1
|
Ke Y, Cao Z, Wang X, Liu D, Fu Y, Chen H, Cheng Y, Guo K, Li Y, Long X, Yang M, Zhao Q. K Ca3.1 Promotes the Migration of Macrophages From Epicardial Adipose Tissue to Induce Vulnerability to Atrial Fibrillation During Rapid Pacing. Can J Cardiol 2024:S0828-282X(24)00916-4. [PMID: 39147322 DOI: 10.1016/j.cjca.2024.08.266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND The relationship between local epicardial adipose tissue (EAT) macrophages and atrial fibrillation (AF) remains unclear. The purpose of this study was to investigate the role of KCa3.1 in the migration of macrophages from EAT to adjacent atrial tissue during rapid pacing. METHODS Part 1: Eighteen beagles were randomly divided into the sham group, pacing group, and pacing + clodronate liposome (CL) group. Part 2: Eighteen beagles were randomly divided into the sham group, pacing group, and pacing + TRAM-34 group. HL-1 cells and RAW264.7 cells were co-cultured to explore the specific migratory mechanism of macrophages. RESULTS Depleting EAT macrophages significantly reduced macrophage infiltration in the adjacent atrium and the induction of AF in canines with rapid atrial pacing. TRAM-34 significantly inhibited the migration of macrophages from EAT to the adjacent atrium and electrical remodelling in canines with rapid atrial pacing. Compared with those of the control HL-1 cells, the secretion of CCL2 and the number of migrating macrophages in pacing HL-1 cells was significantly increased, which could be reversed by TRAM-34. Further in vitro experiments showed that KCa3.1 regulated CCL2 secretion through the p65/STAT3 signalling pathway. CONCLUSIONS Inhibiting myocardial KCa3.1 reduced the migration of EAT macrophages to adjacent atrial muscles caused by rapid atrial pacing, thereby decreasing vulnerability to AF. The mechanism by which KCa3.1 regulates CCL2 may be related to the p65/STAT3 signalling pathway.
Collapse
Affiliation(s)
- Yuanjia Ke
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Zhen Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Xuewen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Dishiwen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Yuntao Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Huiyu Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Yanni Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Kexin Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Yajia Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Xiaojian Long
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
Yuntao F, Jinjun L, Hua Fen L, Huiyu C, Dishiwen L, Zhen C, Wang Y, Wang X, Ke Y, Yanni C, Kexin G, Zhibin P, Mei Y, Zhao Q. Atrial fibroblast-derived exosomal miR-21 upregulate myocardial KCa3.1 via the PI3K-Akt pathway during rapid pacing. Heliyon 2024; 10:e33059. [PMID: 39040331 PMCID: PMC11260968 DOI: 10.1016/j.heliyon.2024.e33059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 07/24/2024] Open
Abstract
Background Fibroblast-derived exosomes can regulate the electrical remodeling of cardiomyocytes, and the intermediate-conductance calcium-activated potassium channel (KCa3.1) is important in atrial electrical remodeling. However, the underlying molecular mechanisms remain unclear. This study aimed to investigate the regulation of cardiac electrophysiology by exosomes linked to KCa3.1. Methods Atrial myocytes (AMs) and atrial fibroblasts were isolated from Sprague-Dawley suckling rats and cultured individually. The cellular atrial fibrillation (AF) model was established via electrical stimulation (1.0 v/cm, 10 Hz), and fibroblast-derived exosomes were isolated via ultracentrifugation. Exosomes were co-cultured with AMs to investigate their influences on KCa3.1 and the underlying mechanisms. Nanoparticle tracking analysis and transmission electron microscopy were used to measure exosome particle sizes and concentrations. Whole-cell patch clamp was applied to record the current density of KCa3.1 and action potential duration (APD). The expression of miR-21-5p was detected by reverse-transcription polymerase chain reaction (RT-PCR). Western blotting or immunofluorescence was used to measure the expression of exosomal markers, Akt phosphorylation, and KCa3.1. Results Rapid pacing promoted the secretion of exosomes from atrial fibroblasts and miR-21-5p expression in atrial fibroblasts and exosomes. KCa3.1 protein expression and current density significantly increased, and APD50 and APD90 were sharply shortened after rapid pacing in AMs. TRAM-34 (KCa3.1 blocker) extended APD and reduced susceptibility to AF. KCa3.1 and P-AKT expressions were amplified after co-culturing AMs with exosomes secreted by atrial fibroblasts. In contrast, the increase in KCa3.1 expression was reversed after the cells were co-cultured with exosomes secreted by atrial fibroblasts that were transfected with miR-21-5p inhibitors or after the use of LY294002, a PI3K/Akt pathway inhibitor. Conclusions Rapid pacing promoted the secretion of exosomes from fibroblasts, and miR-21-5p was upregulated in exosomes. Moreover, the miR-21-5p-enriched exosomes upregulated KCa3.1 expression in AMs via the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Fu Yuntao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liang Jinjun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liu Hua Fen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Chen Huiyu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liu Dishiwen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cao Zhen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Youcheng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xuewen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yuanjia Ke
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cheng Yanni
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Guo Kexin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | | | - Yang Mei
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
3
|
Hennis K, Piantoni C, Biel M, Fenske S, Wahl-Schott C. Pacemaker Channels and the Chronotropic Response in Health and Disease. Circ Res 2024; 134:1348-1378. [PMID: 38723033 PMCID: PMC11081487 DOI: 10.1161/circresaha.123.323250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2024]
Abstract
Loss or dysregulation of the normally precise control of heart rate via the autonomic nervous system plays a critical role during the development and progression of cardiovascular disease-including ischemic heart disease, heart failure, and arrhythmias. While the clinical significance of regulating changes in heart rate, known as the chronotropic effect, is undeniable, the mechanisms controlling these changes remain not fully understood. Heart rate acceleration and deceleration are mediated by increasing or decreasing the spontaneous firing rate of pacemaker cells in the sinoatrial node. During the transition from rest to activity, sympathetic neurons stimulate these cells by activating β-adrenergic receptors and increasing intracellular cyclic adenosine monophosphate. The same signal transduction pathway is targeted by positive chronotropic drugs such as norepinephrine and dobutamine, which are used in the treatment of cardiogenic shock and severe heart failure. The cyclic adenosine monophosphate-sensitive hyperpolarization-activated current (If) in pacemaker cells is passed by hyperpolarization-activated cyclic nucleotide-gated cation channels and is critical for generating the autonomous heartbeat. In addition, this current has been suggested to play a central role in the chronotropic effect. Recent studies demonstrate that cyclic adenosine monophosphate-dependent regulation of HCN4 (hyperpolarization-activated cyclic nucleotide-gated cation channel isoform 4) acts to stabilize the heart rate, particularly during rapid rate transitions induced by the autonomic nervous system. The mechanism is based on creating a balance between firing and recently discovered nonfiring pacemaker cells in the sinoatrial node. In this way, hyperpolarization-activated cyclic nucleotide-gated cation channels may protect the heart from sinoatrial node dysfunction, secondary arrhythmia of the atria, and potentially fatal tachyarrhythmia of the ventricles. Here, we review the latest findings on sinoatrial node automaticity and discuss the physiological and pathophysiological role of HCN pacemaker channels in the chronotropic response and beyond.
