1
|
Lazzerini PE, Boutjdir M. Autoimmune cardiac channelopathies and heart rhythm disorders: A contemporary review. Heart Rhythm 2025; 22:1541-1561. [PMID: 40058514 DOI: 10.1016/j.hrthm.2025.03.1879] [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] [Received: 01/17/2025] [Revised: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Cardiac arrhythmias still represent a major health problem worldwide, at least in part because the fundamental pathogenic mechanisms are not fully understood, thus affecting the efficacy of therapeutic measures. In fact, whereas cardiac arrhythmias are in most cases due to structural heart diseases, the underlying cause remains elusive in a significant number of patients despite intensive investigations even including postmortem examination and molecular autopsy. A large body of data progressively accumulated during the last decade provides strong evidence that autoimmune mechanisms may be involved in a significant number of such unexplained or poorly explained cardiac arrhythmias. Several proarrhythmic anti-cardiac ion channel autoantibodies have been discovered, in all cases able to directly interfere with the electrophysiologic properties of the heart but leading to different arrhythmic phenotypes, including long QT syndrome, short QT syndrome, and atrioventricular block. These autoantibodies, which may develop independent of a history of autoimmune diseases, could help explain a percentage of arrhythmic events of unknown origin, thereby opening new frontiers for diagnosis and treatment of heart rhythm disorders. Based on this evidence, the novel term autoimmune cardiac channelopathies was coined in 2017. Since then, the interest in the field of cardioimmunology has shown a tumultuous growth, so much so that the number of arrhythmogenic anti-ion channel autoantibodies reported has significantly increased, also in association with not previously described arrhythmic phenotypes, such as atrial fibrillation, Brugada syndrome, and ventricular fibrillation/cardiac arrest. Thus, an updated reassessment of this topic, also highlighting perspectives and unmet needs, has become necessary and represents the main objective of this review.
Collapse
Affiliation(s)
- Pietro Enea Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences, Division of Internal Medicine and Geriatrics, Electroimmunology Unit, University of Siena, Siena, Italy.
| | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, SUNY Downstate Health Sciences University, New York, New York; New York University Grossman School of Medicine, New York, New York.
| |
Collapse
|
2
|
Zaniboni M. In silico analysis of ventricular action potential with a current-voltage-time representation: Thresholds, membrane resistance, repolarization reserve. Physiol Rep 2024; 12:e70085. [PMID: 39529595 PMCID: PMC11555294 DOI: 10.14814/phy2.70085] [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: 05/15/2024] [Revised: 09/18/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
The waveform of ventricular action potential (AP) is a key determinant of the cardiac cycle, a marker of beating pathophysiology, and a target for anti-arrhythmic drug design. The information contained in the waveform, though, is limited to the actual dynamics of the AP under consideration. By measuring quasi-instantaneous current-voltage relationships during repolarization, I propose a three-dimensional representation of the ventricular AP which includes potential dynamic responses that the beat can show when electrically perturbed. This representation is described in the case of a numerically reconstructed ventricular AP, but it can be, at least partially, derived in real cardiomyocytes. Simulation allows to disclose the potentialities and the limitations of the approach, that can be extended to any non-cardiac AP. By reporting, at any AP time, the ion current available within the physiological membrane potential range at that time, the representation makes all together available: (1) refractory period, (2) thresholds for eliciting full or calcium-driven APs, (3) threshold for all-or-none repolarization, (4) membrane resistance during repolarization, (5) the safety of membrane repolarization. It provides further evidence of a negative membrane resistance during the late phase of ventricular AP and a quantitative estimate of repolarization reserve (RR), key determinants of repolarization dynamics.
Collapse
Affiliation(s)
- Massimiliano Zaniboni
- Department of Chemistry, Life Sciences and Environmental SustainabilityUniversity of Parma (Italy) ‐ Parco Area Delle ScienzeParmaItaly
| |
Collapse
|
3
|
Wessel N, Kim JS, Joung BY, Ko YG, Dischl D, Gapelyuk A, Lee YH, Kim KW, Park JW, Landmesser U. Magnetocardiography at rest predicts cardiac death in patients with acute chest pain. Front Cardiovasc Med 2023; 10:1258890. [PMID: 38155993 PMCID: PMC10752986 DOI: 10.3389/fcvm.2023.1258890] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023] Open
Abstract
Introduction Sudden cardiac arrest is a major cause of morbidity and mortality worldwide and remains a major public health problem for which better non-invasive prediction tools are needed. Primary preventive therapies, such as implantable cardioverter defibrillators, are not personalized and not predictive. Most of these devices do not deliver life-saving therapy during their lifetime. The individual relationship between fatal arrhythmias and cardiac function abnormalities in predicting cardiac death risk has rarely been explored. Methods We retrospectively analyzed the measurements at rest for 191 patients with acute chest pain (ACP) magnetocardiographically. Our recently introduced analyses are able to detect inhomogeneities of the depolarization and repolarization. Moreover, electrically silent phenomena-intracellular ionic currents as well as vortex currents-can be measured and quantified. All included ACP patients were recruited in 2009 at Yonsei University Hospital and were followed up until 2022. Results During half of the follow-up period (6.5 years), 11 patients died. Out of all the included nine clinical, eight magnetocardiographical, and nine newly introduced magnetoionographical parameters we tested in this study, three parameters revealed themselves to be outstanding at predicting death: heart rate-corrected QT (QTc) prolongation, depression of repolarization current IKr + IKs, and serum creatinine (all significant in Cox regression, p < 0.05). They clearly predicted cardiac death over the 6.5 years duration (sensitivity 90.9%, specificity 85.6%, negative predictive accuracy 99.4%). Cardiac death risk was more than ninefold higher in patients with low repolarization reserve and QTc prolongation in comparison with the remaining patients with ACP (p < 0.001). The non-parametric Kaplan-Meier statistics estimated significantly lower survival functions from their lifetime data (p < 0.001). Discussion To the best of our knowledge, these are the first data linking magnetocardiographical and magnetoionographical parameters and subsequent significant fatal events in people, suggesting structural and functional components to clinical life-threatening ventricular arrhythmogenesis. The findings support investigation of new prevention strategies and herald those new non-invasive techniques as complementary risk stratification tools.
Collapse
Affiliation(s)
- N. Wessel
- Department of Human Medicine, MSB Medical School Berlin GmbH, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Physics, Humboldt Universität zu Berlin, Berlin, Germany
| | - J. S. Kim
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - B. Y. Joung
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Y. G. Ko
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - D. Dischl
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - A. Gapelyuk
- Department of Physics, Humboldt Universität zu Berlin, Berlin, Germany
| | - Y. H. Lee
- Center for Biosignals, KRISS Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
| | - K. W. Kim
- Center for Biosignals, KRISS Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
| | - J. W. Park
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - U. Landmesser
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
4
|
Kingma J, Simard C, Drolet B. Overview of Cardiac Arrhythmias and Treatment Strategies. Pharmaceuticals (Basel) 2023; 16:844. [PMID: 37375791 DOI: 10.3390/ph16060844] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Maintenance of normal cardiac rhythm requires coordinated activity of ion channels and transporters that allow well-ordered propagation of electrical impulses across the myocardium. Disruptions in this orderly process provoke cardiac arrhythmias that may be lethal in some patients. Risk of common acquired arrhythmias is increased markedly when structural heart disease caused by myocardial infarction (due to fibrotic scar formation) or left ventricular dysfunction is present. Genetic polymorphisms influence structure or excitability of the myocardial substrate, which increases vulnerability or risk of arrhythmias in patients. Similarly, genetic polymorphisms of drug-metabolizing enzymes give rise to distinct subgroups within the population that affect specific drug biotransformation reactions. Nonetheless, identification of triggers involved in initiation or maintenance of cardiac arrhythmias remains a major challenge. Herein, we provide an overview of knowledge regarding physiopathology of inherited and acquired cardiac arrhythmias along with a summary of treatments (pharmacologic or non-pharmacologic) used to limit their effect on morbidity and potential mortality. Improved understanding of molecular and cellular aspects of arrhythmogenesis and more epidemiologic studies (for a more accurate portrait of incidence and prevalence) are crucial for development of novel treatments and for management of cardiac arrhythmias and their consequences in patients, as their incidence is increasing worldwide.
Collapse
Affiliation(s)
- John Kingma
- Department of Medicine, Ferdinand Vandry Pavillon, 1050 Av. de la Médecine, Québec City, QC G1V 0A6, Canada
| | - Chantale Simard
- Faculty of Pharmacy Ferdinand Vandry Pavillon, 1050 Av. de la Médecine, Québec City, QC G1V 0A6, Canada
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval 2725 Chemin Sainte-Foy, Québec City, QC G1V 4G5, Canada
| | - Benoît Drolet
- Faculty of Pharmacy Ferdinand Vandry Pavillon, 1050 Av. de la Médecine, Québec City, QC G1V 0A6, Canada
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval 2725 Chemin Sainte-Foy, Québec City, QC G1V 4G5, Canada
| |
Collapse
|
5
|
Kekenes-Huskey PM, Burgess DE, Sun B, Bartos DC, Rozmus ER, Anderson CL, January CT, Eckhardt LL, Delisle BP. Mutation-Specific Differences in Kv7.1 ( KCNQ1) and Kv11.1 ( KCNH2) Channel Dysfunction and Long QT Syndrome Phenotypes. Int J Mol Sci 2022; 23:7389. [PMID: 35806392 PMCID: PMC9266926 DOI: 10.3390/ijms23137389] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
The electrocardiogram (ECG) empowered clinician scientists to measure the electrical activity of the heart noninvasively to identify arrhythmias and heart disease. Shortly after the standardization of the 12-lead ECG for the diagnosis of heart disease, several families with autosomal recessive (Jervell and Lange-Nielsen Syndrome) and dominant (Romano-Ward Syndrome) forms of long QT syndrome (LQTS) were identified. An abnormally long heart rate-corrected QT-interval was established as a biomarker for the risk of sudden cardiac death. Since then, the International LQTS Registry was established; a phenotypic scoring system to identify LQTS patients was developed; the major genes that associate with typical forms of LQTS were identified; and guidelines for the successful management of patients advanced. In this review, we discuss the molecular and cellular mechanisms for LQTS associated with missense variants in KCNQ1 (LQT1) and KCNH2 (LQT2). We move beyond the "benign" to a "pathogenic" binary classification scheme for different KCNQ1 and KCNH2 missense variants and discuss gene- and mutation-specific differences in K+ channel dysfunction, which can predispose people to distinct clinical phenotypes (e.g., concealed, pleiotropic, severe, etc.). We conclude by discussing the emerging computational structural modeling strategies that will distinguish between dysfunctional subtypes of KCNQ1 and KCNH2 variants, with the goal of realizing a layered precision medicine approach focused on individuals.
Collapse
Affiliation(s)
- Peter M. Kekenes-Huskey
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Don E. Burgess
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (D.E.B.); (E.R.R.)
| | - Bin Sun
- Department of Pharmacology, Harbin Medical University, Harbin 150081, China;
| | | | - Ezekiel R. Rozmus
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (D.E.B.); (E.R.R.)
| | - Corey L. Anderson
- Cellular and Molecular Arrythmias Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (C.L.A.); (C.T.J.); (L.L.E.)
| | - Craig T. January
- Cellular and Molecular Arrythmias Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (C.L.A.); (C.T.J.); (L.L.E.)
| | - Lee L. Eckhardt
- Cellular and Molecular Arrythmias Program, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (C.L.A.); (C.T.J.); (L.L.E.)
| | - Brian P. Delisle
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; (D.E.B.); (E.R.R.)
| |
Collapse
|
6
|
Abramochkin DV, Filatova TS, Pustovit KB, Voronina YA, Kuzmin VS, Vornanen M. Ionic currents underlying different patterns of electrical activity in working cardiac myocytes of mammals and non-mammalian vertebrates. Comp Biochem Physiol A Mol Integr Physiol 2022; 268:111204. [PMID: 35346823 DOI: 10.1016/j.cbpa.2022.111204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/19/2022]
Abstract
The orderly contraction of the vertebrate heart is determined by generation and propagation of cardiac action potentials (APs). APs are generated by the integrated activity of time- and voltage-dependent ionic channels which carry inward Na+ and Ca2+ currents, and outward K+ currents. This review compares atrial and ventricular APs and underlying ion currents between different taxa of vertebrates. We have collected literature data and attempted to find common electrophysiological features for two or more vertebrate groups, show differences between taxa and cardiac chambers, and indicate gaps in the existing data. Although electrical excitability of the heart in all vertebrates is based on the same superfamily of channels, there is a vast variability of AP waveforms between atrial and ventricular myocytes, between different species of the same vertebrate class and between endothermic and ectothermic animals. The wide variability of AP shapes is related to species-specific differences in animal size, heart rate, stage of ontogenetic development, excitation-contraction coupling, temperature and oxygen availability. Some of the differences between taxa are related to evolutionary development of genomes, which appear e.g. in the expression of different Na+ and K+ channel orthologues in cardiomyocytes of vertebrates. There is a wonderful variability of AP shapes and underlying ion currents with which electrical excitability of vertebrate heart can be generated depending on the intrinsic and extrinsic conditions of animal body. This multitude of ionic mechanisms provides excellent material for studying how the function of the vertebrate heart can adapt or acclimate to prevailing physiological and environmental conditions.
