51
|
Regions of Highly Recurrent Electrogram Morphology With Low Cycle Length Reflect Substrate for Atrial Fibrillation. JACC. BASIC TO TRANSLATIONAL SCIENCE 2022; 8:68-84. [PMID: 36777167 PMCID: PMC9911322 DOI: 10.1016/j.jacbts.2022.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 11/27/2022]
Abstract
Traditional anatomically guided ablation and attempts to perform electrogram-guided atrial fibrillation (AF) ablation (CFAE, DF, and FIRM) have not been shown to be sufficient treatment for persistent AF. Using biatrial high-density electrophysiologic mapping in a canine rapid atrial pacing model of AF, we systematically investigated the relationship of electrogram morphology recurrence (EMR) (Rec% and CLR) with established AF electrogram parameters and tissue characteristics. Rec% correlates with stability of rotational activity and with the spatial distribution of parasympathetic nerve fibers. These results have indicated that EMR may therefore be a viable therapeutic target in persistent AF.
Collapse
Key Words
- AF, atrial fibrillation
- AI, anisotropy index
- CFAE, complex fractionated atrial electrogram
- CLR, cycle length of the most recurrent electrogram morphology
- DF, dominant frequency
- EGM, electrogram
- EMR, electrogram morphology recurrence
- FFT, fast Fourier transform
- FI, fractionation interval
- FIRM, focal impulse and rotor mapping
- LAA, left atrial appendage
- LAFW, left atrial free wall
- LAT, local activation time
- OI, organization index
- PLA, posterior left atrium
- PV, pulmonary vein
- RAA, right atrial appendage
- RAFW, right atrial free wall
- RAP, rapid atrial pacing
- Rec%, recurrence percentage
- ShEn, Shannon’s entropy
- arrhythmias
- atrial fibrillation
- fibrosis
- mapping
Collapse
|
52
|
Dago M, Crespo-García T, Cámara-Checa A, Rapún J, Rubio-Alarcón M, Marín M, Tamargo J, Caballero R, Delpón E. Empagliflozin and Dapagliflozin Increase Na + and Inward Rectifier K + Current Densities in Human Cardiomyocytes Derived from Induced Pluripotent Stem Cells (hiPSC-CMs). Cells 2022; 11:3707. [PMID: 36496967 PMCID: PMC9738206 DOI: 10.3390/cells11233707] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/23/2022] Open
Abstract
Dapagliflozin (dapa) and empagliflozin (empa) are sodium-glucose cotransporter-2 inhibitors (SGLT2is) that reduce morbidity and mortality in heart failure (HF) patients. Sodium and inward rectifier K+ currents (INa and IK1), carried by Nav1.5 and Kir2.1 channels, respectively, are responsible for cardiac excitability, conduction velocity, and refractoriness. In HF patients, Nav1.5 and Kir2.1 expression are reduced, enhancing risk of arrhythmia. Incubation with dapa or empa (24-h,1 µM) significantly increased INa and IK1 densities recorded in human-induced pluripotent stem cell-cardiomyocytes (hiPSC-CMs) using patch-clamp techniques. Dapa and empa, respectively, shifted to more hyperpolarized potentials the INa activation and inactivation curves. Identical effects were observed in Chinese hamster ovary (CHO) cells that were incubated with dapa or empa and transiently expressed human Nav1.5 channels. Conversely, empa but not dapa significantly increased human Kir2.1 currents in CHO cells. Dapa and empa effects on INa and IK1 were also apparent in Ca-calmodulin kinase II-silenced CHO cells. Cariporide, a Na+/H+ exchanger type 1 (NHE1) inhibitor, did not increase INa or IK1 in hiPSC-CMs. Dapa and empa at therapeutic concentrations increased INa and IK1 in healthy human cardiomyocytes. These SGLT2is could represent a new class of drugs with a novel and long-pursued antiarrhythmic mechanism of action.
Collapse
Affiliation(s)
- María Dago
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Teresa Crespo-García
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Anabel Cámara-Checa
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Josu Rapún
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marcos Rubio-Alarcón
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Marín
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
53
|
Jin X, Amoni M, Gilbert G, Dries E, Doñate Puertas R, Tomar A, Nagaraju CK, Pradhan A, Yule DI, Martens T, Menten R, Vanden Berghe P, Rega F, Sipido K, Roderick HL. InsP 3R-RyR Ca 2+ channel crosstalk facilitates arrhythmias in the failing human ventricle. Basic Res Cardiol 2022; 117:60. [PMID: 36378362 DOI: 10.1007/s00395-022-00967-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/13/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022]
Abstract
Dysregulated intracellular Ca2+ handling involving altered Ca2+ release from intracellular stores via RyR channels underlies both arrhythmias and reduced function in heart failure (HF). Mechanisms linking RyR dysregulation and disease are not fully established. Studies in animals support a role for InsP3 receptor Ca2+ channels (InsP3R) in pathological alterations in cardiomyocyte Ca2+ handling but whether these findings translate to the divergent physiology of human cardiomyocytes during heart failure is not determined. Using electrophysiological and Ca2+ recordings in human ventricular cardiomyocytes, we uncovered that Ca2+ release via InsP3Rs facilitated Ca2+ release from RyR and induced arrhythmogenic delayed after depolarisations and action potentials. InsP3R-RyR crosstalk was particularly increased in HF at RyR clusters isolated from the T-tubular network. Reduced SERCA activity in HF further facilitated the action of InsP3. Nanoscale imaging revealed co-localisation of InsP3Rs with RyRs in the dyad, which was increased in HF, providing a mechanism for augmented Ca2+ channel crosstalk. Notably, arrhythmogenic activity dependent on InsP3Rs was increased in tissue wedges from failing hearts perfused with AngII to promote InsP3 generation. These data indicate a central role for InsP3R-RyR Ca2+ signalling crosstalk in the pro-arrhythmic action of GPCR agonists elevated in HF and the potential for their therapeutic targeting.
Collapse
Affiliation(s)
- Xin Jin
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium.,Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Matthew Amoni
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Guillaume Gilbert
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Eef Dries
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Rosa Doñate Puertas
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Ashutosh Tomar
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Chandan K Nagaraju
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Ankit Pradhan
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - David I Yule
- Department of Pharmacology and Physiology, Medical Center School of Medicine and Dentistry, University of Rochester, 601 Elmwood Avenue, Box 711, Rochester, NY, 14642, USA
| | - Tobie Martens
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000, Leuven, Belgium.,Cell and Tissue Imaging Cluster (CIC), KU Leuven, 3000, Leuven, Belgium
| | - Roxane Menten
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000, Leuven, Belgium.,Cell and Tissue Imaging Cluster (CIC), KU Leuven, 3000, Leuven, Belgium
| | - Filip Rega
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium.,Department of Cardiology and Department of Cardiac Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Karin Sipido
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - H Llewelyn Roderick
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
54
|
Oknińska M, Mączewski M, Mackiewicz U. Ventricular arrhythmias in acute myocardial ischaemia-Focus on the ageing and sex. Ageing Res Rev 2022; 81:101722. [PMID: 36038114 DOI: 10.1016/j.arr.2022.101722] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 01/31/2023]
Abstract
Annually, approximately 17 million people die from cardiovascular diseases worldwide, half of them suddenly. The most common direct cause of sudden cardiac death is ventricular arrhythmia triggered by an acute coronary syndrome (ACS). The study summarizes the knowledge of the mechanisms of arrhythmia onset during ACS in humans and in animal models and factors that may influence the susceptibility to life-threatening arrhythmias during ACS with particular focus on the age and sex. The real impact of age and sex on the arrhythmic susceptibility within the setting of acute ischaemia is masked by the fact that ACSs result from coronary artery disease appearing with age much earlier among men than among women. However, results of researches show that in ageing process changes with potential pro-arrhythmic significance, such as increased fibrosis, cardiomyocyte hypertrophy, decrease number of gap junction channels, disturbances of the intracellular Ca2+ signalling or changes in electrophysiological parameters, occur independently of the development of cardiovascular diseases and are more severe in male individuals. A review of the literature also indicates a marked paucity of research in this area in female and elderly individuals. Greater awareness of sex differences in the aging process could help in the development of personalized prevention methods targeting potential pro-arrhythmic factors in patients of both sexes to reduce mortality during the acute phase of myocardial infarction. This is especially important in an era of aging populations in which women will predominate due to their longer lifespan.
Collapse
Affiliation(s)
- Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland.
| |
Collapse
|
55
|
Sung E, Prakosa A, Zhou S, Berger RD, Chrispin J, Nazarian S, Trayanova NA. Fat infiltration in the infarcted heart as a paradigm for ventricular arrhythmias. NATURE CARDIOVASCULAR RESEARCH 2022; 1:933-945. [PMID: 36589896 PMCID: PMC9802586 DOI: 10.1038/s44161-022-00133-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Infiltrating adipose tissue (inFAT) has been recently found to co-localize with scar in infarcted hearts and may contribute to ventricular arrhythmias (VAs), a life-threatening heart rhythm disorder. However, the contribution of inFAT to VA has not been well-established. We investigated the role of inFAT versus scar in VA through a combined prospective clinical and mechanistic computational study. Using personalized computational heart models and comparing the results from simulations of VA dynamics with measured electrophysiological abnormalities during the clinical procedure, we demonstrate that inFAT, rather than scar, is a primary driver of arrhythmogenic propensity and is frequently present in critical regions of the VA circuit. We determined that, within the VA circuitry, inFAT, as opposed to scar, is primarily responsible for conduction slowing in critical sites, mechanistically promoting VA. Our findings implicate inFAT as a dominant player in infarct-related VA, challenging existing paradigms and opening the door for unexplored anti-arrhythmic strategies.
Collapse
Affiliation(s)
- Eric Sung
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.,Alliance for Cardiovascular Diagnostic and Treatment Innovation, Johns Hopkins University, Baltimore, MD, USA
| | - Adityo Prakosa
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.,Alliance for Cardiovascular Diagnostic and Treatment Innovation, Johns Hopkins University, Baltimore, MD, USA
| | - Shijie Zhou
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.,Alliance for Cardiovascular Diagnostic and Treatment Innovation, Johns Hopkins University, Baltimore, MD, USA
| | - Ronald D. Berger
- Alliance for Cardiovascular Diagnostic and Treatment Innovation, Johns Hopkins University, Baltimore, MD, USA.,Department of Medicine, Division of Cardiology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Jonathan Chrispin
- Alliance for Cardiovascular Diagnostic and Treatment Innovation, Johns Hopkins University, Baltimore, MD, USA.,Department of Medicine, Division of Cardiology, Johns Hopkins Hospital, Baltimore, MD, USA.,These authors jointly supervised this work: Jonathan Chrispin, Saman Nazarian, Natalia A. Trayanova
| | - Saman Nazarian
- Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,These authors jointly supervised this work: Jonathan Chrispin, Saman Nazarian, Natalia A. Trayanova
| | - Natalia A. Trayanova
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.,Alliance for Cardiovascular Diagnostic and Treatment Innovation, Johns Hopkins University, Baltimore, MD, USA.,These authors jointly supervised this work: Jonathan Chrispin, Saman Nazarian, Natalia A. Trayanova.,Correspondence and requests for materials should be addressed to Natalia A. Trayanova.
| |
Collapse
|
56
|
Mechanism and prevention of atrial remodeling and their related genes in cardiovascular disorders. Curr Probl Cardiol 2022; 48:101414. [PMID: 36155200 DOI: 10.1016/j.cpcardiol.2022.101414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022]
Abstract
Atrial fibrillation (AF) is associated with profound structural and functional changes in the atrium. Inflammation mediated atrial fibrosis is one of the key mechanisms in the pathogenesis of AF. The collagen deposition in extracellular matrix (ECM) is mainly mediated by transforming growth factor β1 (TGF-β1) which promotes AF via controlling smads mediated-collagen gene transcription and regulating the balance of metalloproteinases (MMPs)/ tissue inhibitor of metalloproteinases (TIMPs). Although many processes can alter atrial properties and promote AF, animal models and clinical studies have provided insights into two major forms of atrial remodeling: Atrial tachycardia remodeling (ATR), which occurs with rapid atrial tachyarrhythmia's such as AF and atrial flutter, and atrial structural remodeling (ASR), which is associated with CHF and other fibrosis-promoting conditions. The mechanism of atrial remodeling such as atrial enlargement, ultra structural changes of atrial muscle tissue and myocardial interstitial fibrosis in AF is still unclear. At present, many studies focus on calcium overload, renin angiotensin aldosterone system and transforming growth factor β1, that effect on atrial structural remodeling. Recent experimental studies and clinical investigations have provided structural remodeling is important contributor to the AF. This paper reviews the current understanding of the progresses about mechanism of atrial structural remodeling, and highlights the potential therapeutic approaches aimed at attenuating structural remodeling to prevent AF. Now some recent advancements of this area are reviewed in this paper.