Collapse
Affiliation(s)
- Konstantin Hennis
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center Munich, Walter Brendel Centre of Experimental Medicine, Faculty of Medicine (K.H., C.P., C.W.-S.), Ludwig-Maximilians-Universität München, Germany
| | - Chiara Piantoni
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center Munich, Walter Brendel Centre of Experimental Medicine, Faculty of Medicine (K.H., C.P., C.W.-S.), Ludwig-Maximilians-Universität München, Germany
| | - Martin Biel
- Department of Pharmacy, Center for Drug Research (M.B., S.F.), Ludwig-Maximilians-Universität München, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (M.B., S.F.)
| | - Stefanie Fenske
- Department of Pharmacy, Center for Drug Research (M.B., S.F.), Ludwig-Maximilians-Universität München, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (M.B., S.F.)
| | - Christian Wahl-Schott
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center Munich, Walter Brendel Centre of Experimental Medicine, Faculty of Medicine (K.H., C.P., C.W.-S.), Ludwig-Maximilians-Universität München, Germany
| |
Collapse
|
4
|
Burg S, Levi O, Elyagon S, Shapiro S, Murninkas M, Etzion S, Gradwohl G, Makarovsky D, Lichtenstein A, Gordon Y, Attali B, Etzion Y. The SK4 channel allosteric blocker, BA6b9, reduces atrial fibrillation substrate in rats with reduced ejection fraction. PNAS NEXUS 2024; 3:pgae192. [PMID: 38783894 PMCID: PMC11114471 DOI: 10.1093/pnasnexus/pgae192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
Atrial fibrillation (AF), the most common cardiac arrhythmia, is strongly associated with several comorbidities including heart failure (HF). AF in general, and specifically in the context of HF, is progressive in nature and associated with poor clinical outcomes. Current therapies for AF are limited in number and efficacy and do not target the underlying causes of atrial remodeling such as inflammation or fibrosis. We previously identified the calcium-activated SK4 K+ channels, which are preferentially expressed in the atria relative to the ventricles in both rat and human hearts, as attractive druggable target for AF treatment. Here, we examined the ability of BA6b9, a novel allosteric inhibitor of SK4 channels that targets the specific calmodulin-PIP2 binding domain, to alter AF susceptibility and atrial remodeling in a systolic HF rat postmyocardial infarction (post-MI) model. Daily BA6b9 injection (20 mg/kg/day) for 3 weeks starting 1-week post-MI prolonged the atrial effective refractory period, reduced AF induction and duration, and dramatically prevented atrial structural remodeling. In the post-MI left atrium (LA), pronounced upregulation of the SK4 K+ channel was observed, with corresponding increases in collagen deposition, α-SMA levels, and NLRP3 inflammasome expression. Strikingly, BA6b9 treatment reversed these changes while also significantly reducing the lateralization of the atrial connexin Cx43 in the LA of post-MI rats. Our findings indicate that the blockade of SK4 K+ channels using BA6b9 not only favors rhythm control but also remarkably reduces atrial structural remodeling, a property that is highly desirable for novel AF therapies, particularly in patients with comorbid HF.
Collapse
Affiliation(s)
- Shira Burg
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Or Levi
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Sigal Elyagon
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Shir Shapiro
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Michael Murninkas
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Sharon Etzion
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Gideon Gradwohl
- Medical Engineering Unit, The Jerusalem College of Technology, Jerusalem 9116001, Israel
| | - Daria Makarovsky
- Inter-Departmental Core Facility, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Alexandra Lichtenstein
- Inter-Departmental Core Facility, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yaara Gordon
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Bernard Attali
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yoram Etzion
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
5
|
Chen H, Liu H, Liu D, Fu Y, Yao Y, Cao Z, Peng Z, Yang M, Zhao Q. M2 macrophage‑derived exosomes alleviate KCa3.1 channel expression in rapidly paced HL‑1 myocytes via the NF‑κB (p65)/STAT3 signaling pathway. Mol Med Rep 2024; 29:55. [PMID: 38334149 PMCID: PMC10877089 DOI: 10.3892/mmr.2024.13179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
The present study was designed to explore the role of M2 macrophage‑derived exosomes (M2‑exos) on the KCa3.1 channel in a cellular atrial fibrillation (AF) model using rapidly paced HL‑1 myocytes. M2 macrophages and M2‑exos were isolated and identified. MicroRNA (miR)‑146a‑5p levels in M2 macrophages and M2‑exos were quantified using reverse transcription‑quantitative PCR (RT‑qPCR). HL‑1 myocytes were randomly divided into six groups: Control group, pacing group, pacing + coculture group (pacing HL‑1 cells cocultured with M2‑exos), pacing + mimic‑miR‑146a‑5p group, pacing + NC‑miR‑146a‑5p group and pacing + pyrrolidine dithiocarbamate (PDTC; a special blocker of the NF‑κB signaling pathway) group. Transmission electron microscopy, nanoparticle tracking analysis, western blotting, RT‑qPCR and immunohistochemistry were performed in the present study. A whole‑cell clamp was also applied to record the current density of KCa3.1 and action potential duration (APD) in each group. The results revealed that miR‑146a‑5p was highly expressed in both M2 macrophages and M2‑exos. Pacing HL‑1 cells led to a shorter APD, an increased KCa3.1 current density and higher protein levels of KCa3.1, phosphorylated (p‑)NF‑κB p65, p‑STAT3 and IL‑1β compared with the control group. M2‑exos, miR‑146a‑5p‑mimic and PDTC both reduced the protein expression of KCa3.1, p‑NF‑κB p65, p‑STAT3 and IL‑1β and the current density of KCa3.1, resulting in a longer APD in the pacing HL‑1 cells. In conclusion, M2‑exos and their cargo, which comprised miR‑146a‑5p, decreased KCa3.1 expression and IL‑1β secretion in pacing HL‑1 cells via the NF‑κB/STAT3 signaling pathway, limiting the shorter APD caused by rapid pacing.