Collapse
Affiliation(s)
- Denis V Abramochkin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye gory, 1, 12, Moscow 119234, Russia.
| | - Tatiana S Filatova
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye gory, 1, 12, Moscow 119234, Russia
| | - Ksenia B Pustovit
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye gory, 1, 12, Moscow 119234, Russia
| | - Yana A Voronina
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye gory, 1, 12, Moscow 119234, Russia; Laboratory of Cardiac Electrophysiology, National Medical Research Center for Cardiology, 3(rd) Cherepkovskaya str., 15A, Moscow, Russia
| | - Vladislav S Kuzmin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskiye gory, 1, 12, Moscow 119234, Russia; Department of Physiology, Pirogov Russian National Research Medical University, Ostrovityanova str., 1, Moscow, Russia
| | - Matti Vornanen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| |
Collapse
|
7
|
The grapefruit polyphenol naringenin inhibits multiple cardiac ion channels. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:735-740. [PMID: 35412073 DOI: 10.1007/s00210-022-02240-4] [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] [Received: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 01/08/2023]
Abstract
Drinking fresh grapefruit juice is associated with a significant prolongation of the QT segment on the electrocardiogram (ECG) in healthy volunteers. Among the prominent polyphenols contained in citrus fruits and primarily in grapefruit, the flavonoid naringenin is known to be a blocker of the human ether-a-go-go related gene (hERG) potassium channel. Here we hypothesized that naringenin could interfere with other major ion channels shaping the cardiac ventricular action potential (AP). To test this hypothesis, we examined the effects of naringenin on the seven channels comprising the Comprehensive in vitro Pro-Arrhythmia (CiPA) ion channel panel for early arrhythmogenic risk assessment in drug discovery and development. We used automated population patch-clamp of human ion channels heterologously expressed in mammalian cells to evaluate half-maximal inhibitory concentrations (IC50). Naringenin blocked all CiPA ion channels tested with IC50 values in the 30-100 µM concentration-range. The rank-order of channel sensitivity was the following: hERG > Kir2.1 > NaV1.5 (late current) > NaV1.5 (peak current) > KV7.1 > KV4.3 > CaV1.2. This multichannel inhibitory profile of naringenin suggests exercising caution when large amounts of grapefruit juice or other citrus juices enriched in this flavonoid polyphenol are drunk in conjunction with QT prolonging drugs or by carriers of congenital long-QT syndromes.
Collapse
|
8
|
Sher A, Niederer SA, Mirams GR, Kirpichnikova A, Allen R, Pathmanathan P, Gavaghan DJ, van der Graaf PH, Noble D. A Quantitative Systems Pharmacology Perspective on the Importance of Parameter Identifiability. Bull Math Biol 2022; 84:39. [PMID: 35132487 PMCID: PMC8821410 DOI: 10.1007/s11538-021-00982-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 11/30/2021] [Indexed: 12/31/2022]
Abstract
There is an inherent tension in Quantitative Systems Pharmacology (QSP) between the need to incorporate mathematical descriptions of complex physiology and drug targets with the necessity of developing robust, predictive and well-constrained models. In addition to this, there is no “gold standard” for model development and assessment in QSP. Moreover, there can be confusion over terminology such as model and parameter identifiability; complex and simple models; virtual populations; and other concepts, which leads to potential miscommunication and misapplication of methodologies within modeling communities, both the QSP community and related disciplines. This perspective article highlights the pros and cons of using simple (often identifiable) vs. complex (more physiologically detailed but often non-identifiable) models, as well as aspects of parameter identifiability, sensitivity and inference methodologies for model development and analysis. The paper distills the central themes of the issue of identifiability and optimal model size and discusses open challenges.
Collapse
Affiliation(s)
- Anna Sher
- Pfizer Worldwide Research, Development and Medical, Massachusetts, USA.
| | | | - Gary R Mirams
- Centre for Mathematical Medicine and Biology, Mathematical Sciences, University of Nottingham, Nottingham, UK
| | | | - Richard Allen
- Pfizer Worldwide Research, Development and Medical, Massachusetts, USA
| | - Pras Pathmanathan
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Maryland, USA
| | - David J Gavaghan
- Department of Computer Science, University of Oxford, Oxford, UK
| | | | - Denis Noble
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Safe electrophysiologic profile of dexmedetomidine in different experimental arrhythmia models. Sci Rep 2021; 11:23940. [PMID: 34907251 PMCID: PMC8671395 DOI: 10.1038/s41598-021-03364-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022] Open
Abstract
Previous studies suggest an impact of dexmedetomidine on cardiac electrophysiology. However, experimental data is sparse. Therefore, purpose of this study was to investigate the influence of dexmedetomidine on different experimental models of proarrhythmia. 50 rabbit hearts were explanted and retrogradely perfused. The first group (n = 12) was treated with dexmedetomidine in ascending concentrations (3, 5 and 10 µM). Dexmedetomidine did not substantially alter action potential duration (APD) but reduced spatial dispersion of repolarization (SDR) and rendered the action potentials rectangular, resulting in no proarrhythmia. In further 12 hearts, erythromycin (300 µM) was administered to simulate long-QT-syndrome-2 (LQT2). Additional treatment with dexmedetomidine reduced SDR, thereby suppressing torsade de pointes. In the third group (n = 14), 0.5 µM veratridine was added to reduce the repolarization reserve. Further administration of dexmedetomidine did not influence APD, SDR or the occurrence of arrhythmias. In the last group (n = 12), a combination of acetylcholine (1 µM) and isoproterenol (1 µM) was used to facilitate atrial fibrillation. Additional treatment with dexmedetomidine prolonged the atrial APD but did not reduce AF episodes. In this study, dexmedetomidine did not significantly alter cardiac repolarization duration and was not proarrhythmic in different models of ventricular and atrial arrhythmias. Of note, dexmedetomidine might be antiarrhythmic in acquired LQT2 by reducing SDR.
Collapse
|
10
|
Lazzerini PE, Laghi-Pasini F, Boutjdir M, Capecchi PL. Anti-Ro/SSA Antibodies and the Autoimmune Long-QT Syndrome. Front Med (Lausanne) 2021; 8:730161. [PMID: 34552948 PMCID: PMC8450397 DOI: 10.3389/fmed.2021.730161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/11/2021] [Indexed: 01/08/2023] Open
Abstract
Autoimmunity is increasingly recognized as a novel pathogenic mechanism for cardiac arrhythmias. Several arrhythmogenic autoantibodies have been identified, cross-reacting with different types of surface proteins critically involved in the cardiomyocyte electrophysiology, primarily ion channels (autoimmune cardiac channelopathies). Specifically, some of these autoantibodies can prolong the action potential duration leading to acquired long-QT syndrome (LQTS), a condition known to increase the risk of life-threatening ventricular arrhythmias, particularly Torsades de Pointes (TdP). The most investigated form of autoimmune LQTS is associated with the presence of circulating anti-Ro/SSA-antibodies, frequently found in patients with autoimmune diseases (AD), but also in a significant proportion of apparently healthy subjects of the general population. Accumulating evidence indicates that anti-Ro/SSA-antibodies can markedly delay the ventricular repolarization via a direct inhibitory cross-reaction with the extracellular pore region of the human-ether-a-go-go-related (hERG) potassium channel, resulting in a higher propensity for anti-Ro/SSA-positive subjects to develop LQTS and ventricular arrhythmias/TdP. Recent population data demonstrate that the risk of LQTS in subjects with circulating anti-Ro/SSA antibodies is significantly increased independent of a history of overt AD, intriguingly suggesting that these autoantibodies may silently contribute to a number of cases of ventricular arrhythmias and cardiac arrest in the general population. In this review, we highlight the current knowledge in this topic providing complementary basic, clinical and population health perspectives.
Collapse
Affiliation(s)
- Pietro Enea Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Franco Laghi-Pasini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Mohamed Boutjdir
- Veterans Affairs New York Harbor Healthcare System, State University of New York Downstate Medical Center, New York, NY, United States.,New York University School of Medicine, New York, NY, United States
| | - Pier Leopoldo Capecchi
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| |
Collapse
|
11
|
Hommers L, Scherf-Clavel M, Stempel R, Roth J, Falter M, Deckert J, Mattheisen M, Unterecker S, Gawlik M. Antipsychotics in routine treatment are minor contributors to QT prolongation compared to genetics and age. J Psychopharmacol 2021; 35:1127-1133. [PMID: 33779379 PMCID: PMC8436313 DOI: 10.1177/02698811211003477] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Drug-induced prolongation of cardiac repolarization limits the treatment with many psychotropic drugs. Recently, the contribution of polygenic variation to the individual duration of the QT interval was identified. AIMS To explore the interaction between antipsychotic drugs and the individual polygenic influence on the QT interval. METHODS Retrospective analysis of clinical and genotype data of 804 psychiatric inpatients diagnosed with a psychotic disorder. The individual polygenic influence on the QT interval was calculated according to the method of Arking et al. RESULTS Linear regression modelling showed a significant association of the individual polygenic QT interval score (ßstd = 0.176, p < 0.001) and age (ßstd = 0.139, p < 0.001) with the QTc interval corrected according to Fridericia's formula. Sex showed a nominal trend towards significance (ßstd = 0.064, p = 0.064). No association was observed for the number of QT prolonging drugs according to AZCERT taken. Subsample analysis (n = 588) showed a significant association of potassium serum concentrations with the QTc interval (ßstd = -0.104, p = 0.010). Haloperidol serum concentrations were associated with the QTc interval only in single medication analysis (n = 26, ßstd = 0.101, p = 0.004), but not in multivariate regression analysis. No association was observed for aripiprazole, clozapine, quetiapine and perazine, while olanzapine and the sum of risperidone and its metabolite showed a negative association. CONCLUSIONS Individual genetic factors and age are main determinants of the QT interval. Antipsychotic drug serum concentrations within the therapeutic range contribute to QTc prolongation on an individual level.