Collapse
|
57
|
Modulation of KV4.3-KChIP2 Channels by IQM-266: Role of DPP6 and KCNE2. Int J Mol Sci 2022; 23:ijms23169170. [PMID: 36012438 PMCID: PMC9409462 DOI: 10.3390/ijms23169170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/10/2022] [Accepted: 08/13/2022] [Indexed: 11/17/2022] Open
Abstract
The transient outward potassium current (Itof) is generated by the activation of KV4 channels assembled with KChIP2 and other accessory subunits (DPP6 and KCNE2). To test the hypothesis that these subunits modify the channel pharmacology, we analyzed the electrophysiological effects of (3-(2-(3-phenoxyphenyl)acetamido)-2-naphthoic acid) (IQM-266), a new KChIP2 ligand, on the currents generated by KV4.3/KChIP2, KV4.3/KChIP2/DPP6 and KV4.3/KChIP2/KCNE2 channels. CHO cells were transiently transfected with cDNAs codifying for different proteins (KV4.3/KChIP2, KV4.3/KChIP2/DPP6 or KV4.3/KChIP2/KCNE2), and the potassium currents were recorded using the whole-cell patch-clamp technique. IQM-266 decreased the maximum peak of KV4.3/KChIP2, KV4.3/KChIP2/DPP6 and KV4.3/KChIP2/KCNE2 currents, slowing their time course of inactivation in a concentration-, voltage-, time- and use-dependent manner. IQM-266 produced an increase in the charge in KV4.3/KChIP2 channels that was intensified when DPP6 was present and abolished in the presence of KCNE2. IQM-266 induced an activation unblocking effect during the application of trains of pulses to cells expressing KV4.3/KChIP2 and KV4.3/KChIP2/KCNE2, but not in KV4.3/KChIP2/DPP6 channels. Overall, all these results are consistent with a preferential IQM-266 binding to an active closed state of Kv4.3/KChIP2 and Kv4.3/KChIP2/KCNE2 channels, whereas in the presence of DPP6, IQM-266 binds preferentially to an inactivated state. In conclusion, DPP6 and KCNE2 modify the pharmacological response of KV4.3/KChIP2 channels to IQM-266.
Collapse
|
58
|
Wang R, Ye H, Ma L, Wei J, Wang Y, Zhang X, Wang L. Effect of Sacubitril/Valsartan on Reducing the Risk of Arrhythmia: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front Cardiovasc Med 2022; 9:890481. [PMID: 35859597 PMCID: PMC9289747 DOI: 10.3389/fcvm.2022.890481] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022] Open
Abstract
Background and Objective Relevant data of PARADIGM-HF reveals sacubitril/valsartan (SV) therapy led to a greater reduction in the risks of arrhythmia, and sudden cardiac death than angiotensin converting enzyme inhibitor (ACEI)/angiotensin receptor inhibitor (ARB) therapy in HFrEF, however, inconsistent results were reported in subsequent studies. Here, we conduct a meta-analysis of related randomized controlled trials (RCTs) to evaluate the protective effect of SV on reducing the risk of arrhythmias. Methods and Results RCTs focused on the difference in therapeutic outcomes between SV and ACEI/ARB were searched from PUBMED, EMBASE, ClinicalTrials.gov, and Cochrane Library. The results were extracted from each individual study, expressed as binary risk, 95% confidence interval (CI) and relative risk (RR). Sixteen RCTs including 22, 563 patients met the study criteria. Compared with ACEI/ARB therapy, SV therapy did significantly reduce in the risks of severe arrhythmias among patients with heart failure with reduced ejection fraction (HFrEF) (RR 0.83, 95% CI 0.73–0.95, p = 0.006), ventricular tachycardia (VT) among patients with HFrEF (RR 0.69, 95% CI 0.51–0.92, p = 0.01), cardiac arrest among patients with heart failure (HF) (RR 0.52, 95% CI 0.37–0.73, p = 0.0002), cardiac arrest among patients with HFrEF (RR 0.49, 95% CI 0.32–0.76, p = 0.001), cardiac arrest or ventricular fibrillation (VF) among patients with HF (RR 0.63, 95% CI 0.48–0.83, p = 0.001), and cardiac arrest or VF among patients with HFrEF (RR 0.65, 95% CI 0.47–0.89, p = 0.008), but reduced the risks of arrhythmias (RR 0.87, 95% CI 0.74–1.01, p = 0.07), atrial arrhythmias (RR 0.98, 95% CI 0.83–1.16, p = 0.85), and atrial fibrillation (RR 0.98, 95% CI 0.82–1.17, p = 0.82) among all patients with no significant between-group difference. The merged result was robust after sensitivity analysis, and there was no publication bias. Conclusion Our meta-analysis provides evidence that, compared with ACEI/ARB, SV can additionally reduce the risks of most arrhythmias, just the significant differences are revealed in reducing the risks of VT, severe arrhythmias, and cardiac arrest in patients with HFrEF. Besides, the positive effect of SV on VF according to statistical result of combining VF with cardiac arrest in patients with HFrEF is credibility.
Collapse
Affiliation(s)
- Ruxin Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Haowen Ye
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Li Ma
- Department of Functional Examination, Gansu Provincial Maternal and Child Health Hospital, Lanzhou, China
| | - Jinjing Wei
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ying Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaofang Zhang
- Clinical Experimental Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Xiaofang Zhang,
| | - Lihong Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Lihong Wang,
| |
Collapse
|
59
|
Xia R, Tomsits P, Loy S, Zhang Z, Pauly V, Schüttler D, Clauss S. Cardiac Macrophages and Their Effects on Arrhythmogenesis. Front Physiol 2022; 13:900094. [PMID: 35812333 PMCID: PMC9257039 DOI: 10.3389/fphys.2022.900094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/30/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiac electrophysiology is a complex system established by a plethora of inward and outward ion currents in cardiomyocytes generating and conducting electrical signals in the heart. However, not only cardiomyocytes but also other cell types can modulate the heart rhythm. Recently, cardiac macrophages were demonstrated as important players in both electrophysiology and arrhythmogenesis. Cardiac macrophages are a heterogeneous group of immune cells including resident macrophages derived from embryonic and fetal precursors and recruited macrophages derived from circulating monocytes from the bone marrow. Recent studies suggest antiarrhythmic as well as proarrhythmic effects of cardiac macrophages. The proposed mechanisms of how cardiac macrophages affect electrophysiology vary and include both direct and indirect interactions with other cardiac cells. In this review, we provide an overview of the different subsets of macrophages in the heart and their possible interactions with cardiomyocytes under both physiologic conditions and heart disease. Furthermore, we elucidate similarities and differences between human, murine and porcine cardiac macrophages, thus providing detailed information for researchers investigating cardiac macrophages in important animal species for electrophysiologic research. Finally, we discuss the pros and cons of mice and pigs to investigate the role of cardiac macrophages in arrhythmogenesis from a translational perspective.
Collapse
Affiliation(s)
- Ruibing Xia
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Philipp Tomsits
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Simone Loy
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Zhihao Zhang
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Valerie Pauly
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Dominik Schüttler
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | - Sebastian Clauss
- Department of Medicine I, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital Munich, Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| |
Collapse
|
60
|
Young LJ, Antwi-Boasiako S, Ferrall J, Wold LE, Mohler PJ, El Refaey M. Genetic and non-genetic risk factors associated with atrial fibrillation. Life Sci 2022; 299:120529. [PMID: 35385795 PMCID: PMC9058231 DOI: 10.1016/j.lfs.2022.120529] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 12/15/2022]
Abstract
Atrial fibrillation (AF) is the most common arrhythmic disorder and its prevalence in the United States is projected to increase to more than twelve million cases in 2030. AF increases the risk of other forms of cardiovascular disease, including stroke. As the incidence of atrial fibrillation increases dramatically with age, it is paramount to elucidate risk factors underlying AF pathogenesis. Here, we review tissue and cellular pathways underlying AF, as well as critical components that impact AF susceptibility including genetic and environmental risk factors. Finally, we provide the latest information on potential links between SARS-CoV-2 and human AF. Improved understanding of mechanistic pathways holds promise in preventative care and early diagnostics, and also introduces novel targeted forms of therapy that might attenuate AF progression and maintenance.
Collapse
Affiliation(s)
- Lindsay J Young
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Steve Antwi-Boasiako
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Joel Ferrall
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Loren E Wold
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA; College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Peter J Mohler
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA; Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH, USA
| | - Mona El Refaey
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Surgery, Division of Cardiac Surgery, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
61
|
Younis A, Nehoray N, Glikson M, Bodurian C, Nof E, Bragazzi NL, Berger M, Zareba W, Goldenberg I, Beinart R. QTc Dynamics Following Cardioversion for Persistent Atrial Fibrillation. Front Cardiovasc Med 2022; 9:881446. [PMID: 35722129 PMCID: PMC9205203 DOI: 10.3389/fcvm.2022.881446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundCardioversion (CV) for atrial fibrillation (AF) is common. We aimed to assess changes in QTc over time following electrical CV (ECV) for persistent AF, and to compare the benefit of using continuous Holter monitoring vs. conventional follow-up by ECG.MethodsProspective observational cohort study. We comprised 90 patients admitted to our center for elective ECV due to persistent AF who were prospectively enrolled from July 2017 to August 2018. All patients underwent 7-days Holter started prior to ECV. Baseline QTc was defined as median QTc during 1 h post ECV. The primary endpoint was QTc prolongation defined as QTc ≥500 ms, or ≥10% increase (if baseline QTc was >480 ms). Conventional monitoring was defined as 2-h ECG post ECV.ResultsMean age was 67 ± 11 years and 61% were male. Median baseline QTc was 452 ms (IQ range: 431–479 ms) as compared with a maximal median QTc of 474 ms (IQ range: 433–527 ms; p <0.001 for the change in QTc from baseline). Peak median QTc occurred 44 h post ECV. The primary endpoint was met in 3 patients (3%) using conventional monitoring, compared with 39 new patients (43%) using Holter (p <0.001 for comparison). The Holter monitoring was superior to conventional monitoring in detecting clinically significant QTc prolongation (OR = 13; p <0.001).ConclusionsECV of patients with persistent AF was associated with increased transient risk of QTc prolongation in nearly half of the patients. Peak median QTc occurs during end of second day following ECV and prolonged ECG monitoring provides superior detection of significant QTc prolongation compared with conventional monitoring.
Collapse
Affiliation(s)
- Arwa Younis
- Cardiac Electrophysiology and Pacing Section, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, United States
- *Correspondence: Arwa Younis ; orcid.org/0000-0002-2485-5025
| | - Nofrat Nehoray
- Chaim Sheba Medical Center Affiliated to Sackler Medical School, Tel Aviv University, Ramat Gan, Israel
| | - Michael Glikson
- Heart Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Christopher Bodurian
- Clinical Cardiovascular Research Center, University of Rochester, Rochester, NY, United States
| | - Eyal Nof
- Chaim Sheba Medical Center Affiliated to Sackler Medical School, Tel Aviv University, Ramat Gan, Israel
| | - Nicola Luigi Bragazzi
- Laboratory for Industrial and Applied Mathematics, Center for Disease Modeling, York University, Toronto, ON, Canada
| | - Michael Berger
- Chaim Sheba Medical Center Affiliated to Sackler Medical School, Tel Aviv University, Ramat Gan, Israel
| | - Wojciech Zareba
- Clinical Cardiovascular Research Center, University of Rochester, Rochester, NY, United States
| | - Ilan Goldenberg
- Clinical Cardiovascular Research Center, University of Rochester, Rochester, NY, United States
| | - Roy Beinart
- Chaim Sheba Medical Center Affiliated to Sackler Medical School, Tel Aviv University, Ramat Gan, Israel
| |
Collapse
|
62
|
Assimon MM, Pun PH, Al-Khatib SM, Brookhart MA, Gaynes BN, Winkelmayer WC, Flythe JE. Proton pump inhibitors may enhance the risk of citalopram- and escitalopram-associated sudden cardiac death among patients receiving hemodialysis. Pharmacoepidemiol Drug Saf 2022; 31:670-679. [PMID: 35285107 PMCID: PMC9064943 DOI: 10.1002/pds.5428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/17/2022] [Accepted: 03/10/2022] [Indexed: 11/08/2022]
Abstract
PURPOSE Polypharmacy is common in the hemodialysis population and increases the likelihood that patients will be exposed to clinically significant drug-drug interactions. Concurrent use of proton pump inhibitors (PPIs) with citalopram or escitalopram may potentiate the QT-prolonging effects of these selective serotonin reuptake inhibitors through pharmacodynamic and/or pharmacokinetic interactions. METHODS We conducted a retrospective cohort study using data from the U.S. Renal Data System (2007-2017) and a new-user design to examine the differential risk of sudden cardiac death (SCD) associated with citalopram/escitalopram initiation vs. sertraline initiation in the presence and absence of PPI use among adults receiving hemodialysis. We studied 72 559 patients:14 983 (21%) citalopram/escitalopram initiators using a PPI; 26 503 (36%) citalopram/escitalopram initiators not using a PPI;10 779 (15%) sertraline initiators using a PPI; and 20 294 (28%) sertraline initiators not using a PPI (referent). The outcome of interest was 1-year SCD. We used inverse probability of treatment weighted survival models to estimate weighted hazard ratios (HRs) and 95% confidence intervals (CIs). RESULTS Compared with sertraline initiators not using a PPI, citalopram/escitalopram initiators using a PPI had the numerically highest risk of SCD (HR [95% CI] = 1.31 [1.11-1.54]), followed by citalopram/escitalopram initiators not using a PPI (HR [95% CI] = 1.22 [1.06-1.41]). Sertraline initiators using a PPI had a similar risk of SCD compared with those not using a PPI (HR [95% CI] = 1.03 [0.85-1.26]). CONCLUSIONS Existing PPI use may elevate the risk of SCD associated with citalopram or escitalopram initiation among hemodialysis patients.
