1
|
Yao B, Lei Z, Gonçalves MAFV, Sluijter JPG. Integrating Prime Editing and Cellular Reprogramming as Novel Strategies for Genetic Cardiac Disease Modeling and Treatment. Curr Cardiol Rep 2024; 26:1197-1208. [PMID: 39259489 DOI: 10.1007/s11886-024-02118-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/12/2024] [Indexed: 09/13/2024]
Abstract
PURPOSE OF REVIEW This review aims to evaluate the potential of CRISPR-based gene editing tools, particularly prime editors (PE), in treating genetic cardiac diseases. It seeks to answer how these tools can overcome current therapeutic limitations and explore the synergy between PE and induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) for personalized medicine. RECENT FINDINGS Recent advancements in CRISPR technology, including CRISPR-Cas9, base editors, and PE, have demonstrated precise genome correction capabilities. Notably, PE has shown exceptional precision in correcting genetic mutations. Combining PE with iPSC-CMs has emerged as a robust platform for disease modeling and developing innovative treatments for genetic cardiac diseases. The review finds that PE, when combined with iPSC-CMs, holds significant promise for treating genetic cardiac diseases by addressing their root causes. This approach could revolutionize personalized medicine, offering more effective and precise treatments. Future research should focus on refining these technologies and their clinical applications.
Collapse
Affiliation(s)
- Bing Yao
- Experimental Cardiology Laboratory, Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Circulatory Health Research Center, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Zhiyong Lei
- Experimental Cardiology Laboratory, Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Circulatory Health Research Center, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Manuel A F V Gonçalves
- Department of Cell and Chemical Biology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands.
- Regenerative Medicine Center Utrecht, Circulatory Health Research Center, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
2
|
Vouloagkas I, Agbariah A, Zegkos T, Gossios TD, Tziomalos G, Parcharidou D, Didagelos M, Kamperidis V, Ziakas A, Efthimiadis GK. The many faces of SCN5A pathogenic variants: from channelopathy to cardiomyopathy. Heart Fail Rev 2024:10.1007/s10741-024-10459-x. [PMID: 39465469 DOI: 10.1007/s10741-024-10459-x] [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] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
The SCN5A gene encodes the alpha subunit of the cardiac sodium channel, which plays a fundamental role in the generation and propagation of the action potential in the heart muscle. During the past years our knowledge concerning the function of the cardiac sodium channel and the diseases caused by mutations of the SCN5A gene has grown. Although initially SCN5A pathogenic variants were mainly associated with channelopathies, increasing recent evidence suggests an association with structural heart disease in the form of heart muscle disease. The pathways leading to a cardiomyopathic phenotype remain unclear and require further elucidation. The aim of the present review is to provide a concise summary regarding the mechanisms through which SCN5A pathogenic variants result in heart disease, focusing in cardiomyopathy, highlighting along the way the complex role of the SCN5A gene at the intersection of cardiac excitability and contraction networks.
Collapse
Affiliation(s)
- Ioannis Vouloagkas
- Department of Medicine, Cantonal Hospital of Winterthur, Winterthur, Switzerland
| | - Andrea Agbariah
- Department of Cardiology, Università Degli Studi Di Verona, Verona, Italy
| | - Thomas Zegkos
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Thomas D Gossios
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece.
| | - Georgios Tziomalos
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Despoina Parcharidou
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Matthaios Didagelos
- 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Vasileios Kamperidis
- 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Antonios Ziakas
- 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Georgios K Efthimiadis
- Cardiomyopathies Laboratory, 1st Aristotle University of Thessaloniki Cardiology Department, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
3
|
Ágoston M, Kohajda Z, Virág L, Baláti B, Nagy N, Lengyel C, Bitay M, Bogáts G, Vereckei A, Papp JG, Varró A, Jost N. A Comparative Study of the Rapid (I Kr) and Slow (I Ks) Delayed Rectifier Potassium Currents in Undiseased Human, Dog, Rabbit, and Guinea Pig Cardiac Ventricular Preparations. Pharmaceuticals (Basel) 2024; 17:1091. [PMID: 39204196 PMCID: PMC11357539 DOI: 10.3390/ph17081091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/24/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
To understand the large inter-species variations in drug effects on repolarization, the properties of the rapid (IKr) and the slow (IKs) components of the delayed rectifier potassium currents were compared in myocytes isolated from undiseased human donor (HM), dog (DM), rabbit (RM) and guinea pig (GM) ventricles by applying the patch clamp and conventional microelectrode techniques at 37 °C. The amplitude of the E-4031-sensitive IKr tail current measured at -40 mV after a 1 s long test pulse of 20 mV, which was very similar in HM and DM but significant larger in RM and GM. The L-735,821-sensitive IKs tail current was considerably larger in GM than in RM. In HM, the IKs tail was even smaller than in DM. At 30 mV, the IKr component was activated extremely rapidly and monoexponentially in each studied species. The deactivation of the IKr component in HM, DM, and RM measured at -40 mV. After a 30 mV pulse, it was slow and biexponential, while in GM, the IKr tail current was best fitted triexponentially. At 30 mV, the IKs component activated slowly and had an apparent monoxponential time course in HM, DM, and RM. In contrast, in GM, the activation was clearly biexponential. In HM, DM, and RM, IKs component deactivation measured at -40 mV was fast and monoexponential, while in GM, in addition to the fast component, another slower component was also revealed. These results suggest that the IK in HM resembles that measured in DM and RM and considerably differs from that observed in GM. These findings suggest that the dog and rabbit are more appropriate species than the guinea pig for preclinical evaluation of new potential drugs expected to affect cardiac repolarization.
Collapse
Affiliation(s)
- Márta Ágoston
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, P.O. Box 427, 6701 Szeged, Hungary
| | - Zsófia Kohajda
- HUN-REN-SZTE Research Group for Cardiovascular Pharmacology, 6701 Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, P.O. Box 427, 6701 Szeged, Hungary
- Interdisciplinary Research and Development and Innovation Centre of Excellence, University of Szeged, 6720 Szeged, Hungary
| | - Beáta Baláti
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, P.O. Box 427, 6701 Szeged, Hungary
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, P.O. Box 427, 6701 Szeged, Hungary
- HUN-REN-SZTE Research Group for Cardiovascular Pharmacology, 6701 Szeged, Hungary
| | - Csaba Lengyel
- Department of Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Miklós Bitay
- Department of Cardiac Surgery, Albert Szent-Györgyi Medical School, University of Szeged, 6742 Szeged, Hungary
| | - Gábor Bogáts
- Department of Cardiac Surgery, Albert Szent-Györgyi Medical School, University of Szeged, 6742 Szeged, Hungary
| | - András Vereckei
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Julius Gy. Papp
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, P.O. Box 427, 6701 Szeged, Hungary
- HUN-REN-SZTE Research Group for Cardiovascular Pharmacology, 6701 Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, P.O. Box 427, 6701 Szeged, Hungary
- HUN-REN-SZTE Research Group for Cardiovascular Pharmacology, 6701 Szeged, Hungary
- Interdisciplinary Research and Development and Innovation Centre of Excellence, University of Szeged, 6720 Szeged, Hungary
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, P.O. Box 427, 6701 Szeged, Hungary
- HUN-REN-SZTE Research Group for Cardiovascular Pharmacology, 6701 Szeged, Hungary
- Interdisciplinary Research and Development and Innovation Centre of Excellence, University of Szeged, 6720 Szeged, Hungary
| |
Collapse
|
4
|
Byun JY, Han S, Qdaisat A, Park C. Long QT syndrome after using EGFR-TKIs in older patients with advanced non-small cell lung cancer. Expert Opin Drug Saf 2024; 23:1007-1015. [PMID: 38088244 DOI: 10.1080/14740338.2023.2294924] [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: 05/28/2023] [Accepted: 10/31/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Long QT syndrome (LQTS) has been reported in older patients with advanced non-small cell lung cancer (NSCLC) following the use of osimertinib, the third-generation epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI). However, there have not been analytic epidemiology studies on this topic. We aimed to compare the risk of LQTS between osimertinib and first/second-generation EGFR-TKIs in older patients with advanced NSCLC. RESEARCH DESIGN AND METHODS This retrospective observational study used the 2006-2019 Surveillance, Epidemiology, and End Results (SEER)-Medicare data and included older patients with advanced NSCLC who were treated with either osimertinib or first/second-generation EGFR-TKIs during 2007-2017. Inverse probability of treatment weighting (IPTW) was used to balance the two groups with propensity scores estimated based on the patients' socioeconomic and clinical characteristics. Crude incidence rate (IR) and adjusted hazard ratio (HR) of the primary outcome, incident LQTS, were estimated. RESULTS A total of 545 and 1,135 patients were included in the osimertinib and first/second-generation EGFR-TKI groups, which increased to 1,614 and 1,659, respectively, after IPTW. The osimertinib group had a higher IR of LQTS (2.62 per 100 person-years, 95% CI 2.03-3.38) compared to the first/second-generation EGFR-TKI group (1.33 per 100 person-years, 95% CI 0.92-1.92). After adjusting for covariates, the osimertinib group had a higher risk of LQTS than the first/second-generation EGFR-TKI group, with an HR of 1.94 (95% CI 1.23-3.08). The increased LQTS risk in the osimertinib group was even higher in females, whites and patients aged ≥ 75. CONCLUSIONS Given the elevated risk of LQTS associated with osimertinib user, close monitoring for cardiac rhythm irregularities of high-risk patients following initiation of EGFR-TKI is recommended.
Collapse
Affiliation(s)
- Joo-Young Byun
- Health Outcomes division, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Sola Han
- Health Outcomes division, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| | - Aiham Qdaisat
- Department of Emergency Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chanhyun Park
- Health Outcomes division, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
5
|
Stevens-Sostre WA, Flores-Aldama L, Bustos D, Li J, Morais-Cabral JH, Delemotte L, Robertson GA. An intracellular hydrophobic nexus critical for hERG1 channel slow deactivation. Biophys J 2024; 123:2024-2037. [PMID: 38219015 PMCID: PMC11309987 DOI: 10.1016/j.bpj.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/17/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024] Open
Abstract
Slow deactivation is a critical property of voltage-gated K+ channels encoded by the human Ether-à-go-go-Related Gene 1 (hERG). hERG1 channel deactivation is modulated by interactions between intracellular N-terminal Per-Arnt-Sim (PAS) and C-terminal cyclic nucleotide-binding homology (CNBh) domains. The PAS domain is multipartite, comprising a globular domain (gPAS; residues 26-135) and an N-terminal PAS-cap that is further subdivided into an initial unstructured "tip" (residues 1-12) and an amphipathic α-helical region (residues 13-25). Although the PAS-cap tip has long been considered the effector of slow deactivation, how its position near the gating machinery is controlled has not been elucidated. Here, we show that a triad of hydrophobic interactions among the gPAS, PAS-cap α helix, and the CNBh domains is required to support slow deactivation in hERG1. The primary sequence of this "hydrophobic nexus" is highly conserved among mammalian ERG channels but shows key differences to fast-deactivating Ether-à-go-go 1 (EAG1) channels. Combining sequence analysis, structure-directed mutagenesis, electrophysiology, and molecular dynamics simulations, we demonstrate that polar serine substitutions uncover an intermediate deactivation mode that is also mimicked by deletion of the PAS-cap α helix. Molecular dynamics simulation analyses of the serine-substituted channels show an increase in distance among the residues of the hydrophobic nexus, a rotation of the intracellular gating ring, and a retraction of the PAS-cap tip from its receptor site near the voltage sensor domain and channel gate. These findings provide compelling evidence that the hydrophobic nexus coordinates the respective components of the intracellular gating ring and positions the PAS-cap tip to control hERG1 deactivation gating.
Collapse
Affiliation(s)
- Whitney A Stevens-Sostre
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Lisandra Flores-Aldama
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Daniel Bustos
- Centro de Investigación de Estudios Avanzados Del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica Del Maule, Talca, Chile; Laboratorio de Bioinformática y Química Computacional (LBQC), Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica Del Maule, Talca, Chile
| | - Jin Li
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - João H Morais-Cabral
- Instituto de Investigação e Inovação Em Saude da Universidade Do Porto (i3S); Instituto de Biologia Molecular e Celular, Universidade Do Porto, Porto, Portugal
| | - Lucie Delemotte
- KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
| | - Gail A Robertson
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
6
|
Odening KE. Interactions between KCNQ1 and KCNH2 may modulate the long QT type 1 phenotype. Cardiovasc Res 2024; 120:673-674. [PMID: 38630861 DOI: 10.1093/cvr/cvae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 04/19/2024] Open
Affiliation(s)
- Katja E Odening
- Translational Cardiology, Department of Cardiology and Department of Physiology, University Hospital Bern, University of Bern, Bühlplatz 5, 3012 Bern, Switzerland
| |
Collapse
|
7
|
DePriest J, Nixon J. The Use of a Checklist to Optimize Electrolyte Replacement in the ICU. Am J Med Qual 2024; 39:118-122. [PMID: 38713599 DOI: 10.1097/jmq.0000000000000177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2024]
Abstract
Electrolyte replacement protocols are routinely used in intensive care units (ICU) to guide magnesium replacement. Guided by serum levels, these protocols include no patient-specific factors despite a literature showing ICU patients routinely have significant deficits despite normal serum levels. The authors developed a checklist to help identify patients requiring more aggressive magnesium replacement than the electrolyte replacement protocol would provide. The checklist included risk factors for having significant magnesium deficits and for developing arrhythmias. The checklist was retrospectively applied to 364 medical ICU patients. Diabetic patients prescribed outpatient diuretics were defined as the highest-risk population. A total of 88% of patients in this subgroup had normal magnesium levels. Despite averaging 3.4 risk factors per patient, only 3 of 32 patients received magnesium. Applying the checklist would have suggested additional repletion for at least 85% of patients. A checklist can help identify ICU patients who may require more aggressive magnesium supplementation than protocols will provide.
