/2022] Open
Abstract
Long QT syndromes can be either acquired or congenital. Drugs are one of the many etiologies that may induce acquired long QT syndrome. In fact, many drugs frequently used in the clinical setting are a known risk factor for a prolonged QT interval, thus increasing the chances of developing torsade de pointes. The molecular mechanisms involved in the prolongation of the QT interval are common to most medications. However, there is considerable inter-individual variability in drug response, thus making the application of personalized medicine a relevant aspect in long QT syndrome, in order to evaluate the risk of every individual from a pharmacogenetic standpoint.
Collapse
Affiliation(s)
- Daniela Baracaldo-Santamaría
- School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia; (D.B.-S.); (J.M.C.-R.); (C.A.D.-D.)
| | - Kevin Llinás-Caballero
- GENIUROS Research Group, Center for Research in Genetics and Genomics (CIGGUR), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia; (K.L.-C.); (C.M.R.); (D.J.F.-M.)
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia
| | - Julián Miguel Corso-Ramirez
- School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia; (D.B.-S.); (J.M.C.-R.); (C.A.D.-D.)
| | - Carlos Martín Restrepo
- GENIUROS Research Group, Center for Research in Genetics and Genomics (CIGGUR), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia; (K.L.-C.); (C.M.R.); (D.J.F.-M.)
| | | | - Dora Janeth Fonseca-Mendoza
- GENIUROS Research Group, Center for Research in Genetics and Genomics (CIGGUR), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia; (K.L.-C.); (C.M.R.); (D.J.F.-M.)
| | - Carlos Alberto Calderon-Ospina
- GENIUROS Research Group, Center for Research in Genetics and Genomics (CIGGUR), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia; (K.L.-C.); (C.M.R.); (D.J.F.-M.)
| |
Collapse
|
10
|
Sander P, Feng M, Schweitzer MK, Wilting F, Gutenthaler SM, Arduino DM, Fischbach S, Dreizehnter L, Moretti A, Gudermann T, Perocchi F, Schredelseker J. Approved drugs ezetimibe and disulfiram enhance mitochondrial Ca 2+ uptake and suppress cardiac arrhythmogenesis. Br J Pharmacol 2021; 178:4518-4532. [PMID: 34287836 DOI: 10.1111/bph.15630] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 06/21/2021] [Accepted: 06/30/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Treatment of cardiac arrhythmia remains challenging due to severe side effects of common anti-arrhythmic drugs. We previously demonstrated that mitochondrial Ca2+ uptake in cardiomyocytes represents a promising new candidate structure for safer drug therapy. However, druggable agonists of mitochondrial Ca2+ uptake suitable for preclinical and clinical studies are still missing. EXPERIMENTAL APPROACH Herewe screened 727 compounds with a history of use in human clinical trials in a three-step screening approach. As a primary screening platform we used a permeabilized HeLa cell-based mitochondrial Ca2+ uptake assay. Hits were validated in cultured HL-1 cardiomyocytes and finally tested for anti-arrhythmic efficacy in three translational models: a Ca2+ overload zebrafish model and cardiomyocytes of both a mouse model for catecholaminergic polymorphic ventricular tachycardia (CPVT) and induced pluripotent stem cell derived cardiomyocytes from a CPVT patient. KEY RESULTS We identifiedtwo candidate compounds, the clinically approved drugs ezetimibe and disulfiram, which stimulate SR-mitochondria Ca2+ transfer at nanomolar concentrations. This is significantly lower compared to the previously described mitochondrial Ca2+ uptake enhancers (MiCUps) efsevin, a gating modifier of the voltage-dependent anion channel 2, and kaempferol, an agonist of the mitochondrial Ca2+ uniporter. Both substances restored rhythmic cardiac contractions in a zebrafish cardiac arrhythmia model and significantly suppressed arrhythmogenesis in freshly isolated ventricular cardiomyocytes from a CPVT mouse model as well as induced pluripotent stem cell derived cardiomyocytes from a CPVT patient. CONCLUSION AND IMPLICATIONS Taken together we identified ezetimibe and disulfiram as novel MiCUps and efficient suppressors of arrhythmogenesis and as such as, promising candidates for future preclinical and clinical studies.
Collapse
Affiliation(s)
- Paulina Sander
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Michael Feng
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, Neuherberg, Germany
| | - Maria K Schweitzer
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Fabiola Wilting
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sophie M Gutenthaler
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Daniela M Arduino
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, Neuherberg, Germany
| | - Sandra Fischbach
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Lisa Dreizehnter
- I. Department of Medicine, Cardiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Alessandra Moretti
- I. Department of Medicine, Cardiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Partner Site Munich Heart Alliance (MHA), Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany.,Partner Site Munich Heart Alliance (MHA), Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Munich, Germany
| | - Fabiana Perocchi
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center (HDC), Helmholtz Zentrum München, Neuherberg, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| | - Johann Schredelseker
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany.,Partner Site Munich Heart Alliance (MHA), Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK), Munich, Germany
| |
Collapse
|
11
|
Sharif AF, Elsheikh E, Al-Asmari AZ, Gameel DE. Potential Role of Serum S-100β Protein as a Predictor of Cardiotoxicity and Clinical Poor Outcome in Acute Amphetamine Intoxication. Cardiovasc Toxicol 2021; 21:375-386. [PMID: 33423174 DOI: 10.1007/s12012-020-09630-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/29/2020] [Indexed: 11/28/2022]
Abstract
Cardio- and neurotoxicity of amphetamines play an important role in worsening morbidity, making the initial evaluation of the patient's status a potentially lifesaving action. The current study hypothesized that the S-100β serum level could predict the severity of acute amphetamine toxicity and the in-hospital outcome. The current study is a prospective cohort study conducted on 77 patients diagnosed with acute amphetamine exposure and referred to Aseer Poison Control Center, Saudi Arabia. The patients admitted to ICU showed significantly higher serum levels of S-100β in comparison to those not admitted (p < 0.05). Moreover, the S-100β level was significantly elevated among patients with prolonged QTc intervals. Receiver-operating characteristic curve of S-100β serum level as an in-hospital outcome predictor showed that at a cutoff value > 0.430 ug/L, the sensitivity of S-100β serum level as severity predictor was 100%, and the specificity was 74.1%. In conclusion, the current study revealed that the S-100β serum level could be used as an outcome predictor in hospital admission cases due to toxic amphetamine exposure and offers an idea about the cardiac and neuronal involvement. This can help select patients who will benefit most from ICU admission and early management and assess the severity of cases in settings where GC-MS is not available.