Collapse
Affiliation(s)
- Magdalene M. Assimon
- University of North Carolina Kidney Center, Division of Nephrology and Hypertension, Department of Medicine, UNC School of Medicine, Chapel Hill, NC
| | - Patrick H. Pun
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC
- Duke Clinical Research Institute, Durham NC
| | - Sana M. Al-Khatib
- Duke Clinical Research Institute, Durham NC
- Division of Cardiology, Duke University Medical Center, Durham, NC
| | - M. Alan Brookhart
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC
| | - Bradley N. Gaynes
- Department of Psychiatry, UNC School of Medicine, Chapel Hill, NC
- Department of Epidemiology, UNC Gillings School of Global Public Health, Chapel Hill, NC
| | - Wolfgang C. Winkelmayer
- Selzman Institute for Kidney Health, Section of Nephrology, Baylor College of Medicine, Houston, TX
| | - Jennifer E. Flythe
- University of North Carolina Kidney Center, Division of Nephrology and Hypertension, Department of Medicine, UNC School of Medicine, Chapel Hill, NC
- Cecil G. Sheps Center for Health Services Research, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
63
|
Interplay between mineral bone disorder and cardiac damage in acute kidney injury: from Ca 2+ mishandling and preventive role of Klotho in mice to its potential mortality prediction in human. Transl Res 2022; 243:60-77. [PMID: 35077866 DOI: 10.1016/j.trsl.2022.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 12/27/2022]
Abstract
Biomarkers of mineral bone disorders (MBD) including phosphorus, fibroblast growth factor (FGF)-23 and Klotho are strongly altered in patients with acute kidney injury (AKI) who have high cardiac outcomes and mortality rates. However, the crosslink between MBD and cardiac damage after an AKI episode still remains unclear. We tested MBD and cardiac biomarkers in an experimental AKI model after 24 or 72 hours of folic acid injection and we analyzed structural cardiac remodeling, intracellular calcium (Ca2+) dynamics in cardiomyocytes and cardiac rhythm. AKI mice presented high levels of FGF-23, phosphorus and cardiac troponin T and exhibited a cardiac hypertrophy phenotype accompanied by an increase in systolic Ca2+ release 24 hours after AKI. Ca2+ transients and contractile dysfunction were reduced 72 hours after AKI while diastolic sarcoplasmic reticulum Ca2+ leak, pro-arrhythmogenic Ca2+ events and ventricular arrhythmias were increased. These cardiac events were linked to the activation of the calcium/calmodulin-dependent kinase II pathway through the increased phosphorylation of ryanodine receptors and phospholamban specific sites after AKI. Cardiac hypertrophy and the altered intracellular Ca2+ dynamics were prevented in transgenic mice overexpressing Klotho after AKI induction. In a translational retrospective longitudinal clinical study, we determined that combining FGF-23 and phosphorus with cardiac troponin T levels achieved a better prediction of mortality in AKI patients at hospital admission. Thus, monitoring MBD and cardiac damage biomarkers could be crucial to prevent mortality in AKI patients. In this setting, Klotho might be considered as a new cardioprotective therapeutic tool to prevent deleterious cardiac events in AKI conditions.
Collapse
|
64
|
Oshiyama NF, Pereira AHM, Cardoso AC, Franchini KG, Bassani JWM, Bassani RA. Developmental differences in myocardial transmembrane Na + transport: Implications for excitability and Na + handling. J Physiol 2022; 600:2651-2667. [PMID: 35489088 DOI: 10.1113/jp282661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/20/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Previous studies showed that myocardial preparations from immature rats are less sensitive to electrical field stimulation than adult preparations. Freshly-isolated ventricular myocytes from neonatal rats showed lower excitability than adult cells, e.g., less negative threshold membrane potential and greater membrane depolarization required for action potential triggering. In addition to differences in mRNA levels for Na+ channels isoforms and greater Na+ current (INa ) density, Na+ channel voltage-dependence was shifted to the right in immature myocytes, which seems to be sufficient to decrease excitability, according to computer simulations. Only in neonatal myocytes did cyclic activity promote marked cytosolic Na+ accumulation, which was prevented by abolition of systolic Ca2+ transients by blockade of Ca2+ currents. Developmental changes in INa may account for the difference in action potential initiation parameters, but not for cytosolic Na+ accumulation, which seems to be due mainly to Na+ /Ca2+ exchanger-mediated Na+ influx. ABSTRACT Little is currently known about possible developmental changes in myocardial Na+ handling, which may have impact on cell excitability and Ca2+ content. Resting intracellular Na+ concentration ([Na+ ]i ), measured in freshly-isolated rat ventricular myocytes with CoroNa-green, was not significantly different in neonates (3-5 days old) and adults, but electrical stimulation caused marked [Na+ ]i rise only in neonates. Inhibition of L-type Ca2+ current by CdCl2 abolished not only systolic Ca2+ transients, but also activity-dependent intracellular Na+ accumulation in immature cells. This indicates that the main Na+ influx pathway during activity is the Na+ /Ca2+ exchanger, rather than voltage-dependent Na+ current (INa ), which was not affected by CdCl2 . In immature myocytes, INa density was 2-fold greater, inactivation was faster, and the current peak occurred at less negative transmembrane potential (Em ) than in adults. Na+ channel steady-state activation and inactivation curves in neonates showed a rightward shift, which should increase channel availability at diastolic Em , but also require greater depolarization for excitation, which was observed experimentally and reproduced in computer simulations. Ventricular mRNA levels of Nav 1.1, Nav 1.4 and Nav 1.5 pore-forming isoforms were greater in neonate ventricles, while decrease was seen for the β1 subunit. Both molecular and biophysical changes in the channel profile may contribute to the differences in INa density and voltage-dependence, and also to the less negative threshold Em in neonates, compared to adults. The apparently lower excitability in immature ventricle may confer protection against the development of spontaneous activity in this tissue. Abstract figure legend Little is currently known about possible developmental changes in myocardial Na+ transport, which may have impact on cell excitability and other physiological aspects. At the mRNA level, neonatal rat ventricle expresses a greater variety of Na+ channel isoforms than in adults. In immature ventricular cardiomyocytes, Na+ current (INa ) density was greater, but voltage-dependence is shifted to less negative potentials than in adults. This should increase channel availability at diastolic membrane potential, but also require greater depolarization for excitation, which was observed experimentally and reproduced in computer simulation. We also observed that electrical stimulation caused marked intracellular Na+ accumulation only in neonates, which was abolished when Ca2+ transients and the Na+ /Ca2+ exchanger (NCX) were inhibited by Cd2+ + Ni2+ . Thus, it seems that the main Na+ influx pathway during activity in neonates is the NCX, rather than voltage-dependent INa , which was not affected by these blockers. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Natália F Oshiyama
- Department of Biomedical Engineering, School of Electrical and Computer Engineering, University of Campinas, Campinas, SP, Brazil.,National Laboratory for Cell Calcium Study, (LabNECC), Center for Biomedical Engineering, University of Campinas, Campinas, SP, Brazil
| | - Ana H M Pereira
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials (LNBio/CNPEM), Campinas, SP, Brazil
| | - Alisson C Cardoso
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials (LNBio/CNPEM), Campinas, SP, Brazil
| | - Kleber G Franchini
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials (LNBio/CNPEM), Campinas, SP, Brazil.,Department of Internal Medicine, School of Medicine, University of Campinas, Campinas, SP, Brazil
| | - José W M Bassani
- Department of Biomedical Engineering, School of Electrical and Computer Engineering, University of Campinas, Campinas, SP, Brazil.,National Laboratory for Cell Calcium Study, (LabNECC), Center for Biomedical Engineering, University of Campinas, Campinas, SP, Brazil
| | - Rosana A Bassani
- Department of Biomedical Engineering, School of Electrical and Computer Engineering, University of Campinas, Campinas, SP, Brazil.,National Laboratory for Cell Calcium Study, (LabNECC), Center for Biomedical Engineering, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
65
|
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia despite substantial efforts to understand the pathophysiology of the condition and develop improved treatments. Identifying the underlying causative mechanisms of AF in individual patients is difficult and the efficacy of current therapies is suboptimal. Consequently, the incidence of AF is steadily rising and there is a pressing need for novel therapies. Research has revealed that defects in specific molecular pathways underlie AF pathogenesis, resulting in electrical conduction disorders that drive AF. The severity of this so-called electropathology correlates with the stage of AF disease progression and determines the response to AF treatment. Therefore, unravelling the molecular mechanisms underlying electropathology is expected to fuel the development of innovative personalized diagnostic tools and mechanism-based therapies. Moreover, the co-creation of AF studies with patients to implement novel diagnostic tools and therapies is a prerequisite for successful personalized AF management. Currently, various treatment modalities targeting AF-related electropathology, including lifestyle changes, pharmaceutical and nutraceutical therapy, substrate-based ablative therapy, and neuromodulation, are available to maintain sinus rhythm and might offer a novel holistic strategy to treat AF.
Collapse
Affiliation(s)
- Bianca J J M Brundel
- Department of Physiology, Amsterdam University Medical Centers, VU Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands.
| | - Xun Ai
- Department of Physiology and Cell Biology, College of Medicine/Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | | | - Myrthe F Kuipers
- AFIPonline.org, Atrial Fibrillation Innovation Platform, Amsterdam, Netherlands
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | |
Collapse
|
66
|
Liu M, Kang GJ, Dudley SC. Preventing unfolded protein response-induced ion channel dysregulation to treat arrhythmias. Trends Mol Med 2022; 28:443-451. [DOI: 10.1016/j.molmed.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 01/15/2023]
|
67
|
Remme CA. Getting to the heart of rhythm: A century of progress. Physiol Rev 2022; 102:1553-1567. [PMID: 35343827 DOI: 10.1152/physrev.00043.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The human heart beats over eighty thousand times a day, and the average person's heart may have beaten up to 3 billion times by the age of 80. During the early stages of pregnancy, the heart beat provides the first visual and auditory sign of life of the foetus. Conversely, the first audible sound that the foetus is likely to hear is the heart beat of the mother. How fitting then, that at the "birth" Physiological Reviews the very first article published in 1921 written by Eyster and Meek addressed "The origin and conduction of the heart beat".1 In their insightful review, the authors discussed the landmark discoveries made from the mid-19th century on the electrical function of the heart. Now, a hundred years later, at the start of the next century of Physiological Reviews, an update on the huge progress made in the "exciting" field of cardiac electrophysiology is warranted. Guided by a number of excellent reviews published in Physiological Reviews since 1921 as well as a large body of literature, an overview of the important advancements made on the topic is provided here.
Collapse
Affiliation(s)
- Carol Ann Remme
- Amsterdam UMC, location University of Amsterdam, Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
68
|
Attachaipanich T, Chattipakorn SC, Chattipakorn N. Potential roles of sodium-glucose co-transporter 2 inhibitors in attenuating cardiac arrhythmias in diabetes and heart failure. J Cell Physiol 2022; 237:2404-2419. [PMID: 35324001 DOI: 10.1002/jcp.30727] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/02/2022] [Accepted: 03/12/2022] [Indexed: 12/12/2022]
Abstract
Sodium-glucose co-transporter 2 (SGLT-2) inhibitors are antidiabetic drugs that have been shown to exert cardiovascular benefits. Their benefits including a reduction of cardiovascular events and worsening heart failure have been extended to nondiabetic patients with high-risk. Although both heart failure and diabetes are known to increase risk of cardiac arrhythmias, the effects of SGLT-2 inhibitors on arrhythmia reduction and their underlying mechanisms are still not fully understood. This review aims to summarize the current available evidence ranging from basic research to clinical reports regarding the potential benefits of SGLT-2 inhibitors against cardiac arrhythmias. Previous in vitro and in vivo studies using various models including heart failure and diabetes are comprehensively summarized to examine the evidence of how SGLT-2 inhibitors affect cardiac action potential, cellular ion currents, calcium ion homeostasis, and cardiac mitochondrial function. Clinical reports investigating the association between SGLT-2 inhibitors and arrhythmias including atrial fibrillation and ventricular arrhythmias are also comprehensively summarized. Valuable information obtained from this review can be used to encourage further clinical investigations to warrant the potential use of SGLT-2 inhibitors against cardiac arrhythmias in both diabetic and heart failure settings.
Collapse
Affiliation(s)
- Tanawat Attachaipanich
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Research Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
69
|
Gionti V, Scacchi S, Colli Franzone P, Pavarino LF, Dore R, Storti C. Role of Scar and Border Zone Geometry on the Genesis and Maintenance of Re-Entrant Ventricular Tachycardia in Patients With Previous Myocardial Infarction. Front Physiol 2022; 13:834747. [PMID: 35399271 PMCID: PMC8989182 DOI: 10.3389/fphys.2022.834747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/21/2022] [Indexed: 11/23/2022] Open
Abstract
In patients with healed myocardial infarction, the left ventricular ejection fraction is characterized by low sensitivity and specificity in the prediction of future malignant arrhythmias. Thus, there is the need for new parameters in daily practice to perform arrhythmic risk stratification. The aim of this study is to identify some features of proarrhythmic geometric configurations of scars and border zones (BZ), by means of numerical simulations based on left ventricular models derived from post myocardial infarction patients. Two patients with similar clinical characteristics were included in this study. Both patients exhibited left ventricular scars characterized by subendo- and subepicardial BZ and a transmural BZ isthmus. The scar of patient #1 was significantly larger than that of patient #2, whereas the transmural BZ isthmus and the subdendo- and subepicardial BZs of patient #2 were thicker than those of patient #1. Patient #1 was positive at electrophysiologic testing, whereas patient #2 was negative. Based on the cardiac magnetic resonance (CMR) data, we developed a geometric model of the left ventricles of the two patients, taking into account the position, extent, and topological features of scars and BZ. The numerical simulations were based on the anisotropic monodomain model of electrocardiology. In the model of patient #1, sustained ventricular tachycardia (VT) was inducible by an S2 stimulus delivered at any of the six stimulation sites considered, while in the model of patient #2 we were not able to induce sustained VT. In the model of patient #1, making the subendo- and subepicardial BZs as thick as those of patient #2 did not affect the inducibility and maintenance of VT. On the other hand, in the model of patient #2, making the subendo- and subepicardial BZs as thin as those of patient #1 yielded sustained VT. In conclusion, the results show that the numerical simulations have an effective predictive capability in discriminating patients at high arrhythmic risk. The extent of the infarct scar and the presence of transmural BZ isthmuses and thin subendo- and subepicardial BZs promote sustained VT.
