1
|
Cao C, Yang L, Song J, Liu Z, Li H, Li L, Fu J, Liu J. Cardiomyocyte regeneration after infarction: changes, opportunities and challenges. Mol Cell Biochem 2025:10.1007/s11010-025-05251-w. [PMID: 40097887 DOI: 10.1007/s11010-025-05251-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/08/2025] [Indexed: 03/19/2025]
Abstract
Myocardial infarction is a cardiovascular disease that poses a serious threat to human health. The traditional view is that adult mammalian cardiomyocytes have almost no regenerative ability, but recent studies have shown that they have regenerative potential under specific conditions. This article comprehensively describes the research progress of post-infarction cardiomyocyte regeneration, including the characteristics of cardiomyocytes and post-infarction changes, regeneration mechanisms, influencing factors, potential therapeutic strategies, challenges and future development directions, and deeply discusses the specific pathways and targets included in the regeneration mechanism, aiming to provide new ideas and methods for the treatment of myocardial infarction.
Collapse
Affiliation(s)
- Ce Cao
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Lili Yang
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Jianshu Song
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Zixin Liu
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Haoran Li
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Lei Li
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Jianhua Fu
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China
| | - Jianxun Liu
- Beijing Key Laboratory of Chinese Materia Pharmacology, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, 100091, China.
| |
Collapse
|
2
|
Palmen R, Walton M, Wagner J. Pediatric flecainide pharmacogenomics: a roadmap to delivering precision-based care to pediatrics arrhythmias. Front Pharmacol 2024; 15:1477485. [PMID: 39741635 PMCID: PMC11686437 DOI: 10.3389/fphar.2024.1477485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025] Open
Abstract
Flecainide acetate is a Class 1c anti-arrhythmic with a potent sodium voltage gated channel blockade which is utilized for the second-line treatment of tachyarrhythmias in children and adults. Given its narrow therapeutic index, the individualization of drug therapy is of utmost importance for clinicians. Despite efforts to improve anti-arrhythmic drug therapy, there remain knowledge gaps regarding the impact of variation in the genes relevant to flecainide's disposition and response. This variability is compounded in developing children whose drug disposition and response pathways may remain immature. The purpose of this comprehensive review is to outline flecainide's disposition and response pathways while simultaneously highlighting opportunities for prospective investigation in the pediatric population.
Collapse
Affiliation(s)
- Ronald Palmen
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
| | - Mollie Walton
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
- Division of Cardiology, Kansas City, MO, United States
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy, Kansas City, MO, United States
| | - Jonathan Wagner
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
- Division of Cardiology, Kansas City, MO, United States
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy, Kansas City, MO, United States
| |
Collapse
|
3
|
Choi JH, Kim S, Kang OY, Choi SY, Hyun JY, Lee HS, Shin I. Selective fluorescent labeling of cellular proteins and its biological applications. Chem Soc Rev 2024; 53:9446-9489. [PMID: 39109465 DOI: 10.1039/d4cs00094c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Proteins, which are ubiquitous in cells and critical to almost all cellular functions, are indispensable for life. Fluorescence imaging of proteins is key to understanding their functions within their native milieu, as it provides insights into protein localization, dynamics, and trafficking in living systems. Consequently, the selective labeling of target proteins with fluorophores has emerged as a highly active research area, encompassing bioorganic chemistry, chemical biology, and cell biology. Various methods for selectively labeling proteins with fluorophores in cells and tissues have been established and are continually being developed to visualize and characterize proteins. This review highlights research findings reported since 2018, with a focus on the selective labeling of cellular proteins with small organic fluorophores and their biological applications in studying protein-associated biological events. We also discuss the strengths and weaknesses of each labeling approach for their utility in living systems.
Collapse
Affiliation(s)
- Joo Hee Choi
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| | - Sooin Kim
- Department of Chemistry, Sogang University, 04107 Seoul, Republic of Korea.
| | - On-Yu Kang
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
| | - Seong Yun Choi
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
- Pharmaceutical Chemistry, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Ji Young Hyun
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
- Pharmaceutical Chemistry, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Hyun Soo Lee
- Department of Chemistry, Sogang University, 04107 Seoul, Republic of Korea.
| | - Injae Shin
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| |
Collapse
|
4
|
Zhang Y, Chen H, Ma Q, Jia H, Ma H, Du Z, Liu Y, Zhang X, Zhang Y, Guan Y, Ma H. Electrophysiological Mechanism of Catestatin Antiarrhythmia: Enhancement of Ito, IK, and IK1 and Inhibition of ICa-L in Rat Ventricular Myocytes. J Am Heart Assoc 2024; 13:e035415. [PMID: 39158577 PMCID: PMC11963934 DOI: 10.1161/jaha.124.035415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/09/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Cardiovascular disease remains one of the leading causes of death globally. Myocardial ischemia and infarction, in particular, frequently cause disturbances in cardiac electrical activity that can trigger ventricular arrhythmias. We aimed to investigate whether catestatin, an endogenous catecholamine-inhibiting peptide, ameliorates myocardial ischemia-induced ventricular arrhythmias in rats and the underlying ionic mechanisms. METHODS AND RESULTS Adult male Sprague-Dawley rats were randomly divided into control and catestatin groups. Ventricular arrhythmias were induced by ligation of the left anterior descending coronary artery and electrical stimulation. Action potential, transient outward potassium current, delayed rectifier potassium current, inward rectifying potassium current, and L-type calcium current (ICa-L) of rat ventricular myocytes were recorded using a patch-clamp technique. Catestatin notably reduced ventricular arrhythmia caused by myocardial ischemia/reperfusion and electrical stimulation of rats. In ventricular myocytes, catestatin markedly shortened the action potential duration of ventricular myocytes, which was counteracted by potassium channel antagonists TEACl and 4-AP, and ICa-L current channel agonist Bay K8644. In addition, catestatin significantly increased transient outward potassium current, delayed rectifier potassium current, and inward rectifying potassium current density in a concentration-dependent manner. Catestatin accelerated the activation and decelerated the inactivation of the transient outward potassium current channel. Furthermore, catestatin decreased ICa-L current density in a concentration-dependent manner. Catestatin also accelerated the inactivation of the ICa-L channel and slowed down the recovery of ICa-L from inactivation. CONCLUSIONS Catestatin enhances the activity of transient outward potassium current, delayed rectifier potassium current, and inward rectifying potassium current, while suppressing the ICa-L in ventricular myocytes, leading to shortened action potential duration and ultimately reducing the ventricular arrhythmia in rats.
Collapse
MESH Headings
- Animals
- Male
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Rats, Sprague-Dawley
- Chromogranin A/pharmacology
- Chromogranin A/metabolism
- Action Potentials/drug effects
- Peptide Fragments/pharmacology
- Calcium Channels, L-Type/metabolism
- Calcium Channels, L-Type/drug effects
- Arrhythmias, Cardiac/physiopathology
- Arrhythmias, Cardiac/prevention & control
- Arrhythmias, Cardiac/metabolism
- Anti-Arrhythmia Agents/pharmacology
- Heart Ventricles/drug effects
- Heart Ventricles/metabolism
- Heart Ventricles/physiopathology
- Potassium Channels, Inwardly Rectifying/metabolism
- Potassium Channels, Inwardly Rectifying/drug effects
- Disease Models, Animal
- Potassium Channel Blockers/pharmacology
- Rats
- Patch-Clamp Techniques
- Delayed Rectifier Potassium Channels/metabolism
- Delayed Rectifier Potassium Channels/drug effects
- Potassium Channels/metabolism
- Potassium Channels/drug effects
Collapse
Affiliation(s)
- Ying Zhang
- Department of PhysiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Hua Chen
- Department of Cardiovascular Care UnitHebei General HospitalShijiazhuangHebeiChina
| | - Qingmin Ma
- Department of OphthalmologyHebei General HospitalShijiazhuangHebeiChina
| | - Hui Jia
- Department of PhysiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Hongyu Ma
- Department of PhysiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Zishuo Du
- Department of PhysiologyHebei Medical UniversityShijiazhuangHebeiChina
| | - Yan Liu
- Department of EndocrinologyThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Xiangjian Zhang
- Hebei Collaborative Innovation Center for Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
| | - Yi Zhang
- Department of PhysiologyHebei Medical UniversityShijiazhuangHebeiChina
- Hebei Collaborative Innovation Center for Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
| | - Yue Guan
- Department of PhysiologyHebei Medical UniversityShijiazhuangHebeiChina
- Hebei Collaborative Innovation Center for Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
| | - Huijie Ma
- Department of PhysiologyHebei Medical UniversityShijiazhuangHebeiChina
- The Key Laboratory of Neural and Vascular Biology, Ministry of EducationHebei Medical UniversityShijiazhuangHebeiChina
- Key Laboratory of Neurophysiology of Hebei ProvinceShijiazhuangHebeiChina
- Hebei Collaborative Innovation Center for Cardio‐Cerebrovascular DiseaseShijiazhuangHebeiChina
| |
Collapse
|
5
|
Ponce-Balbuena D, Tyrrell DJ, Cruz-Cortés C, Guerrero-Serna G, Da Rocha AM, Herron TJ, Song J, Raza DS, Anumonwo J, Goldstein DR, Espinoza-Fonseca LM. Paradoxical SERCA dysregulation contributes to atrial fibrillation in a model of diet-induced obesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606385. [PMID: 39149279 PMCID: PMC11326153 DOI: 10.1101/2024.08.02.606385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Obesity is a major risk factor for atrial fibrillation (AF) the most common serious cardiac arrhythmia, but the molecular mechanisms underlying diet-induced AF remain unclear. In this study, we subjected mice to a chronic high-fat diet and acute sympathetic activation ('two-hit' model) to study the mechanisms by which diet-induced obesity promotes AF. Surface electrocardiography revealed that diet-induced obesity and sympathetic activation synergize during intracardiac tachypacing to induce AF. At the cellular level, diet-induced obesity and acute adrenergic stimulation facilitate the formation of delayed afterdepolarizations in atrial myocytes, implicating altered Ca2+ dynamics as the underlying cause of AF. We found that diet-induced obesity does not alter the expression of major Ca2+-handling proteins in atria, including the sarcoplasmic reticulum Ca2+-ATPase (SERCA), a major component of beat-to-beat Ca2+ cycling in the heart. Paradoxically, obesity reduces phospholamban phosphorylation, suggesting decreased SERCA activity, yet atrial myocytes from obese mice showed a significantly increased Ca2+ transient amplitude and SERCA-mediated Ca2+ uptake. Adrenergic stimulation further increases the Ca2+ transient amplitude but does not affect Ca2+ reuptake in atrial myocytes from obese mice. Transcriptomics analysis showed that a high-fat diet prompts upregulation of neuronatin, a protein that has been implicated in obesity and is known to stimulate SERCA activity. We propose a mechanism in which obesity primes SERCA for paradoxical activation, and adrenergic stimulation facilitates AF conversion through a Ca2+-induced Ca2+ release gain in atrial myocytes. Overall, this study links obesity, altered Ca2+ signaling, and AF, and targeting this mechanism may prove effective for treating obesity-induced AF.