Collapse
Affiliation(s)
- Jack DePriest
- Department of Internal Medicine, Division of Pulmonary, Sleep and Critical Care Medicine, Prisma Health Midland-University of South Carolina School of Medicine, Columbia, SC
| | - Joanna Nixon
- Department of Pharmacy, University of Kentucky HealthCare, Lexington, KY
| |
Collapse
|
8
|
Zhang H, Tarabanis C, Jethani N, Goldstein M, Smith S, Chinitz L, Ranganath R, Aphinyanaphongs Y, Jankelson L. QTNet: Predicting Drug-Induced QT Prolongation With Artificial Intelligence-Enabled Electrocardiograms. JACC Clin Electrophysiol 2024; 10:956-966. [PMID: 38703162 DOI: 10.1016/j.jacep.2024.01.022] [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: 08/24/2023] [Revised: 01/19/2024] [Accepted: 01/31/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND Prediction of drug-induced long QT syndrome (diLQTS) is of critical importance given its association with torsades de pointes. There is no reliable method for the outpatient prediction of diLQTS. OBJECTIVES This study sought to evaluate the use of a convolutional neural network (CNN) applied to electrocardiograms (ECGs) to predict diLQTS in an outpatient population. METHODS We identified all adult outpatients newly prescribed a QT-prolonging medication between January 1, 2003, and March 31, 2022, who had a 12-lead sinus ECG in the preceding 6 months. Using risk factor data and the ECG signal as inputs, the CNN QTNet was implemented in TensorFlow to predict diLQTS. RESULTS Models were evaluated in a held-out test dataset of 44,386 patients (57% female) with a median age of 62 years. Compared with 3 other models relying on risk factors or ECG signal or baseline QTc alone, QTNet achieved the best (P < 0.001) performance with a mean area under the curve of 0.802 (95% CI: 0.786-0.818). In a survival analysis, QTNet also had the highest inverse probability of censorship-weighted area under the receiver-operating characteristic curve at day 2 (0.875; 95% CI: 0.848-0.904) and up to 6 months. In a subgroup analysis, QTNet performed best among males and patients ≤50 years or with baseline QTc <450 ms. In an external validation cohort of solely suburban outpatient practices, QTNet similarly maintained the highest predictive performance. CONCLUSIONS An ECG-based CNN can accurately predict diLQTS in the outpatient setting while maintaining its predictive performance over time. In the outpatient setting, our model could identify higher-risk individuals who would benefit from closer monitoring.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Population Health, NYU Langone Health, New York University School of Medicine, New York, New York, USA.
| | - Constantine Tarabanis
- Leon H. Charney Division of Cardiology, Cardiac Electrophysiology, NYU Langone Health, New York University School of Medicine, New York, New York, USA
| | - Neil Jethani
- Department of Population Health, NYU Langone Health, New York University School of Medicine, New York, New York, USA; Courant Institute of Mathematical Sciences, New York University, New York, New York, USA
| | - Mark Goldstein
- Courant Institute of Mathematical Sciences, New York University, New York, New York, USA
| | - Silas Smith
- Ronald O. Perelman Department of Emergency Medicine, NYU Langone Health, New York, New York, USA
| | - Larry Chinitz
- Leon H. Charney Division of Cardiology, Cardiac Electrophysiology, NYU Langone Health, New York University School of Medicine, New York, New York, USA
| | - Rajesh Ranganath
- Department of Population Health, NYU Langone Health, New York University School of Medicine, New York, New York, USA; Courant Institute of Mathematical Sciences, New York University, New York, New York, USA
| | - Yindalon Aphinyanaphongs
- Department of Population Health, NYU Langone Health, New York University School of Medicine, New York, New York, USA
| | - Lior Jankelson
- Leon H. Charney Division of Cardiology, Cardiac Electrophysiology, NYU Langone Health, New York University School of Medicine, New York, New York, USA.
| |
Collapse
|
9
|
Mohammed ASA, Mohácsi G, Naveed M, Prorok J, Jost N, Virág L, Baczkó I, Topal L, Varró A. Cellular electrophysiological effects of the citrus flavonoid hesperetin in dog and rabbit cardiac ventricular preparations. Sci Rep 2024; 14:7237. [PMID: 38538818 PMCID: PMC10973458 DOI: 10.1038/s41598-024-57828-y] [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: 01/10/2024] [Accepted: 03/21/2024] [Indexed: 05/01/2024] Open
Abstract
Recent experimental data shows that hesperetin, a citrus flavonoid, affects potassium channels and can prolong the QTc interval in humans. Therefore, in the present study we investigated the effects of hesperetin on various transmembrane ionic currents and on ventricular action potentials. Transmembrane current measurements and action potential recordings were performed by patch-clamp and the conventional microelectrode techniques in dog and rabbit ventricular preparations. At 10 µM concentration hesperetin did not, however, at 30 µM significantly decreased the amplitude of the IK1, Ito, IKr potassium currents. Hesperetin at 3-30 µM significantly and in a concentration-dependent manner reduced the amplitude of the IKs current. The drug significantly decreased the amplitudes of the INaL and ICaL currents at 30 µM. Hesperetin (10 and 30 µM) did not change the action potential duration in normal preparations, however, in preparations where the repolarization reserve had been previously attenuated by 100 nM dofetilide and 1 µg/ml veratrine, caused a moderate but significant prolongation of repolarization. These results suggest that hesperetin at close to relevant concentrations inhibits the IKs outward potassium current and thereby reduces repolarization reserve. This effect in certain specific situations may prolong the QT interval and consequently may enhance proarrhythmic risk.
Collapse
Affiliation(s)
- Aiman Saleh A Mohammed
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi School of Medicine, University of Szeged, Szeged, Hungary
| | - Gábor Mohácsi
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi School of Medicine, University of Szeged, Szeged, Hungary
| | - Muhammad Naveed
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi School of Medicine, University of Szeged, Szeged, Hungary
| | - János Prorok
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi School of Medicine, University of Szeged, Szeged, Hungary
- HUN-REN-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Research Network, Szeged, Hungary
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi School of Medicine, University of Szeged, Szeged, Hungary
- HUN-REN-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Research Network, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi School of Medicine, University of Szeged, Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi School of Medicine, University of Szeged, Szeged, Hungary.
- HUN-REN-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Research Network, Szeged, Hungary.
| | - Leila Topal
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi School of Medicine, University of Szeged, Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi School of Medicine, University of Szeged, Szeged, Hungary.
- HUN-REN-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Research Network, Szeged, Hungary.
| |
Collapse
|
10
|
Fullerton KE, Clark AP, Krogh-Madsen T, Christini DJ. Optimization of a cardiomyocyte model illuminates role of increased INa,L in repolarization reserve. Am J Physiol Heart Circ Physiol 2024; 326:H334-H345. [PMID: 38038718 DOI: 10.1152/ajpheart.00553.2023] [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: 09/05/2023] [Revised: 11/15/2023] [Accepted: 11/19/2023] [Indexed: 12/02/2023]
Abstract
Cardiac ion currents may compensate for each other when one is compromised by a congenital or drug-induced defect. Such redundancy contributes to a robust repolarization reserve that can prevent the development of lethal arrhythmias. Most efforts made to describe this phenomenon have quantified contributions by individual ion currents. However, it is important to understand the interplay between all major ion-channel conductances, as repolarization reserve is dependent on the balance between all ion currents in a cardiomyocyte. Here, a genetic algorithm was designed to derive profiles of nine ion-channel conductances that optimize repolarization reserve in a mathematical cardiomyocyte model. Repolarization reserve was quantified using a previously defined metric, repolarization reserve current, i.e., the minimum constant current to prevent normal action potential repolarization in a cell. The optimization improved repolarization reserve current up to 84% compared to baseline in a human adult ventricular myocyte model and increased resistance to arrhythmogenic insult. The optimized conductance profiles were not only characterized by increased repolarizing current conductances but also uncovered a previously unreported behavior by the late sodium current. Simulations demonstrated that upregulated late sodium increased action potential duration, without compromising repolarization reserve current. The finding was generalized to multiple models. Ultimately, this computational approach, in which multiple currents were studied simultaneously, illuminated mechanistic insights into how the metric's magnitude could be increased and allowed for the unexpected role of late sodium to be elucidated.NEW & NOTEWORTHY Genetic algorithms are typically used to fit models or extract desired parameters from data. Here, we use the tool to produce a ventricular cardiomyocyte model with increased repolarization reserve. Since arrhythmia mitigation is dependent on multiple cardiac ion-channel conductances, study using a comprehensive, unbiased, and systems-level approach is important. The use of this optimization strategy allowed us to find robust profiles that illuminated unexpected mechanistic determinants of key ion-channel conductances in repolarization reserve.
Collapse
Affiliation(s)
- Kristin E Fullerton
- Physiology, Biophysics and Systems Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, New York, United States
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, United States
| | - Alexander P Clark
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States
| | - Trine Krogh-Madsen
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, United States
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York, United States
| | - David J Christini
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, New York, United States
| |
Collapse
|
11
|
Mokrov GV. Multitargeting in cardioprotection: An example of biaromatic compounds. Arch Pharm (Weinheim) 2023; 356:e2300196. [PMID: 37345968 DOI: 10.1002/ardp.202300196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/23/2023]
Abstract
A multitarget drug design approach is actively developing in modern medicinal chemistry and pharmacology, especially with regard to multifactorial diseases such as cardiovascular diseases, cancer, and neurodegenerative diseases. A detailed study of many well-known drugs developed within the single-target approach also often reveals additional mechanisms of their real pharmacological action. One of the multitarget drug design approaches can be the identification of the basic pharmacophore models corresponding to a wide range of the required target ligands. Among such models in the group of cardioprotectors is the linked biaromatic system. This review develops the concept of a "basic pharmacophore" using the biaromatic pharmacophore of cardioprotectors as an example. It presents an analysis of possible biological targets for compounds corresponding to the biaromatic pharmacophore and an analysis of the spectrum of biological targets for the five most known and most studied cardioprotective drugs corresponding to this model, and their involvement in the biological effects of these drugs.
Collapse
|
12
|
Szedlak P, Steele D, Hopkins P. Cardiac muscle physiology. BJA Educ 2023; 23:350-357. [PMID: 37600215 PMCID: PMC10435365 DOI: 10.1016/j.bjae.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2023] [Indexed: 08/22/2023] Open
Affiliation(s)
- P. Szedlak
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | | - P.M. Hopkins
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
- University of Leeds, Leeds, UK
| |
Collapse
|
13
|
Van Den Abeele R, Hendrickx S, Van Nieuwenhuyse E, Dunnink A, Panfilov AV, Vos MA, Wülfers EM, Vandersickel N. Directed graph mapping shows rotors maintain non-terminating and focal sources maintain self-terminating Torsade de Pointes in canine model. Front Physiol 2023; 14:1201260. [PMID: 37565147 PMCID: PMC10411729 DOI: 10.3389/fphys.2023.1201260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/28/2023] [Indexed: 08/12/2023] Open
Abstract
Torsade de Pointes is a polymorphic ventricular tachycardia which is as yet incompletely understood. While the onset of a TdP episode is generally accepted to be caused by triggered activity, the mechanisms for the perpetuation is still under debate. In this study, we analysed data from 54 TdP episodes divided over 5 dogs (4 female, 1 male) with chronic atrioventricular block. Previous research on this dataset showed both reentry and triggered activity to perpetuate the arrhythmia. 13 of those TdP episodes showed reentry as part of the driving mechanism of perpetuating the episode. The remaining 41 episodes were purely ectopic. Reentry was the main mechanism in long-lasting episodes (>14 beats), while focal sources were responsible for maintaining shorter episodes. Building on these results, we re-analysed the data using directed graph mapping This program uses principles from network theory and a combination of positional data and local activation times to identify reentry loops and focal sources within the data. The results of this study are twofold. First, concerning reentry loops, we found that on average non-terminating (NT) episodes (≥10 s) show significantly more simultaneous reentry loops than self-terminating (ST) TdP (<10 s). Non-terminating episodes have on average 2.72 ± 1.48 simultaneous loops, compared to an average of 1.33 ± 0.66 for self-terminating episodes. In addition, each NT episode showed a presence of (bi-)ventricular loops between 10.10% and 69.62% of their total reentry duration. Compared to the ST episodes, only 1 in 4 episodes (25%) showed (bi-)ventricular reentry, lasting only 7.12% of its total reentry duration. This suggests that while focal beats trigger TdP, macro-reentry and multiple simultaneous localized reentries are the major drivers of long-lasting episodes. Second, using heatmaps, we found focal sources to occur in preferred locations, instead of being distributed randomly. This may have implications on treatment if such focal origins can be disabled reliably.