Collapse
Affiliation(s)
- Vincenzo Gionti
- Divisione di Cardiologia, Istituto di Cura Città di Pavia, Pavia, Italy
| | - Simone Scacchi
- Dipartimento di Matematica, Università degli Studi di Milano, Milan, Italy
- *Correspondence: Simone Scacchi
| | | | - Luca F. Pavarino
- Dipartimento di Matematica, Università degli Studi di Pavia, Pavia, Italy
| | - Roberto Dore
- Divisione di Cardiologia, Istituto di Cura Città di Pavia, Pavia, Italy
| | - Cesare Storti
- Divisione di Cardiologia, Istituto di Cura Città di Pavia, Pavia, Italy
| |
Collapse
|
70
|
Tsumoto K, Kurata Y. Bifurcations and Proarrhythmic Behaviors in Cardiac Electrical Excitations. Biomolecules 2022; 12:459. [PMID: 35327651 PMCID: PMC8946197 DOI: 10.3390/biom12030459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/09/2022] [Accepted: 03/14/2022] [Indexed: 12/23/2022] Open
Abstract
The heart is a hierarchical dynamic system consisting of molecules, cells, and tissues, and acts as a pump for blood circulation. The pumping function depends critically on the preceding electrical activity, and disturbances in the pattern of excitation propagation lead to cardiac arrhythmia and pump failure. Excitation phenomena in cardiomyocytes have been modeled as a nonlinear dynamical system. Because of the nonlinearity of excitation phenomena, the system dynamics could be complex, and various analyses have been performed to understand the complex dynamics. Understanding the mechanisms underlying proarrhythmic responses in the heart is crucial for developing new ways to prevent and control cardiac arrhythmias and resulting contractile dysfunction. When the heart changes to a pathological state over time, the action potential (AP) in cardiomyocytes may also change to a different state in shape and duration, often undergoing a qualitative change in behavior. Such a dynamic change is called bifurcation. In this review, we first summarize the contribution of ion channels and transporters to AP formation and our knowledge of ion-transport molecules, then briefly describe bifurcation theory for nonlinear dynamical systems, and finally detail its recent progress, focusing on the research that attempts to understand the developing mechanisms of abnormal excitations in cardiomyocytes from the perspective of bifurcation phenomena.
Collapse
Affiliation(s)
| | - Yasutaka Kurata
- Department of Physiology II, Kanazawa Medical University, Uchinada 920-0293, Japan;
| |
Collapse
|
71
|
Lou J, Chen H, Huang S, Chen P, Yu Y, Chen F. Update on risk factors and biomarkers of sudden unexplained cardiac death. J Forensic Leg Med 2022; 87:102332. [DOI: 10.1016/j.jflm.2022.102332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/21/2022] [Accepted: 03/02/2022] [Indexed: 02/01/2023]
|
72
|
Kelley BP, Chaudry AM, Syed FF. Developing a Mechanistic Approach to Sudden Death Prevention in Mitral Valve Prolapse. J Clin Med 2022; 11:1285. [PMID: 35268384 PMCID: PMC8910972 DOI: 10.3390/jcm11051285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 11/30/2022] Open
Abstract
Sudden cardiac death (SCD) from ventricular fibrillation (VF) can occur in mitral valve prolapse (MVP) in the absence of other comorbidities including mitral regurgitation, heart failure or coronary disease. Although only a small proportion with MVP are at risk, it can affect young, otherwise healthy adults, most commonly premenopausal women, often as the first presentation of MVP. In this review, we discuss arrhythmic mechanisms in MVP and mechanistic approaches for sudden death risk assessment and prevention. We define arrhythmogenic or arrhythmic MVP (AMVP) as MVP associated with complex and frequent ventricular ectopy, and malignant MVP (MMVP) as MVP with high risk of SCD. Factors predisposing to AMVP are myxomatous, bileaflet MVP and mitral annular disjunction (MAD). Data from autopsy, cardiac imaging and electrophysiological studies suggest that ectopy in AMVP is due to inflammation, fibrosis and scarring within the left ventricular (LV) base, LV papillary muscles and Purkinje tissue. Postulated mechanisms include repetitive injury to these regions from systolic papillary muscle stretch and abrupt mitral annular dysmotility (excursion and curling) and diastolic endocardial interaction of redundant mitral leaflets and chordae. Whereas AMVP is seen relatively commonly (up to 30%) in those with MVP, MVP-related SCD is rare (2-4%). However, the proportion at risk (i.e., with MMVP) is unknown. The clustering of cardiac morphological and electrophysiological characteristics similar to AMVP in otherwise idiopathic SCD suggests that MMVP arises when specific arrhythmia modulators allow for VF initiation and perpetuation through action potential prolongation, repolarization heterogeneity and Purkinje triggering. Adequately powered prospective studies are needed to assess strategies for identifying MMVP and the primary prevention of SCD, including ICD implantation, sympathetic modulation and early surgical mitral valve repair. Given the low event rate, a collaborative multicenter approach is essential.
Collapse
Affiliation(s)
- Brian P. Kelley
- Division of Cardiology, University of North Carolina, Chapel Hill, NC 27599, USA;
| | | | - Faisal F. Syed
- Division of Cardiology, University of North Carolina, Chapel Hill, NC 27599, USA;
| |
Collapse
|
73
|
Aminu AJ, Chen W, Yin Z, Kuniewicz M, Walocha J, Perde F, Molenaar P, Iaizzo PA, Dobrzynski H, Atkinson AJ. Novel micro-computed tomography contrast agents to visualise the human cardiac conduction system and surrounding structures in hearts from normal, aged, and obese individuals. TRANSLATIONAL RESEARCH IN ANATOMY 2022. [DOI: 10.1016/j.tria.2022.100175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
74
|
Li DS, Xue GL, Yang JM, Li CZ, Zhang RX, Tian T, Li Z, Shen KW, Guo Y, Liu XN, Wang J, Lu YJ, Pan ZW. Knockout of interleukin-17A diminishes ventricular arrhythmia susceptibility in diabetic mice via inhibiting NF-κB-mediated electrical remodeling. Acta Pharmacol Sin 2022; 43:307-315. [PMID: 33911193 PMCID: PMC8791974 DOI: 10.1038/s41401-021-00659-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 03/16/2021] [Indexed: 02/03/2023] Open
Abstract
Interleukin-17A (IL-17), a potent proinflammatory cytokine, has been shown to participate in cardiac electrical disorders. Diabetes mellitus is an independent risk factor for ventricular arrhythmia. In this study, we investigated the role of IL-17 in ventricular arrhythmia of diabetic mice. Diabetes was induced in both wild-type and IL-17 knockout mice by intraperitoneal injection of streptozotocin (STZ). High-frequency electrical stimuli were delivered into the right ventricle to induce ventricular arrhythmias. We showed that the occurrence rate of ventricular tachycardia was significantly increased in diabetic mice, which was attenuated by IL-17 knockout. We conducted optical mapping on perfused mouse hearts and found that cardiac conduction velocity (CV) was significantly decreased, and action potential duration (APD) was prolonged in diabetic mice, which were mitigated by IL-17 knockout. We performed whole-cell patch clamp recordings from isolated ventricular myocytes, and found that the densities of Ito, INa and ICa,L were reduced, the APDs at 50% and 90% repolarization were increased, and early afterdepolarization (EAD) was markedly increased in diabetic mice. These alterations were alleviated by the knockout of IL-17. Moreover, knockout of IL-17 alleviated the downregulation of Nav1.5 (the pore forming subunit of INa), Cav1.2 (the main component subunit of ICa,L) and KChIP2 (potassium voltage-gated channel interacting protein 2, the regulatory subunit of Ito) in the hearts of diabetic mice. The expression of NF-κB was significantly upregulated in the hearts of diabetic mice, which was suppressed by IL-17 knockout. In neonatal mouse ventricular myocytes, knockdown of NF-κB significantly increased the expression of Nav1.5, Cav1.2 and KChIP2. These results imply that IL-17 may represent a potential target for the development of agents against diabetes-related ventricular arrhythmias.
Collapse
Affiliation(s)
- De-Sheng Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Gen-Long Xue
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ji-Ming Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Chang-Zhu Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Rui-Xin Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Tao Tian
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Zheng Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ke-Wei Shen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yang Guo
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xue-Ning Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jin Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yan-Jie Lu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Zhen-Wei Pan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
75
|
Andelova K, Bacova BS, Sykora M, Hlivak P, Barancik M, Tribulova N. Mechanisms Underlying Antiarrhythmic Properties of Cardioprotective Agents Impacting Inflammation and Oxidative Stress. Int J Mol Sci 2022; 23:1416. [PMID: 35163340 PMCID: PMC8835881 DOI: 10.3390/ijms23031416] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 01/27/2023] Open
Abstract
The prevention of cardiac life-threatening ventricular fibrillation and stroke-provoking atrial fibrillation remains a serious global clinical issue, with ongoing need for novel approaches. Numerous experimental and clinical studies suggest that oxidative stress and inflammation are deleterious to cardiovascular health, and can increase heart susceptibility to arrhythmias. It is quite interesting, however, that various cardio-protective compounds with antiarrhythmic properties are potent anti-oxidative and anti-inflammatory agents. These most likely target the pro-arrhythmia primary mechanisms. This review and literature-based analysis presents a realistic view of antiarrhythmic efficacy and the molecular mechanisms of current pharmaceuticals in clinical use. These include the sodium-glucose cotransporter-2 inhibitors used in diabetes treatment, statins in dyslipidemia and naturally protective omega-3 fatty acids. This approach supports the hypothesis that prevention or attenuation of oxidative and inflammatory stress can abolish pro-arrhythmic factors and the development of an arrhythmia substrate. This could prove a powerful tool of reducing cardiac arrhythmia burden.
Collapse
Affiliation(s)
- Katarina Andelova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| | - Barbara Szeiffova Bacova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| | - Matus Sykora
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| | - Peter Hlivak
- Department of Arrhythmias and Pacing, National Institute of Cardiovascular Diseases, Pod Krásnou Hôrkou 1, 83348 Bratislava, Slovakia;
| | - Miroslav Barancik
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| | - Narcis Tribulova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Dúbravská Cesta 9, 84104 Bratislava, Slovakia; (K.A.); (M.S.); (M.B.)
| |
Collapse
|
76
|
Yıldız A, Vardı N, Parlakpınar H, Ateş B, Çolakoğlu N. Effects of Low- and High-Dose Valproic Acid and Lamotrigine on the Heart in Female Rats. Cardiovasc Toxicol 2022; 22:326-340. [DOI: 10.1007/s12012-021-09714-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/13/2021] [Indexed: 11/03/2022]
|
77
|
Navarro-García JA, Salguero-Bodes R, González-Lafuente L, Martín-Nunes L, Rodríguez-Sánchez E, Bada-Bosch T, Hernández E, Mérida-Herrero E, Praga M, Solís J, Arribas F, Bueno H, Kuro-O M, Fernández-Velasco M, Ruilope LM, Delgado C, Ruiz-Hurtado G. The anti-aging factor Klotho protects against acquired long QT syndrome induced by uremia and promoted by fibroblast growth factor 23. BMC Med 2022; 20:14. [PMID: 35042527 PMCID: PMC8767669 DOI: 10.1186/s12916-021-02209-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/06/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is associated with increased propensity for arrhythmias. In this context, ventricular repolarization alterations have been shown to predispose to fatal arrhythmias and sudden cardiac death. Between mineral bone disturbances in CKD patients, increased fibroblast growth factor (FGF) 23 and decreased Klotho are emerging as important effectors of cardiovascular disease. However, the relationship between imbalanced FGF23-Klotho axis and the development of cardiac arrhythmias in CKD remains unknown. METHODS We carried out a translational approach to study the relationship between the FGF23-Klotho signaling axis and acquired long QT syndrome in CKD-associated uremia. FGF23 levels and cardiac repolarization dynamics were analyzed in patients with dialysis-dependent CKD and in uremic mouse models of 5/6 nephrectomy (Nfx) and Klotho deficiency (hypomorphism), which show very high systemic FGF23 levels. RESULTS Patients in the top quartile of FGF23 levels had a higher occurrence of very long QT intervals (> 490 ms) than peers in the lowest quartile. Experimentally, FGF23 induced QT prolongation in healthy mice. Similarly, alterations in cardiac repolarization and QT prolongation were observed in Nfx mice and in Klotho hypomorphic mice. QT prolongation in Nfx mice was explained by a significant decrease in the fast transient outward potassium (K+) current (Itof), caused by the downregulation of K+ channel 4.2 subunit (Kv4.2) expression. Kv4.2 expression was also significantly reduced in ventricular cardiomyocytes exposed to FGF23. Enhancing Klotho availability prevented both long QT prolongation and reduced Itof current. Likewise, administration of recombinant Klotho blocked the downregulation of Kv4.2 expression in Nfx mice and in FGF23-exposed cardiomyocytes. CONCLUSION The FGF23-Klotho axis emerges as a new therapeutic target to prevent acquired long QT syndrome in uremia by minimizing the predisposition to potentially fatal ventricular arrhythmias and sudden cardiac death in patients with CKD.