Collapse
Affiliation(s)
- Daniela Ponce-Balbuena
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Daniel J. Tyrrell
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Carlos Cruz-Cortés
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Guadalupe Guerrero-Serna
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Andre Monteiro Da Rocha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Todd J. Herron
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Jianrui Song
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Danyal S. Raza
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Justus Anumonwo
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Daniel R. Goldstein
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Microbiology and Immunology, University of Michigan, MI 48109, USA
| | - L. Michel Espinoza-Fonseca
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| |
Collapse
|
6
|
Hauwanga WN, Yau RCC, Goh KS, Castro Ceron JI, Alphonse B, Singh G, Elamin S, Jamched V, Abraham AA, Purvil J, Devan JN, Valentim G, McBenedict B, Lima Pessôa B, Mesquita ET. Management of Long QT Syndrome: A Systematic Review. Cureus 2024; 16:e62592. [PMID: 39027806 PMCID: PMC11257643 DOI: 10.7759/cureus.62592] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
Long QT syndrome (LQTS) is a cardiac disorder characterized by prolonged repolarization of the heart's electrical cycle, which can be observed as an extended QT interval on an electrocardiogram (ECG). The safe and effective management of LQTS often necessitates a multifaceted approach encompassing pharmacological treatment, lifestyle modifications, and, in high-risk cases, the implantation of implantable cardioverter-defibrillators (ICDs). Beta-blockers, particularly nadolol and propranolol, are foundational in treating LQTS, especially for high-risk patients, though ICDs are recommended for those with a history of cardiac arrest or recurrent arrhythmic episodes. Intermediate and low-risk patients are usually managed with medical therapy and regular monitoring. Lifestyle modifications, such as avoiding strenuous physical activities and certain medications, play a critical role. Additionally, psychological support is essential due to the anxiety and depression associated with LQTS. Left cardiac sympathetic denervation (LCSD) offers an alternative for those intolerant to beta-blockers or ICDs. For diagnosis and management, advancements in artificial intelligence (AI) are proving beneficial, enhancing early detection and risk stratification. Despite these developments, significant gaps in understanding the pathophysiology and optimal management strategies for LQTS remain. Future research should focus on refining risk stratification, developing new therapeutic approaches, and generating robust data to guide treatment decisions, ultimately aiming for a personalized medicine approach.
Collapse
Affiliation(s)
- Wilhelmina N Hauwanga
- Family Medicine, Faculty of Medicine, Federal University of the State of Rio de Janeiro, Rio de Janeiro, BRA
| | | | - Kang Suen Goh
- Internal Medicine, Monash University Malaysia, Johor Bahru, MYS
| | | | | | - Gurinder Singh
- Neurosurgery, Fluminense Federal University, Niterói, BRA
| | - Sara Elamin
- Neurosurgery, Fluminense Federal University, Niterói, BRA
| | | | | | - Joshi Purvil
- Neurosurgery, Fluminense Federal University, Niterói, BRA
| | - Jeshua N Devan
- Neurosurgery, Fluminense Federal University, Niterói, BRA
| | | | | | | | | |
Collapse
|
7
|
Pendyala VV, Pribil S, Schaal V, Sharma K, Jagadesan S, Yu L, Kumar V, Guda C, Gao L. Effects of Acute and Chronic Gabapentin Treatment on Cardiovascular Function of Rats. Cells 2023; 12:2705. [PMID: 38067133 PMCID: PMC10706228 DOI: 10.3390/cells12232705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Gabapentin (GBP), a GABA analogue, is primarily used as an anticonvulsant for the treatment of partial seizures and neuropathic pain. Whereas a majority of the side effects are associated with the nervous system, emerging evidence suggests there is a high risk of heart diseases in patients taking GBP. In the present study, we first used a preclinical model of rats to investigate, firstly, the acute cardiovascular responses to GBP (bolus i.v. injection, 50 mg/kg) and secondly the effects of chronic GBP treatment (i.p. 100 mg/kg/day × 7 days) on cardiovascular function and the myocardial proteome. Under isoflurane anesthesia, rat blood pressure (BP), heart rate (HR), and left ventricular (LV) hemodynamics were measured using Millar pressure transducers. The LV myocardium and brain cortex were analyzed by proteomics, bioinformatics, and western blot to explore the molecular mechanisms underlying GBP-induced cardiac dysfunction. In the first experiment, we found that i.v. GBP significantly decreased BP, HR, maximal LV pressure, and maximal and minimal dP/dt, whereas it increased IRP-AdP/dt, Tau, systolic, diastolic, and cycle durations (* p < 0.05 and ** p < 0.01 vs. baseline; n = 4). In the second experiment, we found that chronic GBP treatment resulted in hypotension, bradycardia, and LV systolic dysfunction, with no change in plasma norepinephrine. In the myocardium, we identified 109 differentially expressed proteins involved in calcium pathways, cholesterol metabolism, and galactose metabolism. Notably, we found that calmodulin, a key protein of intracellular calcium signaling, was significantly upregulated by GBP in the heart but not in the brain. In summary, we found that acute and chronic GBP treatments suppressed cardiovascular function in rats, which is attributed to abnormal calcium signaling in cardiomyocytes. These data reveal a novel side effect of GBP independent of the nervous system, providing important translational evidence to suggest that GBP can evoke adverse cardiovascular events by depression of myocardial function.
Collapse
Affiliation(s)
- Ved Vasishtha Pendyala
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA; (V.V.P.); (S.P.); (V.S.); (L.Y.)
| | - Sarah Pribil
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA; (V.V.P.); (S.P.); (V.S.); (L.Y.)
| | - Victoria Schaal
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA; (V.V.P.); (S.P.); (V.S.); (L.Y.)
| | - Kanika Sharma
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA; (K.S.); (V.K.)
| | - Sankarasubramanian Jagadesan
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA; (S.J.); (C.G.)
- Center for Biomedical Informatics Research and Innovation, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA
| | - Li Yu
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA; (V.V.P.); (S.P.); (V.S.); (L.Y.)
| | - Vikas Kumar
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA; (K.S.); (V.K.)
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA; (S.J.); (C.G.)
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA; (S.J.); (C.G.)
- Center for Biomedical Informatics Research and Innovation, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA
| | - Lie Gao
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA; (V.V.P.); (S.P.); (V.S.); (L.Y.)
| |
Collapse
|
8
|
Frosio A, Micaglio E, Polsinelli I, Calamaio S, Melgari D, Prevostini R, Ghiroldi A, Binda A, Carrera P, Villa M, Mastrocinque F, Presi S, Salerno R, Boccellino A, Anastasia L, Ciconte G, Ricagno S, Pappone C, Rivolta I. Unravelling Novel SCN5A Mutations Linked to Brugada Syndrome: Functional, Structural, and Genetic Insights. Int J Mol Sci 2023; 24:15089. [PMID: 37894777 PMCID: PMC10606416 DOI: 10.3390/ijms242015089] [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: 09/08/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Brugada Syndrome (BrS) is a rare inherited cardiac arrhythmia causing potentially fatal ventricular tachycardia or fibrillation, mainly occurring during rest or sleep in young individuals without heart structural issues. It increases the risk of sudden cardiac death, and its characteristic feature is an abnormal ST segment elevation on the ECG. While BrS has diverse genetic origins, a subset of cases can be conducted to mutations in the SCN5A gene, which encodes for the Nav1.5 sodium channel. Our study focused on three novel SCN5A mutations (p.A344S, p.N347K, and p.D349N) found in unrelated BrS families. Using patch clamp experiments, we found that these mutations disrupted sodium currents: p.A344S reduced current density, while p.N347K and p.D349N completely abolished it, leading to altered voltage dependence and inactivation kinetics when co-expressed with normal channels. We also explored the effects of mexiletine treatment, which can modulate ion channel function. Interestingly, the p.N347K and p.D349N mutations responded well to the treatment, rescuing the current density, while p.A344S showed a limited response. Structural analysis revealed these mutations were positioned in key regions of the channel, impacting its stability and function. This research deepens our understanding of BrS by uncovering the complex relationship between genetic mutations, ion channel behavior, and potential therapeutic interventions.
Collapse
Affiliation(s)
- Anthony Frosio
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
| | - Emanuele Micaglio
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- Arrhythmia and Electrophysiology Department, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (F.M.); (A.B.)
| | - Ivan Polsinelli
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
| | - Serena Calamaio
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
| | - Dario Melgari
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
| | - Rachele Prevostini
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
| | - Andrea Ghiroldi
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
| | - Anna Binda
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore, 48, 20900 Monza, Italy;
| | - Paola Carrera
- Laboratory of Clinical Molecular Genetics and Cytogenetics, Unit of Genomics for Diagnosis of Human Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (P.C.); (S.P.)
| | - Marco Villa
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
| | - Flavio Mastrocinque
- Arrhythmia and Electrophysiology Department, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (F.M.); (A.B.)
| | - Silvia Presi
- Laboratory of Clinical Molecular Genetics and Cytogenetics, Unit of Genomics for Diagnosis of Human Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (P.C.); (S.P.)
| | - Raffaele Salerno
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy;
| | - Antonio Boccellino
- Arrhythmia and Electrophysiology Department, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (F.M.); (A.B.)
| | - Luigi Anastasia
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy;
| | - Giuseppe Ciconte
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- Arrhythmia and Electrophysiology Department, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (F.M.); (A.B.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy;
| | - Stefano Ricagno
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- Department of Biosciences, Università degli Studi di Milano, Via Celoria, 26, 20133 Milan, Italy
| | - Carlo Pappone
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- Arrhythmia and Electrophysiology Department, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (F.M.); (A.B.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy;
| | - Ilaria Rivolta
- Institute of Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy; (A.F.); (E.M.); (I.P.); (S.C.); (D.M.); (R.P.); (A.G.); (M.V.); (L.A.); (G.C.); (S.R.); (C.P.)
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore, 48, 20900 Monza, Italy;
| |
Collapse
|
9
|
Ma J, Wang NY, Jagani R, Wang HS. Proarrhythmic toxicity of low dose bisphenol A and its analogs in human iPSC-derived cardiomyocytes and human cardiac organoids through delay of cardiac repolarization. CHEMOSPHERE 2023; 328:138562. [PMID: 37004823 PMCID: PMC10121900 DOI: 10.1016/j.chemosphere.2023.138562] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/07/2023] [Accepted: 03/31/2023] [Indexed: 06/19/2023]
Abstract
Bisphenol A (BPA) and its analogs are common environmental chemicals with many potential adverse health effects. The impact of environmentally relevant low dose BPA on human heart, including cardiac electrical properties, is not understood. Perturbation of cardiac electrical properties is a key arrhythmogenic mechanism. In particular, delay of cardiac repolarization can cause ectopic excitation of cardiomyocytes and malignant arrhythmia. This can occur as a result of genetic mutations (i.e., long QT (LQT) syndrome), or cardiotoxicity of drugs and environmental chemicals. To define the impact of low dose BPA on electrical properties of cardiomyocytes in a human-relevant model system, we examined the rapid effects of 1 nM BPA in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) using patch-clamp and confocal fluorescence imaging. Acute exposure to BPA delayed repolarization and prolonged action potential duration (APD) in hiPSC-CMs through inhibition of the hERG K+ channel. In nodal-like hiPSC-CMs, BPA acutely increased pacing rate through stimulation of the If pacemaker channel. Existing arrhythmia susceptibility determines the response of hiPSC-CMs to BPA. BPA resulted in modest APD prolongation but no ectopic excitation in baseline condition, while rapidly promoted aberrant excitations and tachycardia-like events in myocytes that had drug-simulated LQT phenotype. In hiPSC-CM-based human cardiac organoids, the effects of BPA on APD and aberrant excitation were shared by its analog chemicals, which are often used in "BPA-free" products, with bisphenol AF having the largest effects. Our results reveal that BPA and its analogs have repolarization delay-associated pro-arrhythmic toxicity in human cardiomyocytes, particularly in myocytes that are prone to arrhythmias. The toxicity of these chemicals depends on existing pathophysiological conditions of the heart, and may be particularly pronounced in susceptible individuals. An individualized approach is needed in risk assessment and protection.