Collapse
Affiliation(s)
- Huiyu Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Huafen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Dishiwen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yuntao Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yajun Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhen Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhibin Peng
- Department of Cardiology, Yidu People's Hospital, Yidu, Hubei 443000, P.R. China
| | - Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei 430060, P.R. China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
6
|
Vera OD, Wulff H, Braun AP. Endothelial KCa channels: Novel targets to reduce atherosclerosis-driven vascular dysfunction. Front Pharmacol 2023; 14:1151244. [PMID: 37063294 PMCID: PMC10102451 DOI: 10.3389/fphar.2023.1151244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023] Open
Abstract
Elevated levels of cholesterol in the blood can induce endothelial dysfunction, a condition characterized by impaired nitric oxide production and decreased vasodilatory capacity. Endothelial dysfunction can promote vascular disease, such as atherosclerosis, where macrophages accumulate in the vascular intima and fatty plaques form that impair normal blood flow in conduit arteries. Current pharmacological strategies to treat atherosclerosis mostly focus on lipid lowering to prevent high levels of plasma cholesterol that induce endothelial dysfunction and atherosclerosis. While this approach is effective for most patients with atherosclerosis, for some, lipid lowering is not enough to reduce their cardiovascular risk factors associated with atherosclerosis (e.g., hypertension, cardiac dysfunction, stroke, etc.). For such patients, additional strategies targeted at reducing endothelial dysfunction may be beneficial. One novel strategy to restore endothelial function and mitigate atherosclerosis risk is to enhance the activity of Ca2+-activated K+ (KCa) channels in the endothelium with positive gating modulator drugs. Here, we review the mechanism of action of these small molecules and discuss their ability to improve endothelial function. We then explore how this strategy could mitigate endothelial dysfunction in the context of atherosclerosis by examining how KCa modulators can improve cardiovascular function in other settings, such as aging and type 2 diabetes. Finally, we consider questions that will need to be addressed to determine whether KCa channel activation could be used as a long-term add-on to lipid lowering to augment atherosclerosis treatment, particularly in patients where lipid-lowering is not adequate to improve their cardiovascular health.
Collapse
Affiliation(s)
- O. Daniel Vera
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, United States
| | - Andrew P. Braun
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- *Correspondence: Andrew P. Braun,
| |
Collapse
|
7
|
Arbel Ganon L, Davoodi M, Alexandrovich A, Yaniv Y. Synergy between Membrane Currents Prevents Severe Bradycardia in Mouse Sinoatrial Node Tissue. Int J Mol Sci 2023; 24:ijms24065786. [PMID: 36982861 PMCID: PMC10051777 DOI: 10.3390/ijms24065786] [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: 01/16/2023] [Revised: 03/04/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
Bradycardia is initiated by the sinoatrial node (SAN), which is regulated by a coupled-clock system. Due to the clock coupling, reduction in the 'funny' current (If), which affects SAN automaticity, can be compensated, thus preventing severe bradycardia. We hypothesize that this fail-safe system is an inherent feature of SAN pacemaker cells and is driven by synergy between If and other ion channels. This work aimed to characterize the connection between membrane currents and their underlying mechanisms in SAN cells. SAN tissues were isolated from C57BL mice and Ca2+ signaling was measured in pacemaker cells within them. A computational model of SAN cells was used to understand the interactions between cell components. Beat interval (BI) was prolonged by 54 ± 18% (N = 16) and 30 ± 9% (N = 21) in response to If blockade, by ivabradine, or sodium current (INa) blockade, by tetrodotoxin, respectively. Combined drug application had a synergistic effect, manifested by a BI prolonged by 143 ± 25% (N = 18). A prolongation in the local Ca2+ release period, which reports on the level of crosstalk within the coupled-clock system, was measured and correlated with the prolongation in BI. The computational model predicted that INa increases in response to If blockade and that this connection is mediated by changes in T and L-type Ca2+ channels.
Collapse
Affiliation(s)
- Limor Arbel Ganon
- Laboratory of Bioelectric and Bioenergetic Systems, Faculty of Biomedical Engineering, Technion-IIT, Haifa 3200003, Israel
| | - Moran Davoodi
- Laboratory of Bioelectric and Bioenergetic Systems, Faculty of Biomedical Engineering, Technion-IIT, Haifa 3200003, Israel
| | - Alexandra Alexandrovich
- Laboratory of Bioelectric and Bioenergetic Systems, Faculty of Biomedical Engineering, Technion-IIT, Haifa 3200003, Israel
| | - Yael Yaniv
- Laboratory of Bioelectric and Bioenergetic Systems, Faculty of Biomedical Engineering, Technion-IIT, Haifa 3200003, Israel
| |
Collapse
|
8
|
Li K, Jiang Y, Zeng Y, Zhou Y. Advances in Ion Channel, Non-Desmosomal Variants and Autophagic Mechanisms Implicated in Arrhythmogenic Cardiomyopathy. Curr Issues Mol Biol 2023; 45:2186-2200. [PMID: 36975511 PMCID: PMC10047275 DOI: 10.3390/cimb45030141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/11/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a heterogeneous disorder characterized by the replacement of cardiac myocytes with fibro-fatty tissues, leading to abnormal excitation-contraction (EC) coupling and a range of malignant events, such as ventricular tachycardia (VT), sudden cardiac death/arrest (SCD/A) and heart failure (HF). The concept of ACM has recently been ex-tended to include right ventricular cardiomyopathy (ARVC), left ventricular cardiomyopathy (ALVC) and biventricular cardiomyopathy. ARVC is generally seen as the most common type of ACM. The pathogenesis of ACM involves mutation variants in desmosomal or non-desmosomal gene loci, as well as various external factors, such as intense exercise, stress and infections. Ion channel alterations, autophagy and non-desmosomal variants are also important components in the development of ACM. As clinical practice enters the era of precision therapy, it is important to review recent studies on these topics to better diagnose and treat the molecular phase of ACM.