Collapse
Affiliation(s)
- Asmaa F Sharif
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Tanta University, Tanta, Egypt. .,Clinical Sciences Departement, College of Medicine, Dar Al Uloom University, Riyadh, Saudi Arabia.
| | - Eman Elsheikh
- Cardiology Department, Faculty of Medicine, Tanta University, Tanta, Egypt.,Internal Medicine Department, King Faisal University, Hofuf, Saudi Arabia
| | - Abdullah Z Al-Asmari
- Poison Control Centers and Medical Chemistry Legitimacy South, Aseer, Saudi Arabia.,Poison Control Center, Aseer, Saudi Arabia
| | - Dina El Gameel
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Tanta University, Tanta, Egypt.,Poison Control Center, Aseer, Saudi Arabia
| |
Collapse
|
12
|
Hegyi B, Pölönen RP, Hellgren KT, Ko CY, Ginsburg KS, Bossuyt J, Mercola M, Bers DM. Cardiomyocyte Na + and Ca 2+ mishandling drives vicious cycle involving CaMKII, ROS, and ryanodine receptors. Basic Res Cardiol 2021; 116:58. [PMID: 34648073 PMCID: PMC8516771 DOI: 10.1007/s00395-021-00900-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/08/2021] [Accepted: 09/30/2021] [Indexed: 12/19/2022]
Abstract
Cardiomyocyte Na+ and Ca2+ mishandling, upregulated Ca2+/calmodulin-dependent kinase II (CaMKII), and increased reactive oxygen species (ROS) are characteristics of various heart diseases, including heart failure (HF), long QT (LQT) syndrome, and catecholaminergic polymorphic ventricular tachycardia (CPVT). These changes may form a vicious cycle of positive feedback to promote cardiac dysfunction and arrhythmias. In HF rabbit cardiomyocytes investigated in this study, the inhibition of CaMKII, late Na+ current (INaL), and leaky ryanodine receptors (RyRs) all attenuated the prolongation and increased short-term variability (STV) of action potential duration (APD), but in age-matched controls these inhibitors had no or minimal effects. In control cardiomyocytes, we enhanced RyR leak (by low [caffeine] plus isoproterenol mimicking CPVT) which markedly increased STV and delayed afterdepolarizations (DADs). These proarrhythmic changes were significantly attenuated by both CaMKII inhibition and mitochondrial ROS scavenging, with a slight synergy with INaL inhibition. Inducing LQT by elevating INaL (by Anemone toxin II, ATX-II) caused markedly prolonged APD, increased STV, and early afterdepolarizations (EADs). Those proarrhythmic ATX-II effects were largely attenuated by mitochondrial ROS scavenging, and partially reduced by inhibition of CaMKII and pathological leaky RyRs using dantrolene. In human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) bearing LQT3 mutation SCN5A N406K, dantrolene significantly attenuated cell arrhythmias and APD prolongation. Targeting critical components of the Na+-Ca2+-CaMKII-ROS-INaL arrhythmogenic vicious cycle may exhibit important on-target and also trans-target effects (e.g., INaL and RyR inhibition can alter INaL-mediated LQT3 effects). Incorporating this vicious cycle into therapeutic strategies provides novel integrated insight for treating cardiac arrhythmias and diseases.
Collapse
Affiliation(s)
- Bence Hegyi
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Risto-Pekka Pölönen
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA ,grid.168010.e0000000419368956Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Kim T. Hellgren
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Christopher Y. Ko
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Kenneth S. Ginsburg
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Julie Bossuyt
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| | - Mark Mercola
- grid.168010.e0000000419368956Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, CA 94305 USA
| | - Donald M. Bers
- grid.27860.3b0000 0004 1936 9684Department of Pharmacology, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616 USA
| |
Collapse
|
13
|
Mapping Calcium Dynamics in the Heart of Zebrafish Embryos with Ratiometric Genetically Encoded Calcium Indicators. Int J Mol Sci 2020; 21:ijms21186610. [PMID: 32927644 PMCID: PMC7555812 DOI: 10.3390/ijms21186610] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/23/2022] Open
Abstract
Zebrafish embryos have been proposed as a cost-effective vertebrate model to study heart function. Many fluorescent genetically encoded Ca2+ indicators (GECIs) have been developed, but those with ratiometric readout seem more appropriate to image a moving organ such as the heart. Four ratiometric GECIs based on troponin C, TN-XXL, Twitch-1, Twitch-2B, and Twitch-4 were expressed transiently in the heart of zebrafish embryos. Their emission ratio reported the Ca2+ levels in both the atrium and the ventricle. We measured several kinetic parameters of the Ca2+ transients: systolic and diastolic ratio, the amplitude of the systolic Ca2+ rise, the heart rate, as well as the rise and decay times and slopes. The systolic ratio change decreased in cells expressing high biosensor concentration, possibly caused by Ca2+ buffering. The GECIs were able to report the effect of nifedipine and propranolol on the heart, which resulted in changes in heart rate, diastolic and systolic Ca2+ levels, and Ca2+ kinetics. As a result, Twitch-1 and Twitch-4 (Kd 0.25 and 2.8 µM, respectively) seem the most promising GECIs for generating transgenic zebrafish lines, which could be used for modeling heart disorders, for drug screening, and for cardiotoxicity assessment during drug development.
Collapse
|
14
|
Two-variable nullcline analysis of ionic general equilibrium predicts calcium homeostasis in ventricular myocytes. PLoS Comput Biol 2020; 16:e1007572. [PMID: 32502205 PMCID: PMC7316341 DOI: 10.1371/journal.pcbi.1007572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 06/25/2020] [Accepted: 05/05/2020] [Indexed: 01/16/2023] Open
Abstract
Ventricular contraction is roughly proportional to the amount of calcium released from the Sarcoplasmic Reticulum (SR) during systole. While it is rather straightforward to measure calcium levels and contractibility under different physiological conditions, the complexity of calcium handling during systole and diastole has made the prediction of its release at steady state impossible. Here we approach the problem analyzing the evolution of intracellular and extracellular calcium fluxes during a single beat which is away from homeostatic balance. Using an in-silico subcellular model of rabbit ventricular myocyte, we show that the high dimensional nonlinear problem of finding the steady state can be reduced to a two-variable general equilibrium condition where pre-systolic calcium level in the cytosol and in the SR must fulfill simultaneously two different equalities. This renders calcium homeostasis as a problem that can be studied in terms of its equilibrium structure, leading to precise predictions of steady state from single-beat measurements. We show how changes in ion channels modify the general equilibrium, as shocks would do in general equilibrium macroeconomic models. This allows us to predict when an enhanced entrance of calcium in the cell reduces its contractibility and explain why SERCA gene therapy, a change in calcium handling to treat heart failure, might fail to improve contraction even when it successfully increases SERCA expression. Cardiomyocytes, upon voltage excitation, release calcium, which leads to cell contraction. However, under some pathological conditions, calcium handling is impaired. Recently, SERCA gene therapy, whose aim is to improve Ca2+ sequestration by the Sarcoplasmic Reticulum (SR), has failed to improve the prognosis of patients with Heart Failure. This, together with recent counterintuitive results in calcium handling, has highlighted the need for a framework to understand calcium homeostasis across species and pathologies. We show here that the proper framework is a general equilibrium approach of two independent variables. The development of this framework allows us to find a possible mechanism for the failure of SERCA gene therapy even when it manages to increase Ca SERCA expression.