Collapse
Affiliation(s)
- Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | | | - Ravikumar Jagani
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hong-Sheng Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
10
|
Aweimer A, Mügge A, Akin I, El-Battrawy I. [Asymptomatic channelopathies : Risk stratification and primary prophylaxis]. Herzschrittmacherther Elektrophysiol 2023; 34:101-108. [PMID: 37103573 DOI: 10.1007/s00399-023-00937-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 02/19/2023] [Indexed: 04/28/2023]
Abstract
In general, asymptomatic patients with channelopathies are at increased risk of sudden cardiac death (SCD), due to pathogenic variants in genes encoding ion channels that result in pathological ion currents. Channelopathies include long-QT syndrome (LQTS), Brugada syndrome (BrS), catecholaminergic polymorphic ventricular tachycardia (CPVT), and short-QT syndrome (SQTS). In addition to the patient's clinical presentation, history and clinical tests, the main diagnostic tools are electrocardiography and genetic testing to identify known gene mutations. Early and correct diagnosis as well as further risk stratification of affected individuals and their relatives are paramount for prognosis. The recent availability of risk score calculators for LQTS and BrS allows SCD risk to be accurately estimated. The extent to which these improve patient selection for treatment with an implantable cardioverter-defibrillator (ICD) system is currently unknown. In most cases, initiation of basic therapy in asymptomatic patients in the form of avoidance of triggers, which are usually medication or stressful situations, is sufficient and contributes to risk reduction. In addition, there are other risk-reducing prophylactic measures, such as permanent medication with nonselective β‑ blockers (for LQTS and CPVT) or mexiletine for LQTS3. Patients and their family members should be referred to specialized outpatient clinics for individual risk stratification in the sense of primary prophylaxis.
Collapse
Affiliation(s)
- Assem Aweimer
- Klinik für Kardiologie und Angiologie, Berufsgenossenschaftliches Universitätsklinikum Bergmannsheil Bochum, Ruhr Universität Bochum, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Deutschland.
| | - Andreas Mügge
- Klinik für Kardiologie und Angiologie, Berufsgenossenschaftliches Universitätsklinikum Bergmannsheil Bochum, Ruhr Universität Bochum, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Deutschland
| | - Ibrahim Akin
- I. Medizinische Klinik, Universitätsklinikum Mannheim, Medizinische Fakultät Mannheim, Universität Heidelberg, Mannheim, Deutschland
| | - Ibrahim El-Battrawy
- Klinik für Kardiologie und Angiologie, Berufsgenossenschaftliches Universitätsklinikum Bergmannsheil Bochum, Ruhr Universität Bochum, Bürkle-de-la-Camp-Platz 1, 44789, Bochum, Deutschland
| |
Collapse
|
11
|
Ignatova I, Frolov R, Nymark S. The retinal pigment epithelium displays electrical excitability and lateral signal spreading. BMC Biol 2023; 21:84. [PMID: 37069561 PMCID: PMC10111697 DOI: 10.1186/s12915-023-01559-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 03/10/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND The non-neuronal retinal pigment epithelium (RPE) functions in intimate association with retinal photoreceptors, performing a multitude of tasks critical for maintaining retinal homeostasis and collaborating with retinal glial cells to provide metabolic support and ionic buffering. Accordingly, the RPE has recently been shown to display dynamic properties mediated by an array of ion channels usually more characteristic of astrocytes and excitable cells. The recent discovery of canonical voltage-activated Na+ channels in the RPE and their importance for phagocytosis of photoreceptor outer segments raises a question about their electrogenic function. Here, we performed a detailed electrophysiological analysis related to the functioning of these channels in human embryonic stem cell (hESC)-derived RPE. RESULTS Our studies examining the electrical properties of the hESC-RPE revealed that its membrane mainly displays passive properties in a broad voltage range, with the exception of depolarization-induced spikes caused by voltage-activated Na+ current (INa). Spike amplitude depended on the availability of INa and spike kinetics on the membrane time constant, and the spikes could be largely suppressed by TTX. Membrane resistance fluctuated rapidly and strongly, repeatedly changing over the course of recordings and causing closely correlated fluctuations in resting membrane potential. In a minority of cells, we found delayed secondary INa-like inward currents characterized by comparatively small amplitudes and slow kinetics, which produced secondary depolarizing spikes. Up to three consecutive delayed inward current waves were detected. These currents could be rapidly and reversibly augmented by applying L-type Ca2+ channel blocker nifedipine to diminish influx of calcium and thus increase gap junctional conductance. CONCLUSIONS This work shows, for the first time, that INa and INa-mediated voltage spikes can spread laterally through gap junctions in the monolayer of cells that are traditionally considered non-excitable. Our findings support a potential role of the RPE that goes beyond giving homeostatic support to the retina.
Collapse
Affiliation(s)
- Irina Ignatova
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Soile Nymark
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
12
|
Yan Z, Zhong L, Zhu W, Chung SK, Hou P. Chinese herbal medicine for the treatment of cardiovascular diseases ─ targeting cardiac ion channels. Pharmacol Res 2023; 192:106765. [PMID: 37075871 DOI: 10.1016/j.phrs.2023.106765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of morbidity and mortality, imposing an increasing global health burden. Cardiac ion channels (voltage-gated NaV, CaV, KVs, and others) synergistically shape the cardiac action potential (AP) and control the heartbeat. Dysfunction of these channels, due to genetic mutations, transcriptional or post-translational modifications, may disturb the AP and lead to arrhythmia, a major risk for CVD patients. Although there are five classes of anti-arrhythmic drugs available, they can have varying levels of efficacies and side effects on patients, possibly due to the complex pathogenesis of arrhythmias. As an alternative treatment option, Chinese herbal remedies have shown promise in regulating cardiac ion channels and providing anti-arrhythmic effects. In this review, we first discuss the role of cardiac ion channels in maintaining normal heart function and the pathogenesis of CVD, then summarize the classification of Chinese herbal compounds, and elaborate detailed mechanisms of their efficacy in regulating cardiac ion channels and in alleviating arrhythmia and CVD. We also address current limitations and opportunities for developing new anti-CVD drugs based on Chinese herbal medicines.
Collapse
Affiliation(s)
- Zhenzhen Yan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Ling Zhong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Wandi Zhu
- Cardiovascular Medicine Division and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Sookja Kim Chung
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China; Faculty of Medicine & Faculty of Innovation Engineering at Macau University of Science and Technology, Taipa, Macao SAR, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Panpan Hou
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China; Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute. Zhuhai, Guangdong, China.
| |
Collapse
|
13
|
Abstract
The global burden caused by cardiovascular disease is substantial, with heart disease representing the most common cause of death around the world. There remains a need to develop better mechanistic models of cardiac function in order to combat this health concern. Heart rhythm disorders, or arrhythmias, are one particular type of disease which has been amenable to quantitative investigation. Here we review the application of quantitative methodologies to explore dynamical questions pertaining to arrhythmias. We begin by describing single-cell models of cardiac myocytes, from which two and three dimensional models can be constructed. Special focus is placed on results relating to pattern formation across these spatially-distributed systems, especially the formation of spiral waves of activation. Next, we discuss mechanisms which can lead to the initiation of arrhythmias, focusing on the dynamical state of spatially discordant alternans, and outline proposed mechanisms perpetuating arrhythmias such as fibrillation. We then review experimental and clinical results related to the spatio-temporal mapping of heart rhythm disorders. Finally, we describe treatment options for heart rhythm disorders and demonstrate how statistical physics tools can provide insights into the dynamics of heart rhythm disorders.
Collapse
Affiliation(s)
- Wouter-Jan Rappel
- Department of Physics, University of California San Diego, La Jolla, CA 92037
| |
Collapse
|
14
|
Research Progress on Natural Products’ Therapeutic Effects on Atrial Fibrillation by Regulating Ion Channels. Cardiovasc Ther 2022; 2022:4559809. [PMID: 35387267 PMCID: PMC8964196 DOI: 10.1155/2022/4559809] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/28/2022] [Accepted: 03/03/2022] [Indexed: 11/18/2022] Open
Abstract
Antiarrhythmic drugs (AADs) have a therapeutic effect on atrial fibrillation (AF) by regulating the function of ion channels. However, several adverse effects and high recurrence rates after drug withdrawal seriously affect patients’ medication compliance and clinical prognosis. Thus, safer and more effective drugs are urgently needed. Active components extracted from natural products are potential choices for AF therapy. Natural products like Panax notoginseng (Burk.) F.H. Chen, Sophora flavescens Ait., Stephania tetrandra S. Moore., Pueraria lobata (Willd.) Ohwi var. thomsonii (Benth.) Vaniot der Maesen., and Coptis chinensis Franch. have a long history in the treatment of arrhythmia, myocardial infarction, stroke, and heart failure in China. Based on the classification of chemical structures, this article discussed the natural product components’ therapeutic effects on atrial fibrillation by regulating ion channels, connexins, and expression of related genes, in order to provide a reference for development of therapeutic drugs for atrial fibrillation.
Collapse
|
15
|
Abstract
Long QT syndrome (LQTS) is a cardiovascular disorder characterized by an abnormality in cardiac repolarization leading to a prolonged QT interval and T-wave irregularities on the surface electrocardiogram. It is commonly associated with syncope, seizures, susceptibility to torsades de pointes, and risk for sudden death. LQTS is a rare genetic disorder and a major preventable cause of sudden cardiac death in the young. The availability of therapy for this lethal disease emphasizes the importance of early and accurate diagnosis. Additionally, understanding of the molecular mechanisms underlying LQTS could help to optimize genotype-specific treatments to prevent deaths in LQTS patients. In this review, we briefly summarize current knowledge regarding molecular underpinning of LQTS, in particular focusing on LQT1, LQT2, and LQT3, and discuss novel strategies to study ion channel dysfunction and drug-specific therapies in LQT1, LQT2, and LQT3 syndromes.
Collapse
Affiliation(s)
| | - Isabelle Deschênes
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
16
|
Ozturk N, Uslu S, Ozdemir S. Diabetes-induced changes in cardiac voltage-gated ion channels. World J Diabetes 2021; 12:1-18. [PMID: 33520105 PMCID: PMC7807254 DOI: 10.4239/wjd.v12.i1.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/05/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus affects the heart through various mechanisms such as microvascular defects, metabolic abnormalities, autonomic dysfunction and incompatible immune response. Furthermore, it can also cause functional and structural changes in the myocardium by a disease known as diabetic cardiomyopathy (DCM) in the absence of coronary artery disease. As DCM progresses it causes electrical remodeling of the heart, left ventricular dysfunction and heart failure. Electrophysiological changes in the diabetic heart contribute significantly to the incidence of arrhythmias and sudden cardiac death in diabetes mellitus patients. In recent studies, significant changes in repolarizing K+ currents, Na+ currents and L-type Ca2+ currents along with impaired Ca2+ homeostasis and defective contractile function have been identified in the diabetic heart. In addition, insulin levels and other trophic factors change significantly to maintain the ionic channel expression in diabetic patients. There are many diagnostic tools and management options for DCM, but it is difficult to detect its development and to effectively prevent its progress. In this review, diabetes-associated alterations in voltage-sensitive cardiac ion channels are comprehensively assessed to understand their potential role in the pathophysiology and pathogenesis of DCM.