Collapse
Affiliation(s)
- Kexin Li
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215000, China
- Institution for Hypertension of Soochow University, Suzhou 215000, China
| | - Yufeng Jiang
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215000, China
- Institution for Hypertension of Soochow University, Suzhou 215000, China
| | - Yiyao Zeng
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215000, China
- Institution for Hypertension of Soochow University, Suzhou 215000, China
| | - Yafeng Zhou
- Department of Cardiology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215000, China
- Institution for Hypertension of Soochow University, Suzhou 215000, China
- Correspondence: ; Tel.: +86-512-65955026
| |
Collapse
|
9
|
Li Y, Dinkel H, Pakalniskyte D, Busley AV, Cyganek L, Zhong R, Zhang F, Xu Q, Maywald L, Aweimer A, Huang M, Liao Z, Meng Z, Yan C, Prädel T, Rose L, Moscu‐Gregor A, Hohn A, Yang Z, Qiao L, Mügge A, Zhou X, Akin I, El‐Battrawy I. Novel insights in the pathomechanism of Brugada syndrome and fever-related type 1 ECG changes in a preclinical study using human-induced pluripotent stem cell-derived cardiomyocytes. Clin Transl Med 2023; 13:e1130. [PMID: 36881552 PMCID: PMC9990896 DOI: 10.1002/ctm2.1130] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Brugada syndrome (BrS) is causing sudden cardiac death (SCD) mainly at young age. Studying the underlying mechanisms associated with BrS type I electrocardiogram (ECG) changes in the presence of fever and roles of autophagy for BrS remains lacking. OBJECTIVES We sought to study the pathogenic role of an SCN5A gene variant for BrS with fever-induced type 1 ECG phenotype. In addition, we studied the role of inflammation and autophagy in the pathomechanism of BrS. METHODS Human-induced pluripotent stem cell (hiPSC) lines from a BrS patient harboring a pathogenic variant (c.3148G>A/p. Ala1050Thr) in SCN5A and two healthy donors (non-BrS) and a CRISPR/Cas9 site-corrected cell line (BrS-corr) were differentiated into cardiomyocytes (hiPSC-CMs) for the study. RESULTS Reductions of Nav 1.5 expression, peak sodium channel current (INa ) and upstroke velocity (Vmax ) of action potentials with an increase in arrhythmic events were detected in BrS compared to non-BrS and BrS-corr cells. Increasing the cell culture temperature from 37 to 40°C (fever-like state) exacerbated the phenotypic changes in BrS cells. The fever-effects were enhanced by protein kinase A (PKA) inhibitor but reversed by PKA activator. Lipopolysaccharides (LPS) but not increased temperature up to 40°C enhanced the autophagy level in BrS-hiPSC-CMs by increasing reactive oxidative species and inhibiting PI3K/AKT signalling, and hence exacerbated the phenotypic changes. LPS enhanced high temperature-related effect on peak INa shown in BrS hiPSC-CMs. Effects of LPS and high temperature were not detected in non-BrS cells. CONCLUSIONS The study demonstrated that the SCN5A variant (c.3148G>A/p.Ala1050Thr) caused loss-of-function of sodium channels and increased the channel sensitivity to high temperature and LPS challenge in hiPSC-CMs from a BrS cell line with this variant but not in two non-BrS hiPSC-CM lines. The results suggest that LPS may exacerbate BrS phenotype via enhancing autophagy, whereas fever may exacerbate BrS phenotype via inhibiting PKA-signalling in BrS cardiomyocytes with but probably not limited to this variant.
Collapse
Affiliation(s)
- Yingrui Li
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
- DZHK (German Center for Cardiovascular Research)Partner SiteHeidelberg‐Mannheim and GöttingenMannheimGermany
| | - Hendrik Dinkel
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
- DZHK (German Center for Cardiovascular Research)Partner SiteHeidelberg‐Mannheim and GöttingenMannheimGermany
| | - Dalia Pakalniskyte
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
- DZHK (German Center for Cardiovascular Research)Partner SiteHeidelberg‐Mannheim and GöttingenMannheimGermany
| | - Alexandra Viktoria Busley
- DZHK (German Center for Cardiovascular Research)Partner SiteHeidelberg‐Mannheim and GöttingenMannheimGermany
- Stem Cell UnitClinic for Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | - Lukas Cyganek
- DZHK (German Center for Cardiovascular Research)Partner SiteHeidelberg‐Mannheim and GöttingenMannheimGermany
- Stem Cell UnitClinic for Cardiology and PneumologyUniversity Medical Center GöttingenGöttingenGermany
| | - Rujia Zhong
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
| | - Feng Zhang
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
| | - Qiang Xu
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan ProvinceInstitute of Cardiovascular ResearchSouthwest Medical UniversityLuzhouChina
| | - Lasse Maywald
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
- DZHK (German Center for Cardiovascular Research)Partner SiteHeidelberg‐Mannheim and GöttingenMannheimGermany
| | - Assem Aweimer
- Department of Cardiology and AngiologyBergmannsheil University HospitalsRuhr University of BochumBochumGermany
| | - Mengying Huang
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
| | - Zhenxing Liao
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
| | - Zenghui Meng
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
| | - Chen Yan
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
| | - Timo Prädel
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
- DZHK (German Center for Cardiovascular Research)Partner SiteHeidelberg‐Mannheim and GöttingenMannheimGermany
| | - Lena Rose
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
| | | | - Alyssa Hohn
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
| | - Zhen Yang