Collapse
Affiliation(s)
- Leif Hommers
- Center for Mental Health, University Hospital of Würzburg, Würzburg, Germany
- Interdisciplinary Center for Clinical Research, University Hospital of Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center (CHFC), University Hospital of Würzburg, Würzburg, Germany
- Leif Hommers, Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Margarete-Höppel-Platz 1, Würzburg, 97080, Germany.
| | - Maike Scherf-Clavel
- Center for Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Roberta Stempel
- Center for Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Julian Roth
- Center for Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Matthias Falter
- Center for Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Jürgen Deckert
- Center for Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Manuel Mattheisen
- Center for Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Stefan Unterecker
- Center for Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Micha Gawlik
- Center for Mental Health, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
12
|
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: 0.5] [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
|
13
|
Lazzerini PE, Cartocci A, Qu YS, Saponara S, Furini S, Fusi F, Fabris F, Gamberucci A, El-Sherif N, Cevenini G, Pettini F, Laghi-Pasini F, Acampa M, Bertolozzi I, Capecchi PL, Lazaro D, Boutjdir M. Proton Pump Inhibitors Directly Block hERG-Potassium Channel and Independently Increase the Risk of QTc Prolongation in a Large Cohort of US Veterans. Circ Arrhythm Electrophysiol 2021; 14:e010042. [PMID: 34143643 DOI: 10.1161/circep.121.010042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Pietro Enea Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences (P.E.L., F.L.-P., P.L.C.), University of Siena, Italy
| | - Alessandra Cartocci
- Department of Medical Biotechnologies (A.C., S.F., G.C., F.P.), University of Siena, Italy
| | - Yongxia Sarah Qu
- Research and Development Department, VA New York Harbor Healthcare System, SUNY Downstate Medical Center (Y.S.Q., F.F., N.E.-S., D.L., M.B.).,Department of Cardiology, New York Presbyterian Brooklyn Methodist Hospital (Y.S.Q.)
| | - Simona Saponara
- Department of Life Sciences (S.S.), University of Siena, Italy
| | - Simone Furini
- Department of Medical Biotechnologies (A.C., S.F., G.C., F.P.), University of Siena, Italy
| | - Fabio Fusi
- Department of Biotechnology, Chemistry and Pharmacy (F.F.), University of Siena, Italy
| | | | - Alessandra Gamberucci
- Department of Molecular and Developmental Medicine (A.G.), University of Siena, Italy
| | | | - Gabriele Cevenini
- Department of Medical Biotechnologies (A.C., S.F., G.C., F.P.), University of Siena, Italy.,Research and Development Department, VA New York Harbor Healthcare System, SUNY Downstate Medical Center (Y.S.Q., F.F., N.E.-S., D.L., M.B.)
| | - Francesco Pettini
- Department of Medical Biotechnologies (A.C., S.F., G.C., F.P.), University of Siena, Italy
| | - Franco Laghi-Pasini
- Department of Medical Sciences, Surgery and Neurosciences (P.E.L., F.L.-P., P.L.C.), University of Siena, Italy
| | - Maurizio Acampa
- Research and Development Department, VA New York Harbor Healthcare System, SUNY Downstate Medical Center (Y.S.Q., F.F., N.E.-S., D.L., M.B.).,Department of Neurological and Sensorineural Sciences, Stroke Unit, University Hospital of Siena, Italy (M.A.)
| | - Iacopo Bertolozzi
- Cardiology Intensive Therapy Unit, Department of Internal Medicine, Nuovo Ospedale San Giovanni di Dio, Florence, Italy (I.B.)
| | - Pier Leopoldo Capecchi
- Department of Medical Sciences, Surgery and Neurosciences (P.E.L., F.L.-P., P.L.C.), University of Siena, Italy
| | - Deana Lazaro
- Research and Development Department, VA New York Harbor Healthcare System, SUNY Downstate Medical Center (Y.S.Q., F.F., N.E.-S., D.L., M.B.)
| | - Mohamed Boutjdir
- Research and Development Department, VA New York Harbor Healthcare System, SUNY Downstate Medical Center (Y.S.Q., F.F., N.E.-S., D.L., M.B.).,Department of Medicine, NYU School of Medicine, New York, NY (M.B.)
| |
Collapse
|
14
|
Qiu H, Li HW, Zhang SH, Zhou XG, Li WP. Torsades de pointes episode in a woman with high-grade fever and inflammatory activation: A case report. World J Clin Cases 2021; 9:2899-2907. [PMID: 33969075 PMCID: PMC8058677 DOI: 10.12998/wjcc.v9.i12.2899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/07/2021] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND QT interval prolongation can induce torsades de pointes (TdP), a potentially fatal ventricular arrhythmia. Recently, an increasing number of non-cardiac drugs have been found to cause QT prolongation and/or TdP onset. Moreover, recent findings have demonstrated the key roles of systemic inflammatory activation and fever in promoting long-QT syndrome (LQTS) and TdP development.
CASE SUMMARY A 30-year-old woman was admitted with a moderate to high-grade episodic fever for two weeks. The patient was administered with multiple antibiotics after hospitalization but still had repeating fever and markedly elevated C-reactive protein. Once after a high fever, the patient suddenly lost consciousness, and electrocardiogram (ECG) showed transient TdP onset after frequent premature ventricular contraction. The patient recovered sinus rhythm and consciousness spontaneously, and post-TdP ECG revealed a prolonged QTc interval of 560 ms. The patient’s clinical manifestations and unresponsiveness to the antibiotics led to the final diagnosis of adult-onset Still’s disease (AOSD). There was no evidence of cardiac involvement. After the AOSD diagnosis, discontinuation of antibiotics and immediate initiation of intravenous dexamethasone administration resulted in the normal temperature and QTc interval. The genetic analysis identified that the patient and her father had heterozygous mutations in KCNH2 (c.1370C>T) and AKAP9 (c.7725A>C). During the 2-year follow-up period, the patient had no recurrence of any arrhythmia and maintained normal QTc interval.
CONCLUSION This case study highlights the risk of systemic inflammatory activation and antibiotic-induced TdP/LQTS onset. Genetic analysis should be considered to identify individuals at high risk of developing TdP.
Collapse
Affiliation(s)
- Hui Qiu
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Hong-Wei Li
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Shu-Hong Zhang
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Xiao-Ge Zhou
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wei-Ping Li
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
15
|
Lazzerini PE, Cevenini G, Qu YS, Fabris F, El-Sherif N, Acampa M, Cartocci A, Laghi-Pasini F, Capecchi PL, Boutjdir M, Lazaro D. Risk of QTc Interval Prolongation Associated With Circulating Anti-Ro/SSA Antibodies Among US Veterans: An Observational Cohort Study. J Am Heart Assoc 2021; 10:e018735. [PMID: 33533258 PMCID: PMC7955337 DOI: 10.1161/jaha.120.018735] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Anti‐Sjögren's syndrome‐related antigen A‐antibodies (anti‐Ro/SSA‐antibodies) are responsible for a novel form of acquired long‐QT syndrome, owing to autoimmune‐mediated inhibition of cardiac human ether‐a‐go‐go‐related gene‐potassium channels. However, current evidence derives only from basic mechanistic studies and relatively small sample‐size clinical investigations. Hence, the aim of our study is to estimate the risk of QTc prolongation associated with the presence of anti‐Ro/SSA‐antibodies in a large population of unselected subjects. Methods and Results This is a retrospective observational cohort study using the Veterans Affairs Informatics and Computing Infrastructure. Participants were veterans who were tested for anti‐Ro/SSA status and had an ECG. Descriptive statistics and univariate and multivariate logistic regression analyses were performed to identify risk factors for heart rate‐corrected QT interval (QTc) prolongation. The study population consisted of 7339 subjects (61.4±12.2 years), 612 of whom were anti‐Ro/SSA‐positive (8.3%). Subjects who were anti‐Ro/SSA‐positive showed an increased prevalence of QTc prolongation, in the presence of other concomitant risk factors (crude odds ratios [OR], 1.67 [1.26–2.21] for QTc >470/480 ms; 2.32 [1.54–3.49] for QTc >490 ms; 2.77 [1.66–4.60] for QTc >500 ms), independent of a connective tissue disease history. Adjustments for age, sex, electrolytes, cardiovascular risk factors/diseases, and medications gradually attenuated QTc prolongation estimates, particularly when QT‐prolonging drugs were added to the model. Nevertheless, stepwise‐fully adjusted OR for the higher cutoffs remained significantly increased in anti‐Ro/SSA‐positive subjects, particularly for QTc >500 ms (2.27 [1.34–3.87]). Conclusions Anti‐Ro/SSA‐antibody positivity was independently associated with an increased risk of marked QTc prolongation in a large cohort of US veterans. Our data suggest that within the general population individuals who are anti‐Ro/SSA‐positive may represent a subgroup of patients particularly predisposed to ventricular arrhythmias/sudden cardiac death.
Collapse
Affiliation(s)
| | | | - Yongxia Sarah Qu
- VA New York Harbor Healthcare SystemSUNY Downstate Medical Center New York NY.,Department of Cardiology New York Presbyterian Brooklyn Methodist Hospital Brooklyn NY
| | - Frank Fabris
- VA New York Harbor Healthcare SystemSUNY Downstate Medical Center New York NY
| | - Nabil El-Sherif
- VA New York Harbor Healthcare SystemSUNY Downstate Medical Center New York NY
| | | | | | - Franco Laghi-Pasini
- Department of Medical Sciences Surgery and Neurosciences University of Siena Italy
| | | | - Mohamed Boutjdir
- VA New York Harbor Healthcare SystemSUNY Downstate Medical Center New York NY.,NYU School of Medicine New York NY
| | - Deana Lazaro
- VA New York Harbor Healthcare SystemSUNY Downstate Medical Center New York NY
| |
Collapse
|
16
|
Huang Y, Alsabbagh MW. Estimates of population‐based incidence of malignant arrhythmias associated with medication use—a narrative review. Fundam Clin Pharmacol 2020; 34:418-432. [DOI: 10.1111/fcp.12578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Yichang Huang
- School of Pharmacy Faculty of Science University of Waterloo Room 4015, 10A Victoria St. S. Kitchener ON Canada
| | - Mhd. Wasem Alsabbagh
- School of Pharmacy Faculty of Science University of Waterloo Room 3006, 10A Victoria St. S. Kitchener ON Canada
| |
Collapse
|
17
|
Arrhythmogenic foods – A growing medical problem. Trends Cardiovasc Med 2020; 30:310-312. [DOI: 10.1016/j.tcm.2019.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/12/2019] [Accepted: 08/20/2019] [Indexed: 11/20/2022]
|
18
|
Orvos P, Kohajda Z, Szlovák J, Gazdag P, Árpádffy-Lovas T, Tóth D, Geramipour A, Tálosi L, Jost N, Varró A, Virág L. Evaluation of Possible Proarrhythmic Potency: Comparison of the Effect of Dofetilide, Cisapride, Sotalol, Terfenadine, and Verapamil on hERG and Native IKr Currents and on Cardiac Action Potential. Toxicol Sci 2020; 168:365-380. [PMID: 30561737 DOI: 10.1093/toxsci/kfy299] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The proarrhythmic potency of drugs is usually attributed to the IKr current block. During safety pharmacology testing analysis of IKr in cardiomyocytes was replaced by human ether-a-go-go-related gene (hERG) test using automated patch-clamp systems in stable transfected cell lines. Aim of this study was to compare the effect of proarrhythmic compounds on hERG and IKr currents and on cardiac action potential. The hERG current was measured by using both automated and manual patch-clamp methods on HEK293 cells. The native ion currents (IKr, INaL, ICaL) were recorded from rabbit ventricular myocytes by manual patch-clamp technique. Action potentials in rabbit ventricular muscle and undiseased human donor hearts were studied by conventional microelectrode technique. Dofetilide, cisapride, sotalol, terfenadine, and verapamil blocked hERG channels at 37°C with an IC50 of 7 nM, 18 nM, 343 μM, 165 nM, and 214 nM, respectively. Using manual patch-clamp, the IC50 values of sotalol and terfenadine were 78 µM and 31 nM, respectively. The IC50 values calculated from IKr measurements at 37°C were 13 nM, 26 nM, 52 μM, 54 nM, and 268 nM, respectively. Cisapride, dofetilide, and sotalol excessively lengthened, terfenadine, and verapamil did not influence the action potential duration. Terfenadine significantly inhibited INaL and moderately ICaL, verapamil blocked only ICaL. Automated hERG assays may over/underestimate proarrhythmic risk. Manual patch-clamp has substantially higher sensitivity to certain drugs. Action potential studies are also required to analyze complex multichannel effects. Therefore, manual patch-clamp and action potential experiments should be a part of preclinical safety tests.