Collapse
Affiliation(s)
- Nihal Ozturk
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| | - Serkan Uslu
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| | - Semir Ozdemir
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya 07058, Turkey
| |
Collapse
|
17
|
Kuo MW, Tsai HH, Wang SH, Chen YY, Yu AL, Yu J. Yulink, predicted from evolutionary analysis, is involved in cardiac function. J Biomed Sci 2021; 28:7. [PMID: 33423678 PMCID: PMC7798328 DOI: 10.1186/s12929-020-00701-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 12/21/2020] [Indexed: 11/11/2022] Open
Abstract
Background The comparative evolutionary genomics analysis was used to study the functions of novel Ka/Ks-predicted human exons in a zebrafish model. The Yulink (MIOS, Entrez Gene: 54,468), a conserved gene from zebrafish to human with WD40 repeats at N-terminus, was identified and found to encode an 875 amino acid in human. The biological function of this Yulink gene in cardiomyocytes remains unexplored. The purpose of this study is to determine the involvement of Yulink in the functions of cardiomyocytes and to investigate its molecular regulatory mechanism. Methods Knockdown of Yulink was performed using morpholino or shRNA in zebrafish, mouse HL-1 cardiomyocytes, and human iPSC-derived cardiomyocytes. The expression levels of mRNA and protein were quantified by qPCR and western blots. Other methods including DNA binding, ligand uptake, agonists treatment and Ca2+ imaging assays were used to study the molecular regulatory mechanism by Yulink. Statistical data were shown as mean ± SD or mean ± standard error. Results The knockdown of yulink with three specific morpholinos in zebrafish resulted in cardiac dysfunctions with pericardial edema, decreased heart beats and cardiac output. The Yulink knockdown in mouse HL-1 cardiomyocytes disrupted Ca2+ cycling, reduced DNA binding activity of PPARγ (peroxisome proliferator-activated receptor gamma) and resulted in a reduction of Serca2 (sarcoplasmic reticulum Ca2+ ATPase 2) expression. Expression of Serca2 was up-regulated by PPARγ agonists and down-regulated by PPARγ-shRNA knockdown, suggesting that Yulink regulates SERCA2 expression through PPARγ in mouse HL-1 cardiomyocytes. On the other hand, YULINK, PPARγ or SERCA2 over-expression rescued the phenotypes of Yulink KD cells. In addition, knockdown of YULINK in human iPSC-derived cardiomyocytes also disrupted Ca2+ cycling via decreased SERCA2 expression. Conclusions Overall, our data showed that Yulink is an evolutionarily conserved gene from zebrafish to human. Mechanistically Yulink regulated Serca2 expression in cardiomyocytes, presumably mediated through PPARγ nuclear entry. Deficiency of Yulink in mouse and human cardiomyocytes resulted in irregular Ca2+ cycling, which may contribute to arrhythmogenesis.
Collapse
Affiliation(s)
- Ming-Wei Kuo
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Hsiu-Hui Tsai
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Sheng-Hung Wang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Yi-Yin Chen
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan.,Department of Pediatrics, University of California, San Diego, CA, USA
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan. .,Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
18
|
Established and Emerging Mechanisms in the Pathogenesis of Arrhythmogenic Cardiomyopathy: A Multifaceted Disease. Int J Mol Sci 2020; 21:ijms21176320. [PMID: 32878278 PMCID: PMC7503882 DOI: 10.3390/ijms21176320] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/13/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a heritable myocardial disease that manifests with cardiac arrhythmias, syncope, sudden cardiac death, and heart failure in the advanced stages. The pathological hallmark of ACM is a gradual replacement of the myocardium by fibroadiposis, which typically starts from the epicardium. Molecular genetic studies have identified causal mutations predominantly in genes encoding for desmosomal proteins; however, non-desmosomal causal mutations have also been described, including genes coding for nuclear proteins, cytoskeleton componentsand proteins involved in excitation-contraction coupling. Despite the poor prognosis, currently available treatments can only partially control symptoms and to date there is no effective therapy for ACM. Inhibition of the canonical Wnt/β-catenin pathway and activation of the Hippo and the TGF-β pathways have been implicated in the pathogenesis of ACM. Yet, our understanding of the molecular mechanisms involved in the development of the disease and the cell source of fibroadiposis remains incomplete. Elucidation of the pathogenesis of the disease could facilitate targeted approaches for treatment. In this manuscript we will provide a comprehensive review of the proposed molecular and cellular mechanisms of the pathogenesis of ACM, including the emerging evidence on abnormal calcium homeostasis and inflammatory/autoimmune response. Moreover, we will propose novel hypothesis about the role of epicardial cells and paracrine factors in the development of the phenotype. Finally, we will discuss potential innovative therapeutic approaches based on the growing knowledge in the field.
Collapse
|
19
|
Russo V, Rago A, Carbone A, Bottino R, Ammendola E, Della Cioppa N, Galante D, Golino P, Nigro G. Atrial Fibrillation in COVID-19: From Epidemiological Association to Pharmacological Implications. J Cardiovasc Pharmacol 2020; 76:138-145. [PMID: 32453074 DOI: 10.1097/fjc.0000000000000854] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Coronavirus disease 2019 (COVID-19) outbreak is a public health emergency of international concerns because of a highly pathogenic human coronavirus (HCoV), actually named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Despite much emerging data about the epidemiological association between cardiovascular diseases and COVID-19, little is still known about atrial fibrillation and its optimal management in this clinical contest. The aim of our review is to describe the pharmacological interactions between cardiovascular drugs more commonly used in atrial fibrillation management and experimental COVID-19 therapies, based on EU and US summaries of product characteristics.
Collapse
Affiliation(s)
- Vincenzo Russo
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Monaldi Hospital, Naples, Italy
| | | | - Andreina Carbone
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Monaldi Hospital, Naples, Italy
| | - Roberta Bottino
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Monaldi Hospital, Naples, Italy
| | | | | | - Dario Galante
- Intensive Care Unit-Anesthesiology and Reanimation, Tatarella Hospital, Foggia, Italy
| | - Paolo Golino
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Monaldi Hospital, Naples, Italy
| | - Gerardo Nigro
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli," Monaldi Hospital, Naples, Italy
| |
Collapse
|
20
|
Whittaker DG, Clerx M, Lei CL, Christini DJ, Mirams GR. Calibration of ionic and cellular cardiac electrophysiology models. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1482. [PMID: 32084308 PMCID: PMC8614115 DOI: 10.1002/wsbm.1482] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 12/30/2022]
Abstract
Cardiac electrophysiology models are among the most mature and well-studied mathematical models of biological systems. This maturity is bringing new challenges as models are being used increasingly to make quantitative rather than qualitative predictions. As such, calibrating the parameters within ion current and action potential (AP) models to experimental data sets is a crucial step in constructing a predictive model. This review highlights some of the fundamental concepts in cardiac model calibration and is intended to be readily understood by computational and mathematical modelers working in other fields of biology. We discuss the classic and latest approaches to calibration in the electrophysiology field, at both the ion channel and cellular AP scales. We end with a discussion of the many challenges that work to date has raised and the need for reproducible descriptions of the calibration process to enable models to be recalibrated to new data sets and built upon for new studies. This article is categorized under: Analytical and Computational Methods > Computational Methods Physiology > Mammalian Physiology in Health and Disease Models of Systems Properties and Processes > Cellular Models.
Collapse
Affiliation(s)
- Dominic G. Whittaker
- Centre for Mathematical Medicine & Biology, School of Mathematical SciencesUniversity of NottinghamNottinghamUK
| | - Michael Clerx
- Computational Biology & Health Informatics, Department of Computer ScienceUniversity of OxfordOxfordUK
| | - Chon Lok Lei
- Computational Biology & Health Informatics, Department of Computer ScienceUniversity of OxfordOxfordUK
| | | | - Gary R. Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical SciencesUniversity of NottinghamNottinghamUK
| |
Collapse
|
21
|
Sizemore G, McLaughlin S, Newman M, Brundage K, Ammer A, Martin K, Pugacheva E, Coad J, Mattes MD, Yu HG. Opening large-conductance potassium channels selectively induced cell death of triple-negative breast cancer. BMC Cancer 2020; 20:595. [PMID: 32586284 PMCID: PMC7318490 DOI: 10.1186/s12885-020-07071-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/15/2020] [Indexed: 12/15/2022] Open
Abstract
Background Unlike other breast cancer subtypes that may be treated with a variety of hormonal or targeted therapies, there is a need to identify new, effective targets for triple-negative breast cancer (TNBC). It has recently been recognized that membrane potential is depolarized in breast cancer cells. The primary objective of the study is to explore whether hyperpolarization induced by opening potassium channels may provide a new strategy for treatment of TNBC. Methods Breast cancer datasets in cBioPortal for cancer genomics was used to search for ion channel gene expression. Immunoblots and immunohistochemistry were used for protein expression in culture cells and in the patient tissues. Electrophysiological patch clamp techniques were used to study properties of BK channels in culture cells. Flow cytometry and fluorescence microscope were used for cell viability and cell cycle studies. Ultrasound imaging was used to study xenograft in female NSG mice. Results In large datasets of breast cancer patients, we identified a gene, KCNMA1 (encoding for a voltage- and calcium-dependent large-conductance potassium channel, called BK channel), overexpressed in triple-negative breast cancer patients. Although overexpressed, 99% of channels are closed in TNBC cells. Opening BK channels hyperpolarized membrane potential, which induced cell cycle arrest in G2 phase and apoptosis via caspase-3 activation. In a TNBC cell induced xenograft model, treatment with a BK channel opener significantly slowed tumor growth without cardiac toxicity. Conclusions Our results support the idea that hyperpolarization induced by opening BK channel in TNBC cells can become a new strategy for development of a targeted therapy in TNBC.
Collapse
Affiliation(s)
- Gina Sizemore
- Clinical and Translational Sciences Institute, West Virginia University, Morgantown, USA
| | - Sarah McLaughlin
- Animal Models & Imaging Facility, Cancer Institute, West Virginia University, Morgantown, USA
| | - Mackenzie Newman
- Department of Physiology & Pharmacology, West Virginia University, Morgantown, WV, 26506, USA
| | - Kathleen Brundage
- Department of Microbiology and Cell Biology, Flow Cytometry Facility, West Virginia University, Morgantown, USA
| | - Amanda Ammer
- Animal Models & Imaging Facility, Cancer Institute, West Virginia University, Morgantown, USA
| | - Karen Martin
- Animal Models & Imaging Facility, Cancer Institute, West Virginia University, Morgantown, USA
| | - Elena Pugacheva
- Department of Biochemistry, Cancer Institute, West Virginia University, Morgantown, USA
| | - James Coad
- Department of Pathology, West Virginia University, Morgantown, USA
| | - Malcolm D Mattes
- Department of Radiation Oncology, Cancer Institute, West Virginia University, Morgantown, USA
| | - Han-Gang Yu
- Department of Physiology & Pharmacology, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
22
|
Fouda MA, Ghovanloo MR, Ruben PC. Cannabidiol protects against high glucose-induced oxidative stress and cytotoxicity in cardiac voltage-gated sodium channels. Br J Pharmacol 2020; 177:2932-2946. [PMID: 32077098 DOI: 10.1111/bph.15020] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/23/2019] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Cardiovascular complications are the major cause of mortality in diabetic patients. However, the molecular mechanisms underlying diabetes-associated arrhythmias are unclear. We hypothesized that high glucose could adversely affect Nav 1.5, the major cardiac sodium channel isoform of the heart, at least partially via oxidative stress. We further hypothesized that cannabidiol (CBD), one of the main constituents of Cannabis sativa, through its effects on Nav 1.5, could protect against high glucose-elicited oxidative stress and cytotoxicity. EXPERIMENTAL APPROACH To test these ideas, we used CHO cells transiently co-transfected with cDNA encoding human Nav 1.5 α-subunit under control and high glucose conditions (50 or 100 mM for 24 hr). Several experimental and computational techniques were used, including voltage clamp of heterologous expression systems, cell viability assays, fluorescence assays and action potential modelling. KEY RESULTS High glucose evoked cell death associated with elevation in reactive oxygen species (ROS) right shifted the voltage dependence of conductance and steady-state fast inactivation, and increased persistent current leading to computational prolongation of action potential (hyperexcitability) which could result in long QT3 arrhythmia. CBD mitigated all the deleterious effects provoked by high glucose. Perfusion with lidocaine (a well-known sodium channel inhibitor with antioxidant effects) or co-incubation of Tempol (a well-known antioxidant) elicited protection, comparable to CBD, against the deleterious effects of high glucose. CONCLUSION AND IMPLICATIONS These findings suggest that, through its favourable antioxidant and sodium channel inhibitory effects, CBD may protect against high glucose-induced arrhythmia and cytotoxicity.