Collapse
Affiliation(s)
- Robin Van Den Abeele
- Biophysics Group, Department of Physics and Astronomy, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Sander Hendrickx
- Biophysics Group, Department of Physics and Astronomy, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Enid Van Nieuwenhuyse
- Biophysics Group, Department of Physics and Astronomy, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Albert Dunnink
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Alexander V. Panfilov
- Biophysics Group, Department of Physics and Astronomy, Faculty of Sciences, Ghent University, Ghent, Belgium
- Laboratory of Computational Biology and Medicine, Ural Federal University, Yekaterinburg, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, Moscow, Russia
| | - Marc A. Vos
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Eike M. Wülfers
- Biophysics Group, Department of Physics and Astronomy, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Nele Vandersickel
- Biophysics Group, Department of Physics and Astronomy, Faculty of Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
14
|
ActionPytential: An open source tool for analyzing and visualizing cardiac action potential data. Heliyon 2023; 9:e14440. [PMID: 36967904 PMCID: PMC10031321 DOI: 10.1016/j.heliyon.2023.e14440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023] Open
Abstract
The action potential forms the basis of cardiac pacemaking, conduction, and contraction. Action potentials can be recorded from numerous preparation types, including ventricular or atrial trabecules, Purkinje fibers, isolated cardiac myocytes. Numerous techniques are also available as well, such as the conventional microelectrode and the single-cell current clamp techniques, optical mapping, or in silico modeling. With such a vast array of electrophysiological methods comes an array of available hardware and software solutions. In this work, we present a software with an intuitive graphical user interface, ActionPytential, that enables the analysis of any type of cardiac action potential, regardless of acquisition method or tissue type. In most available software tools, the analysis of continuous (gap-free) recordings often requires manual user interaction to segment the individual action potentials. We provide an automated solution for this, both for slow-response and for externally paced action potentials. As of now, ActionPytential calculates 34 parameters from each action potential. The most often utilized ones, including amplitude, maximal rate of depolarization, and action potential duration values, were validated on 1200 action potentials from human, dog, rabbit, guinea pig, and rat cardiac preparations. We also provide new parameters that were previously only measurable manually, including the position and the depth of the notch in potentials showing a spike-and-dome morphology. Further notable features include a Butterworth-type low-pass filter, the averaging of multiple potentials, automated corrections for baseline drifting, aided manual analysis, high-quality plots, and batch processing for any number of potentials. ActionPytential is available for all major platforms (Windows, MacOS, GNU + Linux, BSD).
Collapse
|
15
|
Asatryan B, Barth AS. Sex-related differences in incidence, phenotype and risk of sudden cardiac death in inherited arrhythmia syndromes. Front Cardiovasc Med 2023; 9:1010748. [PMID: 36684594 PMCID: PMC9845907 DOI: 10.3389/fcvm.2022.1010748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023] Open
Abstract
Inherited Arrhythmia Syndromes (IAS) including long QT and Brugada Syndrome, are characterized by life-threatening arrhythmias in the absence of apparent structural heart disease and are caused by pathogenic variants in genes encoding cardiac ion channels or associated proteins. Studies of large pedigrees of families affected by IAS have demonstrated incomplete penetrance and variable expressivity. Biological sex is one of several factors that have been recognized to modulate disease severity in IAS. There is a growing body of evidence linking sex hormones to the susceptibility to arrhythmias, yet, many sex-specific disease aspects remain underrecognized as female sex and women with IAS are underinvestigated and findings from male-predominant cohorts are often generalized to both sexes with minimal to no consideration of relevant sex-associated differences in prevalence, disease manifestations and outcome. In this review, we highlight current knowledge of sex-related biological differences in normal cardiac electrophysiology and sex-associated factors that influence IAS phenotypes.
Collapse
Affiliation(s)
- Babken Asatryan
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andreas S. Barth
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States,*Correspondence: Andreas S. Barth ✉
| |
Collapse
|
16
|
Krijger Juárez C, Amin AS, Offerhaus JA, Bezzina CR, Boukens BJ. Cardiac Repolarization in Health and Disease. JACC Clin Electrophysiol 2023; 9:124-138. [PMID: 36697193 DOI: 10.1016/j.jacep.2022.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 12/03/2022]
Abstract
Abnormal cardiac repolarization is at the basis of life-threatening arrhythmias in various congenital and acquired cardiac diseases. Dysfunction of ion channels involved in repolarization at the cellular level are often the underlying cause of the repolarization abnormality. The expression pattern of the gene encoding the affected ion channel dictates its impact on the shape of the T-wave and duration of the QT interval, thereby setting the stage for both the occurrence of the trigger and the substrate for maintenance of the arrhythmia. Here we discuss how research into the genetic and electrophysiological basis of repolarization has provided us with insights into cardiac repolarization in health and disease and how this in turn may provide the basis for future improved patient-specific management.
Collapse
Affiliation(s)
- Christian Krijger Juárez
- Department of Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Ahmad S Amin
- Department of Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Joost A Offerhaus
- Department of Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Bastiaan J Boukens
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, the Netherlands; Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
17
|
Del Buono MG, Damonte JI, Moroni F, Ravindra K, Westman P, Chiabrando JG, Bressi E, Li P, Kapoor K, Mao Y, Oakes T, Rodriguez-Miguelez P, Gal TS, Koneru J, Ellenbogen KA, Kron J, Abbate A. QT Prolongation and In-Hospital Ventricular Arrhythmic Complications in Patients With Apical Ballooning Takotsubo Syndrome. JACC Clin Electrophysiol 2022; 8:1500-1510. [PMID: 36543499 DOI: 10.1016/j.jacep.2022.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/03/2022] [Accepted: 08/10/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND Takotsubo syndrome is associated with life threatening arrhythmias, and the apical ballooning pattern is characterized by a peculiar QT prolongation and particularly high-risk of arrhythmias. OBJECTIVES The aim of the study was to determine the association of QT interval on electrocardiogram for ventricular arrhythmic complications in patients with apical ballooning Takotsubo syndrome in a diverse population at a large urban hospital in the U.S. METHODS We reviewed 105 cases of apical ballooning Takotsubo syndrome in patients admitted between 2011 and 2017. Two cardiologists reviewed the electrocardiograms to measure QT interval, adjusted for rate using the Fridericia formula (QTCF), and ventricular arrhythmic complications during the hospitalization. Data are reported as median and interquartile range or number and percentage. RESULTS Of the 105 patients, 86 (82%) were female, and 34 (32%) were self-reported Black or African American. The mean age was 65 years (range: 58-72 years). Left ventricular ejection fraction was 25% (range: 25%-35%). Heart rate was 101 beats/min (range: 83-121 beats/min). Ten (11%) patients experienced a ventricular arrhythmic complication and had significantly longer QTCF (470 [range: 422-543] milliseconds) than did those without complications (417 [range: 383-456] milliseconds, P = 0.031). The area under the curve for QTCF was 0.708 (95% CI: 0.536-0.880; P = 0.031). Twenty-eight (27%) patients had a QTCF ≥460 milliseconds and significantly more arrhythmic complications (21% vs 5%, odds ratio 4.997 [95% CI: 1.288-19.237], P = 0.021). QTCF was an independent predictor of ventricular arrhythmias: odds ratio 1.090 for each 10-millisecond increase in QTCF (95% CI: 1.004-1.183; P = 0.040, corrected for sex). CONCLUSIONS In a diverse population of patients with apical ballooning Takotsubo syndrome admitted to a large urban hospital in the United States, QTCF at admission ≥460 milliseconds identifies patients at high risk for in-hospital arrhythmic complications. Further studies are needed to determine strategies aimed at shortening QT interval to potentially prevent life-threatening arrhythmic events.
Collapse
Affiliation(s)
- Marco Giuseppe Del Buono
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA; Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Juan Ignacio Damonte
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA; Department of Interventional Cardiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Francesco Moroni
- Robert M. Berne Cardiovascular Research Center, and Department of Medicine, Division of Cardiovascular Medicine, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia, USA
| | - Krishna Ravindra
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Peter Westman
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Juan G Chiabrando
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA; Department of Interventional Cardiology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Edoardo Bressi
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Pengyang Li
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Kunal Kapoor
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Yuxuan Mao
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Travis Oakes
- Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Paula Rodriguez-Miguelez
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Tamas S Gal
- Department of Biostatistics, Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jayanthi Koneru
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Kenneth A Ellenbogen
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jordana Kron
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA; Robert M. Berne Cardiovascular Research Center, and Department of Medicine, Division of Cardiovascular Medicine, Heart and Vascular Center, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
18
|
Dahlberg P, Axelsson K, Jensen SM, Lundahl G, Vahedi F, Perkins R, Gransberg L, Bergfeldt L. Accelerated QT adaptation following atropine-induced heart rate increase in LQT1 patients versus healthy controls: A sign of disturbed hysteresis. Physiol Rep 2022; 10:e15487. [PMID: 36324292 PMCID: PMC9630760 DOI: 10.14814/phy2.15487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/08/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023] Open
Abstract
Hysteresis, a ubiquitous regulatory phenomenon, is a salient feature of the adaptation of ventricular repolarization duration to heart rate (HR) change. We therefore compared the QT interval adaptation to rapid HR increase in patients with the long QT syndrome type 1 (LQT1) versus healthy controls because LQT1 is caused by loss-of-function mutations affecting the repolarizing potassium channel current IKs , presumably an important player in QT hysteresis. The study was performed in an outpatient hospital setting. HR was increased in LQT1 patients and controls by administering an intravenous bolus of atropine (0.04 mg/kg body weight) for 30 s. RR and QT intervals were recorded by continuous Frank vectorcardiography. Atropine induced transient expected side effects but no adverse arrhythmias. There was no difference in HR response (RR intervals) to atropine between the groups. Although atropine-induced ΔQT was 48% greater in 18 LQT1 patients than in 28 controls (p < 0.001), QT adaptation was on average 25% faster in LQT1 patients (measured as the time constant τ for the mono-exponential function and the time for 90% of ΔQT; p < 0.01); however, there was some overlap between the groups, possibly a beta-blocker effect. The shorter QT adaptation time to atropine-induced HR increase in LQT1 patients on the group level corroborates the importance of IKs in QT adaptation hysteresis in humans and shows that LQT1 patients have a disturbed ultra-rapid cardiac memory. On the individual level, the QT adaptation time possibly reflects the effect-size of the loss-of-function mutation, but its clinical implications need to be shown.
Collapse
Affiliation(s)
- Pia Dahlberg
- Department of Molecular and Clinical MedicineInstitute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
- Region Västra Götaland, Department of CardiologySahlgrenska University HospitalGothenburgSweden
| | - Karl‐Jonas Axelsson
- Department of Molecular and Clinical MedicineInstitute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
- Region Västra Götaland, Department of CardiologySahlgrenska University HospitalGothenburgSweden
| | - Steen M. Jensen
- Department of Public Health and Clinical Medicine, and Heart CentreUmeå UniversityUmeåSweden
| | - Gunilla Lundahl
- Department of Molecular and Clinical MedicineInstitute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Farzad Vahedi
- Department of Molecular and Clinical MedicineInstitute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
- Region Västra Götaland, Department of CardiologySahlgrenska University HospitalGothenburgSweden
| | - Rosie Perkins
- Department of Molecular and Clinical MedicineInstitute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Lennart Gransberg
- Department of Molecular and Clinical MedicineInstitute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Lennart Bergfeldt
- Department of Molecular and Clinical MedicineInstitute of Medicine, Sahlgrenska Academy, University of GothenburgGothenburgSweden
- Region Västra Götaland, Department of CardiologySahlgrenska University HospitalGothenburgSweden
| |
Collapse
|
19
|
Doldi F, Plagwitz L, Hoffmann LP, Rath B, Frommeyer G, Reinke F, Leitz P, Büscher A, Güner F, Brix T, Wegner FK, Willy K, Hanel Y, Dittmann S, Haverkamp W, Schulze-Bahr E, Varghese J, Eckardt L. Detection of Patients with Congenital and Often Concealed Long-QT Syndrome by Novel Deep Learning Models. J Pers Med 2022; 12:jpm12071135. [PMID: 35887632 PMCID: PMC9323528 DOI: 10.3390/jpm12071135] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction: The long-QT syndrome (LQTS) is the most common ion channelopathy, typically presenting with a prolonged QT interval and clinical symptoms such as syncope or sudden cardiac death. Patients may present with a concealed phenotype making the diagnosis challenging. Correctly diagnosing at-risk patients is pivotal to starting early preventive treatment. Objective: Identification of congenital and often concealed LQTS by utilizing novel deep learning network architectures, which are specifically designed for multichannel time series and therefore particularly suitable for ECG data. Design and Results: A retrospective artificial intelligence (AI)-based analysis was performed using a 12-lead ECG of genetically confirmed LQTS (n = 124), including 41 patients with a concealed LQTS (33%), and validated against a control cohort (n = 161 of patients) without known LQTS or without QT-prolonging drug treatment but any other cardiovascular disease. The performance of a fully convolutional network (FCN) used in prior studies was compared with a different, novel convolutional neural network model (XceptionTime). We found that the XceptionTime model was able to achieve a higher balanced accuracy score (91.8%) than the associated FCN metric (83.6%), indicating improved prediction possibilities of novel AI architectures. The predictive accuracy prevailed independently of age and QTc parameters. Conclusions: In this study, the XceptionTime model outperformed the FCN model for LQTS patients with even better results than in prior studies. Even when a patient cohort with cardiovascular comorbidities is used. AI-based ECG analysis is a promising step for correct LQTS patient identification, especially if common diagnostic measures might be misleading.