Collapse
|
15
|
Abstract
The term phenotype is so commonly used that we often assume that we each mean the same thing. The general definition, the set of observable characteristics of an individual resulting from the interaction of their genotype with the environment, is often left to the eye of the beholder. Whether applied to the multiple levels of biological phenomena or the intact human being, our ability to characterize, classify, and analyze phenotype has been limited by measurement deficits, computing limitations, and a culture that avoids the generalizable. With the advent of modern technology, there is the potential for a revolution in phenotyping, which incorporates old and new in structured ways to dramatically advance basic understanding of biology and behavior and to lead to major improvements in clinical care and public health. This revolution in how we think about phenotypes will require a radical change in the scale at which biomedicine operates with significant changes in the unit of action, which will have far-reaching implications for how care, translation, and discovery are implemented.
Collapse
Affiliation(s)
- Calum A MacRae
- From the One Brave Idea (C.A.M., R.M.C.).,Cardiovascular Medicine Division and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (C.A.M.)
| | - Robert M Califf
- From the One Brave Idea (C.A.M., R.M.C.).,Verily Life Sciences (R.M.C.).,Google Health, South San Francisco and Mountain View, CA (R.M.C.)
| |
Collapse
|
16
|
Morissette P, Polak S, Chain A, Zhai J, Imredy JP, Wildey MJ, Travis J, Fitzgerald K, Fanelli P, Passini E, Rodriguez B, Sannajust F, Regan C. Combining an in silico proarrhythmic risk assay with a tPKPD model to predict QTc interval prolongation in the anesthetized guinea pig assay. Toxicol Appl Pharmacol 2020; 390:114883. [PMID: 31981640 PMCID: PMC7322544 DOI: 10.1016/j.taap.2020.114883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/03/2020] [Accepted: 01/14/2020] [Indexed: 12/12/2022]
Abstract
Human-based in silico models are emerging as important tools to study the effects of integrating inward and outward ion channel currents to predict clinical proarrhythmic risk. The aims of this study were 2-fold: 1) Evaluate the capacity of an in silico model to predict QTc interval prolongation in the in vivo anesthetized cardiovascular guinea pig (CVGP) assay for new chemical entities (NCEs) and; 2) Determine if a translational pharmacokinetic/pharmacodynamic (tPKPD) model can improve the predictive capacity. In silico simulations for NCEs were performed using a population of human ventricular action potential (AP) models. PatchXpress® (PX) or high throughput screening (HTS) ion channel data from respectively n = 73 and n = 51 NCEs were used as inputs for the in silico population. These NCEs were also tested in the CVGP (n = 73). An M5 pruned decision tree-based regression tPKPD model was used to evaluate the concentration at which an NCE is liable to prolong the QTc interval in the CVGP. In silico results successfully predicted the QTc interval prolongation outcome observed in the CVGP with an accuracy/specificity of 85%/73% and 75%/77%, when using PX and HTS ion channel data, respectively. Considering the tPKPD predicted concentration resulting in QTc prolongation (EC5%) increased accuracy/specificity to 97%/95% using PX and 88%/97% when using HTS. Our results support that human-based in silico simulations in combination with tPKPD modeling can provide correlative results with a commonly used early in vivo safety assay, suggesting a path toward more rapid NCE assessment with reduced resources, cycle time, and animal use. Cardiac electrophysiological in silico model predicts QTc interval prolongation in the guinea pig. PKPD model predicts relevant QTc interval prolongation concentration in guinea pig. Combining the models improves the accuracy of predicting guinea pig QTc effects. Combining models accelerates assessment of QTc with lower resources and animal use.
Collapse
Affiliation(s)
- Pierre Morissette
- Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA, USA.
| | - Sebastian Polak
- Certara UK Limited, Simcyp Division, Sheffield, UK; Jagiellonian University Medical College, Faculty of Pharmacy, Krakow, Poland
| | - Anne Chain
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism (PPDM), Merck & Co., Inc., Rahway, NJ, USA
| | - Jin Zhai
- Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA, USA
| | - John P Imredy
- Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA, USA
| | - Mary Jo Wildey
- Pharmacology, Screening and Informatics, Merck & Co., Kenilworth, NJ, USA
| | - Jeffrey Travis
- Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA, USA
| | - Kevin Fitzgerald
- Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA, USA
| | - Patrick Fanelli
- Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA, USA
| | - Elisa Passini
- Computational Cardiovascular Science Group, Department of Computer Science, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Blanca Rodriguez
- Computational Cardiovascular Science Group, Department of Computer Science, BHF Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Frederick Sannajust
- Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA, USA
| | - Christopher Regan
- Safety Assessment & Laboratory Animal Resources (SALAR), Merck & Co., Inc., West Point, PA, USA
| |
Collapse
|
17
|
Kurata Y, Tsumoto K, Hayashi K, Hisatome I, Kuda Y, Tanida M. Multiple Dynamical Mechanisms of Phase-2 Early Afterdepolarizations in a Human Ventricular Myocyte Model: Involvement of Spontaneous SR Ca 2+ Release. Front Physiol 2020; 10:1545. [PMID: 31998140 PMCID: PMC6965073 DOI: 10.3389/fphys.2019.01545] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 12/05/2019] [Indexed: 12/19/2022] Open
Abstract
Early afterdepolarization (EAD) is known to cause lethal ventricular arrhythmias in long QT syndrome (LQTS). In this study, dynamical mechanisms of EAD formation in human ventricular myocytes (HVMs) were investigated using the mathematical model developed by ten Tusscher and Panfilov (Am J Physiol Heart Circ Physiol 291, 2006). We explored how the rapid (IKr) and slow (IKs) components of delayed-rectifier K+ channel currents, L-type Ca2+ channel current (ICa L), Na+/Ca2+ exchanger current (INCX), and intracellular Ca2+ handling via the sarcoplasmic reticulum (SR) contribute to initiation, termination and modulation of phase-2 EADs during pacing in relation to bifurcation phenomena in non-paced model cells. Parameter-dependent dynamical behaviors of the non-paced model cell were determined by calculating stabilities of equilibrium points (EPs) and limit cycles, and bifurcation points to construct bifurcation diagrams. Action potentials (APs) and EADs during pacing were reproduced by numerical simulations for constructing phase diagrams of the paced model cell dynamics. Results are summarized as follows: (1) A modified version of the ten Tusscher-Panfilov model with accelerated ICaL inactivation could reproduce bradycardia-related EADs in LQTS type 2 and β-adrenergic stimulation-induced EADs in LQTS type 1. (2) Two types of EADs with different initiation mechanisms, ICaL reactivation-dependent and spontaneous SR Ca2+ release-mediated EADs, were detected. (3) Termination of EADs (AP repolarization) during pacing depended on the slow activation of IKs. (4) Spontaneous SR Ca2+ releases occurred at higher Ca2+ uptake rates, attributable to the instability of steady-state intracellular Ca2+ concentrations. Dynamical mechanisms of EAD formation and termination in the paced model cell are closely related to stability changes (bifurcations) in dynamical behaviors of the non-paced model cell, but they are model-dependent. Nevertheless, the modified ten Tusscher-Panfilov model would be useful for systematically investigating possible dynamical mechanisms of EAD-related arrhythmias in LQTS.