Collapse
Affiliation(s)
- Mohamed A Fouda
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada.,Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt
| | - Mohammad-Reza Ghovanloo
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
23
|
Newly Discovered Action of HpTx3 from Venom of Heteropoda venatoria on Na v1.7 and Its Pharmacological Implications in Analgesia. Toxins (Basel) 2019; 11:toxins11120680. [PMID: 31757020 PMCID: PMC6950750 DOI: 10.3390/toxins11120680] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
It has been reported that Heteropodatoxin3 (HpTx3), a peptidic neurotoxin purified from the venom of the spider species Heteropoda venatoria, could inhibit Kv4.2 channels. Our present study newly found that HpTx3 also has potent and selective inhibitory action on Nav1.7, with an IC50 of 135.61 ± 12.98 nM. Without effect on the current–voltage (I-V) relationship of Nav1.7, HpTx3 made minor alternation in the voltage-dependence of activation and steady-state inactivation of Nav1.7 (4.15 mV and 7.29 mV, respectively) by interacting with the extracellular S3–S4 loop (S3b–S4 sequence) in domain II and the domain IV of the Nav channel subtype, showing the characteristics of both pore blocker and gate modifier toxin. During the interaction of HpTx3 with the S3b–S4 sequence of Nav1.7, the amino acid residue D in the sequence played a key role. When administered intraperitoneally or intramuscularly, HpTx3 displayed potent analgesic activity in a dose-dependent manner in different mouse pain models induced by formalin, acetic acid, complete Freund’s adjuvant, hot plate, or spared nerve injury, demonstrating that acute, inflammatory, and neuropathic pains were all effectively inhibited by the toxin. In most cases HpTx3 at doses of ≥ 1mg/kg could produce the analgesic effect comparable to that of 1 mg/kg morphine. These results suggest that HpTx3 not only can be used as a molecular probe to investigate ion channel function and pain mechanism, but also has potential in the development of the drugs that treat the Nav1.7 channel-related pain.
Collapse
|
24
|
Freundt JK, Frommeyer G, Spieker T, Wötzel F, Grotthoff JS, Stypmann J, Hempel G, Schäfers M, Jacobs AH, Eckardt L, Lange PS. Histone deacetylase inhibition by Entinostat for the prevention of electrical and structural remodeling in heart failure. BMC Pharmacol Toxicol 2019; 20:16. [PMID: 30841920 PMCID: PMC6404297 DOI: 10.1186/s40360-019-0294-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/26/2019] [Indexed: 12/14/2022] Open
Abstract
Background The development of heart failure is accompanied by complex changes in cardiac electrophysiology and functional properties of cardiomyocytes and fibroblasts. Histone deacetylase (HDAC) inhibitors hold great promise for the pharmaceutical therapy of several malignant diseases. Here, we describe novel effects of the class I HDAC inhibitor Entinostat on electrical and structural remodeling in an in vivo model of pacing induced heart failure. Methods Rabbits were implanted a pacemaker system, subjected to rapid ventricular pacing and treated with Entinostat or placebo, respectively. Following stimulation, rabbit hearts were explanted and subsequently subjected to electrophysiological studies and further immunohistological analyses of left ventricles. Results In vivo, rapid ventricular stimulation caused a significant prolongation of monophasic action potential duration compared to sham hearts (from 173 ± 26 ms to 250 ± 41 ms; cycle length 900 ms; p < 0.05) and an increased incidence of Early afterdepolarisations (+ 150%), while treatment with Entinostat in failing hearts could partially prevent this effect (from 250 ± 41 ms to 170 ± 53 ms, p < 0.05; reduction in EAD by 50%). Entinostat treatment partially restored KCNH2 and Cav1.3 gene expressions in failing hearts, and inhibited the development of cardiac fibrosis in vivo. Conclusion In a rabbit model of heart failure, Entinostat diminishes heart failure related prolongation of repolarization and partially restores KCNH2 and Cav1.3 expression. In addition, Entinostat exerts antifibrotic properties both in vitro and in vivo. Thus, Entinostat might be an interesting candidate for the pharmaceutical therapy of heart failure directed against structural and electrical remodeling. Electronic supplementary material The online version of this article (10.1186/s40360-019-0294-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Johanna K Freundt
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Gerrit Frommeyer
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Tilmann Spieker
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Fabian Wötzel
- Department of Pathology, University Hospital Münster, Münster, Germany
| | | | - Jörg Stypmann
- Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany
| | - Georg Hempel
- Institute for Pharmaceutical and Medical Chemistry, University of Münster, Münster, Germany
| | - Michael Schäfers
- European Institute for Molecular Imaging, University Hospital Münster, Münster, Germany.,Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Andreas H Jacobs
- European Institute for Molecular Imaging, University Hospital Münster, Münster, Germany
| | - Lars Eckardt
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany
| | - Philipp S Lange
- Department of Cardiology II: Electrophysiology, University Hospital Münster, Münster, Germany.
| |
Collapse
|
25
|
Frasier CR, Zhang H, Offord J, Dang LT, Auerbach DS, Shi H, Chen C, Goldman AM, Eckhardt LL, Bezzerides VJ, Parent JM, Isom LL. Channelopathy as a SUDEP Biomarker in Dravet Syndrome Patient-Derived Cardiac Myocytes. Stem Cell Reports 2018; 11:626-634. [PMID: 30146492 PMCID: PMC6135724 DOI: 10.1016/j.stemcr.2018.07.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 07/23/2018] [Accepted: 07/26/2018] [Indexed: 12/31/2022] Open
Abstract
Dravet syndrome (DS) is a severe developmental and epileptic encephalopathy with a high incidence of sudden unexpected death in epilepsy (SUDEP). Most DS patients carry de novo variants in SCN1A, resulting in Nav1.1 haploinsufficiency. Because SCN1A is expressed in heart and in brain, we proposed that cardiac arrhythmia contributes to SUDEP in DS. We generated DS patient and control induced pluripotent stem cell-derived cardiac myocytes (iPSC-CMs). We observed increased sodium current (INa) and spontaneous contraction rates in DS patient iPSC-CMs versus controls. For the subject with the largest increase in INa, cardiac abnormalities were revealed upon clinical evaluation. Generation of a CRISPR gene-edited heterozygous SCN1A deletion in control iPSCs increased INa density in iPSC-CMs similar to that seen in patient cells. Thus, the high risk of SUDEP in DS may result from a predisposition to cardiac arrhythmias in addition to seizures, reflecting expression of SCN1A in heart and brain.
Collapse
Affiliation(s)
- Chad R Frasier
- Department of Pharmacology, University of Michigan Medical School, 2301E MSRB III, Ann Arbor, MI 48109, USA
| | - Helen Zhang
- Department of Neurology, University of Michigan Medical School, 5021 BSRB, Ann Arbor, MI 48109, USA
| | - James Offord
- Department of Pharmacology, University of Michigan Medical School, 2301E MSRB III, Ann Arbor, MI 48109, USA
| | - Louis T Dang
- Division of Pediatric Neurology, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - David S Auerbach
- Department of Pharmacology, University of Michigan Medical School, 2301E MSRB III, Ann Arbor, MI 48109, USA
| | - Huilin Shi
- Department of Neurology, University of Michigan Medical School, 5021 BSRB, Ann Arbor, MI 48109, USA
| | - Chunling Chen
- Department of Pharmacology, University of Michigan Medical School, 2301E MSRB III, Ann Arbor, MI 48109, USA
| | - Alica M Goldman
- Ann Arbor VA Healthcare System, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - L Lee Eckhardt
- Cellular and Molecular Arrhythmia Research Program, Division of Cardiovascular Medicine, University of Wisconsin, Madison, WI 53705, USA
| | - Vassilios J Bezzerides
- Cardiology, Electrophysiology Division, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jack M Parent
- Department of Neurology, University of Michigan Medical School, 5021 BSRB, Ann Arbor, MI 48109, USA; Ann Arbor VA Healthcare System, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Lori L Isom
- Department of Pharmacology, University of Michigan Medical School, 2301E MSRB III, Ann Arbor, MI 48109, USA; Department of Neurology, University of Michigan Medical School, 5021 BSRB, Ann Arbor, MI 48109, USA.
| |
Collapse
|
26
|
Thouta S, Lo G, Grajauskas L, Claydon T. Investigating the state dependence of drug binding in hERG channels using a trapped-open channel phenotype. Sci Rep 2018; 8:4962. [PMID: 29563525 PMCID: PMC5862968 DOI: 10.1038/s41598-018-23346-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/09/2018] [Indexed: 02/06/2023] Open
Abstract
The hERG channel is a key player in repolarization of the cardiac action potential. Pharmacological blockade of hERG channels depletes the cardiac repolarization reserve, increasing the risk of cardiac arrhythmias. The promiscuous nature of drug interactions with hERG presents a therapeutic challenge for drug design and development. Despite considerable effort, the mechanisms of drug binding remain incompletely understood. One proposed mechanism is that high-affinity drug binding preferentially occurs when channels are in the inactivated state. However, this has been difficult to test, since inactivation is rapid in hERG and access to the drug binding site is limited by slower opening of the activation gate. Here, we have directly assessed the role of inactivation in cisparide and terfenadine drug binding in mutant (I663P) hERG channels where the activation gate is trapped-open. We firstly demonstrate the utility of this approach by showing that inactivation, ion selectivity and high affinity drug binding are preserved in I663P mutant channels. We then assess the role of inactivation by applying cisapride and terfenadine at different membrane voltages, which induce varying degrees of inactivation. We show that the extent of block does not correlate with the extent of inactivation. These data suggest that inactivation is not a major determinant of cisapride or terfenadine binding in hERG channels.
Collapse
Affiliation(s)
- Samrat Thouta
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, B.C., Canada
| | - Garman Lo
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, B.C., Canada
| | - Lukas Grajauskas
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, B.C., Canada
| | - Tom Claydon
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, B.C., Canada.
| |
Collapse
|
27
|
Lee SR, Nilius B, Han J. Gaseous Signaling Molecules in Cardiovascular Function: From Mechanisms to Clinical Translation. Rev Physiol Biochem Pharmacol 2018; 174:81-156. [PMID: 29372329 DOI: 10.1007/112_2017_7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Carbon monoxide (CO), hydrogen sulfide (H2S), and nitric oxide (NO) constitute endogenous gaseous molecules produced by specific enzymes. These gases are chemically simple, but exert multiple effects and act through shared molecular targets to control both physiology and pathophysiology in the cardiovascular system (CVS). The gases act via direct and/or indirect interactions with each other in proteins such as heme-containing enzymes, the mitochondrial respiratory complex, and ion channels, among others. Studies of the major impacts of CO, H2S, and NO on the CVS have revealed their involvement in controlling blood pressure and in reducing cardiac reperfusion injuries, although their functional roles are not limited to these conditions. In this review, the basic aspects of CO, H2S, and NO, including their production and effects on enzymes, mitochondrial respiration and biogenesis, and ion channels are briefly addressed to provide insight into their biology with respect to the CVS. Finally, potential therapeutic applications of CO, H2S, and NO with the CVS are addressed, based on the use of exogenous donors and different types of delivery systems.