Collapse
Affiliation(s)
- Florian Doldi
- Department for Cardiology II-Electrophysiology, University Hospital Münster, 48149 Münster, Germany; (L.P.H.); (B.R.); (G.F.); (F.R.); (P.L.); (A.B.); (F.G.); (F.K.W.); (K.W.); (L.E.)
- Correspondence: ; Tel.: +49-251-8344633
| | - Lucas Plagwitz
- Institute of Medical Informatics, University of Münster, 48149 Münster, Germany; (L.P.); (T.B.); (J.V.)
| | - Lea Philine Hoffmann
- Department for Cardiology II-Electrophysiology, University Hospital Münster, 48149 Münster, Germany; (L.P.H.); (B.R.); (G.F.); (F.R.); (P.L.); (A.B.); (F.G.); (F.K.W.); (K.W.); (L.E.)
| | - Benjamin Rath
- Department for Cardiology II-Electrophysiology, University Hospital Münster, 48149 Münster, Germany; (L.P.H.); (B.R.); (G.F.); (F.R.); (P.L.); (A.B.); (F.G.); (F.K.W.); (K.W.); (L.E.)
| | - Gerrit Frommeyer
- Department for Cardiology II-Electrophysiology, University Hospital Münster, 48149 Münster, Germany; (L.P.H.); (B.R.); (G.F.); (F.R.); (P.L.); (A.B.); (F.G.); (F.K.W.); (K.W.); (L.E.)
| | - Florian Reinke
- Department for Cardiology II-Electrophysiology, University Hospital Münster, 48149 Münster, Germany; (L.P.H.); (B.R.); (G.F.); (F.R.); (P.L.); (A.B.); (F.G.); (F.K.W.); (K.W.); (L.E.)
| | - Patrick Leitz
- Department for Cardiology II-Electrophysiology, University Hospital Münster, 48149 Münster, Germany; (L.P.H.); (B.R.); (G.F.); (F.R.); (P.L.); (A.B.); (F.G.); (F.K.W.); (K.W.); (L.E.)
| | - Antonius Büscher
- Department for Cardiology II-Electrophysiology, University Hospital Münster, 48149 Münster, Germany; (L.P.H.); (B.R.); (G.F.); (F.R.); (P.L.); (A.B.); (F.G.); (F.K.W.); (K.W.); (L.E.)
| | - Fatih Güner
- Department for Cardiology II-Electrophysiology, University Hospital Münster, 48149 Münster, Germany; (L.P.H.); (B.R.); (G.F.); (F.R.); (P.L.); (A.B.); (F.G.); (F.K.W.); (K.W.); (L.E.)
| | - Tobias Brix
- Institute of Medical Informatics, University of Münster, 48149 Münster, Germany; (L.P.); (T.B.); (J.V.)
| | - Felix Konrad Wegner
- Department for Cardiology II-Electrophysiology, University Hospital Münster, 48149 Münster, Germany; (L.P.H.); (B.R.); (G.F.); (F.R.); (P.L.); (A.B.); (F.G.); (F.K.W.); (K.W.); (L.E.)
| | - Kevin Willy
- Department for Cardiology II-Electrophysiology, University Hospital Münster, 48149 Münster, Germany; (L.P.H.); (B.R.); (G.F.); (F.R.); (P.L.); (A.B.); (F.G.); (F.K.W.); (K.W.); (L.E.)
| | - Yvonne Hanel
- Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, 48149 Münster, Germany; (Y.H.); (S.D.); (E.S.-B.)
| | - Sven Dittmann
- Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, 48149 Münster, Germany; (Y.H.); (S.D.); (E.S.-B.)
| | - Wilhelm Haverkamp
- Department of Internal Medicine and Cardiology, Charité University Medicine, 10117 Berlin, Germany;
| | - Eric Schulze-Bahr
- Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, 48149 Münster, Germany; (Y.H.); (S.D.); (E.S.-B.)
| | - Julian Varghese
- Institute of Medical Informatics, University of Münster, 48149 Münster, Germany; (L.P.); (T.B.); (J.V.)
| | - Lars Eckardt
- Department for Cardiology II-Electrophysiology, University Hospital Münster, 48149 Münster, Germany; (L.P.H.); (B.R.); (G.F.); (F.R.); (P.L.); (A.B.); (F.G.); (F.K.W.); (K.W.); (L.E.)
| |
Collapse
|
20
|
An X, Lee J, Kim GH, Kim HJ, Pyo HJ, Kwon I, Cho H. Modulation of I Ks channel-PIP 2 interaction by PRMT1 plays a critical role in the control of cardiac repolarization. J Cell Physiol 2022; 237:3069-3079. [PMID: 35580065 PMCID: PMC9543859 DOI: 10.1002/jcp.30775] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/18/2022] [Accepted: 04/25/2022] [Indexed: 11/12/2022]
Abstract
Recent studies have shown that protein arginine methyltransferase 1 (PRMT1) is highly expressed in the human heart, and loss of PRMT1 contributes to cardiac remodeling in the heart failure. However, the functional importance of PRMT1 in cardiac ion channels remains uncertain. The slow activating delayed rectifier K+ (IKs) channel is a cardiac K+ channel composed of KCNQ1 and KCNE1 subunits and is a new therapeutic target for treating lethal arrhythmias in many cardiac pathologies, especially heart failure. Here, we demonstrate that PRMT1 is a critical regulator of the IKs channel and cardiac rhythm. In the guinea pig ventricular myocytes, treatment with furamidine, a PRMT1‐specific inhibitor, prolonged the action potential duration (APD). We further show that this APD prolongation was attributable to IKs reduction. In HEK293T cells expressing human KCNQ1 and KCNE1, inhibiting PRMT1 via furamidine reduced IKs and concurrently decreased the arginine methylation of KCNQ1, a pore‐forming α‐subunit. Evidence presented here indicates that furamidine decreased IKs mainly by lowering the affinity of IKs channels for the membrane phospholipid, phosphatidylinositol 4,5‐bisphosphate (PIP2), which is crucial for pore opening. Finally, applying exogenous PIP2 to cardiomyocytes prevented the furamidine‐induced IKs reduction and APD prolongation. Taken together, these results indicate that PRMT1 positively regulated IKs activity through channel–PIP2 interaction, thereby restricting excessive cardiac action potential.
Collapse
Affiliation(s)
- Xue An
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Jiwon Lee
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Ga Hye Kim
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hyun-Ji Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hyun-Jeong Pyo
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Ilmin Kwon
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| |
Collapse
|
21
|
de Veld L, van der Lely N, Hermans BJM, van Hoof JJ, Wong L, Vink AS. QTc prolongation in adolescents with acute alcohol intoxication. Eur J Pediatr 2022; 181:2757-2770. [PMID: 35482092 PMCID: PMC9192465 DOI: 10.1007/s00431-022-04471-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/30/2022] [Accepted: 04/09/2022] [Indexed: 01/24/2023]
Abstract
In adults, alcohol intoxication is associated with prolongation of the QT interval corrected for heart rate (QTc). The QTc is influenced by age and sex. Although alcohol intoxication is increasingly common in adolescents, there are no data on the prevalence of QTc prolongation in adolescents with alcohol intoxication. This study aimed to determine the prevalence of QTc prolongation in adolescents with alcohol intoxication and identify at-risk adolescents. In this observational study including adolescents aged 10-18 years, heart rate and QT interval were automatically assessed from an electrocardiogram (ECG) at alcohol intoxication using a validated algorithm. The QTc was calculated using both the Bazett formula (QTcB) and Fridericia formula (QTcF). If present, an ECG recorded within 1 year of the date of admission to the emergency department was obtained as a reference ECG. A total of 317 adolescents were included; 13.3% had a QTcB and 7.9% a QTcF longer than the sex- and age-specific 95th-percentile. None of the adolescents had a QTcB or QTcF > 500 ms, but 11.8% of the adolescents with a reference ECG had a QTcB prolongation of > 60 ms, while no adolescents had a QTcF prolongation of > 60 ms. QTc prolongation was mainly attributable to an increase in heart rate rather than QT prolongation, which underlies the differences between QTcB and QTcF. Male sex and hypokalaemia increased the likelihood of QTc prolongation.Conclusion: QTc prolongation was seen in approximately 10% of the adolescents presenting with alcohol intoxication, and although no ventricular arrhythmias were observed in this cohort, QTc prolongation increases the potential for malignant QT-related arrhythmias. Clinicians must be aware of the possibility of QTc prolongation during alcohol intoxication and make an effort to obtain an ECG at presentation, measure the QT interval, and give an adequate assessment of the findings. We advocate admitting adolescents with alcohol intoxication and QTc prolongation. During hospital admission, we recommend limiting exposure to QTc-prolonging medication, increasing potassium levels to a high-normal range (4.5-5.0 mmol/L) and obtaining a reference ECG at discharge.
Collapse
Affiliation(s)
- Loes de Veld
- Erasmus School of Health Policy and Management, Erasmus University, Postbus 1738, 3000, Rotterdam, DR, Netherlands. .,Department of Pediatrics, Reinier de Graaf Hospital, Delft, Netherlands.
| | - Nico van der Lely
- Department of Pediatrics, Reinier de Graaf Hospital, Delft, Netherlands ,Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Ben J. M. Hermans
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Joris J. van Hoof
- Department of Pediatrics, Reinier de Graaf Hospital, Delft, Netherlands
| | - Lichelle Wong
- Department of Pediatrics, Reinier de Graaf Hospital, Delft, Netherlands
| | - Arja Suzanne Vink
- Department of Cardiology, Heart Center, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands ,Department of Pediatric Cardiology, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
22
|
Scrocco C, Bezzina CR, Ackerman MJ, Behr ER. Genetics and genomics of arrhythmic risk: current and future strategies to prevent sudden cardiac death. Nat Rev Cardiol 2021; 18:774-784. [PMID: 34031597 DOI: 10.1038/s41569-021-00555-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/12/2021] [Indexed: 02/04/2023]
Abstract
A genetic risk of sudden cardiac arrest and sudden death due to an arrhythmic cause, known as sudden cardiac death (SCD), has become apparent from epidemiological studies in the general population and in patients with ischaemic heart disease. However, genetic susceptibility to sudden death is greatest in young people and is associated with uncommon, monogenic forms of heart disease. Despite comprehensive pathology and genetic evaluations, SCD remains unexplained in a proportion of young people and is termed sudden arrhythmic death syndrome, which poses challenges to the identification of relatives from affected families who might be at risk of SCD. In this Review, we assess the current understanding of the epidemiology and causes of SCD and evaluate both the monogenic and the polygenic contributions to the risk of SCD in the young and SCD associated with drug therapy. Finally, we analyse the potential clinical role of genomic testing in the prevention of SCD in the general population.
Collapse
Affiliation(s)
- Chiara Scrocco
- Cardiovascular Clinical Academic Group, Molecular and Clinical Sciences Institute, St George's University of London and St George's University Hospitals NHS Foundation Trust, London, UK
| | - Connie R Bezzina
- Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Michael J Ackerman
- Departments of Cardiovascular Medicine, Pediatric and Adolescent Medicine, and Molecular Pharmacology & Experimental Therapeutics; Divisions of Heart Rhythm Services and Pediatric Cardiology, Mayo Clinic, Rochester, MN, USA.,Windland Smith Rice Genetic Heart Rhythm Clinic and the Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Elijah R Behr
- Cardiovascular Clinical Academic Group, Molecular and Clinical Sciences Institute, St George's University of London and St George's University Hospitals NHS Foundation Trust, London, UK.
| |
Collapse
|
23
|
Chen L, He Y, Wang X, Ge J, Li H. Ventricular voltage-gated ion channels: Detection, characteristics, mechanisms, and drug safety evaluation. Clin Transl Med 2021; 11:e530. [PMID: 34709746 PMCID: PMC8516344 DOI: 10.1002/ctm2.530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac voltage-gated ion channels (VGICs) play critical roles in mediating cardiac electrophysiological signals, such as action potentials, to maintain normal heart excitability and contraction. Inherited or acquired alterations in the structure, expression, or function of VGICs, as well as VGIC-related side effects of pharmaceutical drug delivery can result in abnormal cellular electrophysiological processes that induce life-threatening cardiac arrhythmias or even sudden cardiac death. Hence, to reduce possible heart-related risks, VGICs must be acknowledged as important targets in drug discovery and safety studies related to cardiac disease. In this review, we first summarize the development and application of electrophysiological techniques that are employed in cardiac VGIC studies alone or in combination with other techniques such as cryoelectron microscopy, optical imaging and optogenetics. Subsequently, we describe the characteristics, structure, mechanisms, and functions of various well-studied VGICs in ventricular myocytes and analyze their roles in and contributions to both physiological cardiac excitability and inherited cardiac diseases. Finally, we address the implications of the structure and function of ventricular VGICs for drug safety evaluation. In summary, multidisciplinary studies on VGICs help researchers discover potential targets of VGICs and novel VGICs in heart, enrich their knowledge of the properties and functions, determine the operation mechanisms of pathological VGICs, and introduce groundbreaking trends in drug therapy strategies, and drug safety evaluation.