Collapse
Affiliation(s)
- Péter Orvos
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine.,Department of Ophthalmology, University of Szeged, Szeged H-6720, Hungary
| | - Zsófia Kohajda
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine.,MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged H-6720, Hungary
| | - Jozefina Szlovák
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine
| | - Péter Gazdag
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine
| | | | - Dániel Tóth
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine
| | - Amir Geramipour
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine
| | | | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine.,MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged H-6720, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged H-6720, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine.,MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged H-6720, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged H-6720, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine.,MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged H-6720, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged H-6720, Hungary
| |
Collapse
|
19
|
Zhou X, Qu Y, Passini E, Bueno-Orovio A, Liu Y, Vargas HM, Rodriguez B. Blinded In Silico Drug Trial Reveals the Minimum Set of Ion Channels for Torsades de Pointes Risk Assessment. Front Pharmacol 2020; 10:1643. [PMID: 32082155 PMCID: PMC7003137 DOI: 10.3389/fphar.2019.01643] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Torsades de Pointes (TdP) is a type of ventricular arrhythmia which could be observed as an unwanted drug-induced cardiac side effect, and it is associated with repolarization abnormalities in single cells. The pharmacological evaluations of TdP risk in previous years mainly focused on the hERG channel due to its vital role in the repolarization of cardiomyocytes. However, only considering drug effects on hERG led to false positive predictions since the drug action on other ion channels can also have crucial regulatory effects on repolarization. To address the limitation of only evaluating hERG, the Comprehensive in Vitro Proarrhythmia Assay initiative has proposed to systematically integrate drug effects on multiple ion channels into in silico drug trial to improve TdP risk assessment. It is not clear how many ion channels are sufficient for reliable TdP risk predictions, and whether differences in IC50 and Hill coefficient values from independent sources can lead to divergent in silico prediction outcomes. The rationale of this work is to investigate the above two questions using a computationally efficient population of human ventricular cells optimized to favor repolarization abnormality. Our blinded results based on two independent data sources confirm that simulations with the optimized population of human ventricular cell models enable efficient in silico drug screening, and also provide direct observation and mechanistic analysis of repolarization abnormality. Our results show that 1) the minimum set of ion channels required for reliable TdP risk predictions are Nav1.5 (peak), Cav1.2, and hERG; 2) for drugs with multiple ion channel blockage effects, moderate IC50 variations combined with variable Hill coefficients can affect the accuracy of in silico predictions.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Yusheng Qu
- SPARC, Amgen Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Elisa Passini
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Alfonso Bueno-Orovio
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Yang Liu
- GAU, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Hugo M Vargas
- SPARC, Amgen Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Sun AY. Drug Provocation Testing in Brugada Syndrome: A Test of Uncertain Significance. JACC Clin Electrophysiol 2019; 5:513-515. [PMID: 31000107 DOI: 10.1016/j.jacep.2019.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 03/06/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Albert Y Sun
- Section of Cardiac Electrophysiology, Division of Cardiovascular Diseases, Duke University Medical Center, Durham, North Carolina; Section of Cardiac Electrophysiology, Division of Cardiovascular Diseases, Durham Veterans Affairs Medical Center, Durham, North Carolina.
| |
Collapse
|
21
|
Żołek T, Qile M, Kaźmierczak P, Bloothooft M, van der Heyden MAG, Maciejewska D. Drug-likeness of linear pentamidine analogues and their impact on the hERG K+channel – correlation with structural features. RSC Adv 2019; 9:38355-38371. [PMID: 35540224 PMCID: PMC9082326 DOI: 10.1039/c9ra08404e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/15/2019] [Indexed: 01/08/2023] Open
Abstract
The pentamidines with S atoms or sulfanilide groups in the linker have favorable drug-likeness parameters and low toxicity.
Collapse
Affiliation(s)
- Teresa Żołek
- Department of Organic Chemistry
- Faculty of Pharmacy
- Medical University of Warsaw
- 02-097 Warsaw
- Poland
| | - Muge Qile
- Department of Medical Physiology
- Division Heart & Lungs
- University Medical Center Utrecht
- Utrecht
- The Netherlands
| | - Paweł Kaźmierczak
- Department of Organic Chemistry
- Faculty of Pharmacy
- Medical University of Warsaw
- 02-097 Warsaw
- Poland
| | - Meye Bloothooft
- Department of Medical Physiology
- Division Heart & Lungs
- University Medical Center Utrecht
- Utrecht
- The Netherlands
| | - Marcel A. G. van der Heyden
- Department of Medical Physiology
- Division Heart & Lungs
- University Medical Center Utrecht
- Utrecht
- The Netherlands
| | - Dorota Maciejewska
- Department of Organic Chemistry
- Faculty of Pharmacy
- Medical University of Warsaw
- 02-097 Warsaw
- Poland
| |
Collapse
|
22
|
Lazzerini PE, Capecchi PL, El‐Sherif N, Laghi‐Pasini F, Boutjdir M. Emerging Arrhythmic Risk of Autoimmune and Inflammatory Cardiac Channelopathies. J Am Heart Assoc 2018; 7:e010595. [PMID: 30571503 PMCID: PMC6404431 DOI: 10.1161/jaha.118.010595] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | | | - Nabil El‐Sherif
- Veterans Affairs New York Harbor Healthcare SystemState University of New York Downstate Medical CenterNew YorkNY
| | - Franco Laghi‐Pasini
- Department of Medical Sciences, Surgery and NeurosciencesUniversity of SienaItaly
| | - Mohamed Boutjdir
- Veterans Affairs New York Harbor Healthcare SystemState University of New York Downstate Medical CenterNew YorkNY
- New York University School of MedicineNew YorkNY
| |
Collapse
|
23
|
Large-Scale Simulation of the Phenotypical Variability Induced by Loss-of-Function Long QT Mutations in Human Induced Pluripotent Stem Cell Cardiomyocytes. Int J Mol Sci 2018; 19:ijms19113583. [PMID: 30428582 PMCID: PMC6274824 DOI: 10.3390/ijms19113583] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 01/06/2023] Open
Abstract
Loss-of-function long QT (LQT) mutations inducing LQT1 and LQT2 syndromes have been successfully translated to human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) used as disease-specific models. However, their in vitro investigation mainly relies on experiments using small numbers of cells. This is especially critical when working with cells as heterogeneous as hiPSC-CMs. We aim (i) to investigate in silico the ionic mechanisms underlying LQT1 and LQT2 hiPSC-CM phenotypic variability, and (ii) to enable massive in silico drug tests on mutant hiPSC-CMs. We combined (i) data of control and mutant slow and rapid delayed rectifying K+ currents, IKr and IKs respectively, (ii) a recent in silico hiPSC-CM model, and (iii) the population of models paradigm to generate control and mutant populations for LQT1 and LQT2 cardiomyocytes. Our four populations contain from 1008 to 3584 models. In line with the experimental in vitro data, mutant in silico hiPSC-CMs showed prolonged action potential (AP) duration (LQT1: +14%, LQT2: +39%) and large electrophysiological variability. Finally, the mutant populations were split into normal-like hiPSC-CMs (with action potential duration similar to control) and at risk hiPSC-CMs (with clearly prolonged action potential duration). At risk mutant hiPSC-CMs carried higher expression of L-type Ca2+, lower expression of IKr and increased sensitivity to quinidine as compared to mutant normal-like hiPSC-CMs, resulting in AP abnormalities. In conclusion, we were able to reproduce the two most common LQT syndromes with large-scale simulations, which enable investigating biophysical mechanisms difficult to assess in vitro, e.g., how variations of ion current expressions in a physiological range can impact on AP properties of mutant hiPSC-CMs.
Collapse
|
24
|
Talebi S, Azhir A, Argulian E. Stress cardiomyopathy: Provoked chaotic T-wave lability. Ann Noninvasive Electrocardiol 2018; 23:e12544. [DOI: 10.1111/anec.12544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 03/12/2018] [Indexed: 01/08/2023] Open
Affiliation(s)
- Soheila Talebi
- Cardiology Department; Mount-Sinai St. Luke's Hospital; New York NY USA
| | - Alaleh Azhir
- Department of Biomedical Engineering; Johns Hopkins University; Baltimore MD USA
| | - Edgar Argulian
- Cardiology Department; Mount-Sinai St. Luke's Hospital; New York NY USA
| |
Collapse
|
25
|
Ni H, Morotti S, Grandi E. A Heart for Diversity: Simulating Variability in Cardiac Arrhythmia Research. Front Physiol 2018; 9:958. [PMID: 30079031 PMCID: PMC6062641 DOI: 10.3389/fphys.2018.00958] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/29/2018] [Indexed: 12/31/2022] Open
Abstract
In cardiac electrophysiology, there exist many sources of inter- and intra-personal variability. These include variability in conditions and environment, and genotypic and molecular diversity, including differences in expression and behavior of ion channels and transporters, which lead to phenotypic diversity (e.g., variable integrated responses at the cell, tissue, and organ levels). These variabilities play an important role in progression of heart disease and arrhythmia syndromes and outcomes of therapeutic interventions. Yet, the traditional in silico framework for investigating cardiac arrhythmias is built upon a parameter/property-averaging approach that typically overlooks the physiological diversity. Inspired by work done in genetics and neuroscience, new modeling frameworks of cardiac electrophysiology have been recently developed that take advantage of modern computational capabilities and approaches, and account for the variance in the biological data they are intended to illuminate. In this review, we outline the recent advances in statistical and computational techniques that take into account physiological variability, and move beyond the traditional cardiac model-building scheme that involves averaging over samples from many individuals in the construction of a highly tuned composite model. We discuss how these advanced methods have harnessed the power of big (simulated) data to study the mechanisms of cardiac arrhythmias, with a special emphasis on atrial fibrillation, and improve the assessment of proarrhythmic risk and drug response. The challenges of using in silico approaches with variability are also addressed and future directions are proposed.
Collapse
Affiliation(s)
| | | | - Eleonora Grandi
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
26
|
Cheng G, Wu J, Han W, Sun C. F463L increases the potential of dofetilide on human ether-a-go-go-related gene (hERG) channels. Microsc Res Tech 2018; 81:663-668. [PMID: 29573040 DOI: 10.1002/jemt.23021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 02/02/2018] [Accepted: 03/02/2018] [Indexed: 11/11/2022]
Abstract
Mutations in genes related to long QT syndrome (LQTS) is recognized as an independent risk of drug-induced LQTS. We previously screened a mutation F463L in a Chinese patient with LQT2, syncope, and epilepsy. Here, we planned to illustrate how F463L influences the action of dofetilide on hERG channels. F463L-hERG plasmids were transfected into the stable Human Embryonic Kidney 293 (HEK293) cells expressing WT-hERG to generate heterozygous mutant (WT + F463L-hERG). Whole-cell patch clamp and laser confocal scanning microscopy were used to evaluate electrophysiological consequences and the membrane distribution of hERG protein. In comparison of WT-hERG channels exposed to dofetilide, heterozygous F463L-hERG channels showed a reduction in the density of tail currents when exposed amidarone. F463L-hERG also altered the action of dofetilide on the gating properties of hERG channels. Images of dofetilide-treated cells expressing heterozygous F463L showed a severe retention and reduction of protein expression on the membrane compared to WT. In conclusion, dofetilide displays a powerful inhibitory effect on the currents from cells expressing heterozygous F463L, thus showing an additive suppression of currents by F463L with dofetilide.
Collapse
Affiliation(s)
- Gong Cheng
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R.,Cardiovascular Medicine, Shaanxi Provincial People's Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710068, China
| | - Jine Wu
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R
| | - Wenqi Han
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R
| | - Chaofeng Sun
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R
| |
Collapse
|
27
|
Abstract
This study examines the interaction between hERG and Kv4.3. The functional interaction between hERG and Kv4.3, expressed in a heterologous cell line, was studied using patch clamp techniques, western blot, immunofluorescence, and co-immunoprecipitation. Co-expression of Kv4.3 with hERG increased hERG current density (tail current after a step to +10 mV: 26 ± 3 versus 56 ± 7 pA/pF, p < 0.01). Kv4.3 co-expression also increased the protein expression and promoted the membrane localization of hERG. Western blot showed Kv4.3 increased hERG expression by Hsp70. hERG and Kv4.3 co-localized and co-immunoprecipitated in cultured 293 T cells, indicating physical interactions between hERG and Kv4.3 proteins in vitro. In addition, Hsp70 interacted with hERG and Kv4.3 respectively, and formed complexes with hERG and Kv4.3. The α subunit of Ito Kv4.3 can interact with and modify the localization of the α subunit of IKr hERG, thus providing potentially novel insights into the molecular mechanism of the malignant ventricular arrhythmia in heart failure.