Collapse
Affiliation(s)
- José Alberto Navarro-García
- Cardiorenal Translational Laboratory, Institute of Research imas12, Hospital Universitario 12 de Octubre, Avenida de Córdoba s/n, 28041, Madrid, Spain
| | - Rafael Salguero-Bodes
- Cardiology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Laura González-Lafuente
- Cardiorenal Translational Laboratory, Institute of Research imas12, Hospital Universitario 12 de Octubre, Avenida de Córdoba s/n, 28041, Madrid, Spain
| | - Laura Martín-Nunes
- Biomedical Research Institute Alberto Sols (CSIC-UAM)/CIBER-CV, Arturo Duperier 4, 28029, Madrid, Spain
| | - Elena Rodríguez-Sánchez
- Cardiorenal Translational Laboratory, Institute of Research imas12, Hospital Universitario 12 de Octubre, Avenida de Córdoba s/n, 28041, Madrid, Spain
| | - Teresa Bada-Bosch
- Service of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Eduardo Hernández
- Service of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Manuel Praga
- Service of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Jorge Solís
- Cardiology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando Arribas
- Cardiology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Héctor Bueno
- Cardiology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Makoto Kuro-O
- Division of Anti-aging Medicine, Centre for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - María Fernández-Velasco
- IdiPAZ Institute for Health Research/Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, CIBER-CV, Madrid, Spain
| | - Luis Miguel Ruilope
- Cardiorenal Translational Laboratory, Institute of Research imas12, Hospital Universitario 12 de Octubre, Avenida de Córdoba s/n, 28041, Madrid, Spain.,CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain.,European University of Madrid, Madrid, Spain
| | - Carmen Delgado
- Biomedical Research Institute Alberto Sols (CSIC-UAM)/CIBER-CV, Arturo Duperier 4, 28029, Madrid, Spain.
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research imas12, Hospital Universitario 12 de Octubre, Avenida de Córdoba s/n, 28041, Madrid, Spain. .,CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain.
| |
Collapse
|
78
|
Xu C, Zhao Z, Yuan W, Fengping Z, Zhiqiang Y, Xiaoqin Z. Effect of allisartan on blood pressure and left ventricular hypertrophy through Kv1.5 channels in hypertensive rats. Clin Exp Hypertens 2022; 44:199-207. [PMID: 35014584 DOI: 10.1080/10641963.2021.2018597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND The objective of the present work was to study the anti-hypertensive effect of allisartan on blood pressure (BP) and in facilitating left ventricular remodeling through voltage-gated potassium channels (Kv) 1.5 channels. METHODS A total of 30 SD rats were randomly divided into sham operation group, hypertension control group, and allisartan treatment group. Hypertension was induced by renal artery stenosis. The animals of treatment group were administered with allisartan once a day at a dose of 30 mg/kg body weight through an oral gavage for 4 weeks. The heart function of animals post 4 weeks of treatment was evaluated by echocardiography, and the degree of ventricular hypertrophy and cardiomyocyte hypertrophy were evaluated by histomorphology. The expression of Kv1.5 is detected by real-time quantitative polymerase chain reaction while Western blotting was used to detect the protein expression. RESULTS Four weeks after renal artery stenosis, a significant difference was observed in the whole heart ratio, left heart ratio, and cardiomyocyte area between allisartan treatment group and the hypertension control group (P< .01). A significant decrease in BP of allisartan treatment group compared to hypertension control group (P< .01) was observed. The expression of Kv1.5 mRNA was increased significantly (P< .01) in allisartan treatment group compared to hypertension control group. Western blot analysis also confirmed the increased expression of Kv1.5 channel. CONCLUSION The results showed that allisartan lowers BP and improves left ventricular remodeling through increased expression of Kv1.5 mRNA.
Collapse
Affiliation(s)
- Chunfang Xu
- Department of Cardiology, Southern Medical University Affiliated FengXian Hospital, Shanghai, China
| | - Ziying Zhao
- Endoscopy Center, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wang Yuan
- Department of Cardiology, Southern Medical University Affiliated FengXian Hospital, Shanghai, China
| | - Zhao Fengping
- Department of Cardiology, Southern Medical University Affiliated FengXian Hospital, Shanghai, China
| | - Yan Zhiqiang
- Department of Cardiology, Southern Medical University Affiliated FengXian Hospital, Shanghai, China
| | - Zhang Xiaoqin
- Department of Cardiology, Southern Medical University Affiliated FengXian Hospital, Shanghai, China
| |
Collapse
|
79
|
Chen S, Meng G, Doytchinova A, Wong J, Straka S, Lacy J, Li X, Chen PS, Everett Iv TH. Skin Sympathetic Nerve Activity and the Short-Term QT Interval Variability in Patients With Electrical Storm. Front Physiol 2022; 12:742844. [PMID: 35002752 PMCID: PMC8728059 DOI: 10.3389/fphys.2021.742844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/23/2021] [Indexed: 11/18/2022] Open
Abstract
Background: Skin sympathetic nerve activity (SKNA) and QT interval variability are known to be associated with ventricular arrhythmias. However, the relationship between the two remains unclear. Objective: The aim was to test the hypothesis that SKNA bursts are associated with greater short-term variability of the QT interval (STVQT) in patients with electrical storm (ES) or coronary heart disease without arrhythmias (CHD) than in healthy volunteers (HV). Methods: We simultaneously recorded the ECG and SKNA during sinus rhythm in patients with ES (N = 10) and CHD (N = 8) and during cold-water pressor test in HV (N = 12). The QT and QTc intervals were manually marked and calculated within the ECG. The STVQT was calculated and compared to episodes of SKNA burst and non-bursting activity. Results: The SKNA burst threshold for ES and HV was 1.06 ± 1.07 and 1.88 ± 1.09 μV, respectively (p = 0.011). During SKNA baseline and burst, the QT/QTc intervals and STVQT for ES and CHD were significantly higher than those of the HV. In all subjects, SKNA bursts were associated with an increased STVQT (from 6.43 ± 2.99 to 9.40 ± 5.12 ms, p = 0.002 for ES; from 9.48 ± 4.40 to 12.8 ± 5.26 ms, p = 0.016 for CHD; and from 3.81 ± 0.73 to 4.49 ± 1.24 ms, p = 0.016 for HV). The magnitude of increased STVQT in ES (3.33 ± 3.06 ms) and CHD (3.34 ± 2.34 ms) was both higher than that of the HV (0.68 ± 0.84 ms, p = 0.047 and p = 0.020). Conclusion: Compared to non-bursting activity, SKNA bursts were associated with a larger increase in the QTc interval and STVQT in patients with heart disease than in HV.
Collapse
Affiliation(s)
- Songwen Chen
- The Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guannan Meng
- The Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Anisiia Doytchinova
- The Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, United States
| | - Johnson Wong
- The Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Susan Straka
- The Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Julie Lacy
- The Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiaochun Li
- Department of Biostatistics, Indiana University School of Medicine & Richard M. Fairbanks School of Public Health, Indianapolis, IN, United States
| | - Peng-Sheng Chen
- The Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Thomas H Everett Iv
- The Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
80
|
Elsokkari I, Tsuji Y, Sapp JL, Nattel S. Recent insights into mechanisms and clinical approaches to electrical storm. Can J Cardiol 2021; 38:439-453. [PMID: 34979281 DOI: 10.1016/j.cjca.2021.12.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/21/2021] [Accepted: 12/30/2021] [Indexed: 12/14/2022] Open
Abstract
Electrical storm, characterized by repetitive ventricular tachycardia/fibrillation (VT/VF) over a short period, is becoming commoner with widespread use of implantable cardioverter-defibrillator (ICD) therapy. Electrical storm, sometimes called "arrhythmic storm" or "VT-storm", is usually a medical emergency requiring hospitalization and expert management, and significantly affects short- and long-term outcomes. This syndrome typically occurs in patients with underlying structural heart disease (ischemic or non-ischemic cardiomyopathy) or inherited channelopathies. Triggers for electrical storm should be sought but are often unidentifiable. Initial management is dictated by the hemodynamic status, while subsequent management typically involves ICD interrogation and reprogramming to reduce recurrent shocks, identification/management of triggers like electrolyte abnormalities, myocardial ischemia, or decompensated heart failure, and antiarrhythmic-drug therapy or catheter ablation. Sympathetic nervous system activation is central to the initiation and maintenance of arrhythmic storm, so autonomic modulation is a cornerstone of management. Sympathetic inhibition can be achieved with medications (particularly beta-adrenoreceptor blockers), deep sedation, or cardiac sympathetic denervation. More definitive management targets the underlying ventricular arrhythmia substrate to terminate and prevent recurrent arrhythmia. Arrhythmia targeting can be achieved with antiarrhythmic medications, catheter ablation or more novel therapies such as stereotactic radiation therapy that targets the arrhythmic substrate. Mechanistic studies point to adrenergic activation and other direct consequences of ICD-shocks in promoting further arrhythmogenesis and hypocontractility. Here, we review the pathophysiologic mechanisms, clinical features, prognosis, and therapeutic options for electrical storm. We also outline a clinical approach to this challenging and complex condition, along with its mechanistic basis.
Collapse
Affiliation(s)
- Ihab Elsokkari
- University of Sydney, Nepean Blue Mountains local health district, Australia
| | - Yukiomi Tsuji
- Department of Physiology of Visceral Function, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - John L Sapp
- Dalhousie University, Queen Elizabeth II Health Sciences Centre, Halifax, Nova Scotia, Canada.
| | - Stanley Nattel
- Departments of Medicine and Research Center, Montreal Heart Institute and Université de Montréal and Pharmacology and Therapeutics McGill University, Montreal, Quebec, Canada; Institute of Pharmacology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany; IHU LIYRC Institute, Bordeaux, France.
| |
Collapse
|
81
|
Yang C, Zhang Y, Yang B. MIAT, a potent CVD-promoting lncRNA. Cell Mol Life Sci 2021; 79:43. [PMID: 34921634 PMCID: PMC11072732 DOI: 10.1007/s00018-021-04046-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 10/30/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022]
Abstract
The initial identification of long non-coding RNA myocardial infarction associated transcript (MIAT) as a genetic risk factor of myocardial infarction has made this lncRNA (designated as lncR-MIAT here) a focus of intensive studies worldwide. Emerging evidence supports that lncR-MIAT is susceptible in its expression to multiple deleterious factors like angiotensin II, isoproterenol, hypoxia, and infection and is anomaly overexpressed in serum, plasma, blood cells and myocardial tissues under a variety of cardiovascular conditions including myocardial infarction, cardiac hypertrophy, diabetic cardiomyopathy, dilated cardiomyopathy, sepsis cardiomyopathy, atrial fibrillation and microvascular dysfunction. Experimental results consistently demonstrated that upregulation of lncR-MIAT plays active roles in the pathological processes of the cardiovascular system and knockdown of this lncRNA effectively ameliorates the adverse conditions. The available data revealed that lncR-MIAT acts through multiple mechanisms such as competitive endogenous RNA, natural antisense RNA and RNA/protein interactions. Moreover, the functional domains of lncR-MIAT accounting for certain specific cellular functions of the full-length transcript have been identified and characterized. These insights will not only tremendously advance our understanding of lncRNA biology and pathophysiology, but also offer good opportunities for more innovative and precise design of agents that have the potential to be developed into new drugs for better therapy of cardiovascular diseases (CVDs) in the future. Herein, we provide an overview of lncR-MIAT, focusing on its roles in cardiovascular diseases, underline the unique cellular/molecular mechanisms for its actions, and speculate the perspectives about the translational studies on the potential diagnostic and therapeutic applications of lncR-MIAT.
Collapse
Affiliation(s)
- Chao Yang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, People's Republic of China
- Department of Biochemistry, Qiqihar Medical University, Qiqihar, 161000, Heilongjiang, People's Republic of China
| | - Yong Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, People's Republic of China
- Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, 150086, People's Republic of China
| | - Baofeng Yang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, People's Republic of China.
| |
Collapse
|
82
|
Bueno-Beti C, Asimaki A. Histopathological Features and Protein Markers of Arrhythmogenic Cardiomyopathy. Front Cardiovasc Med 2021; 8:746321. [PMID: 34950711 PMCID: PMC8688541 DOI: 10.3389/fcvm.2021.746321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/17/2021] [Indexed: 11/13/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a heritable heart muscle disease characterized by syncope, palpitations, ventricular arrhythmias and sudden cardiac death (SCD) especially in young individuals. It is estimated to affect 1:5,000 individuals in the general population, with >60% of patients bearing one or more mutations in genes coding for desmosomal proteins. Desmosomes are intercellular adhesion junctions, which in cardiac myocytes reside within the intercalated disks (IDs), the areas of mechanical and electrical cell-cell coupling. Histologically, ACM is characterized by fibrofatty replacement of cardiac myocytes predominantly in the right ventricular free wall though left ventricular and biventricular forms have also been described. The disease is characterized by age-related progression, vast phenotypic manifestation and incomplete penetrance, making proband diagnosis and risk stratification of family members particularly challenging. Key protein redistribution at the IDs may represent a specific diagnostic marker but its applicability is still limited by the need for a myocardial sample. Specific markers of ACM in surrogate tissues, such as the blood and the buccal epithelium, may represent a non-invasive, safe and inexpensive alternative for diagnosis and cascade screening. In this review, we shall cover the most relevant biomarkers so far reported and discuss their potential impact on the diagnosis, prognosis and management of ACM.