Collapse
Affiliation(s)
- Yasutaka Kurata
- Department of Physiology II, Kanazawa Medical University, Uchinada, Japan
| | - Kunichika Tsumoto
- Department of Physiology II, Kanazawa Medical University, Uchinada, Japan
| | - Kenshi Hayashi
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Ichiro Hisatome
- Department of Genetic Medicine and Regenerative Therapeutics, Graduate School of Medical Sciences, Tottori University, Yonago, Japan
| | - Yuhichi Kuda
- Department of Physiology II, Kanazawa Medical University, Uchinada, Japan
| | - Mamoru Tanida
- Department of Physiology II, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
18
|
Ginsenoside Rb1 exerts antiarrhythmic effects by inhibiting I Na and I CaL in rabbit ventricular myocytes. Sci Rep 2019; 9:20425. [PMID: 31892729 PMCID: PMC6938504 DOI: 10.1038/s41598-019-57010-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 12/19/2019] [Indexed: 12/18/2022] Open
Abstract
Ginsenoside Rb1 exerts its pharmacological action by regulating sodium, potassium and calcium ion channels in the membranes of nerve cells. These ion channels are also present in cardiomyocytes, but no studies have been reported to date regarding the effects of Rb1 on cardiac sodium currents (INa), L-type calcium currents (ICaL) and action potentials (APs). Additionally, the antiarrhythmic potential of Rb1 has not been assessed. In this study, we used a whole-cell patch clamp technique to assess the effect of Rb1 on these ion channels. The results showed that Rb1 inhibited INa and ICaL, reduced the action potential amplitude (APA) and maximum upstroke velocity (Vmax), and shortened the action potential duration (APD) in a concentration-dependent manner but had no effect on the inward rectifier potassium current (IK1), delayed rectifier potassium current (IK) or resting membrane potential (RMP). We also designed a pathological model at the cellular and organ level to verify the role of Rb1. The results showed that Rb1 abolished high calcium-induced delayed afterdepolarizations (DADs), depressed the increase in intracellular calcium ([Ca2+]i), relieved calcium overload and protected cardiomyocytes. Rb1 can also reduce the occurrence of ventricular premature beats (VPBs) and ventricular tachycardia (VT) in ischemia-reperfusion (I-R) injury.
Collapse
|
19
|
Choi BR, Li W, Terentyev D, Kabakov AY, Zhong M, Rees CM, Terentyeva R, Kim TY, Qu Z, Peng X, Karma A, Koren G. Transient Outward K + Current (I to) Underlies the Right Ventricular Initiation of Polymorphic Ventricular Tachycardia in a Transgenic Rabbit Model of Long-QT Syndrome Type 1. Circ Arrhythm Electrophysiol 2018; 11:e005414. [PMID: 29769222 PMCID: PMC6081959 DOI: 10.1161/circep.117.005414] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 03/21/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Sudden death in long-QT syndrome type 1 (LQT1), an inherited disease caused by loss-of-function mutations in KCNQ1, is triggered by early afterdepolarizations (EADs) that initiate polymorphic ventricular tachycardia (pVT). We investigated ionic mechanisms that underlie pVT in LQT1 using a transgenic rabbit model of LQT1. METHODS Optical mapping, cellular patch clamping, and computer modeling were used to elucidate the mechanisms of EADs in transgenic LQT1 rabbits. RESULTS The results showed that shorter action potential duration in the right ventricle (RV) was associated with focal activity during pVT initiation. RV cardiomyocytes demonstrated higher incidence of EADs under 50 nmol/L isoproterenol. Voltage-clamp studies revealed that the transient outward potassium current (Ito) magnitude was 28% greater in RV associated with KChiP2 but with no differences in terms of calcium-cycling kinetics and other sarcolemmal currents. Perfusing with the Ito blocker 4-aminopyridine changed the initial focal sites of pVT from the RV to the left ventricle, corroborating the role of Ito in pVT initiation. Computer modeling showed that EADs occur preferentially in the RV because of the larger conductance of the slow-inactivating component of Ito, which repolarizes the membrane potential sufficiently rapidly to allow reactivation of ICa,L before IKr has had sufficient time to activate. CONCLUSIONS Ito heterogeneity creates both triggers and an arrhythmogenic substrate in LQT1. In the absence of IKs, Ito interactions with ICa,L and IKr promote EADs in the RV while prolonging action potential duration in the left ventricle. This heterogeneity of action potential enhances dispersion of refractoriness and facilitates conduction blocks that initiate pVTs.