Collapse
|
28
|
Trenor B, Cardona K, Saiz J, Noble D, Giles W. Cardiac action potential repolarization revisited: early repolarization shows all-or-none behaviour. J Physiol 2017; 595:6599-6612. [PMID: 28815597 PMCID: PMC5663823 DOI: 10.1113/jp273651] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/09/2017] [Indexed: 12/15/2022] Open
Abstract
In healthy mammalian hearts the action potential (AP) waveform initiates and modulates each contraction, or heartbeat. As a result, AP height and duration are key physiological variables. In addition, rate-dependent changes in ventricular AP duration (APD), and variations in APD at a fixed heart rate are both reliable biomarkers of electrophysiological stability. Present guidelines for the likelihood that candidate drugs will increase arrhythmias rely on small changes in APD and Q-T intervals as criteria for safety pharmacology decisions. However, both of these measurements correspond to the final repolarization of the AP. Emerging clinical evidence draws attention to the early repolarization phase of the action potential (and the J-wave of the ECG) as an additional important biomarker for arrhythmogenesis. Here we provide a mechanistic background to this early repolarization syndrome by summarizing the evidence that both the initial depolarization and repolarization phases of the cardiac action potential can exhibit distinct time- and voltage-dependent thresholds, and also demonstrating that both can show regenerative all-or-none behaviour. An important consequence of this is that not all of the dynamics of action potential repolarization in human ventricle can be captured by data from single myocytes when these results are expressed as 'repolarization reserve'. For example, the complex pattern of cell-to-cell current flow that is responsible for AP conduction (propagation) within the mammalian myocardium can change APD and the Q-T interval of the electrocardiogram alter APD stability, and modulate responsiveness to pharmacological agents (such as Class III anti-arrhythmic drugs).
Collapse
Affiliation(s)
- Beatriz Trenor
- Centro de Investigación e BioingenieríaUniversitat Politècnica de ValènciaValenciaSpain
| | - Karen Cardona
- Centro de Investigación e BioingenieríaUniversitat Politècnica de ValènciaValenciaSpain
| | - Javier Saiz
- Centro de Investigación e BioingenieríaUniversitat Politècnica de ValènciaValenciaSpain
| | - Denis Noble
- University Laboratory of PhysiologyUniversity of OxfordOxfordOX1 3PTUK
| | - Wayne Giles
- Faculties of Kinesiology and MedicineUniversity of CalgaryCalgaryAlbertaCanadaT2N 1N4
| |
Collapse
|
29
|
Bhuiyan TA, Graff C, Kanters JK, Melgaard J, Toft E, Kääb S, Struijk JJ. A History of Drug‐Induced Torsades de Pointes Is Associated With T‐wave Morphological Abnormalities. Clin Pharmacol Ther 2017; 103:1100-1106. [DOI: 10.1002/cpt.886] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/07/2017] [Accepted: 09/14/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Tanveer A. Bhuiyan
- Department of Health Science and TechnologyAalborg UniversityAalborg Denmark
| | - Claus Graff
- Department of Health Science and TechnologyAalborg UniversityAalborg Denmark
| | - Jørgen K. Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical SciencesUniversity of CopenhagenCopenhagen Denmark
| | - Jacob Melgaard
- Department of Health Science and TechnologyAalborg UniversityAalborg Denmark
| | - Egon Toft
- College of Medicine, Qatar UniversityDoha Qatar
| | - Stefan Kääb
- Medizinische Klinik und Poliklinik I, University Hospital Munich, Ludvig Maximilians University, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site: Munich Heart AllianceMunich Germany
| | - Johannes J. Struijk
- Department of Health Science and TechnologyAalborg UniversityAalborg Denmark
| |
Collapse
|
30
|
Papp R, Bett GCL, Lis A, Rasmusson RL, Baczkó I, Varró A, Salama G. Genomic upregulation of cardiac Cav1.2α and NCX1 by estrogen in women. Biol Sex Differ 2017; 8:26. [PMID: 28807015 PMCID: PMC5557418 DOI: 10.1186/s13293-017-0148-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/04/2017] [Indexed: 01/08/2023] Open
Abstract
Background Women have a higher risk of lethal arrhythmias than men in long QT syndrome type 2 (LQTS2), but the mechanisms remain uncertain due to the limited availability of healthy control human tissue. We have previously reported that in female rabbits, estrogen increases arrhythmia risk in drug-induced LQTS2 by upregulating L-type Ca2+ (ICa,L) and sodium-calcium exchange (INCX) currents at the base of the epicardium by a genomic mechanism. This study investigates if the effects of estrogen on rabbit ICa,L and INCX apply to human hearts. Methods Postmortem human left ventricular tissue samples were probed with selective antibodies for regional heterogeneities of ion channel protein expression and compared to rabbit myocardium. Functionally, ICa,L and INCX were measured from female and male cardiomyocytes derived from human induced pluripotent stem cells (iPS-CMs) with the voltage-clamp technique from control and estrogen-treated iPS-CMs. Results In women (n = 12), Cav1.2α (primary subunit of the L-type calcium channel protein 1) and NCX1 (sodium-calcium exchange protein) levels were higher at the base than apex of the epicardium (40 ± 14 and 81 ± 30%, respectively, P < 0.05), but not in men (n = 6) or postmenopausal women (n = 6). Similarly, in cardiomyocytes derived from female human iPS-CMs, estrogen (1 nM, 1–2 days) increased ICa,L (31%, P < 0.05) and INCX (7.5-fold, − 90 mV, P < 0.01) and their mRNA levels (P < 0.05). Moreover, in male human iPS-CMs, estrogen failed to alter ICa,L and INCX. Conclusions The results show that estrogen upregulates cardiac ICa,L and INCX in women through genomic mechanisms that account for sex differences in Ca2+ handling and spatial heterogeneities of repolarization due to base-apex heterogeneities of Cav1.2α and NCX1. By analogy with rabbit studies, these effects account for human sex-difference in arrhythmia risk.
Collapse
Affiliation(s)
- Rita Papp
- Department of Bioengineering and the Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Current Address: Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Glenna C L Bett
- Center for Cellular and Systems Electrophysiology, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA.,Department of Physiology and Biophysics, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA.,Obstetrics-Gynecology, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
| | - Agnieszka Lis
- Center for Cellular and Systems Electrophysiology, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA.,Department of Physiology and Biophysics, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
| | - Randall L Rasmusson
- Center for Cellular and Systems Electrophysiology, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA.,Department of Physiology and Biophysics, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary.,MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged, Hungary
| | - Guy Salama
- Department of Bioengineering and the Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA. .,Department of Bioengineering and the Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, 3550 Terrace Street, S628 Scaife Hall, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
31
|
Salem JE, Germain M, Hulot JS, Voiriot P, Lebourgeois B, Waldura J, Tregouet DA, Charbit B, Funck-Brentano C. GENomE wide analysis of sotalol-induced IKr inhibition during ventricular REPOLarization, "GENEREPOL study": Lack of common variants with large effect sizes. PLoS One 2017; 12:e0181875. [PMID: 28800628 PMCID: PMC5553738 DOI: 10.1371/journal.pone.0181875] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/07/2017] [Indexed: 12/19/2022] Open
Abstract
Many drugs used for non-cardiovascular and cardiovascular purposes, such as sotalol, have the side effect of prolonging cardiac repolarization, which can trigger life-threatening cardiac arrhythmias by inhibiting the potassium-channel IKr (KCNH2). On the electrocardiogram (ECG), IKr inhibition induces an increase in QTc and Tpeak-Tend (TpTe) interval and a decrease of T wave maximal amplitude (TAmp). These changes vary markedly between subjects, suggesting the existence of predisposing genetic factors. 990 healthy individuals, prospectively challenged with an oral 80mg sotalol dose, were monitored for changes in ventricular repolarization on ECG between baseline and 3 hours post dosing. QTc and TpTe increased by 5.5±3.5% and 15±19.6%, respectively, and TAmp decreased by 13.2±15.5%. A principal-component analysis derived from the latter ECG changes was performed. A random subsample of 489 individuals were subjected to a genome-wide-association analysis where 8,306,856 imputed single nucleotide polymorphisms (SNPs) were tested for association with QTc, TpTe and TAmp modulations, as well their derived principal-components, to search for common genetic variants associated with sotalol-induced IKr inhibition. None of the studied SNPs reached the statistical threshold for genome-wide significance. This study supports the lack of common variants with larger effect sizes than one would expect based on previous ECG genome-wide-association studies. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov NCT00773201.
Collapse
Affiliation(s)
- Joe-Elie Salem
- Sorbonne-Universités, UPMC Univ Paris 06, INSERM, UMRS-1166, Institute of Cardio metabolism and Nutrition (ICAN), Paris, France
- AP-HP, CIC-1421-Paris-Est, Pitié-Salpêtrière Hospital, Paris, France
| | - Marine Germain
- AP-HP, CIC-1421-Paris-Est, Pitié-Salpêtrière Hospital, Paris, France
| | - Jean-Sébastien Hulot
- Sorbonne-Universités, UPMC Univ Paris 06, INSERM, UMRS-1166, Institute of Cardio metabolism and Nutrition (ICAN), Paris, France
- AP-HP, CIC-1421-Paris-Est, Pitié-Salpêtrière Hospital, Paris, France
| | | | - Bruno Lebourgeois
- Sorbonne-Universités, UPMC Univ Paris 06, INSERM, UMRS-1166, Institute of Cardio metabolism and Nutrition (ICAN), Paris, France
| | | | - David-Alexandre Tregouet
- Sorbonne-Universités, UPMC Univ Paris 06, INSERM, UMRS-1166, Institute of Cardio metabolism and Nutrition (ICAN), Paris, France
| | - Beny Charbit
- AP-HP, CIC-1421-Paris-Est, Pitié-Salpêtrière Hospital, Paris, France
| | - Christian Funck-Brentano
- Sorbonne-Universités, UPMC Univ Paris 06, INSERM, UMRS-1166, Institute of Cardio metabolism and Nutrition (ICAN), Paris, France
- AP-HP, CIC-1421-Paris-Est, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
32
|
Winbo A, Stattin EL, Westin IM, Norberg A, Persson J, Jensen SM, Rydberg A. Sex is a moderator of the association between NOS1AP sequence variants and QTc in two long QT syndrome founder populations: a pedigree-based measured genotype association analysis. BMC MEDICAL GENETICS 2017; 18:74. [PMID: 28720088 PMCID: PMC5516337 DOI: 10.1186/s12881-017-0435-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 07/06/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Sequence variants in the NOS1AP gene have repeatedly been reported to influence QTc, albeit with moderate effect sizes. In the long QT syndrome (LQTS), this may contribute to the substantial QTc variance seen among carriers of identical pathogenic sequence variants. Here we assess three non-coding NOS1AP sequence variants, chosen for their previously reported strong association with QTc in normal and LQTS populations, for association with QTc in two Swedish LQT1 founder populations. METHODS This study included 312 individuals (58% females) from two LQT1 founder populations, whereof 227 genotype positive segregating either Y111C (n = 148) or R518* (n = 79) pathogenic sequence variants in the KCNQ1 gene, and 85 genotype negatives. All were genotyped for NOS1AP sequence variants rs12143842, rs16847548 and rs4657139, and tested for association with QTc length (effect size presented as mean difference between derived and wildtype, in ms), using a pedigree-based measured genotype association analysis. Mean QTc was obtained by repeated manual measurement (preferably in lead II) by one observer using coded 50 mm/s standard 12-lead ECGs. RESULTS A substantial variance in mean QTc was seen in genotype positives 476 ± 36 ms (Y111C 483 ± 34 ms; R518* 462 ± 34 ms) and genotype negatives 433 ± 24 ms. Female sex was significantly associated with QTc prolongation in all genotype groups (p < 0.001). In a multivariable analysis including the entire study population and adjusted for KCNQ1 genotype, sex and age, NOS1AP sequence variants rs12143842 and rs16847548 (but not rs4657139) were significantly associated with QT prolongation, +18 ms (p = 0.0007) and +17 ms (p = 0.006), respectively. Significant sex-interactions were detected for both sequent variants (interaction term r = 0.892, p < 0.001 and r = 0.944, p < 0.001, respectively). Notably, across the genotype groups, when stratified by sex neither rs12143842 nor rs16847548 were significantly associated with QTc in females (both p = 0.16) while in males, a prolongation of +19 ms and +8 ms (p = 0.002 and p = 0.02) was seen in multivariable analysis, explaining up to 23% of QTc variance in all males. CONCLUSIONS Sex was identified as a moderator of the association between NOS1AP sequence variants and QTc in two LQT1 founder populations. This finding may contribute to QTc sex differences and affect the usefulness of NOS1AP as a marker for clinical risk stratification in LQTS.