Collapse
Affiliation(s)
- Sung Ryul Lee
- Department of Convergence Biomedical Science, Cardiovascular and Metabolic Disease Center, College of Medicine, Inje University, Busan, Republic of Korea
| | - Bernd Nilius
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea.
| |
Collapse
|
28
|
Lee HJ, Choi JS, Choi BH, Hahn SJ. Effects of norquetiapine, the active metabolite of quetiapine, on cloned hERG potassium channels. Neurosci Lett 2017; 664:66-73. [PMID: 29133173 DOI: 10.1016/j.neulet.2017.11.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 12/25/2022]
Abstract
Quetiapine is an atypical antipsychotic drug that is widely used for the treatment of schizophrenia. It is mainly metabolized by a cytochrome P450 system in the liver. Norquetiapine is a major active metabolite in humans with a pharmacological profile that differs distinctly from that of quetiapine. We used the whole-cell patch-clamp technique to investigate the effects of norquetiapine on hERG channels that are stably expressed in HEK cells. Quetiapine and norquetiapine inhibited the hERG tail currents at -50mV in a concentration-dependent manner with IC50 values of 8.3 and 10.8μM, respectively, which suggested equal potency. The block of hERG currents by norquetiapine was voltage-dependent with a steep increase over a range of voltages for channel activation. However, at more depolarized potentials where the channels were fully activated, the block by norquetiapine was voltage-independent. The steady-state inactivation curve of the hERG currents was shifted to the hyperpolarizing direction in the presence of norquetiapine. Norquetiapine did not produce a use-dependent block. A fast application of norquetiapine inhibited the hERG current elicited by a 5s depolarizing pulse to +60mV, which fully inactivated the hERG currents, suggesting an inactivated-state block. During a repolarizing pulse wherein the hERG current was slowly deactivated, albeit remaining in an open state, a fast application of norquetiapine rapidly and reversibly inhibited the open state of the hERG current. Our results indicated that quetiapine and norquetiapine had equal potency in inhibiting hERG tail currents. Norquetiapine inhibited the hERG current by preferentially interacting with the open and/or inactivated states of the channels.
Collapse
Affiliation(s)
- Hong Joon Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin-Sung Choi
- College of Pharmacy, Integrated Research Institute of Pharmaceutical, The Catholic University of Korea, Gyeonggi-do, Republic of Korea
| | - Bok Hee Choi
- Department of Pharmacology, Institute for Medical Science, Chonbuk National University Medical School, Jeonju, Jeonbuk 54097, Republic of Korea
| | - Sang June Hahn
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea.
| |
Collapse
|
29
|
Ahmed M, Jalily Hasani H, Ganesan A, Houghton M, Barakat K. Modeling the human Na v1.5 sodium channel: structural and mechanistic insights of ion permeation and drug blockade. Drug Des Devel Ther 2017; 11:2301-2324. [PMID: 28831242 PMCID: PMC5552146 DOI: 10.2147/dddt.s133944] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Abnormalities in the human Nav1.5 (hNav1.5) voltage-gated sodium ion channel (VGSC) are associated with a wide range of cardiac problems and diseases in humans. Current structural models of hNav1.5 are still far from complete and, consequently, their ability to study atomistic interactions of this channel is very limited. Here, we report a comprehensive atomistic model of the hNav1.5 ion channel, constructed using homology modeling technique and refined through long molecular dynamics simulations (680 ns) in the lipid membrane bilayer. Our model was comprehensively validated by using reported mutagenesis data, comparisons with previous models, and binding to a panel of known hNav1.5 blockers. The relatively long classical MD simulation was sufficient to observe a natural sodium permeation event across the channel's selectivity filters to reach the channel's central cavity, together with the identification of a unique role of the lysine residue. Electrostatic potential calculations revealed the existence of two potential binding sites for the sodium ion at the outer selectivity filters. To obtain further mechanistic insight into the permeation event from the central cavity to the intracellular region of the channel, we further employed "state-of-the-art" steered molecular dynamics (SMD) simulations. Our SMD simulations revealed two different pathways through which a sodium ion can be expelled from the channel. Further, the SMD simulations identified the key residues that are likely to control these processes. Finally, we discuss the potential binding modes of a panel of known hNav1.5 blockers to our structural model of hNav1.5. We believe that the data presented here will enhance our understanding of the structure-property relationships of the hNav1.5 ion channel and the underlying molecular mechanisms in sodium ion permeation and drug interactions. The results presented here could be useful for designing safer drugs that do not block the hNav1.5 channel.
Collapse
Affiliation(s)
| | | | | | - Michael Houghton
- Li Ka Shing Institute of Virology
- Li Ka Shing Applied Virology Institute
- Department of Medical Microbiology and Immunology, Katz Centre for Health Research, University of Alberta, Edmonton, AB, Canada
| | - Khaled Barakat
- Faculty of Pharmacy and Pharmaceutical Sciences
- Li Ka Shing Institute of Virology
- Li Ka Shing Applied Virology Institute
| |
Collapse
|
30
|
Rozier K, Bondarenko VE. Distinct physiological effects of β1- and β2-adrenoceptors in mouse ventricular myocytes: insights from a compartmentalized mathematical model. Am J Physiol Cell Physiol 2017; 312:C595-C623. [DOI: 10.1152/ajpcell.00273.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/03/2017] [Accepted: 01/18/2017] [Indexed: 01/08/2023]
Abstract
The β1- and β2-adrenergic signaling systems play different roles in the functioning of cardiac cells. Experimental data show that the activation of the β1-adrenergic signaling system produces significant inotropic, lusitropic, and chronotropic effects in the heart, whereas the effects of the β2-adrenergic signaling system is less apparent. In this paper, a comprehensive compartmentalized experimentally based mathematical model of the combined β1- and β2-adrenergic signaling systems in mouse ventricular myocytes is developed to simulate the experimental findings and make testable predictions of the behavior of the cardiac cells under different physiological conditions. Simulations describe the dynamics of major signaling molecules in different subcellular compartments; kinetics and magnitudes of phosphorylation of ion channels, transporters, and Ca2+ handling proteins; modifications of action potential shape and duration; and [Ca2+]i and [Na+]i dynamics upon stimulation of β1- and β2-adrenergic receptors (β1- and β2-ARs). The model reveals physiological conditions when β2-ARs do not produce significant physiological effects and when their effects can be measured experimentally. Simulations demonstrated that stimulation of β2-ARs with isoproterenol caused a marked increase in the magnitude of the L-type Ca2+ current, [Ca2+]i transient, and phosphorylation of phospholamban only upon additional application of pertussis toxin or inhibition of phosphodiesterases of type 3 and 4. The model also made testable predictions of the changes in magnitudes of [Ca2+]i and [Na+]i fluxes, the rate of decay of [Na+]i concentration upon both combined and separate stimulation of β1- and β2-ARs, and the contribution of phosphorylation of PKA targets to the changes in the action potential and [Ca2+]i transient.
Collapse
Affiliation(s)
- Kelvin Rozier
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia; and
| | - Vladimir E. Bondarenko
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia; and
- Neuroscience Institute, Georgia State University, Atlanta, Georgia
| |
Collapse
|
31
|
Propagation of parametric uncertainty for the K+ channel model in mouse ventricular myocytes. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2017; 2016:5587-5590. [PMID: 28269521 DOI: 10.1109/embc.2016.7591993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cardiac potassium (K+) channel plays an important role in cardiac electrical signaling. Mathematical models have been widely used to investigate the effects of K+ channels on cardiac functions. However, the model of K+ channel involves parametric uncertainties, which can be induced by fitting the model's parameters that best capture experimental data. Since the prediction of cardiac functions are highly parameter-dependent, it is critical to quantify the influence of parametric uncertainty on the model responses to provide the more reliable predictions. This paper presents a new method to efficiently propagate the uncertainty on the model's parameters of K+ channel to the gating variables as well as the current density. In this way, we can estimate the model predictions and their corresponding confidence intervals simultaneously. A generalized polynomial chaos (gPC) expansion approximating the parametric uncertainty is used in combination with the physical models to quantify and propagate the parametric uncertainties onto the modeled predictions of steady state activation and steady state inactivation of the K+ channel. Using Galerkin projection, the variation (i.e., confidence interval) of the gating variables resulting from the uncertainty of model parameters can then be estimated in a computationally efficient fashion. As compared with the Monte Carlo (MC) simulations, the proposed methodology shows it's advantageous in terms of computational efficiency and accuracy, thus demonstrating the potential for dealing with more complicated cardiac models.
Collapse
|
32
|
Carnevali L, Rivara S, Nalivaiko E, Thayer JF, Vacondio F, Mor M, Sgoifo A. Pharmacological inhibition of FAAH activity in rodents: A promising pharmacological approach for psychological—cardiac comorbidity? Neurosci Biobehav Rev 2017; 74:444-452. [DOI: 10.1016/j.neubiorev.2016.04.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/19/2016] [Accepted: 04/20/2016] [Indexed: 01/09/2023]
|
33
|
Kithcart A, MacRae CA. Using Zebrafish for High-Throughput Screening of Novel Cardiovascular Drugs. JACC Basic Transl Sci 2017; 2:1-12. [PMID: 30167552 PMCID: PMC6113531 DOI: 10.1016/j.jacbts.2017.01.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 01/17/2017] [Accepted: 01/17/2017] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases remain a major challenge for modern drug discovery. The diseases are chronic, complex, and the result of sophisticated interactions between genetics and environment involving multiple cell types and a host of systemic factors. The clinical events are often abrupt, and the diseases may be asymptomatic until a highly morbid event. Target selection is often based on limited information, and though highly specific agents are often identified in screening, their final efficacy is often compromised by unanticipated systemic responses, a narrow therapeutic index, or substantial toxicities. Our understanding of complexity of cardiovascular disease has grown dramatically over the past 2 decades, and the range of potential disease mechanisms now includes pathways previously thought only tangentially involved in cardiac or vascular disease. Despite these insights, the majority of active cardiovascular agents derive from a remarkably small number of classes of agents and target a very limited number of pathways. These agents have often been used initially for particular indications and then discovered serendipitously to have efficacy in other cardiac disorders or in a manner unrelated to their original mechanism of action. In this review, the rationale for in vivo screening is described, and the utility of the zebrafish for this approach and for complementary work in functional genomics is discussed. Current limitations of the model in this setting and the need for careful validation in new disease areas are also described. An overview is provided of the complex mechanisms underlying most clinical cardiovascular diseases, and insight is offered into the limits of single downstream pathways as drug targets. The zebrafish is introduced as a model organism, in particular for cardiovascular biology. Potential approaches to overcoming the hurdles to drug discovery in the face of complex biology are discussed, including in vivo screening of zebrafish genetic disease models.
Collapse
Affiliation(s)
- Aaron Kithcart
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,The Broad Institute of MIT and Harvard, Harvard Stem Cell Institute, Boston, Massachusetts
| | - Calum A MacRae
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,The Broad Institute of MIT and Harvard, Harvard Stem Cell Institute, Boston, Massachusetts
| |
Collapse
|
34
|
Finlay M, Harmer SC, Tinker A. The control of cardiac ventricular excitability by autonomic pathways. Pharmacol Ther 2017; 174:97-111. [PMID: 28223225 DOI: 10.1016/j.pharmthera.2017.02.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Central to the genesis of ventricular cardiac arrhythmia are variations in determinants of excitability. These involve individual ionic channels and transporters in cardiac myocytes but also tissue factors such as variable conduction of the excitation wave, fibrosis and source-sink mismatch. It is also known that in certain diseases and particularly the channelopathies critical events occur with specific stressors. For example, in hereditary long QT syndrome due to mutations in KCNQ1 arrhythmic episodes are provoked by exercise and in particular swimming. Thus not only is the static substrate important but also how this is modified by dynamic signalling events associated with common physiological responses. In this review, we examine the regulation of ventricular excitability by signalling pathways from a cellular and tissue perspective in an effort to identify key processes, effectors and potential therapeutic approaches. We specifically focus on the autonomic nervous system and related signalling pathways.