Collapse
Affiliation(s)
- Bum-Rak Choi
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence (B.-R.C., W.L., D.T., A.Y.K., R.T., T.Y.K., G.K.).
| | - Weiyan Li
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence (B.-R.C., W.L., D.T., A.Y.K., R.T., T.Y.K., G.K.)
| | - Dmitry Terentyev
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence (B.-R.C., W.L., D.T., A.Y.K., R.T., T.Y.K., G.K.)
| | - Anatoli Y Kabakov
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence (B.-R.C., W.L., D.T., A.Y.K., R.T., T.Y.K., G.K.)
| | - Mingwang Zhong
- Department of Physics, Northeastern University, Boston MA (M.Z., C.M.R., A.K.)
| | - Colin M Rees
- Department of Physics, Northeastern University, Boston MA (M.Z., C.M.R., A.K.)
| | - Radmila Terentyeva
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence (B.-R.C., W.L., D.T., A.Y.K., R.T., T.Y.K., G.K.)
| | - Tae Yun Kim
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence (B.-R.C., W.L., D.T., A.Y.K., R.T., T.Y.K., G.K.)
| | - Zhilin Qu
- Department of Medicine (Cardiology), University of California, Los Angeles (Z.Q.)
| | - Xuwen Peng
- Department of Comparative Medicine, Pennsylvania State University College of Medicine, Hershey (X.P.)
| | - Alain Karma
- Department of Physics, Northeastern University, Boston MA (M.Z., C.M.R., A.K.)
| | - Gideon Koren
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence (B.-R.C., W.L., D.T., A.Y.K., R.T., T.Y.K., G.K.).
| |
Collapse
|
20
|
Liu W, Kim TY, Huang X, Liu MB, Koren G, Choi BR, Qu Z. Mechanisms linking T-wave alternans to spontaneous initiation of ventricular arrhythmias in rabbit models of long QT syndrome. J Physiol 2018; 596:1341-1355. [PMID: 29377142 DOI: 10.1113/jp275492] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/23/2018] [Indexed: 01/23/2023] Open
Abstract
KEY POINTS T-wave alternans (TWA) and T-wave lability (TWL) are precursors of ventricular arrhythmias in long QT syndrome; however, the mechanistic link remains to be clarified. Computer simulations show that action potential duration (APD) prolongation and slowed heart rates promote APD alternans and chaos, manifesting as TWA and TWL, respectively. Regional APD alternans and chaos can exacerbate pre-existing or induce de novo APD dispersion, which combines with enhanced ICa,L to result in premature ventricular complexes (PVCs) originating from the APD gradient region. These PVCs can directly degenerate into re-entrant arrhythmias without the need for an additional tissue substrate or further exacerbate the APD dispersion to cause spontaneous initiation of ventricular arrhythmias. Experiments conducted in transgenic long QT rabbits show that PVC alternans occurs at slow heart rates, preceding spontaneous intuition of ventricular arrhythmias. ABSTRACT T-wave alternans (TWA) and irregular beat-to-beat T-wave variability or T-wave lability (TWL), the ECG manifestations of action potential duration (APD) alternans and variability, are precursors of ventricular arrhythmias in long QT syndromes. TWA and TWL in patients tend to occur at normal heart rates and are usually potentiated by bradycardia. Whether or how TWA and TWL at normal or slow heart rates are causally linked to arrhythmogenesis remains unknown. In the present study, we used computer simulations and experiments of a transgenic rabbit model of long QT syndrome to investigate the underlying mechanisms. Computer simulations showed that APD prolongation and slowed heart rates caused early afterdepolarization-mediated APD alternans and chaos, manifesting as TWA and TWL, respectively. Regional APD alternans and chaos exacerbated pre-existing APD dispersion and, in addition, APD chaos could also induce APD dispersion de novo via chaos desynchronization. Increased APD dispersion, combined with substantially enhanced ICa,L , resulted in a tissue-scale dynamical instability that gave rise to the spontaneous occurrence of unidirectionally propagating premature ventricular complexes (PVCs) originating from the APD gradient region. These PVCs could directly degenerate into re-entrant arrhythmias without the need for an additional tissue substrate or could block the following sinus beat to result in a longer RR interval, which further exacerbated the APD dispersion giving rise to the spontaneous occurrence of ventricular arrhythmias. Slow heart rate-induced PVC alternans was observed in experiments of transgenic LQT2 rabbits under isoproterenol, which was associated with increased APD dispersion and spontaneous occurrence of ventricular arrhythmias, in agreement with the theoretical predictions.
Collapse
Affiliation(s)
- Weiqing Liu
- Department of Medicine, University of California, Los Angeles, California, USA.,School of Science, Jiangxi University of Science and Technology, Ganzhou, China
| | - Tae Yun Kim
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Xiaodong Huang
- Department of Medicine, University of California, Los Angeles, California, USA.,Department of Physics, South China University of Technology, Guangzhou, China
| | - Michael B Liu
- Department of Medicine, University of California, Los Angeles, California, USA
| | - Gideon Koren
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Bum-Rak Choi
- Cardiovascular Research Center, Division of Cardiology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Zhilin Qu
- Department of Medicine, University of California, Los Angeles, California, USA.,Department of Biomathematics, University of California, Los Angeles, California, USA
| |
Collapse
|
21
|
Estradiol up-regulates L-type Ca 2+ channels via membrane-bound estrogen receptor/phosphoinositide-3-kinase/Akt/cAMP response element-binding protein signaling pathway. Heart Rhythm 2018; 15:741-749. [PMID: 29330129 DOI: 10.1016/j.hrthm.2018.01.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND In long QT syndrome type 2, women are more prone than men to the lethal arrhythmia torsades de pointes. We previously reported that 17β-estradiol (E2) up-regulates L-type Ca2+ channels and current (ICa,L) (∼30%) in rabbit ventricular myocytes by a classic genomic mechanism mediated by estrogen receptor-α (ERα). In long QT syndrome type 2 (IKr blockade or bradycardia), the higher Ca2+ influx via ICa,L causes Ca2+ overload, spontaneous sarcoplasmic reticulum Ca2+ release, and reactivation of ICa,L that triggers early afterdepolarizations and torsades de pointes. OBJECTIVE The purpose of this study was to investigate the molecular mechanisms whereby E2 up-regulates ICa,L, which are poorly understood. METHODS H9C2 and rat myocytes were incubated with E2 ± ER antagonist, or inhibitors of downstream transcription factors, for 24 hours, followed by western blots of Cav1.2α1C and voltage-clamp measurements of ICa,L. RESULTS Incubation of H9C2 cells with E2 (10-100 nM) increased ICa,L density and Cav1.2α1C expression, which were suppressed by the ER antagonist ICI182,780 (1 μM). Enhanced ICa,L and Cav1.2α1C expression by E2 was suppressed by inhibitors of phosphoinositide-3-kinase (Pi3K) (30 μM LY294002; P <.05) and Akt (5 μM MK2206) but not of mitogen-activated protein kinase (5 μM U0126) or protein kinase A (1 μM KT5720). E2 incubation increased p-CREB via the Pi3K/Akt pathway, reached a peak in 20 minutes (3-fold), and leveled off to 1.5-fold 24 hours later. Furthermore, a CREB decoy oligonucleotide inhibited E2-induced Cav1.2α1C expression, whereas membrane-impermeable E2 (E2-bovine serum albumin) was equally effective at Cav1.2α1C up-regulation as E2. CONCLUSION Estradiol up-regulates Cav1.2α1C and ICa,L via plasma membrane ER and by activating Pi3K, Akt, and CREB signaling. The promoter regions of the CACNA1C gene (human-rabbit-rat) contain adjacent/overlapping binding sites for p-CREB and ERα, which suggests a synergistic regulation by these pathways.