Collapse
Affiliation(s)
- Lulan Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| | - Yue He
- Department of CardiologyShanghai Xuhui District Central Hospital & Zhongshan‐xuhui HospitalShanghaiChina
| | - Xiangdong Wang
- Institute of Clinical Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| | - Hua Li
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
24
|
Beer D, Isakadze N, McClellan R, Calkins H, Barth AS. Acquired Long QT and Ventricular Arrhythmias in the Setting of Acute Inflammation: A Case Series. JACC Case Rep 2021; 3:1103-1107. [PMID: 34471893 PMCID: PMC8314122 DOI: 10.1016/j.jaccas.2021.04.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/21/2021] [Accepted: 04/27/2021] [Indexed: 01/08/2023]
Abstract
We report a case series of 4 patients with transient marked QTc prolongation and ventricular arrhythmias in the setting of inflammation with very high ferritin levels. Three patients were positive for coronavirus disease-2019. In the setting of an acute rise in inflammatory markers, electrocardiography screening for QTc prolongation is warranted. (Level of Difficulty: Beginner.)
Collapse
Affiliation(s)
- Dominik Beer
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nino Isakadze
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rebecca McClellan
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hugh Calkins
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andreas S Barth
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
25
|
Reiffel JA. Antiarrhythmic Drugs for Atrial Fibrillation: Selected Features of Ventricular Repolarization That Facilitate Proarrhythmic Torsades de Pointes and Favor Inpatient Initiation. J Innov Card Rhythm Manag 2021; 12:4600-4605. [PMID: 34327046 PMCID: PMC8313184 DOI: 10.19102/icrm.2021.120704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 01/15/2021] [Indexed: 12/18/2022] Open
Abstract
The management approaches to patients with atrial fibrillation (AF) include rhythm-control strategies for those patients who are symptomatic despite rate control and for selected others in whom sinus rhythm is necessary for reasons beyond current symptoms (including commercial pilots, those who are felt likely to develop symptoms as comorbidities progress, and more). First-line therapies among the rhythm-control options are antiarrhythmic drugs (AADs). For many AADs, their initiation in-hospital is either a requirement or strongly advised- especially when the patient is in AF. This article explores some of the rationale behind this requirement to give clinicians a better understanding of the reasons for this undesired inconvenience.
Collapse
|
26
|
Filatova TS, Abramochkin DV, Pavlova NS, Pustovit KB, Konovalova OP, Kuzmin VS, Dobrzynski H. Repolarizing potassium currents in working myocardium of Japanese quail: a novel translational model for cardiac electrophysiology. Comp Biochem Physiol A Mol Integr Physiol 2021; 255:110919. [DOI: 10.1016/j.cbpa.2021.110919] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/06/2021] [Accepted: 02/06/2021] [Indexed: 12/14/2022]
|
27
|
Trachsel DS, Calloe K, J Rgensen E, Lunddahl CS, Pedersen PJ, Kanters JRK, Klaerke DA, Buhl R. Evaluation of electrocardiographic repolarization parameters after administration of trimethoprim-sulfadiazine, detomidine, or their combination in horses. Am J Vet Res 2021; 82:207-217. [PMID: 33629897 DOI: 10.2460/ajvr.82.3.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To determine whether administration of trimethoprim-sulfadiazine (TMS), detomidine (DET), or TMS plus DET would be associated with changes in ECG repolarization parameters in horses. ANIMALS 9 healthy adult horses. PROCEDURES Each horse received 4 treatments in a blinded, randomized, crossover study design as follows: TMS, 16 to 24 mg/kg, IV; DET, 0.015 to 0.02 mg/kg, IV; TMS plus DET; and saline (0.9% NaCl) solution. Surface ECG traces were obtained over 24 hours, and repolarization parameters were measured at predefined time points after each treatment and compared with a 2-way ANOVA for repeated measures. RESULTS Heart rate-corrected QT intervals (QTc) were significantly increased after administration of DET (mean ± SD difference in QTc, 36.57 ± 23.07 milliseconds; increase of 7%) and TMS plus DET (44.96 ± 29.16 milliseconds; increase of 9%), compared with baseline (before treatment) values and values after administration of saline solution. Saline solution and TMS alone did not affect QTc. CONCLUSIONS AND CLINICAL RELEVANCE Administration of DET or TMS plus DET was associated with a significant and possibly clinically relevant prolongation of QTc, with prolongation of 7% to 9%, a range that is considered as a risk factor for the development of cardiac arrhythmias in people. Results were unexpected because DET is considered to be a safe sedative for horses.
Collapse
|
28
|
Berger FA, van der Sijs H, van Gelder T, Kuijper AFM, van den Bemt PMLA, Becker ML. Comparison of two algorithms to support medication surveillance for drug-drug interactions between QTc-prolonging drugs. Int J Med Inform 2020; 145:104329. [PMID: 33181445 DOI: 10.1016/j.ijmedinf.2020.104329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/09/2020] [Accepted: 11/01/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND QTc-prolongation is an independent risk factor for developing life-threatening arrhythmias. Risk management of drug-induced QTc-prolongation is complex and digital support tools could be of assistance. Bindraban et al. and Berger et al. developed two algorithms to identify patients at risk for QTc-prolongation. OBJECTIVE The main aim of this study was to compare the performances of these algorithms for managing QTc-prolonging drug-drug interactions (QT-DDIs). MATERIALS AND METHODS A retrospective data analysis was performed. A dataset was created from QT-DDI alerts generated for in- and outpatients at a general teaching hospital between November 2016 and March 2018. ECGs recorded within 7 days of the QT-DDI alert were collected. Main outcomes were the performance characteristics of both algorithms. QTc-intervals of > 500 ms on the first ECG after the alert were taken as outcome parameter, to which the performances were compared. Secondary outcome was the distribution of risk scores in the study cohort. RESULTS In total, 10,870 QT-DDI alerts of 4987 patients were included. ECGs were recorded in 26.2 % of the QT-DDI alerts. Application of the algorithms resulted in area under the ROC-curves of 0.81 (95 % CI 0.79-0.84) for Bindraban et al. and 0.73 (0.70-0.75) for Berger et al. Cut-off values of ≥ 3 and ≥ 6 led to sensitivities of 85.7 % and 89.1 %, and specificities of 60.8 % and 44.3 % respectively. CONCLUSIONS Both algorithms showed good discriminative abilities to identify patients at risk for QTc-prolongation when using ≥ 2 QTc-prolonging drugs. Implementation of digital algorithms in clinical decision support systems could support the risk management of QT-DDIs.
Collapse
Affiliation(s)
- Florine A Berger
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Heleen van der Sijs
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - T van Gelder
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Aaf F M Kuijper
- Department of Cardiology, Spaarne Gasthuis, Hoofddorp, the Netherlands
| | - Patricia M L A van den Bemt
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, the Netherlands
| | | |
Collapse
|
29
|
Wan H, Selvaggio G, Pearlstein RA. Toward in vivo-relevant hERG safety assessment and mitigation strategies based on relationships between non-equilibrium blocker binding, three-dimensional channel-blocker interactions, dynamic occupancy, dynamic exposure, and cellular arrhythmia. PLoS One 2020; 15:e0234946. [PMID: 33147278 PMCID: PMC7641409 DOI: 10.1371/journal.pone.0234946] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/16/2020] [Indexed: 12/26/2022] Open
Abstract
The human ether-a-go-go-related voltage-gated cardiac ion channel (commonly known as hERG) conducts the rapid outward repolarizing potassium current in cardiomyocytes (IKr). Inadvertent blockade of this channel by drug-like molecules represents a key challenge in pharmaceutical R&D due to frequent overlap between the structure-activity relationships of hERG and many primary targets. Building on our previous work, together with recent cryo-EM structures of hERG, we set about to better understand the energetic and structural basis of promiscuous blocker-hERG binding in the context of Biodynamics theory. We propose a two-step blocker binding process consisting of: The initial capture step: diffusion of a single fully solvated blocker copy into a large cavity lined by the intra-cellular cyclic nucleotide binding homology domain (CNBHD). Occupation of this cavity is a necessary but insufficient condition for ion current disruption.The IKr disruption step: translocation of the captured blocker along the channel axis, such that: The head group, consisting of a quasi-rod-shaped moiety, projects into the open pore, accompanied by partial de-solvation of the binding interface.One tail moiety packs along a kink between the S6 helix and proximal C-linker helix adjacent to the intra-cellular entrance of the pore, likewise accompanied by mutual de-solvation of the binding interface (noting that the association barrier is comprised largely of the total head + tail group de-solvation cost).Blockers containing a highly planar moiety that projects into a putative constriction zone within the closed channel become trapped upon closing, as do blockers terminating prior to this region.A single captured blocker copy may conceivably associate and dissociate to/from the pore many times before exiting the CNBHD cavity. Lastly, we highlight possible flaws in the current hERG safety index (SI), and propose an alternate in vivo-relevant strategy factoring in: Benefit/risk.The predicted arrhythmogenic fractional hERG occupancy (based on action potential (AP) simulations of the undiseased human ventricular cardiomyocyte).Alteration of the safety threshold due to underlying disease.Risk of exposure escalation toward the predicted arrhythmic limit due to patient-to-patient pharmacokinetic (PK) variability, drug-drug interactions, overdose, and use for off-label indications in which the hERG safety parameters may differ from their on-label counterparts.
Collapse
Affiliation(s)
- Hongbin Wan
- Global Discovery Chemistry, Computer-Aided Drug Discovery, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States of America
| | - Gianluca Selvaggio
- Global Discovery Chemistry, Computer-Aided Drug Discovery, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States of America
| | - Robert A. Pearlstein
- Global Discovery Chemistry, Computer-Aided Drug Discovery, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States of America
| |
Collapse
|
30
|
Varró A, Tomek J, Nagy N, Virág L, Passini E, Rodriguez B, Baczkó I. Cardiac transmembrane ion channels and action potentials: cellular physiology and arrhythmogenic behavior. Physiol Rev 2020; 101:1083-1176. [PMID: 33118864 DOI: 10.1152/physrev.00024.2019] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiac arrhythmias are among the leading causes of mortality. They often arise from alterations in the electrophysiological properties of cardiac cells and their underlying ionic mechanisms. It is therefore critical to further unravel the pathophysiology of the ionic basis of human cardiac electrophysiology in health and disease. In the first part of this review, current knowledge on the differences in ion channel expression and properties of the ionic processes that determine the morphology and properties of cardiac action potentials and calcium dynamics from cardiomyocytes in different regions of the heart are described. Then the cellular mechanisms promoting arrhythmias in congenital or acquired conditions of ion channel function (electrical remodeling) are discussed. The focus is on human-relevant findings obtained with clinical, experimental, and computational studies, given that interspecies differences make the extrapolation from animal experiments to human clinical settings difficult. Deepening the understanding of the diverse pathophysiology of human cellular electrophysiology will help in developing novel and effective antiarrhythmic strategies for specific subpopulations and disease conditions.