Collapse
Affiliation(s)
- Malcolm Finlay
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M6BQ, UK
| | - Stephen C Harmer
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M6BQ, UK
| | - Andrew Tinker
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M6BQ, UK.
| |
Collapse
|
35
|
Fernández-Falgueras A, Sarquella-Brugada G, Brugada J, Brugada R, Campuzano O. Cardiac Channelopathies and Sudden Death: Recent Clinical and Genetic Advances. BIOLOGY 2017; 6:7. [PMID: 28146053 PMCID: PMC5372000 DOI: 10.3390/biology6010007] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/17/2017] [Accepted: 01/20/2017] [Indexed: 12/19/2022]
Abstract
Sudden cardiac death poses a unique challenge to clinicians because it may be the only symptom of an inherited heart condition. Indeed, inherited heart diseases can cause sudden cardiac death in older and younger individuals. Two groups of familial diseases are responsible for sudden cardiac death: cardiomyopathies (mainly hypertrophic cardiomyopathy, dilated cardiomyopathy, and arrhythmogenic cardiomyopathy) and channelopathies (mainly long QT syndrome, Brugada syndrome, short QT syndrome, and catecholaminergic polymorphic ventricular tachycardia). This review focuses on cardiac channelopathies, which are characterized by lethal arrhythmias in the structurally normal heart, incomplete penetrance, and variable expressivity. Arrhythmias in these diseases result from pathogenic variants in genes encoding cardiac ion channels or associated proteins. Due to a lack of gross structural changes in the heart, channelopathies are often considered as potential causes of death in otherwise unexplained forensic autopsies. The asymptomatic nature of channelopathies is cause for concern in family members who may be carrying genetic risk factors, making the identification of these genetic factors of significant clinical importance.
Collapse
Affiliation(s)
| | | | - Josep Brugada
- Arrhythmias Unit, Hospital Sant Joan de Déu, University of Barcelona, Barcelona 08950, Spain.
| | - Ramon Brugada
- Cardiovascular Genetics Center, IDIBGI, Girona 17190, Spain.
- Medical Sciences Department, School of Medicine, University of Girona, Girona 17071, Spain.
- Familial Cardiomyopathies Unit, Hospital Josep Trueta, Girona 17007, Spain.
| | - Oscar Campuzano
- Cardiovascular Genetics Center, IDIBGI, Girona 17190, Spain.
- Medical Sciences Department, School of Medicine, University of Girona, Girona 17071, Spain.
| |
Collapse
|
36
|
Vornanen M. Electrical Excitability of the Fish Heart and Its Autonomic Regulation. FISH PHYSIOLOGY 2017. [DOI: 10.1016/bs.fp.2017.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
37
|
|
38
|
Dorian P, Mangat I, Pinter A, Korley V. The Burden of Atrial Fibrillation: Should We Abandon Antiarrhythmic Drug Therapy? J Cardiovasc Pharmacol Ther 2016; 9:257-62. [PMID: 15678244 DOI: 10.1177/107424840400900405] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia, exacting a substantial toll in cardiovascular morbidity and mortality. Until recently, the prevailing philosophy has been that restoration and maintenance of normal sinus rhythm, as opposed to control of ventricular response rate, was the optimal approach to treatment of AF. A series of landmark trials (AFFIRM, RACE, STAF, and PIAF) have called this strategy into question, suggesting outcomes are equivalent with both approaches. These data do not mean that rhythm control is not beneficial, but highlight the limitations of current therapies to achieve and maintain sinus rhythm. Limitations of the rhythm-control strategy may be related to our difficulty in accurately documenting symptomatic benefit from this approach, the lack of efficacy and excessive adverse-effect burden associated with currently available antiarrhythmic agents, and selection biases in the enrollment of patients in clinical trials of rhythm control versus rate control, making the trials incompletely representative of the population eligible for therapy. New pharmacologic agents under development feature increased atrial selectivity or multi-channel-blocking properties (or both). As a result, these compounds may be more effective in prolonging atrial refractoriness and may also have reduced proarrhythmic potential. It is premature to abandon the concept of rhythm control in AF until we have trials designed to include younger and highly symptomatic patients, more sensitive tools to measure symptomatic improvement, and safer, more effective antiarrhythmic agents.
Collapse
Affiliation(s)
- Paul Dorian
- Division of Cardiology, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.
| | | | | | | |
Collapse
|
39
|
Kennedy A, Finlay DD, Guldenring D, Bond R, Moran K, McLaughlin J. The Cardiac Conduction System: Generation and Conduction of the Cardiac Impulse. Crit Care Nurs Clin North Am 2016; 28:269-79. [PMID: 27484656 DOI: 10.1016/j.cnc.2016.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In this article, the authors outline the key components behind the automated generation of the cardiac impulses and the effect these impulses have on cardiac myocytes. Also, a description of the key components of the normal cardiac conduction system is provided, including the sinoatrial node, the atrioventricular node, the His bundle, the bundle branches, and the Purkinje network. Finally, an outline of how each stage of the cardiac conduction system is represented on the electrocardiogram is described, allowing the reader of the electrocardiogram to translate background information about the normal cardiac conduction system to everyday clinical practice.
Collapse
Affiliation(s)
- Alan Kennedy
- The Engineering Research Institute (ERI), School of Engineering, University of Ulster at Jordanstown, Newtownabbey, County Antrim BT37 0QB, Northern Ireland.
| | - Dewar D Finlay
- The Engineering Research Institute (ERI), School of Engineering, University of Ulster at Jordanstown, Newtownabbey, County Antrim BT37 0QB, Northern Ireland
| | - Daniel Guldenring
- The Engineering Research Institute (ERI), School of Engineering, University of Ulster at Jordanstown, Newtownabbey, County Antrim BT37 0QB, Northern Ireland
| | - Raymond Bond
- Computing Science Research Instititute, School of Computing and Mathematics, University of Ulster, Room 16G06, Jordanstown Campus, Shore Road, Newtownabbey BT37 0QB, County Antrim, Northern Ireland
| | - Kieran Moran
- School of Health and Human Performance, Dublin City University, Room XG05, Glanevin, Dublin 9, Ireland
| | - James McLaughlin
- The Engineering Research Institute (ERI), School of Engineering, University of Ulster at Jordanstown, Newtownabbey, County Antrim BT37 0QB, Northern Ireland
| |
Collapse
|
40
|
Tse G, Lai ETH, Yeo JM, Tse V, Wong SH. Mechanisms of Electrical Activation and Conduction in the Gastrointestinal System: Lessons from Cardiac Electrophysiology. Front Physiol 2016; 7:182. [PMID: 27303305 PMCID: PMC4885840 DOI: 10.3389/fphys.2016.00182] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/06/2016] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal (GI) tract is an electrically excitable organ system containing multiple cell types, which coordinate electrical activity propagating through this tract. Disruption in its normal electrophysiology is observed in a number of GI motility disorders. However, this is not well characterized and the field of GI electrophysiology is much less developed compared to the cardiac field. The aim of this article is to use the established knowledge of cardiac electrophysiology to shed light on the mechanisms of electrical activation and propagation along the GI tract, and how abnormalities in these processes lead to motility disorders and suggest better treatment options based on this improved understanding. In the first part of the article, the ionic contributions to the generation of GI slow wave and the cardiac action potential (AP) are reviewed. Propagation of these electrical signals can be described by the core conductor theory in both systems. However, specifically for the GI tract, the following unique properties are observed: changes in slow wave frequency along its length, periods of quiescence, synchronization in short distances and desynchronization over long distances. These are best described by a coupled oscillator theory. Other differences include the diminished role of gap junctions in mediating this conduction in the GI tract compared to the heart. The electrophysiology of conditions such as gastroesophageal reflux disease and gastroparesis, and functional problems such as irritable bowel syndrome are discussed in detail, with reference to ion channel abnormalities and potential therapeutic targets. A deeper understanding of the molecular basis and physiological mechanisms underlying GI motility disorders will enable the development of better diagnostic and therapeutic tools and the advancement of this field.
Collapse
Affiliation(s)
- Gary Tse
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong KongHong Kong, China
| | - Eric Tsz Him Lai
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong KongHong Kong, China
| | - Jie Ming Yeo
- School of Medicine, Imperial College LondonLondon, UK
| | - Vivian Tse
- Department of Physiology, McGill UniversityMontreal, QC, Canada
| | - Sunny Hei Wong
- Department of Medicine and Therapeutics, Institute of Digestive Disease, LKS Institute of Health Sciences, Chinese University of Hong KongHong Kong, China
| |
Collapse
|
41
|
Zhao Y, Gao F, Zhang Y, Wang H, Zhu J, Chang L, Du Z, Zhang Y. Shensong Yangxin capsules prevent ischemic arrhythmias by prolonging action potentials and alleviating Ca2+ overload. Mol Med Rep 2016; 13:5185-92. [PMID: 27122298 DOI: 10.3892/mmr.2016.5203] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 04/07/2016] [Indexed: 11/06/2022] Open
Abstract
Shensong Yangxin capsules (SSYX) are an effective traditional Chinese medicine that has been used to treat coronary heart disease clinically. The present study aimed to establish whether SSYX prevent ischemic arrhythmias in rats, and to explore the underlying mechanisms. Male rats were pretreated with distilled water, SSYX and amiodarone for one week. Acute myocardial ischemia (AMI) was performed to induce ischemic arrhythmias. The incidence and severity of ischemic arrhythmias were evaluated. The action potential, transient outward K+ current (Ito) and inward rectifier K+ current (IK1) of rat cardiomyocytes were measured using the patch‑clamp technique. The intracellular Ca2+ concentration of the cardiomyocytes was measured using a laser scanning confocal microscope. The results revealed that SSYX lowered the incidence of arrhythmia markedly during AMI. Furthermore, SSYX delayed the appearance, and reduced the severity, of ischemic arrhythmias compared with the control. In addition, SSYX markedly decreased the ratio of the myocardial infarction region to the whole heart. In an in vitro study, SSYX prolonged the action potential duration of rat cardiomyocytes, and inhibited Ito and IK1 markedly. Additionally, SSYX inhibited Ca2+ elevation induced by KCl in cardiomyocytes. These results suggested that SSYX prevents ischemic arrhythmia, and the underlying mechanism responsible for this process may include prolonging the action potential and alleviating Ca2+ overload.
Collapse
Affiliation(s)
- Yixiu Zhao
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Feng Gao
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yong Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Hongtao Wang
- State Key Laboratory of Collateral Disease Research and Innovation Medicine, Hebei Yiling Pharmaceutical Research Institute, Shijiazhuang, Hebei 050000, P.R. China
| | - Jiuxin Zhu
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Liping Chang
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Zhimin Du
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yan Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
42
|
Carnevali L, Vacondio F, Rossi S, Macchi E, Spadoni G, Bedini A, Neumann ID, Rivara S, Mor M, Sgoifo A. Cardioprotective effects of fatty acid amide hydrolase inhibitor URB694, in a rodent model of trait anxiety. Sci Rep 2015; 5:18218. [PMID: 26656183 PMCID: PMC4677398 DOI: 10.1038/srep18218] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/02/2015] [Indexed: 12/12/2022] Open
Abstract
In humans, chronic anxiety represents an independent risk factor for cardiac arrhythmias and sudden death. Here we evaluate in male Wistar rats bred for high (HAB) and low (LAB) anxiety-related behavior, as well as non-selected (NAB) animals, the relationship between trait anxiety and cardiac electrical instability and investigate whether pharmacological augmentation of endocannabinoid anandamide-mediated signaling exerts anxiolytic-like and cardioprotective effects. HAB rats displayed (i) a higher incidence of ventricular tachyarrhythmias induced by isoproterenol, and (ii) a larger spatial dispersion of ventricular refractoriness assessed by means of an epicardial mapping protocol. In HAB rats, acute pharmacological inhibition of the anandamide-degrading enzyme, fatty acid amide hydrolase (FAAH), with URB694 (0.3 mg/kg), (i) decreased anxiety-like behavior in the elevated plus maze, (ii) increased anandamide levels in the heart, (iii) reduced isoproterenol-induced occurrence of ventricular tachyarrhythmias, and (iv) corrected alterations of ventricular refractoriness. The anti-arrhythmic effect of URB694 was prevented by pharmacological blockade of the cannabinoid type 1 (CB1), but not of the CB2, receptor. These findings suggest that URB694 exerts anxiolytic-like and cardioprotective effects in HAB rats, the latter via anandamide-mediated activation of CB1 receptors. Thus, pharmacological inhibition of FAAH might be a viable pharmacological strategy for the treatment of anxiety-related cardiac dysfunction.