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
| | - Lin Qiao
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
| | - Andreas Mügge
- Department of Cardiology and AngiologyBergmannsheil University HospitalsRuhr University of BochumBochumGermany
| | - Xiaobo Zhou
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
- DZHK (German Center for Cardiovascular Research)Partner SiteHeidelberg‐Mannheim and GöttingenMannheimGermany
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan ProvinceInstitute of Cardiovascular ResearchSouthwest Medical UniversityLuzhouChina
| | - Ibrahim Akin
- First Department of MedicineFaculty of MedicineUniversity Medical Centre Mannheim (UMM)Heidelberg UniversityMannheimGermany
- DZHK (German Center for Cardiovascular Research)Partner SiteHeidelberg‐Mannheim and GöttingenMannheimGermany
| | - Ibrahim El‐Battrawy
- Department of Cardiology and AngiologyBergmannsheil University HospitalsRuhr University of BochumBochumGermany
| |
Collapse
|
10
|
Zhao H, Wang X, Tang Y, Zhao Q, Huang C. Inhibition of intermittent calcium-activated potassium channel (SK4) attenuates Ang II-induced hypertrophy of human-induced stem cell-derived cardiomyocytes via targeting Ras-Raf-MEK1/2-ERK1/2 and CN-NFAT signaling pathways. Cell Biol Int 2023; 47:480-491. [PMID: 36273427 DOI: 10.1002/cbin.11948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/20/2022] [Accepted: 10/15/2022] [Indexed: 01/17/2023]
Abstract
Cardiac hypertrophy caused by angiotensin II (Ang II) is essential for the pathological process of heart failure. The intermediate calcium-activated potassium channel (SK4) has been shown to be involved in the process of the inflammatory response, cell proliferation, and apoptosis. However, the role of SK4 in cardiac hypertrophy has not been elucidated. Cardiac hypertrophy in human-induced pluripotent stem cells-derived cardiomyocytes (HiPSC-CMs) was induced by Ang II. Cells were transfected with SK4 adenovirus or treated with SK4 inhibitor (TRAM-34). TUNEL staining was used to assess the levels of apoptosis. Real-time polymerase chain reaction and Western blot analysis were used to measure messenger RNA (mRNA) and protein levels, respectively. The present results showed that SK4 expression was upregulated in HiPSC-CMs stimulated by Ang II. The downregulation of SK4 by a specific inhibitor TRAM-34 markedly ameliorated cardiac hypertrophy (reflected by the mRNA levels of atrial natriuretic peptide, brain natriuretic peptide, and β-myosin heavy chain) and apoptosis (reflected by the level of Caspase 3, Bax, and Bcl-2) induced by Ang II treatment. The action of SK4 in cardiac hypertrophy was mediated by Ras-Raf-mitogen-activated protein kinases 1/2 (MEK1/2)-extracellular-regulated protein kinases 1/2 (ERK1/2) and calcineurin (CN)-nuclear factors of activated T cells (NFAT) activation. Our studies demonstrated that inhibition of SK4 significantly alleviated cardiac hypertrophy induced by Ang II in hiPSC-CMs by targeting Ras-Raf-MEK1/2-ERK1/2 signaling and CN-NFAT signaling pathway. Our studies suggest that SK4 may serve as a potential therapeutic target that could delay hypertrophy.
Collapse
Affiliation(s)
- Hongyi Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P. R. China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P. R. China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P. R. China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P. R. China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Cardiovascular Research Institute, Wuhan University, Wuhan, P. R. China.,Hubei Key Laboratory of Cardiology, Wuhan, P. R. China
| |
Collapse
|
11
|
Liu D, Chen H, Fu Y, Yao Y, He S, Wang Y, Cao Z, Wang X, Yang M, Zhao Q. KCa3.1 Promotes Proinflammatory Exosome Secretion by Activating AKT/Rab27a in Atrial Myocytes during Rapid Pacing. Cardiovasc Ther 2023; 2023:3939360. [PMID: 37035755 PMCID: PMC10079387 DOI: 10.1155/2023/3939360] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Purpose The aim of this study was to investigate the role of the medium-conductance calcium-activated potassium channel (KCNN4, KCa3.1) in the secretion of proinflammatory exosomes by atrial myocytes. Methods Eighteen beagles were randomly divided into the sham group (n = 6), pacing group (n = 6), and pacing+TRAM-34 group (n = 6). Electrophysiological data, such as the effective refractory period, atrial fibrillation (AF) induction, and AF duration, were collected by programmed stimulation. Atrial tissues were subjected to hematoxylin and eosin, Masson's trichrome, and immunofluorescence staining. The expression of KCa3.1 and Rab27a was assessed by immunohistochemistry and western blotting. The downstream signaling pathways involved in KCa3.1 were examined by rapid pacing or overexpressing KCNN4 in HL-1 cells. Results Atrial rapid pacing significantly induced electrical remodeling, inflammation, fibrosis, and exosome secretion in the canine atrium, while TRAM-34 (KCa3.1 blocker) inhibited these changes. Compared with those in control HL-1 cells, the levels of exosome markers and inflammatory factors were increased in pacing HL-1 cells. Furthermore, the levels of CD68 and iNOS in macrophages incubated with exosomes derived from HL-1 cells were higher in the pacing-exo group than in the control group. More importantly, KCa3.1 regulated exosome secretion through the AKT/Rab27a signaling pathway. Similarly, inhibiting the downstream signaling pathway of KCa3.1 significantly inhibited exosome secretion. Conclusions KCa3.1 promotes proinflammatory exosome secretion through the AKT/Rab27a signaling pathway. Inhibiting the KCa3.1/AKT/Rab27a signaling pathway reduces myocardial tissue structural remodeling in AF.