Collapse
Affiliation(s)
- András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - Jakub Tomek
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Cardiovascular Pharmacology Research Group, Hungarian Academy of Sciences, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Elisa Passini
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
31
|
Hummel JP, Akar JG. Reply to: “QT prolongation with hydroxychloroquine and azithromycin for the treatment of COVID‐19: The need for pharmacogenetic insights”. J Cardiovasc Electrophysiol 2020; 31:2795. [DOI: 10.1111/jce.14721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 11/29/2022]
Affiliation(s)
- James P. Hummel
- Department of Internal Medicine Cardiovascular Medicine, Yale School of Medicine New Haven Connecticut USA
| | - Joseph G. Akar
- Department of Internal Medicine Cardiovascular Medicine, Yale School of Medicine New Haven Connecticut USA
| |
Collapse
|
32
|
Varga RS, Hornyik T, Husti Z, Kohajda Z, Krajsovszky G, Nagy N, Jost N, Virág L, Tálosi L, Mátyus P, Varró A, Baczkó I. Antiarrhythmic and cardiac electrophysiological effects of SZV-270, a novel compound with combined Class I/B and Class III effects, in rabbits and dogs. Can J Physiol Pharmacol 2020; 99:89-101. [PMID: 32970956 DOI: 10.1139/cjpp-2020-0412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Cardiovascular diseases are the leading causes of mortality. Sudden cardiac death is most commonly caused by ventricular fibrillation (VF). Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia and a major cause of stroke and heart failure. Pharmacological management of VF and AF remains suboptimal due to limited efficacy of antiarrhythmic drugs and their ventricular proarrhythmic adverse effects. In this study, the antiarrhythmic and cardiac cellular electrophysiological effects of SZV-270, a novel compound, were investigated in rabbit and canine models. SZV-270 significantly reduced the incidence of VF in rabbits subjected to coronary artery occlusion/reperfusion and reduced the incidence of burst-induced AF in a tachypaced conscious canine model of AF. SZV-270 prolonged the frequency-corrected QT interval, lengthened action potential duration and effective refractory period in ventricular and atrial preparations, blocked I Kr in isolated cardiomyocytes (Class III effects), and reduced the maximum rate of depolarization (V max) at cycle lengths smaller than 1000 ms in ventricular preparations (Class I/B effect). Importantly, SZV-270 did not provoke Torsades de Pointes arrhythmia in an anesthetized rabbit proarrhythmia model characterized by impaired repolarization reserve. In conclusion, SZV-270 with its combined Class I/B and III effects can prevent reentry arrhythmias with reduced risk of provoking drug-induced Torsades de Pointes.
Collapse
Affiliation(s)
- Richárd S Varga
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| | - Tibor Hornyik
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| | - Zoltán Husti
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| | - Zsófia Kohajda
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - Gábor Krajsovszky
- Department of Organic Chemistry, Semmelweis University, Budapest, Hungary
| | - Norbert Nagy
- MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| | - László Tálosi
- Department of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Péter Mátyus
- Department of Organic Chemistry, Semmelweis University, Budapest, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary.,MTA-SZTE Research Group of Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| |
Collapse
|
33
|
Pászti B, Prorok J, Magyar T, Árpádffy-Lovas T, Györe B, Topál L, Gazdag P, Szlovák J, Naveed M, Jost N, Nagy N, Varró A, Virág L, Koncz I. Cardiac electrophysiological effects of ibuprofen in dog and rabbit ventricular preparations: possible implication to enhanced proarrhythmic risk. Can J Physiol Pharmacol 2020; 99:102-109. [PMID: 32937079 DOI: 10.1139/cjpp-2020-0386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ibuprofen is a widely used nonsteroidal anti-inflammatory drug, which has recently been associated with increased cardiovascular risk, but its electrophysiological effects have not yet been properly studied in isolated cardiac preparations. We studied the effects of ibuprofen on action potential characteristics and several transmembrane ionic currents using the conventional microelectrode technique and the whole-cell configuration of the patch-clamp technique on cardiac preparations and enzymatically isolated ventricular myocytes. In dog (200 µM; n = 6) and rabbit (100 µM; n = 7) papillary muscles, ibuprofen moderately but significantly prolonged repolarization at 1 Hz stimulation frequency. In dog Purkinje fibers, repolarization was abbreviated and maximal rate of depolarization was depressed in a frequency-dependent manner. Levofloxacin (40 µM) alone did not alter repolarization, but augmented the ibuprofen-evoked repolarization lengthening in rabbit preparations (n = 7). In dog myocytes, ibuprofen (250 µM) did not significantly influence IK1, but decreased the amplitude of Ito and IKr potassium currents by 28.2% (60 mV) and 15.2% (20 mV), respectively. Ibuprofen also depressed INaL and ICa currents by 19.9% and 16.4%, respectively. We conclude that ibuprofen seems to be free from effects on action potential parameters at lower concentrations. However, at higher concentrations it may alter repolarization reserve, contributing to the observed proarrhythmic risk in patients.
Collapse
Affiliation(s)
- Bence Pászti
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - János Prorok
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - Tibor Magyar
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Tamás Árpádffy-Lovas
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Balázs Györe
- Department of Oral Surgery, Faculty of Dentistry, University of Szeged, Szeged, Hungary
| | - Leila Topál
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Péter Gazdag
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Jozefina Szlovák
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Muhammad Naveed
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary.,MTA-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,MTA-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary.,MTA-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary.,MTA-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - István Koncz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
34
|
Shugg T, Hudmon A, Overholser BR. Neurohormonal Regulation of I Ks in Heart Failure: Implications for Ventricular Arrhythmogenesis and Sudden Cardiac Death. J Am Heart Assoc 2020; 9:e016900. [PMID: 32865116 PMCID: PMC7726975 DOI: 10.1161/jaha.120.016900] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Heart failure (HF) results in sustained alterations in neurohormonal signaling, including enhanced signaling through the sympathetic nervous system and renin-angiotensin-aldosterone system pathways. While enhanced sympathetic nervous system and renin-angiotensin-aldosterone system activity initially help compensate for the failing myocardium, sustained signaling through these pathways ultimately contributes to HF pathophysiology. HF remains a leading cause of mortality, with arrhythmogenic sudden cardiac death comprising a common mechanism of HF-related death. The propensity for arrhythmia development in HF occurs secondary to cardiac electrical remodeling that involves pathological regulation of ventricular ion channels, including the slow component of the delayed rectifier potassium current, that contribute to action potential duration prolongation. To elucidate a mechanistic explanation for how HF-mediated electrical remodeling predisposes to arrhythmia development, a multitude of investigations have investigated the specific regulatory effects of HF-associated stimuli, including enhanced sympathetic nervous system and renin-angiotensin-aldosterone system signaling, on the slow component of the delayed rectifier potassium current. The objective of this review is to summarize the current knowledge related to the regulation of the slow component of the delayed rectifier potassium current in response to HF-associated stimuli, including the intracellular pathways involved and the specific regulatory mechanisms.
Collapse
Affiliation(s)
- Tyler Shugg
- Division of Clinical PharmacologyIndiana University School of MedicineIndianapolisIN
| | - Andy Hudmon
- Department of Medicinal Chemistry and Molecular PharmacologyPurdue University College of PharmacyWest LafayetteIN
| | - Brian R. Overholser
- Division of Clinical PharmacologyIndiana University School of MedicineIndianapolisIN
- Department of Pharmacy PracticePurdue University College of PharmacyIndianapolisIN
| |
Collapse
|
35
|
Berger FA, van der Sijs H, Becker ML, van Gelder T, van den Bemt PMLA. Development and validation of a tool to assess the risk of QT drug-drug interactions in clinical practice. BMC Med Inform Decis Mak 2020; 20:171. [PMID: 32703198 PMCID: PMC7376881 DOI: 10.1186/s12911-020-01181-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The exact risk of developing QTc-prolongation when using a combination of QTc-prolonging drugs is still unknown, making it difficult to interpret these QT drug-drug interactions (QT-DDIs). A tool to identify high-risk patients is needed to support healthcare providers in handling automatically generated alerts in clinical practice. The main aim of this study was to develop and validate a tool to assess the risk of QT-DDIs in clinical practice. METHODS A model was developed based on risk factors associated with QTc-prolongation determined in a prospective study on QT-DDIs in a university medical center inthe Netherlands. The main outcome measure was QTc-prolongation defined as a QTc interval > 450 ms for males and > 470 ms for females. Risk points were assigned to risk factors based on their odds ratios. Additional risk factors were added based on a literature review. The ability of the model to predict QTc-prolongation was validated in an independent dataset obtained from a general teaching hospital against QTc-prolongation as measured by an ECG as the gold standard. Sensitivities, specificities, false omission rates, accuracy and Youden's index were calculated. RESULTS The model included age, gender, cardiac comorbidities, hypertension, diabetes mellitus, renal function, potassium levels, loop diuretics, and QTc-prolonging drugs as risk factors. Application of the model to the independent dataset resulted in an area under the ROC-curve of 0.54 (95% CI 0.51-0.56) when QTc-prolongation was defined as > 450/470 ms, and 0.59 (0.54-0.63) when QTc-prolongation was defined as > 500 ms. A cut-off value of 6 led to a sensitivity of 76.6 and 83.9% and a specificity of 28.5 and 27.5% respectively. CONCLUSIONS A clinical decision support tool with fair performance characteristics was developed. Optimization of this tool may aid in assessing the risk associated with QT-DDIs. TRIAL REGISTRATION No trial registration, MEC-2015-368.
Collapse
Affiliation(s)
- Florine A Berger
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015, GD, Rotterdam, the Netherlands.
| | - Heleen van der Sijs
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015, GD, Rotterdam, the Netherlands
| | - Matthijs L Becker
- Pharmacy Foundation of Haarlem Hospitals, Haarlem, the Netherlands
- Department of Clinical Pharmacy, Spaarne Gasthuis, Haarlem, the Netherlands
| | - Teun van Gelder
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015, GD, Rotterdam, the Netherlands
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Patricia M L A van den Bemt
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015, GD, Rotterdam, the Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
36
|
Giudicessi JR, Noseworthy PA, Friedman PA, Ackerman MJ. Urgent Guidance for Navigating and Circumventing the QTc-Prolonging and Torsadogenic Potential of Possible Pharmacotherapies for Coronavirus Disease 19 (COVID-19). Mayo Clin Proc 2020; 95:1213-1221. [PMID: 32359771 PMCID: PMC7141471 DOI: 10.1016/j.mayocp.2020.03.024] [Citation(s) in RCA: 286] [Impact Index Per Article: 71.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 02/08/2023]
Abstract
As the coronavirus disease 19 (COVID-19) global pandemic rages across the globe, the race to prevent and treat this deadly disease has led to the "off-label" repurposing of drugs such as hydroxychloroquine and lopinavir/ritonavir, which have the potential for unwanted QT-interval prolongation and a risk of drug-induced sudden cardiac death. With the possibility that a considerable proportion of the world's population soon could receive COVID-19 pharmacotherapies with torsadogenic potential for therapy or postexposure prophylaxis, this document serves to help health care professionals mitigate the risk of drug-induced ventricular arrhythmias while minimizing risk of COVID-19 exposure to personnel and conserving the limited supply of personal protective equipment.
Collapse
Affiliation(s)
- John R Giudicessi
- Department of Cardiovascular Medicine, Clinician-Investigator Training Program, Mayo Clinic, Rochester, MN; Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Mayo Clinic, Rochester, MN
| | - Peter A Noseworthy
- Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Mayo Clinic, Rochester, MN
| | - Paul A Friedman
- Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Mayo Clinic, Rochester, MN
| | - Michael J Ackerman
- Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Mayo Clinic, Rochester, MN; Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN; Department of Molecular Pharmacology and Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN.
| |
Collapse
|
37
|
Kistamás K, Veress R, Horváth B, Bányász T, Nánási PP, Eisner DA. Calcium Handling Defects and Cardiac Arrhythmia Syndromes. Front Pharmacol 2020; 11:72. [PMID: 32161540 PMCID: PMC7052815 DOI: 10.3389/fphar.2020.00072] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/24/2020] [Indexed: 12/13/2022] Open
Abstract
Calcium ions (Ca2+) play a major role in the cardiac excitation-contraction coupling. Intracellular Ca2+ concentration increases during systole and falls in diastole thereby determining cardiac contraction and relaxation. Normal cardiac function also requires perfect organization of the ion currents at the cellular level to drive action potentials and to maintain action potential propagation and electrical homogeneity at the tissue level. Any imbalance in Ca2+ homeostasis of a cardiac myocyte can lead to electrical disturbances. This review aims to discuss cardiac physiology and pathophysiology from the elementary membrane processes that can cause the electrical instability of the ventricular myocytes through intracellular Ca2+ handling maladies to inherited and acquired arrhythmias. Finally, the paper will discuss the current therapeutic approaches targeting cardiac arrhythmias.
Collapse
Affiliation(s)
- Kornél Kistamás
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Roland Veress
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Bányász
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter P Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Dental Physiology, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - David A Eisner
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
38
|
Late sodium current in human, canine and guinea pig ventricular myocardium. J Mol Cell Cardiol 2020; 139:14-23. [DOI: 10.1016/j.yjmcc.2019.12.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/18/2019] [Accepted: 12/25/2019] [Indexed: 02/06/2023]
|
39
|
Pourrier M, Fedida D. The Emergence of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes (hiPSC-CMs) as a Platform to Model Arrhythmogenic Diseases. Int J Mol Sci 2020; 21:ijms21020657. [PMID: 31963859 PMCID: PMC7013748 DOI: 10.3390/ijms21020657] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/13/2022] Open
Abstract
There is a need for improved in vitro models of inherited cardiac diseases to better understand basic cellular and molecular mechanisms and advance drug development. Most of these diseases are associated with arrhythmias, as a result of mutations in ion channel or ion channel-modulatory proteins. Thus far, the electrophysiological phenotype of these mutations has been typically studied using transgenic animal models and heterologous expression systems. Although they have played a major role in advancing the understanding of the pathophysiology of arrhythmogenesis, more physiological and predictive preclinical models are necessary to optimize the treatment strategy for individual patients. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have generated much interest as an alternative tool to model arrhythmogenic diseases. They provide a unique opportunity to recapitulate the native-like environment required for mutated proteins to reproduce the human cellular disease phenotype. However, it is also important to recognize the limitations of this technology, specifically their fetal electrophysiological phenotype, which differentiates them from adult human myocytes. In this review, we provide an overview of the major inherited arrhythmogenic cardiac diseases modeled using hiPSC-CMs and for which the cellular disease phenotype has been somewhat characterized.