Collapse
Affiliation(s)
- Annika Winbo
- Department of Clinical Sciences, Pediatrics, Umeå University, 90187, Umeå, Sweden. .,Department of Physiology, University of Auckland, Auckland, New Zealand.
| | - Eva-Lena Stattin
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ida Maria Westin
- Department of Medical Biosciences, Medical and Clinical Genetics, Umeå University, Umeå, 90185, Sweden
| | - Anna Norberg
- Department of Medical Biosciences, Medical and Clinical Genetics, Umeå University, Umeå, 90185, Sweden
| | - Johan Persson
- Department of Clinical Sciences, Pediatrics, Umeå University, 90187, Umeå, Sweden
| | - Steen M Jensen
- Department of Public Health and Clinical Medicine, Heart Centre, Umeå University, Umeå, 90185, Sweden
| | - Annika Rydberg
- Department of Clinical Sciences, Pediatrics, Umeå University, 90187, Umeå, Sweden
| |
Collapse
|
33
|
Liang P, Sallam K, Wu H, Li Y, Itzhaki I, Garg P, Zhang Y, Vermglinchan V, Lan F, Gu M, Gong T, Zhuge Y, He C, Ebert AD, Sanchez-Freire V, Churko J, Hu S, Sharma A, Lam CK, Scheinman MM, Bers DM, Wu JC. Patient-Specific and Genome-Edited Induced Pluripotent Stem Cell-Derived Cardiomyocytes Elucidate Single-Cell Phenotype of Brugada Syndrome. J Am Coll Cardiol 2017; 68:2086-2096. [PMID: 27810048 DOI: 10.1016/j.jacc.2016.07.779] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 06/29/2016] [Accepted: 07/27/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Brugada syndrome (BrS), a disorder associated with characteristic electrocardiogram precordial ST-segment elevation, predisposes afflicted patients to ventricular fibrillation and sudden cardiac death. Despite marked achievements in outlining the organ level pathophysiology of the disorder, the understanding of human cellular phenotype has lagged due to a lack of adequate human cellular models of the disorder. OBJECTIVES The objective of this study was to examine single cell mechanism of Brugada syndrome using induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). METHODS This study recruited 2 patients with type 1 BrS carrying 2 different sodium voltage-gated channel alpha subunit 5 variants as well as 2 healthy control subjects. We generated iPSCs from their skin fibroblasts by using integration-free Sendai virus. We used directed differentiation to create purified populations of iPSC-CMs. RESULTS BrS iPSC-CMs showed reductions in inward sodium current density and reduced maximal upstroke velocity of action potential compared with healthy control iPSC-CMs. Furthermore, BrS iPSC-CMs demonstrated increased burden of triggered activity, abnormal calcium (Ca2+) transients, and beating interval variation. Correction of the causative variant by genome editing was performed, and resultant iPSC-CMs showed resolution of triggered activity and abnormal Ca2+ transients. Gene expression profiling of iPSC-CMs showed clustering of BrS compared with control subjects. Furthermore, BrS iPSC-CM gene expression correlated with gene expression from BrS human cardiac tissue gene expression. CONCLUSIONS Patient-specific iPSC-CMs were able to recapitulate single-cell phenotype features of BrS, including blunted inward sodium current, increased triggered activity, and abnormal Ca2+ handling. This novel human cellular model creates future opportunities to further elucidate the cellular disease mechanism and identify novel therapeutic targets.
Collapse
Affiliation(s)
- Ping Liang
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California; The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, China.
| | - Karim Sallam
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Haodi Wu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Yingxin Li
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Ilanit Itzhaki
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Priyanka Garg
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Ying Zhang
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Vittavat Vermglinchan
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Feng Lan
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Mingxia Gu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Tingyu Gong
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California; The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Zhuge
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Chunjiang He
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Antje D Ebert
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Veronica Sanchez-Freire
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Jared Churko
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Shijun Hu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Arun Sharma
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Chi Keung Lam
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Melvin M Scheinman
- Department of Medicine, Division of Cardiology, University of California, San Francisco, California
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, California
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiovascular Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
34
|
Lorberbaum T, Sampson KJ, Chang JB, Iyer V, Woosley RL, Kass RS, Tatonetti NP. Coupling Data Mining and Laboratory Experiments to Discover Drug Interactions Causing QT Prolongation. J Am Coll Cardiol 2017; 68:1756-1764. [PMID: 27737742 DOI: 10.1016/j.jacc.2016.07.761] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 07/11/2016] [Accepted: 07/12/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND QT interval-prolonging drug-drug interactions (QT-DDIs) may increase the risk of life-threatening arrhythmia. Despite guidelines for testing from regulatory agencies, these interactions are usually discovered after drugs are marketed and may go undiscovered for years. OBJECTIVES Using a combination of adverse event reports, electronic health records (EHR), and laboratory experiments, the goal of this study was to develop a data-driven pipeline for discovering QT-DDIs. METHODS 1.8 million adverse event reports were mined for signals indicating a QT-DDI. Using 1.6 million electrocardiogram results from 380,000 patients in our institutional EHR, these putative interactions were either refuted or corroborated. In the laboratory, we used patch-clamp electrophysiology to measure the human ether-à-go-go-related gene (hERG) channel block (the primary mechanism by which drugs prolong the QT interval) to evaluate our top candidate. RESULTS Both direct and indirect signals in the adverse event reports provided evidence that the combination of ceftriaxone (a cephalosporin antibiotic) and lansoprazole (a proton-pump inhibitor) will prolong the QT interval. In the EHR, we found that patients taking both ceftriaxone and lansoprazole had significantly longer QTc intervals (up to 12 ms in white men) and were 1.4 times more likely to have a QTc interval above 500 ms. In the laboratory, we found that, in combination and at clinically relevant concentrations, these drugs blocked the hERG channel. As a negative control, we evaluated the combination of lansoprazole and cefuroxime (another cephalosporin), which lacked evidence of an interaction in the adverse event reports. We found no significant effect of this pair in either the EHR or in the electrophysiology experiments. Class effect analyses suggested this interaction was specific to lansoprazole combined with ceftriaxone but not with other cephalosporins. CONCLUSIONS Coupling data mining and laboratory experiments is an efficient method for identifying QT-DDIs. Combination therapy of ceftriaxone and lansoprazole is associated with increased risk of acquired long QT syndrome.
Collapse
Affiliation(s)
- Tal Lorberbaum
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York; Department of Biomedical Informatics, Columbia University, New York, New York
| | - Kevin J Sampson
- Department of Pharmacology, Columbia University, New York, New York
| | - Jeremy B Chang
- Department of Biomedical Informatics, Columbia University, New York, New York
| | - Vivek Iyer
- Department of Cardiology, Columbia University, New York, New York
| | | | - Robert S Kass
- Department of Pharmacology, Columbia University, New York, New York
| | | |
Collapse
|
35
|
Vandersickel N, Van Nieuwenhuyse E, Seemann G, Panfilov AV. Spatial Patterns of Excitation at Tissue and Whole Organ Level Due to Early Afterdepolarizations. Front Physiol 2017; 8:404. [PMID: 28690545 PMCID: PMC5479889 DOI: 10.3389/fphys.2017.00404] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 05/29/2017] [Indexed: 01/08/2023] Open
Abstract
Early after depolarizations (EAD) occur in many pathological conditions, such as congenital or acquired channelopathies, drug induced arrhythmias, and several other situations that are associated with increased arrhythmogenicity. In this paper we present an overview of the relevant computational studies on spatial EAD dynamics in 1D, 2D, and in 3D anatomical models and discuss the relation of EADs to cardiac arrhythmias. We also discuss unsolved problems and highlight new lines of research in this area.
Collapse
Affiliation(s)
| | | | - Gunnar Seemann
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg · Bad Krozingen, Medical Center, University of FreiburgFreiburg, Germany.,Faculty of Medicine, University of FreiburgFreiburg, Germany
| | | |
Collapse
|
36
|
Bossu A, Varkevisser R, Beekman HDM, Houtman MJC, van der Heyden MAG, Vos MA. Short-term Variability of Repolarization Is Superior to Other Repolarization Parameters in the Evaluation of Diverse Antiarrhythmic Interventions in the Chronic Atrioventricular Block Dog. J Cardiovasc Pharmacol 2017; 69:398-407. [PMID: 28574954 PMCID: PMC5464753 DOI: 10.1097/fjc.0000000000000488] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Short-term variability (STV), to quantify beat-to-beat variability of repolarization, is a surrogate parameter that reliably identifies proarrhythmic risk in preclinical models. Examples include not only the use in the chronic atrioventricular block (CAVB) dog model whereby it was developed but also in vulnerable patients with heart failure or drug-induced long QT syndrome. In the CAVB dog model, STV can specifically distinguish between safe and unsafe drugs in proarrhythmic screening. Conversely, this dog model also offers the possibility to evaluate antiarrhythmic strategies in a setting of Torsades de Pointes (TdP) induction with a standard IKr inhibitor. The different antiarrhythmic interventions studied in suppression and prevention of drug-induced TdP in vivo in the CAVB dog model and in vitro in canine ventricular cardiomyocytes are described in this overview. We provide evidence that STV predicts the magnitude of antiarrhythmic effect against TdP better than other repolarization parameters in both suppression and prevention conditions. Moreover, suppression and prevention experiments revealed the same level of antiarrhythmic efficacy, whereas cellular experiments seem more sensitive in comparison with drug testing in vivo. Together, these observations suggest that STV could be used as a consistent indicator to rank efficacy of antiarrhythmic interventions in a number of conditions.
Collapse
Affiliation(s)
- Alexandre Bossu
- Department of Medical Physiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | | | | | | | | |
Collapse
|
37
|
Schlit AF, Delaunois A, Colomar A, Claudio B, Cariolato L, Boev R, Valentin JP, Peters C, Sloan VS, Bentz JWG. Risk of QT prolongation and torsade de pointes associated with exposure to hydroxyzine: re-evaluation of an established drug. Pharmacol Res Perspect 2017; 5:e00309. [PMID: 28480041 PMCID: PMC5415947 DOI: 10.1002/prp2.309] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 03/09/2017] [Indexed: 01/10/2023] Open
Abstract
Several noncardiac drugs have been linked to cardiac safety concerns, highlighting the importance of post‐marketing surveillance and continued evaluation of the benefit‐risk of long‐established drugs. Here, we examine the risk of QT prolongation and/or torsade de pointes (TdP) associated with the use of hydroxyzine, a first generation sedating antihistamine. We have used a combined methodological approach to re‐evaluate the cardiac safety profile of hydroxyzine, including: (1) a full review of the sponsor pharmacovigilance safety database to examine real‐world data on the risk of QT prolongation and/or TdP associated with hydroxyzine use and (2) nonclinical electrophysiological studies to examine concentration‐dependent effects of hydroxyzine on a range of human cardiac ion channels. Based on a review of pharmacovigilance data between 14th December 1955 and 1st August 2016, we identified 59 reports of QT prolongation and/or TdP potentially linked to hydroxyzine use. Aside from intentional overdose, all cases involved underlying medical conditions or concomitant medications that constituted at least 1 additional risk factor for such events. The combination of cardiovascular disorders plus concomitant treatment of drugs known to induce arrhythmia was identified as the greatest combined risk factor. Parallel patch‐clamp studies demonstrated hydroxyzine concentration‐dependent inhibition of several human cardiac ion channels, including the ether‐a‐go‐go‐related gene (hERG) potassium ion channels. Results from this analysis support the listing of hydroxyzine as a drug with “conditional risk of TdP” and are in line with recommendations to limit hydroxyzine use in patients with known underlying risk factors for QT prolongation and/or TdP.
Collapse
Affiliation(s)
| | | | - Aurore Colomar
- UCB Pharma Brussels Belgium.,Present address: Aurore Colomar, Université de Mons Mons Belgium
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The QT interval on surface electrocardiograms provides a model of a multicomponent integrated readout of many biological systems, including ion channels, modulatory subunits, signaling systems that modulate their activity, and mechanisms that regulate the expression of their responsible genes. The problem of drug exposure causing exaggerated QT interval prolongation and torsades de pointes highlights the multicomponent nature of cardiac repolarization and the way in which simple perturbations can yield exaggerated responses. Future directions will involve cellular approaches coupled to evolving technologies that can interrogate multicellular systems and provide a sophisticated view of mechanisms in this previously idiosyncratic drug reaction.