Collapse
Affiliation(s)
| | - Angeliki Asimaki
- Molecular and Clinical Sciences Research Institute, St. George's University of London, London, United Kingdom
| |
Collapse
|
83
|
Husti Z, Varró A, Baczkó I. Arrhythmogenic Remodeling in the Failing Heart. Cells 2021; 10:cells10113203. [PMID: 34831426 PMCID: PMC8623396 DOI: 10.3390/cells10113203] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic heart failure is a clinical syndrome with multiple etiologies, associated with significant morbidity and mortality. Cardiac arrhythmias, including ventricular tachyarrhythmias and atrial fibrillation, are common in heart failure. A number of cardiac diseases including heart failure alter the expression and regulation of ion channels and transporters leading to arrhythmogenic electrical remodeling. Myocardial hypertrophy, fibrosis and scar formation are key elements of arrhythmogenic structural remodeling in heart failure. In this article, the mechanisms responsible for increased arrhythmia susceptibility as well as the underlying changes in ion channel, transporter expression and function as well as alterations in calcium handling in heart failure are discussed. Understanding the mechanisms of arrhythmogenic remodeling is key to improving arrhythmia management and the prevention of sudden cardiac death in patients with heart failure.
Collapse
Affiliation(s)
- Zoltán Husti
- Department of Pharmacology and Pharmacotherapy, University of Szeged, 6720 Szeged, Hungary; (Z.H.); (A.V.)
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6720 Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, 6720 Szeged, Hungary; (Z.H.); (A.V.)
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6720 Szeged, Hungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, 6720 Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, 6720 Szeged, Hungary; (Z.H.); (A.V.)
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6720 Szeged, Hungary
- Correspondence:
| |
Collapse
|
84
|
Sung RK, Boyden PA, Higuchi S, Scheinman M. Diagnosis and Management of Complex Reentrant Arrhythmias Involving the His-Purkinje System. Arrhythm Electrophysiol Rev 2021; 10:190-197. [PMID: 34777824 PMCID: PMC8576512 DOI: 10.15420/aer.2021.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/13/2021] [Indexed: 11/04/2022] Open
Abstract
The His-Purkinje system is a network of bundles and fibres comprised of specialised cells that allow for coordinated, synchronous activation of the ventricles. Although the histology and physiology of the His-Purkinje system have been studied for more than a century, its role in ventricular arrhythmias has recently been discovered with the ongoing elucidation of the mechanisms leading to both benign and life-threatening arrhythmias. Studies of Purkinje-cell electrophysiology show multiple mechanisms responsible for ventricular arrhythmias, including enhanced automaticity, triggered activity and reentry. The variation in functional properties of Purkinje cells in different areas of the His-Purkinje system underlie the propensity for reentry within Purkinje fibres in structurally normal and abnormal hearts. Catheter ablation is an effective therapy in nearly all forms of reentrant arrhythmias involving Purkinje tissue. However, identifying those at risk of developing fascicular arrhythmias is not yet possible. Future research is needed to understand the precise molecular and functional changes resulting in these arrhythmias.
Collapse
Affiliation(s)
| | | | - Satoshi Higuchi
- University of California San Francisco, San Francisco, CA, US
| | | |
Collapse
|
85
|
Assimon MM, Pun PH, Wang LCH, Al-Khatib SM, Brookhart MA, Weber DJ, Winkelmayer WC, Flythe JE. Analysis of Respiratory Fluoroquinolones and the Risk of Sudden Cardiac Death Among Patients Receiving Hemodialysis. JAMA Cardiol 2021; 7:75-83. [PMID: 34668928 DOI: 10.1001/jamacardio.2021.4234] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Importance Respiratory fluoroquinolone antibiotics are some of the most common medications with QT interval-prolonging potential prescribed to patients with hemodialysis-dependent kidney failure-individuals who have a very high risk of sudden cardiac death (SCD). To date, there have been no large-scale, population-specific studies evaluating the cardiac safety of respiratory fluoroquinolones in the hemodialysis population. Objective To investigate the cardiac safety of respiratory fluoroquinolones among individuals with hemodialysis-dependent kidney failure. Design, Setting, and Participants A retrospective cohort study examining safety using an active comparator new-user design was conducted using administrative claims data from a US-wide kidney failure registry from January 1, 2007, to December 31, 2016, including 264 968 Medicare beneficiaries receiving in-center maintenance hemodialysis. Data analysis was performed from January 4 to August 16, 2021. Exposures Respiratory fluoroquinolone (levofloxacin or moxifloxacin) vs amoxicillin-based (amoxicillin or amoxicillin with clavulanic acid) antibiotic treatment. Main Outcomes and Measures Sudden cardiac death within 5 days of outpatient initiation of a study antibiotic. Inverse probability of treatment-weighted survival models to estimate hazard ratios (HRs), risk differences (RDs), and corresponding 95% CIs. Death due to a cause other than SCD was treated as a competing event. Fracture was considered as a negative control outcome. Results The study cohort included 264 968 unique in-center hemodialysis patients and 626 322 study antibiotic treatment episodes: 251 726 respiratory fluoroquinolone treatment episodes (40.2%) and 374 596 amoxicillin-based treatment episodes (59.8%). Of the 264 968 patients, 135 236 (51.0%) were men, and the mean (SD) age was 61 (15) years. Respiratory fluoroquinolone vs amoxicillin-based antibiotic treatment was associated with a higher relative and absolute 5-day risk of SCD (weighted HR, 1.95; 95% CI, 1.57-2.41; and weighted RD per 100 000 treatment episodes, 44.0; 95% CI, 31.0-59.2). Respiratory fluoroquinolone vs amoxicillin-based antibiotic treatment was not associated with the 5-day risk of fracture. Conclusions and Relevance In this study, compared with amoxicillin-based antibiotic treatment, respiratory fluoroquinolone treatment was associated with a higher short-term risk of SCD among patients with hemodialysis-dependent kidney failure. This finding suggests that decisions between the use of respiratory fluoroquinolones and amoxicillin-based antibiotics should be individualized, with prescribers considering both the clinical benefits and potential cardiac risks.
Collapse
Affiliation(s)
- Magdalene M Assimon
- University of North Carolina Kidney Center, Division of Nephrology and Hypertension, Department of Medicine, UNC School of Medicine, Chapel Hill
| | - Patrick H Pun
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina.,Duke Clinical Research Institute, Durham, North Carolina
| | - Lily Chin-Hua Wang
- Cecil G. Sheps Center for Health Services Research, University of North Carolina, Chapel Hill
| | - Sana M Al-Khatib
- Duke Clinical Research Institute, Durham, North Carolina.,Division of Cardiology, Duke University Medical Center, Durham, North Carolina
| | - M Alan Brookhart
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| | - David J Weber
- Division of Division of Infectious Diseases, Department of Medicine, UNC School of Medicine, Chapel Hill, North Carolina
| | - Wolfgang C Winkelmayer
- Selzman Institute for Kidney Health, Section of Nephrology, Baylor College of Medicine, Houston, Texas
| | - Jennifer E Flythe
- University of North Carolina Kidney Center, Division of Nephrology and Hypertension, Department of Medicine, UNC School of Medicine, Chapel Hill.,Cecil G. Sheps Center for Health Services Research, University of North Carolina, Chapel Hill
| |
Collapse
|
86
|
Tóth N, Soós A, Váradi A, Hegyi P, Tinusz B, Vágvölgyi A, Orosz A, Solymár M, Polyák A, Varró A, Farkas AS, Nagy N. Effect of ivabradine in heart failure: a meta-analysis of heart failure patients with reduced versus preserved ejection fraction. Can J Physiol Pharmacol 2021; 99:1159-1174. [PMID: 34636643 DOI: 10.1139/cjpp-2020-0700] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In clinical trials of heart failure reduced ejection fraction (HFrEF), ivabradine seemed to be an effective heart rate lowering agent associated with lower risk of cardiovascular death. In contrast, ivabradine failed to improve cardiovascular outcomes in heart failure preserved ejection fraction (HFpEF) despite the significant effect on heart rate. This meta-analysis is the first to compare the effects of ivabradine on heart rate and mortality parameters in HFpEF versus HFrEF. We screened three databases: PubMed, Embase, and Cochrane Library. The outcomes of these studies were mortality, reduction in heart rate, and left ventricular function improvement. We compared the efficacy of ivabradine treatment in HFpEF versus HFrEF. Heart rate analysis of pooled data showed decrease in both HFrEF (-17.646 beats/min) and HFpEF (-11.434 beats/min), and a tendency to have stronger bradycardic effect in HFrEF (p = 0.094) in randomized clinical trials. Left ventricular ejection fraction analysis revealed significant improvement in HFrEF (5.936, 95% CI: [4.199-7.672], p < 0.001) when compared with placebo (p < 0.001). We found that ivabradine significantly improves left ventricular performance in HFrEF, at the same time it exerts a tendency to have improved bradycardic effect in HFrEF. These disparate effects of ivabradine and the higher prevalence of non-cardiac comorbidities in HFpEF may explain the observed beneficial effects in HFrEF and the unchanged outcomes in HFpEF patients after ivabradine treatment.
Collapse
Affiliation(s)
- Noémi Tóth
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School University of Szeged, Dóm Square 12, Szeged 6720, Hungary
| | - Alexandra Soós
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti Street, Pécs 7624, Hungary
| | - Alex Váradi
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti Street, Pécs 7624, Hungary
| | - Péter Hegyi
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti Street, Pécs 7624, Hungary
| | - Benedek Tinusz
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti Street, Pécs 7624, Hungary.,First Department of Medicine, Medical School, University of Pécs, Ifjúság Street 13, Pécs 7624, Hungary
| | - Anna Vágvölgyi
- Department of Internal Medicine, Albert Szent-Györgyi Medical School University of Szeged, Kálvária sgt. 57, Szeged 6720, Hungary
| | - Andrea Orosz
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School University of Szeged, Dóm Square 12, Szeged 6720, Hungary
| | - Margit Solymár
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti Street, Pécs 7624, Hungary
| | - Alexandra Polyák
- Department of Internal Medicine, Albert Szent-Györgyi Medical School University of Szeged, Kálvária sgt. 57, Szeged 6720, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School University of Szeged, Dóm Square 12, Szeged 6720, Hungary.,ELKH-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| | - Attila S Farkas
- Department of Internal Medicine, Albert Szent-Györgyi Medical School University of Szeged, Kálvária sgt. 57, Szeged 6720, Hungary
| | - Norbert Nagy
- Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School University of Szeged, Dóm Square 12, Szeged 6720, Hungary.,ELKH-SZTE Research Group of Cardiovascular Pharmacology, Szeged, Hungary
| |
Collapse
|
87
|
Yoshiki K, Sasagawa Y, Shimojima M, Takeshita Y, Takata S, Hayashi Y, Takamura T, Tachibana O, Nakada M. Thyrotropin-secreting pituitary adenomas induce left atrial enlargement with subclinical atrial fibrillation: an echocardiographic study. Pituitary 2021; 24:778-786. [PMID: 34009499 DOI: 10.1007/s11102-021-01154-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE Thyrotropin-secreting pituitary adenoma (TSHoma) is rare but occasionally causes cardiovascular complications such as atrial fibrillation (AF) due to hyperthyroidism. Graves' disease (GD) is a common hyperthyroid condition often associated with subclinical AF. Some reports have shown echocardiographic changes in patients with GD. We aimed to evaluate the preoperative cardiac function in patients with TSHomas and compared the results among patients with TSHomas and GD and control subjects. METHODS Patients with TSHomas (n = 6) and GD (n = 20) were compared with control subjects with normal cardiac function (n = 20) based on echocardiographic findings. The average age, sex, and proportions of patients with a history of diabetes mellitus and hypertension were equal in each group, and the AF prevalence was matched in patients with TSHomas and GD. The values of left ventricular end-diastolic diameter (LVEDd), left ventricular end-systolic diameter (LVEDs), left ventricular ejection fraction (LVEF), and left atrial diameter (LAD) were used to assess cardiac function. RESULTS In echocardiography, LAD showed a significant difference between patients with TSHomas and control subjects (p = 0.026). The mean LAD values were 36.9 ± 7.1, 38.2 ± 8.9, and 28.7 ± 3.9 mm for patients with TSHomas and GD and control subjects, respectively. There were no significant differences in other echocardiographic parameters among the groups. Before treatment, serum thyroid hormone levels (free triiodothyronine and thyroxin) were not significantly different among patients with TSHomas and GD. CONCLUSION We found that patients with TSHomas or GD had enlarged LADs. This finding suggests that AF may be more hidden in patients with TSHomas than previously reported.
Collapse
Affiliation(s)
- Kenji Yoshiki
- Department of Neurosurgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Yasuo Sasagawa
- Department of Neurosurgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan.
| | - Masaya Shimojima
- Department of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Yumie Takeshita
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Sho Takata
- Department of Neurosurgery, Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Yasuhiko Hayashi
- Department of Neurosurgery, Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Osamu Tachibana
- Department of Neurosurgery, Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Mitsutoshi Nakada
- Department of Neurosurgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| |
Collapse
|
88
|
Jost N, Christ T, Magyar J. New Strategies for the Treatment of Atrial Fibrillation. Pharmaceuticals (Basel) 2021; 14:ph14090926. [PMID: 34577626 PMCID: PMC8466466 DOI: 10.3390/ph14090926] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia in the clinical practice. It significantly contributes to the morbidity and mortality of the elderly population. Over the past 25-30 years intense effort in basic research has advanced the understanding of the relationship between the pathophysiology of AF and atrial remodelling. Nowadays it is clear that the various forms of atrial remodelling (electrical, contractile and structural) play crucial role in initiating and maintaining the persistent and permanent types of AF. Unlike in ventricular fibrillation, in AF rapid ectopic firing originating from pulmonary veins and re-entry mechanism may induce and maintain (due to atrial remodelling) this complex cardiac arrhythmia. The present review presents and discusses in detail the latest knowledge on the role of remodelling in AF. Special attention is paid to novel concepts and pharmacological targets presumably relevant to the drug treatment of atrial fibrillation.