Collapse
Affiliation(s)
- Dishiwen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Huiyu Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yuntao Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yajun Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Shanqing He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Youcheng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Zhen Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Xuewen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| |
Collapse
|
12
|
Guarina L, Moghbel AN, Pourhosseinzadeh MS, Cudmore RH, Sato D, Clancy CE, Santana LF. Biological noise is a key determinant of the reproducibility and adaptability of cardiac pacemaking and EC coupling. J Gen Physiol 2022; 154:e202012613. [PMID: 35482009 PMCID: PMC9059386 DOI: 10.1085/jgp.202012613] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/16/2022] [Accepted: 04/07/2022] [Indexed: 12/23/2022] Open
Abstract
Each heartbeat begins with the generation of an action potential in pacemaking cells in the sinoatrial node. This signal triggers contraction of cardiac muscle through a process termed excitation-contraction (EC) coupling. EC coupling is initiated in dyadic structures of cardiac myocytes, where ryanodine receptors in the junctional sarcoplasmic reticulum come into close apposition with clusters of CaV1.2 channels in invaginations of the sarcolemma. Cooperative activation of CaV1.2 channels within these clusters causes a local increase in intracellular Ca2+ that activates the juxtaposed ryanodine receptors. A salient feature of healthy cardiac function is the reliable and precise beat-to-beat pacemaking and amplitude of Ca2+ transients during EC coupling. In this review, we discuss recent discoveries suggesting that the exquisite reproducibility of this system emerges, paradoxically, from high variability at subcellular, cellular, and network levels. This variability is attributable to stochastic fluctuations in ion channel trafficking, clustering, and gating, as well as dyadic structure, which increase intracellular Ca2+ variance during EC coupling. Although the effects of these large, local fluctuations in function and organization are sometimes negligible at the macroscopic level owing to spatial-temporal summation within and across cells in the tissue, recent work suggests that the "noisiness" of these intracellular Ca2+ events may either enhance or counterintuitively reduce variability in a context-dependent manner. Indeed, these noisy events may represent distinct regulatory features in the tuning of cardiac contractility. Collectively, these observations support the importance of incorporating experimentally determined values of Ca2+ variance in all EC coupling models. The high reproducibility of cardiac contraction is a paradoxical outcome of high Ca2+ signaling variability at subcellular, cellular, and network levels caused by stochastic fluctuations in multiple processes in time and space. This underlying stochasticity, which counterintuitively manifests as reliable, consistent Ca2+ transients during EC coupling, also allows for rapid changes in cardiac rhythmicity and contractility in health and disease.
Collapse
Affiliation(s)
- Laura Guarina
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| | - Ariana Neelufar Moghbel
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| | | | - Robert H. Cudmore
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| | - Daisuke Sato
- Department of Pharmacology, University of California Davis School of Medicine, Davis, CA
| | - Colleen E. Clancy
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| | - Luis Fernando Santana
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA
| |
Collapse
|
13
|
Allosteric inhibitors targeting the calmodulin-PIP2 interface of SK4 K + channels for atrial fibrillation treatment. Proc Natl Acad Sci U S A 2022; 119:e2202926119. [PMID: 35969786 PMCID: PMC9407317 DOI: 10.1073/pnas.2202926119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Ca2+-activated SK4 K+ channel is gated by Ca2+-calmodulin (CaM) and is expressed in immune cells, brain, and heart. A cryoelectron microscopy (cryo-EM) structure of the human SK4 K+ channel recently revealed four CaM molecules per channel tetramer, where the apo CaM C-lobe and the holo CaM N-lobe interact with the proximal carboxyl terminus and the linker S4-S5, respectively, to gate the channel. Here, we show that phosphatidylinositol 4-5 bisphosphate (PIP2) potently activates SK4 channels by docking to the boundary of the CaM-binding domain. An allosteric blocker, BA6b9, was designed to act to the CaM-PIP2-binding domain, a previously untargeted region of SK4 channels, at the interface of the proximal carboxyl terminus and the linker S4-S5. Site-directed mutagenesis, molecular docking, and patch-clamp electrophysiology indicate that BA6b9 inhibits SK4 channels by interacting with two specific residues, Arg191 and His192 in the linker S4-S5, not conserved in SK1-SK3 subunits, thereby conferring selectivity and preventing the Ca2+-CaM N-lobe from properly interacting with the channel linker region. Immunohistochemistry of the SK4 channel protein in rat hearts showed a widespread expression in the sarcolemma of atrial myocytes, with a sarcomeric striated Z-band pattern, and a weaker occurrence in the ventricle but a marked incidence at the intercalated discs. BA6b9 significantly prolonged atrial and atrioventricular effective refractory periods in rat isolated hearts and reduced atrial fibrillation induction ex vivo. Our work suggests that inhibition of SK4 K+ channels by targeting drugs to the CaM-PIP2-binding domain provides a promising anti-arrhythmic therapy.
Collapse
|
14
|
Ma Y, Fu Y, Wang Y, Yang M, Yao Y, He S, Liu D, Cao Z, Wang X, Tang Y, Zhao Q, Huang C. Blocking Intermediate-Conductance Calcium-Activated Potassium Channels in the Macrophages Around Ganglionated Plexi Suppresses Atrial Fibrillation Vulnerability in Canines With Rapid Atrial Pacing. Front Physiol 2022; 13:837412. [PMID: 35431996 PMCID: PMC9010666 DOI: 10.3389/fphys.2022.837412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Previous studies have indicated that ganglionated plexi (GP) function influences atrial fibrillation (AF) vulnerability, and intermediate-conductance calcium-activated potassium channels (SK4) have a close relationship with cardiomyocyte automaticity and the induction of AF. However, the effects of the SK4 inhibitor on GP function and AF vulnerability are unknown. Eighteen beagles were randomly divided into a control group (n = 6), rapid atrial pacing (RAP) group (n = 6), and triarylmethane-34 (TRAM-34, an SK4 inhibitor) group (n = 6). TRAM-34 (0.3 ml, 15 mmol/L) and saline were locally injected into GPs in the TRAM-34 group dogs and dogs from the other groups, respectively. After that, dogs in the RAP and TRAM-34 groups were subjected to RAP, and the neural activity of anterior right GP (ARGP) and atrial electrophysiology were measured. The levels of inflammatory cytokines and function of macrophages in the ARGP were measured in the three groups. At 10 min after TRAM-34 injection, ARGP activity and atrial electrophysiology did not significantly change. The atrial pacing shortened effective refractory period (ERP) values at all sites and increased the AF vulnerability and ARGP neural activity, while TRAM-34 reversed these changes. The levels of CD68 + cells, induced nitric oxide synthase (iNOS), interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α in the ARGP tissues were higher in the RAP group and TRAM-34 group than they were in the control group. Furthermore, the levels of the CD68 + cells, iNOS, and inflammatory cytokines in the ARGP tissues were higher in the pacing group than those in the TRAM-34 group. Based on these results, administration of TRAM-34 into the atrial GP can suppress GP activity and AF vulnerability during atrial pacing. The effects of TRAM-34 might be related to macrophage polarization and the inflammatory response of GP.