Collapse
Affiliation(s)
- Dan M Roden
- Oates Institute for Experimental Therapeutics, Vanderbilt University School of Medicine, 1285 MRB IV, Nashville, TN 37232-0575, USA.
| |
Collapse
|
39
|
Stillitano F, Hansen J, Kong CW, Karakikes I, Funck-Brentano C, Geng L, Scott S, Reynier S, Wu M, Valogne Y, Desseaux C, Salem JE, Jeziorowska D, Zahr N, Li R, Iyengar R, Hajjar RJ, Hulot JS. Modeling susceptibility to drug-induced long QT with a panel of subject-specific induced pluripotent stem cells. eLife 2017; 6:e19406. [PMID: 28134617 PMCID: PMC5279943 DOI: 10.7554/elife.19406] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 12/08/2016] [Indexed: 12/18/2022] Open
Abstract
A large number of drugs can induce prolongation of cardiac repolarization and life-threatening cardiac arrhythmias. The prediction of this side effect is however challenging as it usually develops in some genetically predisposed individuals with normal cardiac repolarization at baseline. Here, we describe a platform based on a genetically diverse panel of induced pluripotent stem cells (iPSCs) that reproduces susceptibility to develop a cardiotoxic drug response. We generated iPSC-derived cardiomyocytes from patients presenting in vivo with extremely low or high changes in cardiac repolarization in response to a pharmacological challenge with sotalol. In vitro, the responses to sotalol were highly variable but strongly correlated to the inter-individual differences observed in vivo. Transcriptomic profiling identified dysregulation of genes (DLG2, KCNE4, PTRF, HTR2C, CAMKV) involved in downstream regulation of cardiac repolarization machinery as underlying high sensitivity to sotalol. Our findings offer novel insights for the development of iPSC-based screening assays for testing individual drug reactions.
Collapse
Affiliation(s)
- Francesca Stillitano
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Jens Hansen
- Department of Pharmacology and Systems Therapeutics, Systems Biology Center, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Chi-Wing Kong
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Ioannis Karakikes
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Christian Funck-Brentano
- Sorbonne Universités, UPMC Univ Paris 06, AP-HP, INSERM, CIC-1421, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Lin Geng
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Stuart Scott
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, United States
| | | | - Ma Wu
- Cellectis Stem Cells, Paris, France
| | | | | | - Joe-Elie Salem
- Sorbonne Universités, UPMC Univ Paris 06, AP-HP, INSERM, CIC-1421, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Dorota Jeziorowska
- Sorbonne Universités, UPMC Univ Paris 06, AP-HP, INSERM, CIC-1421, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Noël Zahr
- Sorbonne Universités, UPMC Univ Paris 06, AP-HP, INSERM, CIC-1421, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Ronald Li
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, United States
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Stockholm, Sweden
- Dr. Li Dak-Sum Centre, The University of Hong Kong – Karolinska Institutet Collaboration in Regenerative Medicine, Pokfulam, Hong Kong
| | - Ravi Iyengar
- Department of Pharmacology and Systems Therapeutics, Systems Biology Center, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Roger J Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Jean-Sébastien Hulot
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, United States
- Sorbonne Universités, UPMC Univ Paris 06, AP-HP, INSERM, CIC-1421, Institute of Cardiometabolism and Nutrition, Paris, France
| |
Collapse
|
40
|
Tse G, Wong ST, Tse V, Lee YT, Lin HY, Yeo JM. Cardiac dynamics: Alternans and arrhythmogenesis. J Arrhythm 2016; 32:411-417. [PMID: 27761166 PMCID: PMC5063258 DOI: 10.1016/j.joa.2016.02.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 01/28/2016] [Accepted: 02/22/2016] [Indexed: 12/23/2022] Open
Abstract
Pre-existing heterogeneities present in cardiac tissue are essential for maintaining the normal electrical and mechanical functions of the heart. Exacerbation of such heterogeneities or the emergence of dynamic factors can produce repolarization alternans, which are beat-to-beat alternations in the action potential time course. Traditionally, this was explained by restitution, but additional factors, such as cardiac memory, calcium handling dynamics, refractory period restitution, and mechano-electric feedback, are increasingly recognized as the underlying causes. The aim of this article is to review the mechanisms that generate cardiac repolarization alternans and convert spatially concordant alternans to the more arrhythmogenic spatially discordant alternans. This is followed by a discussion on how alternans generate arrhythmias in a number of clinical scenarios, and concluded by an outline of future therapeutic targets for anti-arrhythmic therapy.
Collapse
Affiliation(s)
- Gary Tse
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Sheung Ting Wong
- Faculty of Medicine, Imperial College London, SW7 2AZ London, UK
| | - Vivian Tse
- Department of Physiology, McGill University, Canada
| | - Yee Ting Lee
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Hiu Yu Lin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Jie Ming Yeo
- Faculty of Medicine, Imperial College London, SW7 2AZ London, UK
| |
Collapse
|
41
|
Passini E, Mincholé A, Coppini R, Cerbai E, Rodriguez B, Severi S, Bueno-Orovio A. Mechanisms of pro-arrhythmic abnormalities in ventricular repolarisation and anti-arrhythmic therapies in human hypertrophic cardiomyopathy. J Mol Cell Cardiol 2016; 96:72-81. [PMID: 26385634 PMCID: PMC4915817 DOI: 10.1016/j.yjmcc.2015.09.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 08/05/2015] [Accepted: 09/11/2015] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Hypertrophic cardiomyopathy (HCM) is a cause of sudden arrhythmic death, but the understanding of its pro-arrhythmic mechanisms and an effective pharmacological treatment are lacking. HCM electrophysiological remodelling includes both increased inward and reduced outward currents, but their role in promoting repolarisation abnormalities remains unknown. The goal of this study is to identify key ionic mechanisms driving repolarisation abnormalities in human HCM, and to evaluate anti-arrhythmic effects of single and multichannel inward current blocks. METHODS Experimental ionic current, action potential (AP) and Ca(2+)-transient (CaT) recordings were used to construct populations of human non-diseased and HCM AP models (n=9118), accounting for inter-subject variability. Simulations were conducted for several degrees of selective and combined inward current block. RESULTS Simulated HCM cardiomyocytes exhibited prolonged AP and CaT, diastolic Ca(2+) overload and decreased CaT amplitude, in agreement with experiments. Repolarisation abnormalities in HCM models were consistently driven by L-type Ca(2+) current (ICaL) re-activation, and ICaL block was the most effective intervention to normalise repolarisation and diastolic Ca(2+), but compromised CaT amplitude. Late Na(+) current (INaL) block partially abolished repolarisation abnormalities, with small impact on CaT. Na(+)/Ca(2+) exchanger (INCX) block effectively restored repolarisation and CaT amplitude, but increased Ca(2+) overload. Multichannel block increased efficacy in normalising repolarisation, AP biomarkers and CaT amplitude compared to selective block. CONCLUSIONS Experimentally-calibrated populations of human AP models identify ICaL re-activation as the key mechanism for repolarisation abnormalities in HCM, and combined INCX, INaL and ICaL block as effective anti-arrhythmic therapies also able to partially reverse the HCM electrophysiological phenotype.
Collapse
Affiliation(s)
- Elisa Passini
- Department of Computer Science, University of Oxford, Oxford OX13QD, United Kingdom; Department of Electrical, Electronic and Information Engineering, University of Bologna, Cesena 47521, Italy
| | - Ana Mincholé
- Department of Computer Science, University of Oxford, Oxford OX13QD, United Kingdom
| | - Raffaele Coppini
- Department NEUROFARBA, University of Florence, Florence 50139, Italy
| | - Elisabetta Cerbai
- Department NEUROFARBA, University of Florence, Florence 50139, Italy
| | - Blanca Rodriguez
- Department of Computer Science, University of Oxford, Oxford OX13QD, United Kingdom
| | - Stefano Severi
- Department of Electrical, Electronic and Information Engineering, University of Bologna, Cesena 47521, Italy
| | - Alfonso Bueno-Orovio
- Department of Computer Science, University of Oxford, Oxford OX13QD, United Kingdom.
| |
Collapse
|
42
|
Szepesváry E, Kaski JP. Genetic testing for inheritable cardiac channelopathies. Br J Hosp Med (Lond) 2016; 77:294-302. [DOI: 10.12968/hmed.2016.77.5.294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cardiac channelopathies are linked to an increased risk of ventricular arrhythmia and sudden death. This article reviews the clinical characteristics and genetic basis of common cardiac ion-channel diseases, highlights some genotype–phenotype correlations, and summarizes genetic testing for inheritable cardiac channelopathies.
Collapse
Affiliation(s)
- Eszter Szepesváry
- Clinical Fellow in the Inherited Cardiovascular Diseases Unit, Great Ormond Street Hospital, London
| | - Juan Pablo Kaski
- Consultant Paediatric Cardiologist in the Inherited Cardiovascular Diseases Unit, Great Ormond Street Hospital, London WC1N 3JH
| |
Collapse
|
43
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Cardiac-specific miRNA in cardiogenesis, heart function, and cardiac pathology (with focus on myocardial infarction). J Mol Cell Cardiol 2016; 94:107-121. [PMID: 27056419 DOI: 10.1016/j.yjmcc.2016.03.015] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 02/09/2016] [Accepted: 03/24/2016] [Indexed: 12/21/2022]
Abstract
Cardiac miRNAs (miR-1, miR133a, miR-208a/b, and miR-499) are abundantly expressed in the myocardium. They play a central role in cardiogenesis, heart function and pathology. While miR-1 and miR-133a predominantly control early stages of cardiogenesis supporting commitment of cardiac-specific muscle lineage from embryonic stem cells and mesodermal precursors, miR-208 and miR-499 are involved in the late cardiogenic stages mediating differentiation of cardioblasts to cardiomyocytes and fast/slow muscle fiber specification. In the heart, miR-1/133a control cardiac conductance and automaticity by regulating all phases of the cardiac action potential. miR-208/499 located in introns of the heavy chain myosin genes regulate expression of sarcomeric contractile proteins. In cardiac pathology including myocardial infarction (MI), expression of cardiac miRNAs is markedly altered that leads to deleterious effects associated with heart wounding, arrhythmia, increased apoptosis, fibrosis, hypertrophy, and tissue remodeling. In acute MI, circulating levels of cardiac miRNAs are significantly elevated making them to be a promising diagnostic marker for early diagnosis of acute MI. Great cardiospecific capacity of these miRNAs is very helpful for enhancing regenerative properties and survival of stem cell and cardiac progenitor transplants and for reprogramming of mature non-cardiac cells to cardiomyocytes.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Molecular Genetic Diagnostics and Cell Biology, Division of Laboratory Medicine, Institute of Pediatrics, Research Center for Children's Health, 119991 Moscow, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia; Department of Biophysics, Biological Faculty, Moscow State University, Moscow 119991, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow 121609, Russia
| | - Yuri V Bobryshev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia; Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; School of Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia.
| |
Collapse
|
44
|
α1-Syntrophin Variant Identified in Drug-Induced Long QT Syndrome Increases Late Sodium Current. PLoS One 2016; 11:e0152355. [PMID: 27028743 PMCID: PMC4814026 DOI: 10.1371/journal.pone.0152355] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 03/14/2016] [Indexed: 12/19/2022] Open
Abstract
Drug-induced long-QT syndrome (diLQTS) is often due to drug block of IKr, especially in genetically susceptible patients with subclinical mutations in the IKr-encoding KCHN2. Few variants in the cardiac NaV1.5 Na+ channel complex have been associated with diLQTS. We tested whether a novel SNTA1 (α1-syntrophin) variant (p.E409Q) found in a patient with diLQTS increases late sodium current (INa-L), thereby providing a disease mechanism. Electrophysiological studies were performed in HEK293T cells co-expressing human NaV1.5/nNOS/PMCA4b with either wild type (WT) or SNTA1 variants (A390V-previously reported in congenital LQTS; and E409Q); and in adult rat ventricular cardiomyocytes infected with SNTA1 expressing adenoviruses (WT or one of the two SNTA1 variants). In HEK293T cells and in cardiomyocytes, there was no significant difference in the peak INa densities among the SNTA1 WT and variants. However, both variants increased INa-L (% of peak current) in HEK293T cells (0.58±0.10 in WT vs. 0.90±0.11 in A390V, p = 0.048; vs. 0.88±0.07 in E409Q, p = 0.023). In cardiomyocytes, INa-L was significantly increased by E409Q, but not by A390V compared to WT (0.49±0.14 in WT vs.0.94±0.23 in A390V, p = 0.099; vs. 1.12±0.24 in E409Q, p = 0.019). We demonstrated that a novel SNTA1 variant is likely causative for diLQTS by augmenting INa-L. These data suggest that variants within the NaV1.5-interacting α1-syntrophin are a potential mechanism for diLQTS, thereby expanding the concept that variants within congenital LQTS loci can cause diLQTS.