Collapse
Affiliation(s)
- Marc Pourrier
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
- IonsGate Preclinical Services Inc., Vancouver, BC V6T 1Z3, Canada
- Correspondence:
| | - David Fedida
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
| |
Collapse
|
40
|
Piccirillo G, Moscucci F, Pofi R, D'Alessandro G, Minnetti M, Isidori AM, Francomano D, Lenzi A, Puddu PE, Alexandre J, Magrì D, Aversa A. Changes in left ventricular repolarization after short-term testosterone replacement therapy in hypogonadal males. J Endocrinol Invest 2019; 42:1051-1065. [PMID: 30838540 PMCID: PMC6692303 DOI: 10.1007/s40618-019-01026-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 02/14/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIM Evidences suggest that androgen deficiency is associated with sudden cardiac death (SCD). Our purpose was to analyse some electrocardiographic (ECG) markers of repolarization phase in hypogonadal patients either at baseline or after testosterone replacement therapy (TRT). PATIENTS AND METHODS Baseline and after 6 months of testosterone replacement therapy, 14 hypogonadal patients and 10 age-matched controls underwent a short-term ECG recordings at rest and immediately after a maximal exercise test. The following ECG parameters have been collected: QTe (the interval between the q wave the end of T wave), QTp (the interval between the q wave and the peak of T wave), and Te (the interval between the peak and the end of T wave). RESULTS At baseline, in the hypogonadal patients, corrected QTe and QTp values were longer at rest than in the controls at rest (p < 0.05), whereas, during the recovery phase, only the QTp remained significantly longer (p < 0.05). After TRT, hypogonadal patients showed an improvement only in Te (p < 0.05). Conversely, any difference between hypogonadal patients and control subjects was found with respect to the markers of temporal dispersion of repolarization phases, except for a worse QTp → Te coherence (p = 0.001) obtained during the recovery phase. CONCLUSIONS In conclusion, at rest, hypogonadal patients suffer from a stable increase in the myocardial repolarization phase without an increase in its temporal dispersion and, hence, the SCD risk seems to be low.
Collapse
Affiliation(s)
- G Piccirillo
- Dipartimento di Scienze Cardiovascolari, Respiratorie, Nefrologiche, Anestesiologiche e Geriatriche, Policlinico Umberto I, "La Sapienza" University of Rome, Viale del Policlinico, 00185, Rome, Italy
| | - F Moscucci
- Dipartimento di Scienze Cardiovascolari, Respiratorie, Nefrologiche, Anestesiologiche e Geriatriche, Policlinico Umberto I, "La Sapienza" University of Rome, Viale del Policlinico, 00185, Rome, Italy.
| | - R Pofi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - G D'Alessandro
- Dipartimento di Scienze Cardiovascolari, Respiratorie, Nefrologiche, Anestesiologiche e Geriatriche, Policlinico Umberto I, "La Sapienza" University of Rome, Viale del Policlinico, 00185, Rome, Italy
| | - M Minnetti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - A M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - D Francomano
- Division of Internal Medicine and Endocrinology, Madonna delle Grazie Hospital, Velletri, Rome, Italy
| | - A Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - P E Puddu
- Dipartimento di Scienze Cardiovascolari, Respiratorie, Nefrologiche, Anestesiologiche e Geriatriche, Policlinico Umberto I, "La Sapienza" University of Rome, Viale del Policlinico, 00185, Rome, Italy
- EA 4650, Signalisation, électrophysiologie et imagerie des lésions d'ischémie reperfusion myocardique, Université de Caen, Normandie, France
| | - J Alexandre
- EA 4650, Signalisation, électrophysiologie et imagerie des lésions d'ischémie reperfusion myocardique, Université de Caen, Normandie, France
- Department of Pharmacology, CHU Caen, Caen, France
| | - D Magrì
- Dipartimento di Medicina Clinica e Molecolare, S. Andrea Hospital, "Sapienza" University of Rome, Rome, Italy
| | - A Aversa
- Department of Experimental and Clinical Medicine, University of Catanzaro « Magna Grecia », Catanzaro, Italy
| |
Collapse
|
41
|
van Mil A, Balk GM, Neef K, Buikema JW, Asselbergs FW, Wu SM, Doevendans PA, Sluijter JPG. Modelling inherited cardiac disease using human induced pluripotent stem cell-derived cardiomyocytes: progress, pitfalls, and potential. Cardiovasc Res 2018; 114:1828-1842. [PMID: 30169602 PMCID: PMC6887927 DOI: 10.1093/cvr/cvy208] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/06/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022] Open
Abstract
In the past few years, the use of specific cell types derived from induced pluripotent stem cells (iPSCs) has developed into a powerful approach to investigate the cellular pathophysiology of numerous diseases. Despite advances in therapy, heart disease continues to be one of the leading causes of death in the developed world. A major difficulty in unravelling the underlying cellular processes of heart disease is the extremely limited availability of viable human cardiac cells reflecting the pathological phenotype of the disease at various stages. Thus, the development of methods for directed differentiation of iPSCs to cardiomyocytes (iPSC-CMs) has provided an intriguing option for the generation of patient-specific cardiac cells. In this review, a comprehensive overview of the currently published iPSC-CM models for hereditary heart disease is compiled and analysed. Besides the major findings of individual studies, detailed methodological information on iPSC generation, iPSC-CM differentiation, characterization, and maturation is included. Both, current advances in the field and challenges yet to overcome emphasize the potential of using patient-derived cell models to mimic genetic cardiac diseases.
Collapse
Affiliation(s)
- Alain van Mil
- Division Heart and Lungs, Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Internal Mail No G03.550, GA Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Geerthe Margriet Balk
- Division Heart and Lungs, Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Internal Mail No G03.550, GA Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Klaus Neef
- Division Heart and Lungs, Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Internal Mail No G03.550, GA Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jan Willem Buikema
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Folkert W Asselbergs
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Faculty of Population Health Sciences, Institute of Cardiovascular Science, University College London, London, UK
- Durrer Center for Cardiovascular Research, Netherlands Heart Institute, Utrecht, the Netherlands
- Farr Institute of Health Informatics Research and Institute of Health Informatics, University College London, London, UK
| | - Sean M Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Pieter A Doevendans
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Joost P G Sluijter
- Division Heart and Lungs, Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Internal Mail No G03.550, GA Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
42
|
Landaw J, Qu Z. Control of voltage-driven instabilities in cardiac myocytes with memory. CHAOS (WOODBURY, N.Y.) 2018; 28:113122. [PMID: 30501225 PMCID: PMC6274634 DOI: 10.1063/1.5040854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/29/2018] [Indexed: 06/09/2023]
Abstract
Sudden cardiac death is known to be associated with dynamical instabilities in the heart, and thus control of dynamical instabilities is considered a potential therapeutic strategy. Different control methods were developed previously, including time-delayed feedback pacing control and constant diastolic interval pacing control. Experimental, theoretical, and simulation studies have examined the efficacy of these control methods in stabilizing action potential dynamics. In this study, we apply these control methods to control complex action potential (AP) dynamics under two diseased conditions: early repolarization syndrome and long QT syndrome, in which voltage-driven instabilities occur in the presence of short-term cardiac memory. In addition, we also develop a feedback pacing method to stabilize these instabilities. We perform theoretical analyses using iterated map models and carry out numerical simulations of AP models. We show that under the normal condition where the memory effect is minimal, all three methods can effectively control the action potential duration (APD) dynamics. Under the two diseased conditions where the memory effect is exacerbated, constant diastolic pacing control is least effective, while the feedback pacing control is most effective. Under a very strong memory effect, all three methods fail to stabilize the voltage-driven instabilities. The failure of effective control is due to memory and the all-or-none AP dynamics which results in very steep changes in APD.
Collapse
Affiliation(s)
- Julian Landaw
- Department of Medicine (Cardiology), University of California, Los Angeles, California 90095, USA
| | - Zhilin Qu
- Department of Medicine (Cardiology), University of California, Los Angeles, California 90095, USA
| |
Collapse
|
43
|
Abstract
Women have a longer QT interval than men, which appears to evolve after puberty suggesting that sex hormones have an influence on cardiac electrophysiology. Sex hormones do in fact regulate cardiac ion channels via genomic and nongenomic pathways. Women are at greater risk for life-threatening arrhythmias under conditions that prolong the QT interval. In addition, women exhibit greater sensitivity to QT interval–prolonging drugs. Female sex has also an impact on propensity to cardiovascular disease, including atrial fibrillation. However, ex vivo recorded atrial action potentials (APs) from female and male patients in atrial fibrillation did not exhibit significant differences in shape, except that APs from women had slower upstroke velocity. It is concluded that sex-related differences should be taken into account not only in the clinics, but also in basic research.
Collapse
Affiliation(s)
- Ursula Ravens
- Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg • Bad Krozingen, Medical Faculty, University of Freiburg, Germany; Institute of Physiology, Medical Faculty Carl Gustav Carus, TU Dresden, Germany
- Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg • Bad Krozingen, Medical Faculty, University of Freiburg, Germany; Institute of Physiology, Medical Faculty Carl Gustav Carus, TU Dresden, Germany
| |
Collapse
|
44
|
Piccirillo G, Moscucci F, Mastropietri F, Di Iorio C, Mariani MV, Fabietti M, Stricchiola GM, Parrotta I, Sardella G, Mancone M, Magrì D. Possible predictive role of electrical risk score on transcatheter aortic valve replacement outcomes in older patients: preliminary data. Clin Interv Aging 2018; 13:1657-1667. [PMID: 30237702 PMCID: PMC6138964 DOI: 10.2147/cia.s170226] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Objectives To evaluate the predicative power of the electrical risk score (ERS), a noninvasive and inexpensive test obtained by means of a standard 12-lead electrocardiogram (ECG), in a cohort of elderly patients who had undergone transcatheter aortic valve replacement (TAVR). Methods Survivors and non-survivors after TAVR at 1-year follow-up were compared in respect to the pre-procedural ERS as well as a number of other clinical and instrumental variables. ERS is composed of seven simple ECG markers: heart rate (>75 bpm); QRS duration (>110 ms); left ventricular hypertrophy (Sokolow–Lyon criteria); delayed QRS transition zone (≥ V4); frontal QRS-T angle (>90°); long QTBazett (>450 ms for men and >460 in women) or JTBazett (330 ms for men and >340 ms for women); and long Tpeak to Tend interval (Tp-e) (>89 ms). The trial was registered in ClinicalTrials.gov as NCT03145376. Results A total of 40 patients were evaluated. During the follow-up, the all-cause mortality rate was 25% (ten patients) with 15% of cardiovascular death (six patients). The ERS was the strongest predictor of all-cause (odds ratio 3.73, 95% CI: 1.44–9.66, P<0.05) or cardiovascular (odds ratio 3.95, 95% CI: 1.09–14.27, P<0.05) mortality. Receiver operating characteristic curves showed that ERS had the widest significant sensitivity-specificity area under the curve (AUC) predicting all-cause (AUC: 0.855, P<0.05) or cardiovascular mortality (AUC: 0.908, P<0.05). Conclusion ERS seems to be a useful noninvasive tool able to stratify the risk of mortality in 1-year follow-up of TAVR patients. These findings, however, require larger trials to be confirmed.