Collapse
Affiliation(s)
- Yazhe Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yuntao Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Youcheng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yajun Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanqing He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Dishiwen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- *Correspondence: Qingyan Zhao, ; Congxin Huang,
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
- *Correspondence: Qingyan Zhao, ; Congxin Huang,
| |
Collapse
|
15
|
Wang Y, Yao Y, Ma Y, He S, Yang M, Cao Z, Liudi S, Fu Y, Chen H, Wang X, Huang C, Zhao Q. Blockade of SK4 channels suppresses atrial fibrillation by attenuating atrial fibrosis in canines with prolonged atrial pacing. Int J Med Sci 2022; 19:1995-2007. [PMID: 36483596 PMCID: PMC9724246 DOI: 10.7150/ijms.69626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 10/28/2022] [Indexed: 11/24/2022] Open
Abstract
Background: We previously found that intermediate conductance Ca2+-activated K+ channel (SK4) might be an important target in atrial fibrillation (AF). Objective: To investigate the role of SK4 in AF maintenance. Methods: Twenty beagles were randomly assigned to the sham group (n=6), pacing group (n=7), and pacing+TRAM-34 group (n=7). Rapid atrial pacing continued for 7 days in the pacing and TRAM-34 groups. During the pacing, the TRAM-34 group received TRAM-34 intravenous injection (10 mg/Kg) 3 times per day. Atrial fibroblasts isolated from canines were treated with angiotensin II or adenovirus carrying the SK4 gene (Ad-SK4) to overexpress SK4 channels. Results: TRAM-34 treatment significantly suppressed the increased intra-atrial conducting time (CT) and AF duration in canines after rapid atrial pacing (P<0.05). Compared with the sham group, the expression of SK4 in atria was higher in the pacing group, which was associated with an increased number of myofibroblasts and levels of extracellular matrix in atrium (all P<0.05), and this effect was reversed by TRAM-34 treatment (all P<0.05). In atrial fibroblasts, the increased expression of SK4 induced by angiotensin II stimulation or Ad-SK4 transfection contributed to higher levels of P38, ERK1/2 and their downstream factors c-Jun and c-Fos, leading to the increased expression of α-SMA (all P<0.05), and all these increases were markedly reduced by TRAM-34 treatment. Conclusion: SK4 blockade suppressed AF by attenuating cardiac fibroblast activity and atrial fibrosis, which was realized through not only a decrease in fibrogenic factors but also inhibition of fibrotic signaling pathways.
Collapse
Affiliation(s)
- Youcheng Wang
- Xiaogan Hospital Affiliated to Wuhan University of Science and Technology, Xiaogan, China.,Department of Cardiology, Renmin Hospital of Wuhan University Wuhan City, Wuchang, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yajun Yao
- Department of Cardiology, Renmin Hospital of Wuhan University Wuhan City, Wuchang, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yazhe Ma
- Department of Cardiology, Renmin Hospital of Wuhan University Wuhan City, Wuchang, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanqing He
- Department of Cardiology, Renmin Hospital of Wuhan University Wuhan City, Wuchang, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University Wuhan City, Wuchang, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen Cao
- Department of Cardiology, Renmin Hospital of Wuhan University Wuhan City, Wuchang, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shiwen Liudi
- Department of Cardiology, Renmin Hospital of Wuhan University Wuhan City, Wuchang, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yuntao Fu
- Department of Cardiology, Renmin Hospital of Wuhan University Wuhan City, Wuchang, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Huiyu Chen
- Department of Cardiology, Renmin Hospital of Wuhan University Wuhan City, Wuchang, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University Wuhan City, Wuchang, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University Wuhan City, Wuchang, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University Wuhan City, Wuchang, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
16
|
He S, Wang Y, Yao Y, Cao Z, Yin J, Zi L, Chen H, Fu Y, Wang X, Zhao Q. Inhibition of KCa3.1 Channels Suppresses Atrial Fibrillation via the Attenuation of Macrophage Pro-inflammatory Polarization in a Canine Model With Prolonged Rapid Atrial Pacing. Front Cardiovasc Med 2021; 8:656631. [PMID: 34136541 PMCID: PMC8200470 DOI: 10.3389/fcvm.2021.656631] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/04/2021] [Indexed: 12/04/2022] Open
Abstract
Aims: To investigate the role of KCa3. 1 inhibition in macrophage pro-inflammatory polarization and vulnerability to atrial fibrillation (AF) in a canine model with prolonged rapid atrial pacing. Materials and Methods: Twenty beagle dogs (weighing 8–10 kg) were randomly assigned to a sham group (n = 6), pacing group (n = 7) and pacing+TRAM-34 group (n = 7). An experimental model of AF was established by rapid pacing. TRAM-34 was administered to the Pacing+TRAM-34 group by slow intravenous injection (10 mg/kg), 3 times each day. After 7 days of pacing, the electrophysiology was measured in vivo. The levels of interleukin-1β (IL-1β), monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), CD68, c-Fos, p38, and NF-κB p65 in both atriums were measured by Western blotting, and the levels of inducible nitric oxide synthase (iNOS) and arginase1 (Arg-1) were measured by real-time PCR. Macrophage and KCa3.1 in macrophage in the atrium were quantized following double labeled immunofluorescent. Results: Greater inducibility of AF, an extended duration of AF and lower atrial effective refractory period (AERP) were observed in the pacing group compared with those in the sham group. Both CD68-labeled macrophage and the expression of KCa3.1 in macrophage were elevated in the pacing group and inhibited by TRAM-34, led to higher iNOS expression, lower Arg-1 expression, elevated levels of IL-1β, MCP-1, and TNF-α in the atria, which could be reversed by TRAM-34 treatment (all P < 0.01). KCa3.1 channels were possibly activated via the p38/AP-1/NF-κB signaling pathway. Conclusions: Inhibition of KCa3.1 suppresses vulnerability to AF by attenuating macrophage pro-inflammatory polarization and inflammatory cytokine secretion in a canine model with prolonged rapid atrial pacing.