Collapse
Affiliation(s)
- Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6725 Szeged, Hungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, 6725 Szeged, Hungary
- Correspondence:
| | - Torsten Christ
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - János Magyar
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
- Department of Sport Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
89
|
Zangoue M, Zangouei AS, Mojarrad M, Moghbeli M. MicroRNAs as the critical regulators of protein kinases in prostate and bladder cancers. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00190-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
Bladder cancer (BCa) and prostate cancer (PCa) are frequent urothelial and genital malignancies with a high ratio of morbidity and mortality which are more common among males. Since BCa and PCa cases are mainly diagnosed in advanced stages with clinical complications, it is required to introduce the efficient early detection markers. Protein kinases are critical factors involved in various cellular processes such as cell growth, motility, differentiation, and metabolism. Deregulation of protein kinases can be frequently observed through the neoplastic transformation and tumor progression. Therefore, kinases are required to be regulated via different genetic and epigenetic processes. MicroRNAs (miRNAs) are among the critical factors involved in epigenetic regulation of protein kinases. Since miRNAs are noninvasive and more stable factors in serum and tissues compared with mRNAs, they can be used as efficient diagnostic markers for the early detection of PCa and BCa.
Main body
In present review, we have summarized all of the reported miRNAs that have been associated with regulation of protein kinases in bladder and prostate cancers.
Conclusions
For the first time, this review highlights the miRNAs as critical factors in regulation of protein kinases during prostate and bladder cancers which paves the way of introducing a noninvasive kinase-specific panel of miRNAs for the early detection of these malignancies. It was observed that the class VIII receptors of tyrosine kinases and non-receptor tyrosine kinases were the most frequent targets for the miRNAs in bladder and prostate cancers, respectively.
Collapse
|
90
|
Zhang T, Wu Y, Hu Z, Xing W, Kun LV, Wang D, Hu N. Small-Molecule Integrated Stress Response Inhibitor Reduces Susceptibility to Postinfarct Atrial Fibrillation in Rats via the Inhibition of Integrated Stress Responses. J Pharmacol Exp Ther 2021; 378:197-206. [PMID: 34215702 DOI: 10.1124/jpet.121.000491] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 06/23/2021] [Indexed: 11/22/2022] Open
Abstract
Phosphorylation of the eukaryotic translation initiation factor 2 α-subunit, which subsequently upregulates activating transcription factor 4 (ATF4), is the core event in the integrated stress response (ISR) pathway. Previous studies indicate phosphorylation of eukaryotic translation initiation factor 2 ɑ-subunit in atrial tissue in response to atrial fibrillation (AF). This study investigated the role of ISR pathway in experimental AF by using a small-molecule ISR inhibitor (ISRIB). Accordingly, rats were subjected to coronary artery occlusion to induce myocardial infarction (MI), or sham operation, and received either trans-ISRIB (2 mg/kg/d, i.p.) or vehicle for seven days. Thereafter, animals were subjected to the AF inducibility test by transesophageal rapid burst pacing followed by procurement of left atrium (LA) for assessment of atrial fibrosis, inflammatory indices, autophagy-related proteins, ISR activation, ion channel, and connexin 43 expression. Results showed a significant increase in the AF vulnerability and the activation of ISR in LA as evidenced by enhanced eukaryotic translation initiation factor 2 ɑ-subunit phosphorylation. ISRIB treatment suppressed upregulation of ATF4, fibrosis as indexed by determination of α-smooth muscle actin and collagen levels, inflammatory macrophage infiltration (i.e., CD68 and inducible nitric oxide synthase/CD68-positive macrophage), and autophagy as determined by expression of light chain 3. Further, ISRIB treatment reversed the expression of relevant ion channel (i.e., the voltage-gated sodium channel 1.5 , L-type voltage-dependent calcium channel 1.2, and voltage-activated A-type potassium ion channel 4.3) and connexin 43 remodeling. Collectively, the results suggest that the ISR is a key pathway in pathogenesis of AF, post-MI, and represents a novel target for treatment of AF. SIGNIFICANCE STATEMENT: The activation of integrated stress response (ISR) pathway as evidenced by enhanced eukaryotic translation initiation factor 2 ɑ-subunit phosphorylation in left atrium plays a key role in atrial fibrillation (AF). ISR inhibitor (ISRIB) reduces AF occurrence and atrial proarrhythmogenic substrate. The beneficial action of ISRIB may be mediated by suppressing ISR pathway-related cardiac fibrosis, inflammatory macrophage infiltration, autophagy, and restoring the expression of ion channel and connexin 43. This study suggests a key dysfunctional role for ISR in pathogenesis of AF with implications for novel treatment.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Gerontology (T.Z., Y.W., Z.H., W.X., D.W., N.H.) and Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (W.X., K.L., D.W., N.H.), First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, Anhui, China; Department of Psychology, Wannan Medical College, Wuhu, Anhui, China (T.Z.); and Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland (N.H.)
| | - Yong Wu
- Department of Gerontology (T.Z., Y.W., Z.H., W.X., D.W., N.H.) and Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (W.X., K.L., D.W., N.H.), First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, Anhui, China; Department of Psychology, Wannan Medical College, Wuhu, Anhui, China (T.Z.); and Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland (N.H.)
| | - Zhengtao Hu
- Department of Gerontology (T.Z., Y.W., Z.H., W.X., D.W., N.H.) and Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (W.X., K.L., D.W., N.H.), First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, Anhui, China; Department of Psychology, Wannan Medical College, Wuhu, Anhui, China (T.Z.); and Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland (N.H.)
| | - Wen Xing
- Department of Gerontology (T.Z., Y.W., Z.H., W.X., D.W., N.H.) and Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (W.X., K.L., D.W., N.H.), First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, Anhui, China; Department of Psychology, Wannan Medical College, Wuhu, Anhui, China (T.Z.); and Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland (N.H.)
| | - L V Kun
- Department of Gerontology (T.Z., Y.W., Z.H., W.X., D.W., N.H.) and Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (W.X., K.L., D.W., N.H.), First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, Anhui, China; Department of Psychology, Wannan Medical College, Wuhu, Anhui, China (T.Z.); and Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland (N.H.)
| | - Deguo Wang
- Department of Gerontology (T.Z., Y.W., Z.H., W.X., D.W., N.H.) and Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (W.X., K.L., D.W., N.H.), First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, Anhui, China; Department of Psychology, Wannan Medical College, Wuhu, Anhui, China (T.Z.); and Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland (N.H.)
| | - Nengwei Hu
- Department of Gerontology (T.Z., Y.W., Z.H., W.X., D.W., N.H.) and Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (W.X., K.L., D.W., N.H.), First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, Anhui, China; Department of Psychology, Wannan Medical College, Wuhu, Anhui, China (T.Z.); and Department of Pharmacology & Therapeutics and Institute of Neuroscience, Trinity College, Dublin, Ireland (N.H.)
| |
Collapse
|
91
|
Aiba T. Ischemia-induced premature ventricular complexes: Is it still complex? Heart Rhythm 2021; 18:1988-1989. [PMID: 34428560 DOI: 10.1016/j.hrthm.2021.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 08/18/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Takeshi Aiba
- Department of Clinical Laboratory Medicine and Genetics, National Cerebral and Cardiovascular Center, Osaka, Japan; Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan.
| |
Collapse
|
92
|
Marian AJ, Asatryan B, Wehrens XHT. Genetic basis and molecular biology of cardiac arrhythmias in cardiomyopathies. Cardiovasc Res 2021; 116:1600-1619. [PMID: 32348453 DOI: 10.1093/cvr/cvaa116] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/09/2020] [Accepted: 04/21/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiac arrhythmias are common, often the first, and sometimes the life-threatening manifestations of hereditary cardiomyopathies. Pathogenic variants in several genes known to cause hereditary cardiac arrhythmias have also been identified in the sporadic cases and small families with cardiomyopathies. These findings suggest a shared genetic aetiology of a subset of hereditary cardiomyopathies and cardiac arrhythmias. The concept of a shared genetic aetiology is in accord with the complex and exquisite interplays that exist between the ion currents and cardiac mechanical function. However, neither the causal role of cardiac arrhythmias genes in cardiomyopathies is well established nor the causal role of cardiomyopathy genes in arrhythmias. On the contrary, secondary changes in ion currents, such as post-translational modifications, are common and contributors to the pathogenesis of arrhythmias in cardiomyopathies through altering biophysical and functional properties of the ion channels. Moreover, structural changes, such as cardiac hypertrophy, dilatation, and fibrosis provide a pro-arrhythmic substrate in hereditary cardiomyopathies. Genetic basis and molecular biology of cardiac arrhythmias in hereditary cardiomyopathies are discussed.
Collapse
Affiliation(s)
- Ali J Marian
- Department of Medicine, Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| | - Babken Asatryan
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Xander H T Wehrens
- Department of Biophysics and Molecular Physiology, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
93
|
Oknińska M, Paterek A, Bierła J, Czarnowska E, Mączewski M, Mackiewicz U. Effect of age and sex on the incidence of ventricular arrhythmia in a rat model of acute ischemia. Biomed Pharmacother 2021; 142:111983. [PMID: 34392089 DOI: 10.1016/j.biopha.2021.111983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND The impact of sex and age on the arrhythmic susceptibility within the setting of acute ischemia is masked by the fact that acute coronary events result from coronary artery disease appearing with age much earlier among men than among women. METHODS AND RESULTS LAD ligation or sham operations were performed in rats of both sexes at the age 3 and 24 months. An ECG was recorded continuously for 6 h after the operation. The number of early and late premature ventricular beats (PVBs), episodes of ventricular tachycardia (VT) and fibrillation (VF), heart rate, QRS, QT and Tpeak-Tend duration were analysed. Epicardial action potentials were recorded in vivo, Ca2+ signaling was evaluated in isolated cardiomyocytes, fibrosis and connexin-43 expression and localization were measured in the septum. PVBs, VT and VF episodes are much more common in older males than in young males and females independently from their age. Fibrosis with varying intensity in different muscle layers, hypertrophy of cardiomyocytes, reduced number of gap junctions and their appearance on the lateral myocyte membrane, QT prolongation, increase transmural dispersion of repolarisation and a decreased function of SERCA2a may increase the propensity to arrhythmia within the setting of acute ischemia. CONCLUSION We show that the male sex, especially in case of older individuals is a strong predictor of increased arrhythmic susceptibility within the acute ischemia setting regardless of its impact on the occurrence of cardiovascular diseases. A personalized sex-dependent prevention treatment is needed to reduce the mortality in acute phases of myocardial infarction.
Collapse
Affiliation(s)
- Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Joanna Bierła
- Department of Pathology, The Children's Memorial Health Institute, Aleja Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Elżbieta Czarnowska
- Department of Pathology, The Children's Memorial Health Institute, Aleja Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland.
| |
Collapse
|
94
|
Yang PS, Kim D, Jang E, Yu HT, Kim TH, Sung JH, Pak HN, Lee MH, Joung B. Risk of sick sinus syndrome in patients diagnosed with atrial fibrillation: A population-based cohort. J Cardiovasc Electrophysiol 2021; 32:2704-2714. [PMID: 34379851 DOI: 10.1111/jce.15202] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/25/2021] [Accepted: 07/30/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Sinoatrial node dysfunction and atrial fibrillation (AF) frequently coexist and interact with each other, often to initiate and perpetuate each other. OBJECTIVE To determine the effect of AF on the incidence and risk of sick sinus syndrome (SSS). METHODS The association of incident AF with the development of incident SSS was assessed from 2004 to 2014 in 302 229 SSS- and pacemaker-free subjects aged ≥60 years in the Korea National Health Insurance Service-Senior cohort. RESULTS During an observation period of 1 854 800 person-years, incident AF was observed in a total of 12 797 subjects (0.69%/year). The incidence of SSS was 3.4 and 0.2 per 1000 person-years in the propensity score-matched incident AF and no-AF groups, respectively. After adjustment, the significantly increased risk of SSS was observed in the incident AF group, with a hazard ratio (HR) of 13.4 (95% confidence interval [CI]: 8.4-21.4). This finding was consistently observed after censoring for heart failure (HR: 16.0; 95% CI: 9.2-28.0) or heart failure/myocardial infarction (HR: 16.6; 95% CI: 9.3-29.7). Incident AF also was associated with an increased risk of pacemaker implantation related with both SSS (HR: 21.8; 95% CI: 8.7-18.4) and atrioventricular (AV) block (HR: 9.5; 95% CI: 4.9-18.4). These results were consistent regardless of sex and comorbidities. CONCLUSION Incident AF was associated with more than 10 times increased risk of SSS in an elderly population regardless of comorbidities. The risk of pacemaker implantations related with both sinus node dysfunction and AV block was increased in the elderly population with incident AF.