Collapse
|
22
|
Krogh-Madsen T, Jacobson AF, Ortega FA, Christini DJ. Global Optimization of Ventricular Myocyte Model to Multi-Variable Objective Improves Predictions of Drug-Induced Torsades de Pointes. Front Physiol 2017; 8:1059. [PMID: 29311985 PMCID: PMC5742183 DOI: 10.3389/fphys.2017.01059] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 12/04/2017] [Indexed: 01/22/2023] Open
Abstract
In silico cardiac myocyte models present powerful tools for drug safety testing and for predicting phenotypical consequences of ion channel mutations, but their accuracy is sometimes limited. For example, several models describing human ventricular electrophysiology perform poorly when simulating effects of long QT mutations. Model optimization represents one way of obtaining models with stronger predictive power. Using a recent human ventricular myocyte model, we demonstrate that model optimization to clinical long QT data, in conjunction with physiologically-based bounds on intracellular calcium and sodium concentrations, better constrains model parameters. To determine if the model optimized to congenital long QT data better predicts risk of drug-induced long QT arrhythmogenesis, in particular Torsades de Pointes risk, we tested the optimized model against a database of known arrhythmogenic and non-arrhythmogenic ion channel blockers. When doing so, the optimized model provided an improved risk assessment. In particular, we demonstrate an elimination of false-positive outcomes generated by the baseline model, in which simulations of non-torsadogenic drugs, in particular verapamil, predict action potential prolongation. Our results underscore the importance of currents beyond those directly impacted by a drug block in determining torsadogenic risk. Our study also highlights the need for rich data in cardiac myocyte model optimization and substantiates such optimization as a method to generate models with higher accuracy of predictions of drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Trine Krogh-Madsen
- Greenberg Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States.,Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States.,Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Anna F Jacobson
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Francis A Ortega
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Graduate School, New York, NY, United States
| | - David J Christini
- Greenberg Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States.,Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States.,Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
23
|
Colman MA, Perez Alday EA, Holden AV, Benson AP. Trigger vs. Substrate: Multi-Dimensional Modulation of QT-Prolongation Associated Arrhythmic Dynamics by a hERG Channel Activator. Front Physiol 2017; 8:757. [PMID: 29046643 PMCID: PMC5632683 DOI: 10.3389/fphys.2017.00757] [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] [Received: 08/02/2017] [Accepted: 09/19/2017] [Indexed: 11/13/2022] Open
Abstract
Background: Prolongation of the QT interval of the electrocardiogram (ECG), underlain by prolongation of the action potential duration (APD) at the cellular level, is linked to increased vulnerability to cardiac arrhythmia. Pharmacological management of arrhythmia associated with QT prolongation is typically achieved through attempting to restore APD to control ranges, reversing the enhanced vulnerability to Ca2+-dependent afterdepolarisations (arrhythmia triggers) and increased transmural dispersion of repolarisation (arrhythmia substrate) associated with APD prolongation. However, such pharmacological modulation has been demonstrated to have limited effectiveness. Understanding the integrative functional impact of pharmacological modulation requires simultaneous investigation of both the trigger and substrate. Methods: We implemented a multi-scale (cell and tissue) in silico approach using a model of the human ventricular action potential, integrated with a model of stochastic 3D spatiotemporal Ca2+ dynamics, and parameter modification to mimic prolonged QT conditions. We used these models to examine the efficacy of the hERG activator MC-II-157c in restoring APD to control ranges, examined its effects on arrhythmia triggers and substrates, and the interaction of these arrhythmia triggers and substrates. Results: QT prolongation conditions promoted the development of spontaneous release events underlying afterdepolarisations during rapid pacing. MC-II-157c applied to prolonged QT conditions shortened the APD, inhibited the development of afterdepolarisations and reduced the probability of afterdepolarisations manifesting as triggered activity in single cells. In tissue, QT prolongation resulted in an increased transmural dispersion of repolarisation, which manifested as an increased vulnerable window for uni-directional conduction block. In some cases, MC-II-157c further increased the vulnerable window through its effects on INa. The combination of stochastic release event modulation and transmural dispersion of repolarisation modulation by MC-II-157c resulted in an integrative behavior wherein the arrhythmia trigger is reduced but the arrhythmia substrate is increased, leading to variable and non-linear overall vulnerability to arrhythmia. Conclusion: The relative balance of reduced trigger and increased substrate underlies a multi-dimensional role of MC-II-157c in modulation of cardiac arrhythmia vulnerability associated with prolonged QT interval.
Collapse
Affiliation(s)
- Michael A Colman
- School of Biomedical Sciences and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Erick A Perez Alday
- Division of Cardiovascular Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Arun V Holden
- School of Biomedical Sciences and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Alan P Benson
- School of Biomedical Sciences and Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
24
|
Wilson D, Ermentrout B, Němec J, Salama G. A model of cardiac ryanodine receptor gating predicts experimental Ca 2+-dynamics and Ca 2+-triggered arrhythmia in the long QT syndrome. CHAOS (WOODBURY, N.Y.) 2017; 27:093940. [PMID: 28964110 DOI: 10.1063/1.5000711] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Abnormal Ca2+ handling is well-established as the trigger of cardiac arrhythmia in catecholaminergic polymorphic ventricular tachycardia and digoxin toxicity, but its role remains controversial in Torsade de Pointes (TdP), the arrhythmia associated with the long QT syndrome (LQTS). Recent experimental results show that early afterdepolarizations (EADs) that initiate TdP are caused by spontaneous (non-voltage-triggered) Ca2+ release from Ca2+-overloaded sarcoplasmic reticulum (SR) rather than the activation of the L-type Ca2+-channel window current. In bradycardia and long QT type 2 (LQT2), a second, non-voltage triggered cytosolic Ca2+ elevation increases gradually in amplitude, occurs before overt voltage instability, and then precedes the rise of EADs. Here, we used a modified Shannon-Puglisi-Bers model of rabbit ventricular myocytes to reproduce experimental Ca2+ dynamics in bradycardia and LQT2. Abnormal systolic Ca2+-oscillations and EADs caused by SR Ca2+-release are reproduced in a modified 0-dimensional model, where 3 gates in series control the ryanodine receptor (RyR2) conductance. Two gates control RyR2 activation and inactivation and sense cytosolic Ca2+ while a third gate senses luminal junctional SR Ca2+. The model predicts EADs in bradycardia and low extracellular [K+] and cessation of SR Ca2+-release terminate salvos of EADs. Ca2+-waves, systolic cell-synchronous Ca2+-release, and multifocal diastolic Ca2+ release seen in subcellular Ca2+-mapping experiments are observed in the 2-dimensional version of the model. These results support the role of SR Ca2+-overload, abnormal SR Ca2+-release, and the subsequent activation of the electrogenic Na+/Ca2+-exchanger as the mechanism of TdP. The model offers new insights into the genesis of cardiac arrhythmia and new therapeutic strategies.