Collapse
Affiliation(s)
- Gianfranco Piccirillo
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiologic, and Geriatric Sciences, Policlinico Umberto I, "La Sapienza" University of Rome, Rome, Italy,
| | - Federica Moscucci
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiologic, and Geriatric Sciences, Policlinico Umberto I, "La Sapienza" University of Rome, Rome, Italy,
| | - Fabiola Mastropietri
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiologic, and Geriatric Sciences, Policlinico Umberto I, "La Sapienza" University of Rome, Rome, Italy,
| | - Claudia Di Iorio
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiologic, and Geriatric Sciences, Policlinico Umberto I, "La Sapienza" University of Rome, Rome, Italy,
| | - Marco Valerio Mariani
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiologic, and Geriatric Sciences, Policlinico Umberto I, "La Sapienza" University of Rome, Rome, Italy,
| | - Marcella Fabietti
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiologic, and Geriatric Sciences, Policlinico Umberto I, "La Sapienza" University of Rome, Rome, Italy,
| | - Gaetana M Stricchiola
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiologic, and Geriatric Sciences, Policlinico Umberto I, "La Sapienza" University of Rome, Rome, Italy,
| | - Ilaria Parrotta
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiologic, and Geriatric Sciences, Policlinico Umberto I, "La Sapienza" University of Rome, Rome, Italy,
| | - Gennaro Sardella
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiologic, and Geriatric Sciences, Policlinico Umberto I, "La Sapienza" University of Rome, Rome, Italy,
| | - Massimo Mancone
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiologic, and Geriatric Sciences, Policlinico Umberto I, "La Sapienza" University of Rome, Rome, Italy,
| | - Damiano Magrì
- Department of Molecular and Clinical Medicine, S. Andrea Hospital, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
45
|
Kume S, Shimomura T, Tateyama M, Kubo Y. Two mutations at different positions in the CNBH domain of the hERG channel accelerate deactivation and impair the interaction with the EAG domain. J Physiol 2018; 596:4629-4650. [PMID: 30086184 DOI: 10.1113/jp276208] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/06/2018] [Indexed: 12/24/2022] Open
Abstract
KEY POINTS In the human ether-a-go-go related gene (hERG) channel, both the ether-a-go-go (EAG) domain in the N-terminal and the cyclic nucleotide (CN) binding homology (CNBH) domain in the C-terminal cytoplasmic region are known to contribute to the characteristic slow deactivation. Mutations of Phe860 in the CNBH domain, reported to fill the CN binding pocket, accelerate the deactivation and decrease the fluorescence resonance energy transfer (FRET) efficiencies between the EAG and CNBH domains. An electrostatic interaction between Arg696 and Asp727 in the C-linker domain, critical for HCN and CNG channels, is not formed in the hERG channel. Mutations of newly identified electrostatically interacting pair, Asp727 in the C-linker and Arg752 in the CNBH domains, accelerate the deactivation and decrease FRET efficiency. Voltage-dependent changes in FRET efficiency were not detected. These results suggest that the acceleration of the deactivation by mutations of C-terminal domains is a result of the lack of interaction between the EAG and CNBH domains. ABSTRACT The human ether-a-go-go related gene (hERG) channel shows characteristic slow deactivation, and the contribution of both of the N-terminal cytoplasmic ether-a-go-go (EAG) domain and the C-terminal cytoplasmic cyclic nucleotide (CN) binding homology (CNBH) domain is well known. The interaction between these domains is known to be critical for slow deactivation. We analysed the effects of mutations in the CNBH domain and its upstream C-linker domain on slow deactivation and the interaction between the EAG and CNBH domains by electrophysiological and fluorescence resonance energy transfer (FRET) analyses using Xenopus oocyte and HEK293T cell expression systems. We first observed that mutations of Phe860 in the CNBH domain, which is reported to fill the CN binding pocket as an intrinsic ligand, accelerate deactivation and eliminate the inter-domain interaction. Next, we observed that the salt bridge between Arg696 and Asp727 in the C-linker domain, which is reported to be critical for the function of CN-regulated channels, is not formed. We newly identified an electrostatically interacting pair critical for slow deactivation: Asp727 and Arg752 in the CNBH domain. Their mutations also impaired the inter-domain interaction. Taking these results together, both mutations of the intrinsic ligand (Phe860) and a newly identified salt bridge pair (Asp727 and Arg752) in the hERG channel accelerated deactivation and also decreased the interaction between the EAG and CNBH domains. Voltage-dependent changes in FRET efficiency between the two domains were not detected. The results suggest that the CNBH domain contributes to slow deactivation of the hERG channel by a mechanism involving the EAG domain.
Collapse
Affiliation(s)
- Shinichiro Kume
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Science, SOKENDAI, Hayama, Japan.,Present address: Department of Pathophysiology, Oita University School of Medicine, Yufu, Oita, Japan
| | - Takushi Shimomura
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Science, SOKENDAI, Hayama, Japan
| | - Michihiro Tateyama
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Science, SOKENDAI, Hayama, Japan
| | - Yoshihiro Kubo
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Science, SOKENDAI, Hayama, Japan
| |
Collapse
|
46
|
Garg P, Oikonomopoulos A, Chen H, Li Y, Lam CK, Sallam K, Perez M, Lux RL, Sanguinetti MC, Wu JC. Genome Editing of Induced Pluripotent Stem Cells to Decipher Cardiac Channelopathy Variant. J Am Coll Cardiol 2018; 72:62-75. [PMID: 29957233 PMCID: PMC6050025 DOI: 10.1016/j.jacc.2018.04.041] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 03/13/2018] [Accepted: 04/12/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND The long QT syndrome (LQTS) is an arrhythmogenic disorder of QT interval prolongation that predisposes patients to life-threatening ventricular arrhythmias such as Torsades de pointes and sudden cardiac death. Clinical genetic testing has emerged as the standard of care to identify genetic variants in patients suspected of having LQTS. However, these results are often confounded by the discovery of variants of uncertain significance (VUS), for which there is insufficient evidence of pathogenicity. OBJECTIVES The purpose of this study was to demonstrate that genome editing of patient-specific induced pluripotent stem cells (iPSCs) can be a valuable approach to delineate the pathogenicity of VUS in cardiac channelopathy. METHODS Peripheral blood mononuclear cells were isolated from a carrier with a novel missense variant (T983I) in the KCNH2 (LQT2) gene and an unrelated healthy control subject. iPSCs were generated using an integration-free Sendai virus and differentiated to iPSC-derived cardiomyocytes (CMs). RESULTS Whole-cell patch clamp recordings revealed significant prolongation of the action potential duration (APD) and reduced rapidly activating delayed rectifier K+ current (IKr) density in VUS iPSC-CMs compared with healthy control iPSC-CMs. ICA-105574, a potent IKr activator, enhanced IKr magnitude and restored normal action potential duration in VUS iPSC-CMs. Notably, VUS iPSC-CMs exhibited greater propensity to proarrhythmia than healthy control cells in response to high-risk torsadogenic drugs (dofetilide, ibutilide, and azimilide), suggesting a compromised repolarization reserve. Finally, the selective correction of the causal variant in iPSC-CMs using CRISPR/Cas9 gene editing (isogenic control) normalized the aberrant cellular phenotype, whereas the introduction of the homozygous variant in healthy control cells recapitulated hallmark features of the LQTS disorder. CONCLUSIONS The results suggest that the KCNH2T983I VUS may be classified as potentially pathogenic.
Collapse
Affiliation(s)
- Priyanka Garg
- Stanford Cardiovascular Institute and Department of Medicine, Division of Cardiology, Stanford University, Stanford, California
| | - Angelos Oikonomopoulos
- Stanford Cardiovascular Institute and Department of Medicine, Division of Cardiology, Stanford University, Stanford, California
| | - Haodong Chen
- Stanford Cardiovascular Institute and Department of Medicine, Division of Cardiology, Stanford University, Stanford, California
| | - Yingxin Li
- Stanford Cardiovascular Institute and Department of Medicine, Division of Cardiology, Stanford University, Stanford, California
| | - Chi Keung Lam
- Stanford Cardiovascular Institute and Department of Medicine, Division of Cardiology, Stanford University, Stanford, California
| | - Karim Sallam
- Stanford Cardiovascular Institute and Department of Medicine, Division of Cardiology, Stanford University, Stanford, California
| | - Marco Perez
- Stanford Cardiovascular Institute and Department of Medicine, Division of Cardiology, Stanford University, Stanford, California; Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine, Stanford University, Stanford, California
| | - Robert L Lux
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah
| | - Michael C Sanguinetti
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Joseph C Wu
- Stanford Cardiovascular Institute and Department of Medicine, Division of Cardiology, Stanford University, Stanford, California.
| |
Collapse
|
47
|
Arcos L, Rincón C, Vanegas D, Medina R. Tormenta eléctrica y torsade de pointes asociados a tratamiento con antimoniales en un paciente con leishmaniasis cutánea. REVISTA COLOMBIANA DE CARDIOLOGÍA 2018. [DOI: 10.1016/j.rccar.2017.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
48
|
Duan J, Tao J, Zhai M, Li C, Zhou N, Lv J, Wang L, Lin L, Bai R. Anticancer drugs-related QTc prolongation, torsade de pointes and sudden death: current evidence and future research perspectives. Oncotarget 2018; 9:25738-25749. [PMID: 29876021 PMCID: PMC5986642 DOI: 10.18632/oncotarget.25008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 03/13/2018] [Indexed: 01/08/2023] Open
Abstract
Anticancer drugs may have proarrhythmic effects including drug-induced QT interval prolongation, which is of particular importance because it can lead to a fatal polymorphic ventricular tachycardia termed torsade de pointes (TdP). QT interval prolongation and TdP are rare life-threatening untoward effects of anticancer therapy, particularly with arsenic trioxides and anthracyclines, and even some novel molecular targeted drugs touted as 'tumor specific'. Several factors that affect myocardial repolarization can further increase the risk of TdP. This article reviews the mechanism of QT interval prolongation, risk factors for TdP and the QT toxicity of anticancer drugs as well as its management. Specific attention should be paid to high-risk populations such as patients with underlying heart diseases, electrolyte imbalance and bradycardia. To minimize the occurrence of QT interval prolongation and TdP, it is advisable to conduct a careful risk factor assessment before antitumor therapy. To this end, several new biomarkers have been introduced to predict TdP triggering and recent studies have pointed out the potential clinical relevance of genetic testing.
Collapse
Affiliation(s)
- Jialin Duan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jingwen Tao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Maocai Zhai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Chengpeng Li
- Department of Cardiology, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, P.R. China
| | - Ning Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jiagao Lv
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Lin Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Li Lin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Rong Bai
- Department of Cardiology, An Zhen Hospital, Capital Medical University, Beijing, P.R. China.,Texas Cardiac Arrhythmia Institute at St. David's Medical Center, Austin, TX, USA
| |
Collapse
|
49
|
Landaw J, Qu Z. Memory-induced nonlinear dynamics of excitation in cardiac diseases. Phys Rev E 2018; 97:042414. [PMID: 29758700 PMCID: PMC6542282 DOI: 10.1103/physreve.97.042414] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Indexed: 11/07/2022]
Abstract
Excitable cells, such as cardiac myocytes, exhibit short-term memory, i.e., the state of the cell depends on its history of excitation. Memory can originate from slow recovery of membrane ion channels or from accumulation of intracellular ion concentrations, such as calcium ion or sodium ion concentration accumulation. Here we examine the effects of memory on excitation dynamics in cardiac myocytes under two diseased conditions, early repolarization and reduced repolarization reserve, each with memory from two different sources: slow recovery of a potassium ion channel and slow accumulation of the intracellular calcium ion concentration. We first carry out computer simulations of action potential models described by differential equations to demonstrate complex excitation dynamics, such as chaos. We then develop iterated map models that incorporate memory, which accurately capture the complex excitation dynamics and bifurcations of the action potential models. Finally, we carry out theoretical analyses of the iterated map models to reveal the underlying mechanisms of memory-induced nonlinear dynamics. Our study demonstrates that the memory effect can be unmasked or greatly exacerbated under certain diseased conditions, which promotes complex excitation dynamics, such as chaos. The iterated map models reveal that memory converts a monotonic iterated map function into a nonmonotonic one to promote the bifurcations leading to high periodicity and chaos.
Collapse
Affiliation(s)
- Julian Landaw
- Department of Medicine (Cardiology), University of California, Los Angeles, California 90095, USA and Department of Biomathematics, University of California, Los Angeles, California 90095, USA
| | - Zhilin Qu
- Department of Medicine (Cardiology), University of California, Los Angeles, California 90095, USA and Department of Biomathematics, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
50
|
Zaniboni M. Short-term action potential memory and electrical restitution: A cellular computational study on the stability of cardiac repolarization under dynamic pacing. PLoS One 2018; 13:e0193416. [PMID: 29494628 PMCID: PMC5832261 DOI: 10.1371/journal.pone.0193416] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 02/09/2018] [Indexed: 01/24/2023] Open
Abstract
Electrical restitution (ER) is a major determinant of repolarization stability and, under fast pacing rate, it reveals memory properties of the cardiac action potential (AP), whose dynamics have never been fully elucidated, nor their ionic mechanisms. Previous studies have looked at ER mainly in terms of changes in AP duration (APD) when the preceding diastolic interval (DI) changes and described dynamic conditions where this relationship shows hysteresis which, in turn, has been proposed as a marker of short-term AP memory and repolarization stability. By means of numerical simulations of a non-propagated human ventricular AP, we show here that measuring ER as APD versus the preceding cycle length (CL) provides additional information on repolarization dynamics which is not contained in the companion formulation. We focus particularly on fast pacing rate conditions with a beat-to-beat variable CL, where memory properties emerge from APD vs CL and not from APD vs DI and should thus be stored in APD and not in DI. We provide an ion-currents characterization of such conditions under periodic and random CL variability, and show that the memory stored in APD plays a stabilizing role on AP repolarization under pacing rate perturbations. The gating kinetics of L-type calcium current seems to be the main determinant of this safety mechanism. We also show that, at fast pacing rate and under otherwise identical pacing conditions, a periodically beat-to-beat changing CL is more effective than a random one in stabilizing repolarization. In summary, we propose a novel view of short-term AP memory, differentially stored between systole and diastole, which opens a number of methodological and theoretical implications for the understanding of arrhythmia development.
Collapse
Affiliation(s)
- Massimiliano Zaniboni
- Department of Chemistry, Life Sciences and Environmental Sustainability - University of Parma Parco Area delle Scienze, Parma, Italy
- Center of Excellence for Toxicological Research (CERT) - University of Parma, Parma, Italy
- * E-mail:
| |
Collapse
|