Collapse
Affiliation(s)
- Pil-Sung Yang
- Department of Cardiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Daehoon Kim
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eunsun Jang
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hee Tae Yu
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae-Hoon Kim
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung-Hoon Sung
- Department of Cardiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Hui-Nam Pak
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Moon-Hyoung Lee
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Boyoung Joung
- Division of Cardiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
95
|
Hegyi B, Shimkunas R, Jian Z, Izu LT, Bers DM, Chen-Izu Y. Mechanoelectric coupling and arrhythmogenesis in cardiomyocytes contracting under mechanical afterload in a 3D viscoelastic hydrogel. Proc Natl Acad Sci U S A 2021; 118:e2108484118. [PMID: 34326268 PMCID: PMC8346795 DOI: 10.1073/pnas.2108484118] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The heart pumps blood against the mechanical afterload from arterial resistance, and increased afterload may alter cardiac electrophysiology and contribute to life-threatening arrhythmias. However, the cellular and molecular mechanisms underlying mechanoelectric coupling in cardiomyocytes remain unclear. We developed an innovative patch-clamp-in-gel technology to embed cardiomyocytes in a three-dimensional (3D) viscoelastic hydrogel that imposes an afterload during regular myocyte contraction. Here, we investigated how afterload affects action potentials, ionic currents, intracellular Ca2+ transients, and cell contraction of adult rabbit ventricular cardiomyocytes. We found that afterload prolonged action potential duration (APD), increased transient outward K+ current, decreased inward rectifier K+ current, and increased L-type Ca2+ current. Increased Ca2+ entry caused enhanced Ca2+ transients and contractility. Moreover, elevated afterload led to discordant alternans in APD and Ca2+ transient. Ca2+ alternans persisted under action potential clamp, indicating that the alternans was Ca2+ dependent. Furthermore, all these afterload effects were significantly attenuated by inhibiting nitric oxide synthase 1 (NOS1). Taken together, our data reveal a mechano-chemo-electrotransduction (MCET) mechanism that acutely transduces afterload through NOS1-nitric oxide signaling to modulate the action potential, Ca2+ transient, and contractility. The MCET pathway provides a feedback loop in excitation-Ca2+ signaling-contraction coupling, enabling autoregulation of contractility in cardiomyocytes in response to afterload. This MCET mechanism is integral to the individual cardiomyocyte (and thus the heart) to intrinsically enhance its contractility in response to the load against which it has to do work. While this MCET is largely compensatory for physiological load changes, it may also increase susceptibility to arrhythmias under excessive pathological loading.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Rafael Shimkunas
- Department of Pharmacology, University of California, Davis, CA 95616
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Zhong Jian
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Leighton T Izu
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Ye Chen-Izu
- Department of Pharmacology, University of California, Davis, CA 95616;
- Department of Biomedical Engineering, University of California, Davis, CA 95616
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, CA 95616
| |
Collapse
|
96
|
Excitation and Contraction of the Failing Human Heart In Situ and Effects of Cardiac Resynchronization Therapy: Application of Electrocardiographic Imaging and Speckle Tracking Echo-Cardiography. HEARTS 2021. [DOI: 10.3390/hearts2030027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Despite the success of cardiac resynchronization therapy (CRT) for treating heart failure (HF), the rate of nonresponders remains 30%. Improvements to CRT require understanding of reverse remodeling and the relationship between electrical and mechanical measures of synchrony. The objective was to utilize electrocardiographic imaging (ECGI, a method for noninvasive cardiac electrophysiology mapping) and speckle tracking echocardiography (STE) to study the physiology of HF and reverse remodeling induced by CRT. We imaged 30 patients (63% male, mean age 63.7 years) longitudinally using ECGI and STE. We quantified CRT-induced remodeling of electromechanical parameters and evaluated a novel index, the electromechanical delay (EMD, the delay from activation to peak contraction). We also measured dyssynchrony using ECGI and STE and compared their effectiveness for predicting response to CRT. EMD values were elevated in HF patients compared to controls. However, the EMD values were dependent on the activation sequence (CRT-paced vs. un-paced), indicating that the EMD is not intrinsic to the local tissue, but is influenced by factors such as opposing wall contractions. After 6 months of CRT, patients had increased contraction in native rhythm compared to baseline pre-CRT (baseline: −8.55%, 6 months: −10.14%, p = 0.008). They also had prolonged repolarization at the location of the LV pacing lead. The pre-CRT delay between mean lateral LV and RV electrical activation time was the best predictor of beneficial reduction in LV end systolic volume by CRT (Spearman’s Rho: −0.722, p < 0.001); it outperformed mechanical indices and 12-lead ECG criteria. HF patients have abnormal EMD. The EMD depends upon the activation sequence and is not predictive of response to CRT. ECGI-measured LV activation delay is an effective index for CRT patient selection. CRT causes persistent improvements in contractile function.
Collapse
|
97
|
Boscamp NS, Liberman L, Silver ES. Anodal stimulation in pediatric patients with permanent epicardial ventricular pacemakers. PACING AND CLINICAL ELECTROPHYSIOLOGY: PACE 2021; 44:1593-1598. [PMID: 34287953 DOI: 10.1111/pace.14323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/08/2021] [Accepted: 07/18/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVES We aimed to measure the frequency and factors associated with anodal stimulation in a pediatric population with epicardial pacing leads. BACKGROUND In bipolar pacemakers, capture of the myocardium typically occurs at the cathode. However, AS with capture at the anode has been described. This has not been described in epicardial pacemakers. METHODS Retrospective data were collected from patients ≤ 21 years of age with permanent bipolar epicardial ventricular pacemakers from 1/2017 to 1/2018. AS was defined as a clear change on surface ECG in at least one of the 12 leads assessed by two blinded pediatric electrophysiologists. RESULTS Twenty-four bipolar leads in 23 patients were included in the study. One patient had both biventricular leads tested. Median age was 7.1 years (IQR 5.0-10.9), weight was 20.9 kg (IQR 16.5-33.5), and 65% were male. Testing was performed at a median of 2.8 years (IQR 1.6-6.1) after implant. Congenital heart disease was present in 57%. Complete heart block was the pacemaker indication in 78%. AS was identified in 16/24 (67%) of leads tested. Identification of AS was associated with presence of congenital heart disease (p = 0.004) and 3DD between electrodes (p = 0.04). CONCLUSIONS AS is common in pediatric patients and was associated with a history of congenital heart disease and greater estimated 3DD between electrodes. The prevalent nature of AS may allow clinicians to utilize existing pacemakers as multisite pacing systems.
Collapse
Affiliation(s)
- Nicholas S Boscamp
- Division of Pediatric Cardiology, Morgan Stanley Children's Hospital of New York, Columbia University Irving Medical Center, New York, New York, USA
| | - Leonardo Liberman
- Division of Pediatric Cardiology, Morgan Stanley Children's Hospital of New York, Columbia University Irving Medical Center, New York, New York, USA
| | - Eric S Silver
- Division of Pediatric Cardiology, Morgan Stanley Children's Hospital of New York, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
98
|
Histone deacetylase 2-dependent ventricular electrical remodeling in a porcine model of early heart failure. Life Sci 2021; 281:119769. [PMID: 34186046 DOI: 10.1016/j.lfs.2021.119769] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/29/2022]
Abstract
AIMS Heart failure (HF) is linked to electrical remodeling that promotes ventricular arrhythmias. Underlying molecular signaling is insufficiently understood, in particular concerning patients with early disease stages. Previous observations suggest a key role for epigenetic mechanisms in cardiac remodeling processes. We hypothesized that histone deacetylases (HDACs) 1 and 2 contribute to cellular electrophysiological dysregulation in ventricular cardiomyocytes during HF development. MATERIALS AND METHODS HDAC and ion channel expression was quantified in a porcine model of early HF induced by short-term atrial tachypacing, resulting in atrial fibrillation with rapid ventricular rate response. Anti-Hdac1 and anti-Hdac2 siRNA treatment was employed in neonatal murine cardiomyocytes (NMCM) to study effects of HDACs on ion channel mRNA expression and action potential duration (APD). KEY FINDINGS Early HF was characterized by mild reduction of left ventricular ejection fraction, prolonged QTc intervals, and increased ventricular effective refractory periods. Delayed repolarization was linked to significant downregulation of HDAC2 in left ventricular (LV) tissue. In addition, there was a tendency towards reduced transcript expression of KCNJ2/Kir2.1 K+ channels. In NMCM, knock-down of Hdac2 recapitulated AP prolongation. Finally, siRNA-mediated suppression of Hdac2 reduced Kcnh2/Kv11.1 K+ channel expression. SIGNIFICANCE Suppression of HDAC2 is linked to ventricular electrical remodeling of APD and ion channel expression in early stages of heart failure. This previously unrecognized mechanism may serve as basis for future approaches to prevention and treatment of ventricular arrhythmias.
Collapse
|
99
|
Enhanced BDNF Actions Following Acute Hypoxia Facilitate HIF-1α-Dependent Upregulation of Cav3-T-Type Ca 2+ Channels in Rat Cardiomyocytes. MEMBRANES 2021; 11:membranes11070470. [PMID: 34202148 PMCID: PMC8307968 DOI: 10.3390/membranes11070470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 01/19/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) has recently been recognized as a cardiovascular regulator particularly in the diseased condition, including coronary artery disease, heart failure, cardiomyopathy, and hypertension. Here, we investigate the role of BDNF on the T-type Ca2+ channel, Cav3.1 and Cav3.2, in rat neonatal cardiomyocytes exposed to normoxia (21% O2) and acute hypoxia (1% O2) in vitro for up to 3 h. The exposure of cardiomyocytes to hypoxia (1 h, 3 h) caused a significant upregulation of the mRNAs for hypoxia-inducible factor 1α (Hif1α), Cav3.1, Cav3.2 and Bdnf, but not tropomyosin-related kinase receptor B (TrkB). The upregulation of Cav3.1 and Cav3.2 caused by hypoxia was completely halted by small interfering RNA (siRNA) targeting Hif1a (Hif1a-siRNA) or Bdnf (Bdnf-siRNA). Immunocytochemical staining data revealed a distinct upregulation of Cav3.1- and Cav3.2-proteins caused by hypoxia in cardiomyocytes, which was markedly suppressed by Bdnf-siRNA. These results unveiled a novel regulatory action of BDNF on the T-type Ca2+ channels expression through the HIF-1α-dependent pathway in cardiomyocytes.
Collapse
|
100
|
Farag NE, El-Kherbetawy MK, Ismail HM, Abdelrady AM, Toraih EA, Abdelbasset WK, Lashine RM, EL-dosoky M, Abed SY, Ibraheem KM, Fawzy MS, Zaitone SA. Differential Effect of Three Macrolide Antibiotics on Cardiac Pathology and Electrophysiology in a Myocardial Infarction Rat Model: Influence on Sodium Nav1.5 Channel Expression. Pharmaceuticals (Basel) 2021; 14:ph14070597. [PMID: 34206182 PMCID: PMC8308720 DOI: 10.3390/ph14070597] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/19/2022] Open
Abstract
Macrolides were reported to have cardiotoxic effects presented mainly by electrocardiogram (ECG) changes with increased risk in cardiac patients. We aimed to determine the impact of three macrolides, azithromycin, clarithromycin and erythromycin, on cardiac electrophysiology, cardiac enzyme activities, histopathological changes, and sodium voltage-gated alpha subunit 5 (Nav1.5) channel expression. We used eight experimental groups of male albino rats: vehicle, azithromycin (100 mg/kg), clarithromycin (100 mg/kg), erythromycin (100 mg/kg), MI + vehicle, MI + azithromycin (100 mg/kg), MI + clarithromycin (100 mg/kg) and MI + erythromycin (100 mg/kg); each group received chronic oral doses of the vehicle/drugs for seven weeks. ECG abnormalities and elevated serum cardiac enzymes were observed particularly in rats with AMI compared to healthy rats. Microscopic examination revealed elevated pathology scores for rats treated with clarithromycin in both experiments following treatment with erythromycin in healthy rats. Although rats with MI did not show further elevations in fibrosis score on treatment with macrolides, they produced significant fibrosis in healthy rats. Downregulation of cardiac Nav1.5 transcript was observed following macrolides treatment in both groups (healthy rats and rats with MI). In conclusion, the current findings suggested the potential cardiotoxic effects of chronic doses of macrolide antibiotics in rats with MI as manifested by abnormal ECG changes and pathological findings in addition to downregulation of Nav1.5 channels. Furthermore, in the current dose ranges, azithromycin produced the least toxicity compared to clarithromycin and erythromycin.
Collapse
Affiliation(s)
- Noha E. Farag
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
- Department of Physiology, College of Medicine, Taif University, Taif 21974, Saudi Arabia
| | | | - Hussein M. Ismail
- Department of Cardiology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | | | - Eman A. Toraih
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA;
- Genetics Unit, Histology and Cell Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj 16278, Saudi Arabia;
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza 12613, Egypt
| | - Rehab M. Lashine
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt;
| | - Mohammed EL-dosoky
- Department of Neuroscience Technology, College of Applied Medical Science in Jubail, Imam Abdulrahman Bin Faisal University, Jubail 35816, Saudi Arabia;
| | - Sally Yussef Abed
- Department of Respiratory Care, College of Applied Medical Science in Jubail, Imam Abdulrahman Bin Faisal University, Jubail 35816, Saudi Arabia;
| | - Khalid M. Ibraheem
- Department of Anesthesia Technology, College of Applied Medical Sciences in Jubail, Imam Abdulrahman Bin Faisal University, Jubail 35816, Saudi Arabia;
| | - Manal S. Fawzy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 1321, Saudi Arabia
- Correspondence: (M.S.F.); or (S.A.Z.); Tel.: +20-1008584720 (M.S.F.); +20-1068916396 (S.A.Z.)
| | - Sawsan A. Zaitone
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 714, Saudi Arabia
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
- Correspondence: (M.S.F.); or (S.A.Z.); Tel.: +20-1008584720 (M.S.F.); +20-1068916396 (S.A.Z.)
| |
Collapse
|