Collapse
Affiliation(s)
| | | | - Stefano Rossi
- Department of Life Sciences, University of Parma, Italy
| | - Emilio Macchi
- Department of Life Sciences, University of Parma, Italy
| | - Gilberto Spadoni
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Italy
| | - Annalida Bedini
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Italy
| | - Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Germany
| | | | - Marco Mor
- Department of Pharmacy, University of Parma, Italy
| | - Andrea Sgoifo
- Department of Neuroscience, University of Parma, Italy
| |
Collapse
|
43
|
Abstract
Maintaining adequate tissue perfusion depends on a variety of factors, all of which can be influenced by xenobiotics (substances foreign to the body, including pharmaceuticals, chemicals, and natural compounds). Volume status, systemic vascular resistance, myocardial contractility, and cardiac rhythm all play a significant role in ensuring hemodynamic stability and proper cardiovascular function. Direct effects on the nervous system, the vasculature, or the heart itself as well as indirect metabolic effects may play a significant role in the development of cardiotoxicity. This article is dedicated to discussion of the disruption of cardiovascular physiology by xenobiotics.
Collapse
|
44
|
[Long QT syndrome. History, genetics, clinical symptoms, causes and therapy]. Anaesthesist 2015; 64:586-95. [PMID: 26250931 DOI: 10.1007/s00101-015-0068-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The long QT syndrome is caused by a change in cardiac repolarization due to functional ion channel defects. A differentiation is made between a congenital (cLQTS) and an acquired (aLQTS) form of the disease. The disease results in the name-giving prolongation of the QT interval in the electrocardiogram and represents a predisposition for cardiac arrhythmia and sudden cardiac death. This article summarizes the current knowledge on the history, pathophysiology, clinical symptoms and therapy of cLQTS and aLQTS. This knowledge of pathophysiological features of the symptoms allows the underlying anesthesiological approach for individualized perioperative concepts for patients suffering from LQTS to be derived.
Collapse
|
45
|
Yang KC, Kyle JW, Makielski JC, Dudley SC. Mechanisms of sudden cardiac death: oxidants and metabolism. Circ Res 2015; 116:1937-55. [PMID: 26044249 PMCID: PMC4458707 DOI: 10.1161/circresaha.116.304691] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 02/09/2015] [Indexed: 02/07/2023]
Abstract
Ventricular arrhythmia is the leading cause of sudden cardiac death (SCD). Deranged cardiac metabolism and abnormal redox state during cardiac diseases foment arrhythmogenic substrates through direct or indirect modulation of cardiac ion channel/transporter function. This review presents current evidence on the mechanisms linking metabolic derangement and excessive oxidative stress to ion channel/transporter dysfunction that predisposes to ventricular arrhythmias and SCD. Because conventional antiarrhythmic agents aiming at ion channels have proven challenging to use, targeting arrhythmogenic metabolic changes and redox imbalance may provide novel therapeutics to treat or prevent life-threatening arrhythmias and SCD.
Collapse
Affiliation(s)
- Kai-Chien Yang
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.)
| | - John W Kyle
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.)
| | - Jonathan C Makielski
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.).
| | - Samuel C Dudley
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.).
| |
Collapse
|
46
|
Willis BC, Ponce-Balbuena D, Jalife J. Protein assemblies of sodium and inward rectifier potassium channels control cardiac excitability and arrhythmogenesis. Am J Physiol Heart Circ Physiol 2015; 308:H1463-73. [PMID: 25862830 DOI: 10.1152/ajpheart.00176.2015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/06/2015] [Indexed: 02/07/2023]
Abstract
The understanding of how cardiac ion channels function in the normal and the diseased heart has greatly increased over the last four decades thanks to the advent of patch-clamp technology and, more recently, the emergence of genetics, as well as cellular and molecular cardiology. However, our knowledge of how these membrane-embedded proteins physically interact with each other within macromolecular complexes remains incomplete. This review focuses on how the main cardiac inward sodium channel (NaV1.5) and the strong inward rectifier potassium channel (Kir2.1) function within macromolecular complexes to control cardiac excitability. It has become increasingly clear that these two important ion channel proteins physically interact with multiple other protein partners and with each other from early stages of protein trafficking and targeting through membrane anchoring, recycling, and degradation. Recent findings include compartmentalized regulation of NaV1.5 channel expression and function through a PDZ (postsynaptic density protein, Drosophila disc large tumor suppressor, and zonula occludens-1 protein) domain-binding motif, and interaction of caveolin-3 with Kir2.1 and ankyrin-G as a molecular platform for NaV1.5 signaling. At the cardiomyocyte membrane, NaV1.5 and Kir2.1 interact through at least two distinct PDZ domain-scaffolding proteins (synapse-associated protein-97 and α1-syntrophin), thus modulating reciprocally their cell-surface expression at two different microdomains. Emerging evidence also shows that inheritable mutations in plakophilin-2, ankyrin-G, dystrophin, syntrophin, synapse-associated protein-97, and caveolin-3, among others, modify functional expression and/or localization in the cardiac cell of NaV1.5, Kir2.1 or both to give rise to arrhythmogenic diseases. Unveiling the mechanistic underpinnings of macromolecular interactions should increase our understanding of inherited and acquired arrhythmogenic cardiac diseases and may lead to advances in therapy.
Collapse
Affiliation(s)
- B Cicero Willis
- Department of Internal Medicine and Center for Arrhythmia Research, University of Michigan, Ann Arbor, Michigan; and
| | - Daniela Ponce-Balbuena
- Department of Internal Medicine and Center for Arrhythmia Research, University of Michigan, Ann Arbor, Michigan; and
| | - José Jalife
- Department of Internal Medicine and Center for Arrhythmia Research, University of Michigan, Ann Arbor, Michigan; and Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| |
Collapse
|
47
|
Vonderlin N, Fischer F, Zitron E, Seyler C, Scherer D, Thomas D, Katus HA, Scholz EP. Anesthetic drug midazolam inhibits cardiac human ether-à-go-go-related gene channels: mode of action. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:867-77. [PMID: 25733807 PMCID: PMC4338257 DOI: 10.2147/dddt.s72765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Midazolam is a short-acting benzodiazepine that is in wide clinical use as an anxiolytic, sedative, hypnotic, and anticonvulsant. Midazolam has been shown to inhibit ion channels, including calcium and potassium channels. So far, the effects of midazolam on cardiac human ether-à-go-go-related gene (hERG) channels have not been analyzed. The inhibitory effects of midazolam on heterologously expressed hERG channels were analyzed in Xenopus oocytes using the double-electrode voltage clamp technique. We found that midazolam inhibits hERG channels in a concentration-dependent manner, yielding an IC50 of 170 μM in Xenopus oocytes. When analyzed in a HEK 293 cell line using the patch-clamp technique, the IC50 was 13.6 μM. Midazolam resulted in a small negative shift of the activation curve of hERG channels. However, steady-state inactivation was not significantly affected. We further show that inhibition is state-dependent, occurring within the open and inactivated but not in the closed state. There was no frequency dependence of block. Using the hERG pore mutants F656A and Y652A we provide evidence that midazolam uses a classical binding site within the channel pore. Analyzing the subacute effects of midazolam on hERG channel trafficking, we further found that midazolam does not affect channel surface expression. Taken together, we show that the anesthetic midazolam is a low-affinity inhibitor of cardiac hERG channels without additional effects on channel surface expression. These data add to the current understanding of the pharmacological profile of the anesthetic midazolam.
Collapse
Affiliation(s)
- Nadine Vonderlin
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
| | - Fathima Fischer
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
| | - Edgar Zitron
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany ; German Centre for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Claudia Seyler
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Scherer
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
| | - Dierk Thomas
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany ; German Centre for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Hugo A Katus
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany ; German Centre for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Eberhard P Scholz
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
48
|
Design and synthesis of a novel series of histamine H3 receptor antagonists through a scaffold hopping strategy. Bioorg Med Chem 2015; 23:429-38. [DOI: 10.1016/j.bmc.2014.12.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/08/2014] [Accepted: 12/16/2014] [Indexed: 01/06/2023]
|
49
|
Ito J. [Steroid hormones' genomic and non-genomic actions on cardiac voltage-gated calcium channels]. Nihon Yakurigaku Zasshi 2014; 144:206-210. [PMID: 25381888 DOI: 10.1254/fpj.144.206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
|
50
|
Vijayakumar R, Silva JNA, Desouza KA, Abraham RL, Strom M, Sacher F, Van Hare GF, Haïssaguerre M, Roden DM, Rudy Y. Electrophysiologic substrate in congenital Long QT syndrome: noninvasive mapping with electrocardiographic imaging (ECGI). Circulation 2014; 130:1936-1943. [PMID: 25294783 DOI: 10.1161/circulationaha.114.011359] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Congenital Long QT syndrome (LQTS) is an arrhythmogenic disorder that causes syncope and sudden death. Although its genetic basis has become well-understood, the mechanisms whereby mutations translate to arrhythmia susceptibility in the in situ human heart have not been fully defined. We used noninvasive ECG imaging to map the cardiac electrophysiological substrate and examine whether LQTS patients display regional heterogeneities in repolarization, a substrate that promotes arrhythmogenesis. METHODS AND RESULTS Twenty-five subjects (9 LQT1, 9 LQT2, 5 LQT3, and 2 LQT5) with genotype and phenotype positive LQTS underwent ECG imaging. Seven normal subjects provided control. Epicardial maps of activation, recovery times, activation-recovery intervals, and repolarization dispersion were constructed. Activation was normal in all patients. However, recovery times and activation-recovery intervals were prolonged relative to control, indicating delayed repolarization and abnormally long action potential duration (312±30 ms versus 235±21 ms in control). Activation-recovery interval prolongation was spatially heterogeneous, with repolarization gradients much steeper than control (119±19 ms/cm versus 2.0±2.0 ms/cm). There was variability in steepness and distribution of repolarization gradients between and within LQTS types. Repolarization gradients were steeper in symptomatic patients (130±27 ms/cm in 12 symptomatic patients versus 98±19 ms/cm in 13 asymptomatic patients; P<0.05). CONCLUSIONS LQTS patients display regions with steep repolarization dispersion caused by localized action potential duration prolongation. This defines a substrate for reentrant arrhythmias, not detectable by surface ECG. Steeper dispersion in symptomatic patients suggests a possible role for ECG imaging in risk stratification.
Collapse
Affiliation(s)
- Ramya Vijayakumar
- Cardiac Bioelectricity and Arrhythmia Center, Washington University in St. Louis, Saint Louis, MO.,Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO
| | - Jennifer N A Silva
- Cardiac Bioelectricity and Arrhythmia Center, Washington University in St. Louis, Saint Louis, MO.,Division of Pediatric Cardiology, Washington University School of Medicine/St. Louis Children's Hospital, St. Louis, MO
| | - Kavit A Desouza
- Cardiovascular diseases, Mount Sinai St. Luke's Roosevelt, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Robert L Abraham
- Department of Medicine and the Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN
| | | | - Frederic Sacher
- Bordeaux University Hospital, LIRYC institute, Pessac, France
| | - George F Van Hare
- Cardiac Bioelectricity and Arrhythmia Center, Washington University in St. Louis, Saint Louis, MO.,Division of Pediatric Cardiology, Washington University School of Medicine/St. Louis Children's Hospital, St. Louis, MO
| | | | - Dan M Roden
- Department of Medicine and the Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN
| | - Yoram Rudy
- Cardiac Bioelectricity and Arrhythmia Center, Washington University in St. Louis, Saint Louis, MO.,Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO.,Division of Pediatric Cardiology, Washington University School of Medicine/St. Louis Children's Hospital, St. Louis, MO.,Department of Medicine (Cardiology), Washington University School of Medicine/Barnes-Jewish Hospital, St. Louis, MO
| |
Collapse
|