Collapse
|
45
|
McCauley M, Vallabhajosyula S, Darbar D. Proarrhythmic and Torsadogenic Effects of Potassium Channel Blockers in Patients. Card Electrophysiol Clin 2016; 8:481-93. [PMID: 27261836 DOI: 10.1016/j.ccep.2016.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The most common arrhythmia requiring drug treatment is atrial fibrillation (AF), which affects 2 to 5 million Americans and continues to be a major cause of morbidity and increased mortality. Despite recent advances in catheter-based and surgical therapies, antiarrhythmic drugs continue to be the mainstay of therapy for most patients with symptomatic AF. However, many antiarrhythmics block the rapid component of the cardiac delayed rectifier potassium current (IKr) as a major mechanism of action, and marked QT prolongation and pause-dependent polymorphic ventricular tachycardia (torsades de pointes) are major class toxicities.
Collapse
Affiliation(s)
- Mark McCauley
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 South Wood Street, Suite 920 (MC715), Chicago, IL 60612, USA
| | - Sharath Vallabhajosyula
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 South Wood Street, Suite 920 (MC715), Chicago, IL 60612, USA
| | - Dawood Darbar
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 South Wood Street, Suite 920 (MC715), Chicago, IL 60612, USA.
| |
Collapse
|
46
|
Anderson WD, Makadia HK, Vadigepalli R. Molecular variability elicits a tunable switch with discrete neuromodulatory response phenotypes. J Comput Neurosci 2016; 40:65-82. [PMID: 26621106 PMCID: PMC4867553 DOI: 10.1007/s10827-015-0584-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 10/28/2015] [Accepted: 11/02/2015] [Indexed: 01/08/2023]
Abstract
Recent single cell studies show extensive molecular variability underlying cellular responses. We evaluated the impact of molecular variability in the expression of cell signaling components and ion channels on electrophysiological excitability and neuromodulation. We employed a computational approach that integrated neuropeptide receptor-mediated signaling with electrophysiology. We simulated a population of neurons in which expression levels of a neuropeptide receptor and multiple ion channels were simultaneously varied within a physiological range. We analyzed the effects of variation on the electrophysiological response to a neuropeptide stimulus. Our results revealed distinct response patterns associated with low versus high receptor levels. Neurons with low receptor levels showed increased excitability and neurons with high receptor levels showed reduced excitability. These response patterns were separated by a narrow receptor level range forming a separatrix. The position of this separatrix was dependent on the expression levels of multiple ion channels. To assess the relative contributions of receptor and ion channel levels to the response profiles, we categorized the responses into six phenotypes based on response kinetics and magnitude. We applied several multivariate statistical approaches and found that receptor and channel expression levels influence the neuromodulation response phenotype through a complex though systematic mapping. Our analyses extended our understanding of how cellular responses to neuromodulation vary as a function of molecular expression. Our study showed that receptor expression and biophysical state interact with distinct relative contributions to neuronal excitability.
Collapse
Affiliation(s)
- Warren D Anderson
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, 1020 Locust St, Philadelphia, PA, 19107, USA
- Graduate program in Neuroscience, Thomas Jefferson University, 1020 Locust St, Philadelphia, PA, 19107, USA
- Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust St, Philadelphia, PA, 19107, USA
| | - Hirenkumar K Makadia
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, 1020 Locust St, Philadelphia, PA, 19107, USA
- Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust St, Philadelphia, PA, 19107, USA
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, 1020 Locust St, Philadelphia, PA, 19107, USA.
- Graduate program in Neuroscience, Thomas Jefferson University, 1020 Locust St, Philadelphia, PA, 19107, USA.
- Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust St, Philadelphia, PA, 19107, USA.
| |
Collapse
|
47
|
Sara JD, Sugrue A, Kremen V, Qiang B, Sapir Y, Attia ZI, Ackerman MJ, Friedman PA, Lerman A, Noseworthy PA. Electrocardiographic predictors of coronary microvascular dysfunction in patients with non-obstructive coronary artery disease: Utility of a novel T wave analysis program. Int J Cardiol 2016; 203:601-6. [PMID: 26580336 DOI: 10.1016/j.ijcard.2015.10.228] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/28/2015] [Accepted: 10/30/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND Coronary microvascular dysfunction (CMD) is linked to adverse cardiovascular events. Definitive diagnosis of CMD requires invasive provocative testing during angiography. We developed and tested a novel computerized T wave analysis tool to identify electrocardiographic signatures of CMD. METHODS 1552 patients underwent an invasive assessment of coronary microvascular function. Patients with interpretable pre-procedural ECGs were divided into 2 age and sex matched groups (n=261 in each group, 75% female): normal microvascular function, CFR>2.5 (CFR+), and abnormal microvascular function, CFR ≤ 2.5 (CFR-). ECGs were evaluated using a novel T wave program that quantified subtle changes in T wave morphology. RESULTS T wave repolarization parameters were significantly different between patients with normal and abnormal microvascular function. The top 3 features in males comprised of T wave area in V6 (CFR+: 10091.4 mV(2) vs. CFR-: 8152.3 mV(2), p<0.05); T1 Y-center of gravity in lead II (CFR+: 17.8 mV vs. CFR-: 22.4, p<0.005) and T Peak-T End in lead II (CFR+: 97.6 msec vs. CFR-: 91.1 msec, p<0.05). These could identify the presence of an abnormal CFR with 74 ± 0.2% accuracy. In females, the top 3 features were T wave right slope lead V6 (CFR+: -2489.1 mV/msec vs. CFR-: -2352.3 mV/msec, p<0.005); Amplitude in V6 (CFR+: 190.4 mV vs. 172.7 mV, p=0.05) and Y-center of gravity in lead V1 (CFR+: 33.3 vs. CFR-: 40.0, p=0.001). These features could identify the presence of an abnormal CFR with 67 ± 0.3% accuracy. CONCLUSION Our data demonstrates that a computer-based repolarization measurement tool may identify electrocardiographic signatures of CMD.
Collapse
Affiliation(s)
- Jaskanwal D Sara
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA
| | - Alan Sugrue
- Division of Internal Medicine, Mayo College of Medicine, Rochester, MN, USA
| | - Vaclav Kremen
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA; Czech Institute of Informatics, Robotics, and Cybernetics, Czech Technical University in Prague, Czech Republic
| | - Bo Qiang
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA
| | - Yehu Sapir
- Electrical and Computer Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Zachi I Attia
- Electrical and Computer Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Michael J Ackerman
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA
| | - Paul A Friedman
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA
| | - Amir Lerman
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA
| | - Peter A Noseworthy
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA.
| |
Collapse
|
48
|
Sara JD, Lennon RJ, Ackerman MJ, Friedman PA, Noseworthy PA, Lerman A. Coronary microvascular dysfunction is associated with baseline QTc prolongation amongst patients with chest pain and non-obstructive coronary artery disease. J Electrocardiol 2015; 49:87-93. [PMID: 26620729 DOI: 10.1016/j.jelectrocard.2015.10.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Coronary microvascular dysfunction (CMD) causes ischemia and is linked to adverse cardiovascular events. Acute transmural ischemia is associated with QT prolongation, but whether CMD affects repolarization is unknown. The aim of this study was to determine if CMD is associated with prolongation of resting heart rate corrected QT interval (QTc). METHODS In patients presenting to the catheterization laboratory with chest pain and non-obstructive coronary artery disease (CAD) at angiography, coronary flow reserve (CFR) in response to intracoronary adenosine was measured and compared to baseline to give a CFR ratio. The Bazett's-derived QTc was manually derived from patients' 12-lead ECG obtained prior to the procedure. QTc was compared between patients with normal and abnormal (CFR ratio≤2.5) coronary microvascular function. RESULTS Of the 926 patients included in this study, 281 patients (30%) had CMD (mean age 53.2 years [SD 12.7], 25% male). QTc was significantly longer in those with an abnormal CFR response to adenosine (median [Q1, Q3] ms: 420 [409, 438] vs. 416 [405, 432]; p value<0.001) and patients in the lowest quartile of CFR had a significantly longer QTc compared to those in the highest quartile (median [Q1, Q3] ms: 420 [409, 439] vs. 413 [402, 426]; p<0.001). In a linear regression model adjusting for age and sex, CMD was associated with an increase in QTc of 3.09 ms (p=0.055). CONCLUSION Our data suggest that CMD may be associated with an increase in baseline QTc, however the precise clinical relevance of this finding needs to be better investigated in larger clinical studies.
Collapse
Affiliation(s)
- Jaskanwal D Sara
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA
| | - Ryan J Lennon
- Division of Biomedical Statistics and Informatics, Mayo College of Medicine, Rochester, MN, USA
| | - Michael J Ackerman
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA
| | - Paul A Friedman
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA
| | - Peter A Noseworthy
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA
| | - Amir Lerman
- Division of Cardiovascular Diseases, Mayo College of Medicine, Rochester, MN, USA.
| |
Collapse
|
49
|
Farrugia A, Keyser C, Hollard C, Raul J, Muller J, Ludes B. Targeted next generation sequencing application in cardiac channelopathies: Analysis of a cohort of autopsy-negative sudden unexplained deaths. Forensic Sci Int 2015; 254:5-11. [DOI: 10.1016/j.forsciint.2015.06.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 05/22/2015] [Accepted: 06/24/2015] [Indexed: 12/19/2022]
|
50
|
Sallam K, Li Y, Sager PT, Houser SR, Wu JC. Finding the rhythm of sudden cardiac death: new opportunities using induced pluripotent stem cell-derived cardiomyocytes. Circ Res 2015; 116:1989-2004. [PMID: 26044252 DOI: 10.1161/circresaha.116.304494] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sudden cardiac death is a common cause of death in patients with structural heart disease, genetic mutations, or acquired disorders affecting cardiac ion channels. A wide range of platforms exist to model and study disorders associated with sudden cardiac death. Human clinical studies are cumbersome and are thwarted by the extent of investigation that can be performed on human subjects. Animal models are limited by their degree of homology to human cardiac electrophysiology, including ion channel expression. Most commonly used cellular models are cellular transfection models, which are able to mimic the expression of a single-ion channel offering incomplete insight into changes of the action potential profile. Induced pluripotent stem cell-derived cardiomyocytes resemble, but are not identical, adult human cardiomyocytes and provide a new platform for studying arrhythmic disorders leading to sudden cardiac death. A variety of platforms exist to phenotype cellular models, including conventional and automated patch clamp, multielectrode array, and computational modeling. Induced pluripotent stem cell-derived cardiomyocytes have been used to study long QT syndrome, catecholaminergic polymorphic ventricular tachycardia, hypertrophic cardiomyopathy, and other hereditary cardiac disorders. Although induced pluripotent stem cell-derived cardiomyocytes are distinct from adult cardiomyocytes, they provide a robust platform to advance the science and clinical care of sudden cardiac death.
Collapse
Affiliation(s)
- Karim Sallam
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Yingxin Li
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Philip T Sager
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.)
| | - Steven R Houser
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.).
| | - Joseph C Wu
- From the Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute (K.S., Y.L., P.T.S., J.C.W.), Institute of Stem Cell Biology and Regenerative Medicine (K.S., Y.L., J.C.W.), Stanford University School of Medicine, CA; and Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, Philadelphia, PA (S.R.H.).
| |
Collapse
|