Collapse
Affiliation(s)
- Dan Wilson
- Department of Mathematics, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Bard Ermentrout
- Department of Mathematics, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Jan Němec
- Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Guy Salama
- Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| |
Collapse
|
25
|
Al-Zaiti SS, Alrawashdeh M, Martin-Gill C, Callaway C, Mortara D, Nemec J. Evaluation of beat-to-beat ventricular repolarization lability from standard 12-lead ECG during acute myocardial ischemia. J Electrocardiol 2017; 50:717-724. [PMID: 28916174 DOI: 10.1016/j.jelectrocard.2017.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Acute myocardial ischemia is a common cause of ventricular arrhythmias, yet recent ECG methods predicting susceptibility to ventricular tachyarrhythmia have not been fully evaluated during spontaneous ischemia. We sought to evaluate the clinical utility of alternans and non-alternans components of repolarization variability from the standard 10-second 12-lead ECG signals to risk stratify patients with acute chest pain. METHODS We enrolled consecutive, non-traumatic, chest pain patients transported through Emergency Medical Services (EMS) to three tertiary care hospitals with cardiac catheterization lab capabilities in Pittsburgh, PA. ECG signals were manually annotated by an electrophysiologist, then automatically processed using a custom-written software. Both T wave alternans (TWA) and non-alternans repolarization variability (NARV) were calculated using the absolute RMS differences over the repolarization window between odd/even averaged beats and between consecutive averaged pairs, respectively. The primary study outcome was the presence of acute myocardial infarction (AMI) documented by cardiac angiography. RESULTS After excluding patients with secondary repolarization changes (n=123) and those with excessive noise (n=90), our final sample included 537 patients (age 57±16years, 56% males). Patients with AMI (n=47, 9%) had higher TWA and NARV values (p<0.01). Mean RR correlated with TWA, and noise measures correlated with TWA and NARV, after adjusting for potential confounders. There was a high collinearity between TWA and NARV, and each was separately predictive of AMI after controlling for number of analyzed beats, noise measures, and other clinical variables. CONCLUSIONS Despite limitations imposed by signal quality, TWA and NARV are higher in patients with AMI, even after correction for potential confounders. The clinical value of TWA and NARV derived from standard ECG using our time-domain RMS method is questionable due to the small number of beats and significant noise.
Collapse
Affiliation(s)
- Salah S Al-Zaiti
- Department of Acute and Tertiary Care Nursing, University of Pittsburgh, Pittsburgh, PA, United States; Department of Emergency Medicine, University of Pittsburgh, Pittsburgh, PA, United States.
| | - Mohammad Alrawashdeh
- Department of Acute and Tertiary Care Nursing, University of Pittsburgh, Pittsburgh, PA, United States
| | - Christian Martin-Gill
- Department of Emergency Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Clifton Callaway
- Department of Emergency Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - David Mortara
- UCSF School of Nursing, San Francisco, CA, United States
| | - Jan Nemec
- Department of Cardiac Electrophysiology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
26
|
Schrantee A, Václavů L, Reneman L, Verberne HJ, Booij J, Tan HL. QT prolongation by dexamphetamine: Does experience matter? J Cardiovasc Electrophysiol 2017; 28:912-916. [PMID: 28452189 DOI: 10.1111/jce.13235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/23/2017] [Accepted: 04/12/2017] [Indexed: 12/01/2022]
Abstract
INTRODUCTION Case reports of life-threatening cardiac arrhythmias and sudden cardiac arrest (SCA) among amphetamine users have raised serious concerns about the cardiac safety of this class of drugs. This is important in light of the high prevalence of dexamphetamine (dAMPH) prescription for attention-deficit/hyperactivity disorder (ADHD), and its rising use as a recreational drug. The objective was to investigate electrocardiogram (ECG) parameters upon intravenous administration of a single dAMPH dose in habitual recreational dAMPH users (users) and healthy gender/age/ intelligence-quotient-matched controls (non-users). METHODS AND RESULTS ECG recordings were made in 18 users and 18 non-users during administration of dAMPH (0.3 mg/kg body weight). Baseline ECG was normal in both groups. dAMPH elicited increased heart rate and corrected QT time (QTc) prolongation in both groups (all P < 0.001, QTc = 502 in one individual). QTc prolongation was attenuated in users compared to non-users, exhibiting a significant interaction effect (P = 0.04). CONCLUSION SCA associated with amphetamine use may be related to its QTc prolonging effects, particularly during first-time use. These observations may provide a rationale for conducting ECG analysis immediately after the first-time use of amphetamines, as this could potentially unmask vulnerable individuals.
Collapse
Affiliation(s)
- Anouk Schrantee
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Lena Václavů
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Liesbeth Reneman
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hein J Verberne
- Department of Nuclear Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan Booij
- Department of Nuclear Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hanno L Tan
- Department of Cardiology, Heart Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Wei XH, Yu SD, Ren L, Huang SH, Yang QM, Wang P, Chu YP, Yang W, Ding YS, Huo Y, Wu L. Inhibition of late sodium current suppresses calcium-related ventricular arrhythmias by reducing the phosphorylation of CaMK-II and sodium channel expressions. Sci Rep 2017; 7:981. [PMID: 28428622 PMCID: PMC5430524 DOI: 10.1038/s41598-017-01056-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 03/20/2017] [Indexed: 12/19/2022] Open
Abstract
Cardiac arrhythmias associated with intracellular calcium inhomeostasis are refractory to antiarrhythmic therapy. We hypothesized that late sodium current (I Na) contributed to the calcium-related arrhythmias. Monophasic action potential duration at 90% completion of repolarization (MAPD90) was significantly increased and ventricular arrhythmias were observed in hearts with increased intracellular calcium concentration ([Ca2+]i) by using Bay K 8644, and the increase became greater in hearts treated with a combination of ATX-II and Bay K 8644 compared to Bay K 8644 alone. The prolongations caused by Bay K 8644 and frequent episodes of ventricular tachycardias, both in absence and presence of ATX-II, were significantly attenuated or abolished by late I Na inhibitors TTX and eleclazine. In rabbit ventricular myocytes, Bay K 8644 increased I CaL density, calcium transient and myocyte contraction. TTX and eleclazine decreased the amplitude of late I Na, the reverse use dependence of MAPD90 at slower heart rate, and attenuated the increase of intracellular calcium transient and myocyte contraction. TTX diminished the phosphorylation of CaMKII-δ and Nav 1.5 in hearts treated with Bay K 8644 and ATX-II. In conclusion, late I Na contributes to ventricular arrhythmias and its inhibition is plausible to treat arrhythmias in hearts with increased [Ca2+]i.
Collapse
Affiliation(s)
- Xiao-Hong Wei
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Shan-Dong Yu
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Lu Ren
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Si-Hui Huang
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Qiao-Mei Yang
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Ping Wang
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China
| | - Yan-Peng Chu
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Wei Yang
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Yan-Sheng Ding
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China
| | - Yong Huo
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China. .,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China.
| | - Lin Wu
- Department of Cardiology, Peking University First Hospital, Beijing, 100034, China. .,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, 100191, China.
| |
Collapse
|
28
|
Quinn TA, Ripplinger CM. Recent developments in biophysics & molecular biology of heart rhythm. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 120:1-2. [PMID: 26777585 DOI: 10.1016/j.pbiomolbio.2016.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Affiliation(s)
- T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Canada.
| | | |
Collapse
|
29
|
Zile MA, Trayanova NA. Rate-dependent force, intracellular calcium, and action potential voltage alternans are modulated by sarcomere length and heart failure induced-remodeling of thin filament regulation in human heart failure: A myocyte modeling study. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2015; 120:270-80. [PMID: 26724571 DOI: 10.1016/j.pbiomolbio.2015.12.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/24/2015] [Accepted: 12/23/2015] [Indexed: 01/26/2023]
Abstract
Microvolt T-wave alternans (MTWA) testing identifies heart failure patients at risk for lethal ventricular arrhythmias at near-resting heart rates (<110 beats per minute). Since pressure alternans occurs simultaneously with MTWA and has a higher signal to noise ratio, it may be a better predictor of arrhythmia, although the mechanism remains unknown. Therefore, we investigated the relationship between force alternans (FORCE-ALT), the cellular manifestation of pressure alternans, and action potential voltage alternans (APV-ALT), the cellular driver of MTWA. Our goal was to uncover the mechanisms linking APV-ALT and FORCE-ALT in failing human myocytes and to investigate how the link between those alternans was affected by pacing rate and by physiological conditions such as sarcomere length and heart failure induced-remodeling of mechanical parameters. To achieve this, a mechanically-based, strongly coupled human electromechanical myocyte model was constructed. Reducing the sarcoplasmic reticulum calcium uptake current (Iup) to 27% was incorporated to simulate abnormal calcium handling in human heart failure. Mechanical remodeling was incorporated to simulate altered thin filament activation and crossbridge (XB) cycling rates. A dynamical pacing protocol was used to investigate the development of intracellular calcium concentration ([Ca]i), voltage, and active force alternans at different pacing rates. FORCE-ALT only occurred in simulations incorporating reduced Iup, demonstrating that alternans in the intracellular calcium concentration (CA-ALT) induced FORCE-ALT. The magnitude of FORCE-ALT was found to be largest at clinically relevant pacing rates (<110 bpm), where APV-ALT was smallest. We found that the magnitudes of FORCE-ALT, CA-ALT and APV-ALT were altered by heart failure induced-remodeling of mechanical parameters and sarcomere length due to the presence of myofilament feedback. These findings provide important insight into the relationship between heart-failure-induced electrical and mechanical alternans and how they are altered by physiological conditions at near-resting heart rates.
Collapse
Affiliation(s)
- Melanie A Zile
- Institute for Computational Medicine and Department of Biomedical Engineering at Johns Hopkins University, 3400N Charles St, 316 Hackerman Hall, Baltimore, MD 21218, USA.
| | - Natalia A Trayanova
- Institute for Computational Medicine and Department of Biomedical Engineering at Johns Hopkins University, 3400N Charles St, 316 Hackerman Hall, Baltimore, MD 21218, USA.
| |
Collapse
|
30
|
Lee W, Mann SA, Windley MJ, Imtiaz MS, Vandenberg JI, Hill AP. In silico assessment of kinetics and state dependent binding properties of drugs causing acquired LQTS. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2015; 120:89-99. [PMID: 26713558 DOI: 10.1016/j.pbiomolbio.2015.12.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/06/2015] [Accepted: 12/16/2015] [Indexed: 11/24/2022]
Abstract
The Kv11.1 or hERG potassium channel is responsible for one of the major repolarising currents (IKr) in cardiac myocytes. Drug binding to hERG can result in reduction in IKr, action potential prolongation, acquired long QT syndrome and fatal cardiac arrhythmias. The current guidelines for pre-clinical assessment of drugs in development is based on the measurement of the drug concentration that causes 50% current block, i.e., IC50. However, drugs with the same apparent IC50 may have very different kinetics of binding and unbinding, as well as different affinities for the open and inactivated states of Kv11.1. Therefore, IC50 measurements may not reflect the true risk of drug induced arrhythmias. Here we have used an in silico approach to test the hypothesis that drug binding kinetics and differences in state-dependent affinity will influence the extent of cardiac action potential prolongation independent of apparent IC50 values. We found, in general that drugs with faster overall kinetics and drugs with higher affinity for the open state relative to the inactivated state cause more action potential prolongation. These characteristics of drug-hERG interaction are likely to be more arrhythmogenic but cannot be predicted by IC50 measurement alone. Our results suggest that the pre-clinical assessment of Kv11.1-drug interactions should include descriptions of the kinetics and state dependence of drug binding. Further, incorporation of this information into sophisticated in silico models should be able to better predict arrhythmia risk and therefore more accurately assess safety of new drugs in development.
Collapse
Affiliation(s)
- William Lee
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, NSW 2052, Australia
| | - Stefan A Mann
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, NSW 2052, Australia
| | - Monique J Windley
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia
| | - Mohammad S Imtiaz
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, NSW 2052, Australia
| | - Jamie I Vandenberg
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, NSW 2052, Australia
| | - Adam P Hill
- Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, NSW 2052, Australia.
| |
Collapse
|