Collapse
Affiliation(s)
- Shanqing He
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Youcheng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yajun Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Junkui Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liuliu Zi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Huiyu Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yuntao Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
17
|
Targeting of Potassium Channels in Cardiac Arrhythmias. Trends Pharmacol Sci 2021; 42:491-506. [PMID: 33858691 DOI: 10.1016/j.tips.2021.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
Cardiomyocytes are endowed with a complex repertoire of ion channels, responsible for the generation of action potentials (APs), travelling waves of electrical excitation, propagating throughout the heart and leading to cardiac contractions. Cardiac AP waveforms are shaped by a striking diversity of K+ channels. The pivotal role of K+ channels in cardiac health and disease is underscored by the dramatic impact that K+ channel dysfunction has on cardiac arrhythmias. The development of drugs targeted to specific K+ channels is expected to provide an optimized approach to antiarrhythmic therapy. Here, we review the functional roles of cardiac potassium channels under normal and diseased states. We survey current antiarrhythmic drugs (AADs) targeted to voltage-gated and Ca2+-activated K+ channels and highlight future research opportunities.
Collapse
|
18
|
Wallace MJ, El Refaey M, Mesirca P, Hund TJ, Mangoni ME, Mohler PJ. Genetic Complexity of Sinoatrial Node Dysfunction. Front Genet 2021; 12:654925. [PMID: 33868385 PMCID: PMC8047474 DOI: 10.3389/fgene.2021.654925] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
The pacemaker cells of the cardiac sinoatrial node (SAN) are essential for normal cardiac automaticity. Dysfunction in cardiac pacemaking results in human sinoatrial node dysfunction (SND). SND more generally occurs in the elderly population and is associated with impaired pacemaker function causing abnormal heart rhythm. Individuals with SND have a variety of symptoms including sinus bradycardia, sinus arrest, SAN block, bradycardia/tachycardia syndrome, and syncope. Importantly, individuals with SND report chronotropic incompetence in response to stress and/or exercise. SND may be genetic or secondary to systemic or cardiovascular conditions. Current management of patients with SND is limited to the relief of arrhythmia symptoms and pacemaker implantation if indicated. Lack of effective therapeutic measures that target the underlying causes of SND renders management of these patients challenging due to its progressive nature and has highlighted a critical need to improve our understanding of its underlying mechanistic basis of SND. This review focuses on current information on the genetics underlying SND, followed by future implications of this knowledge in the management of individuals with SND.
Collapse
Affiliation(s)
- Michael J. Wallace
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Mona El Refaey
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Pietro Mesirca
- CNRS, INSERM, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, France
- Laboratory of Excellence ICST, Montpellier, France
| | - Thomas J. Hund
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, United States
| | - Matteo E. Mangoni
- CNRS, INSERM, Institut de Génomique Fonctionnelle, Université de Montpellier, Montpellier, France
- Laboratory of Excellence ICST, Montpellier, France
| | - Peter J. Mohler
- Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
19
|
Mesirca P, Fedorov VV, Hund TJ, Torrente AG, Bidaud I, Mohler PJ, Mangoni ME. Pharmacologic Approach to Sinoatrial Node Dysfunction. Annu Rev Pharmacol Toxicol 2021; 61:757-778. [PMID: 33017571 PMCID: PMC7790915 DOI: 10.1146/annurev-pharmtox-031120-115815] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The spontaneous activity of the sinoatrial node initiates the heartbeat. Sino-atrial node dysfunction (SND) and sick sinoatrial (sick sinus) syndrome are caused by the heart's inability to generate a normal sinoatrial node action potential. In clinical practice, SND is generally considered an age-related pathology, secondary to degenerative fibrosis of the heart pacemaker tissue. However, other forms of SND exist, including idiopathic primary SND, which is genetic, and forms that are secondary to cardiovascular or systemic disease. The incidence of SND in the general population is expected to increase over the next half century, boosting the need to implant electronic pacemakers. During the last two decades, our knowledge of sino-atrial node physiology and of the pathophysiological mechanisms underlying SND has advanced considerably. This review summarizes the current knowledge about SND mechanisms and discusses the possibility of introducing new pharmacologic therapies for treating SND.
Collapse
Affiliation(s)
- Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| | - Vadim V Fedorov
- Frick Center for Heart Failure and Arrhythmia at the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Wexner Medical Center, Columbus, Ohio 43210, USA
| | - Thomas J Hund
- Frick Center for Heart Failure and Arrhythmia at the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Angelo G Torrente
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| | - Isabelle Bidaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| | - Peter J Mohler
- Frick Center for Heart Failure and Arrhythmia at the Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Wexner Medical Center, Columbus, Ohio 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio 43210, USA
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 34096 Montpellier, France;
- LabEx Ion Channels Science and Therapeutics (ICST), 06560 Nice, France
| |
Collapse
|
20
|
Weisbrod D. Small and Intermediate Calcium Activated Potassium Channels in the Heart: Role and Strategies in the Treatment of Cardiovascular Diseases. Front Physiol 2020; 11:590534. [PMID: 33329039 PMCID: PMC7719780 DOI: 10.3389/fphys.2020.590534] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022] Open
Abstract
Calcium-activated potassium channels are a heterogeneous family of channels that, despite their different biophysical characteristics, structures, and pharmacological signatures, play a role of transducer between the ubiquitous intracellular calcium signaling and the electric variations of the membrane. Although this family of channels was extensively described in various excitable and non-excitable tissues, an increasing amount of evidences shows their functional role in the heart. This review aims to focus on the physiological role and the contribution of the small and intermediate calcium-activated potassium channels in cardiac pathologies.
Collapse
|
21
|
Cardiac Fibroblast-Induced Pluripotent Stem Cell-Derived Exosomes as a Potential Therapeutic Mean for Heart Failure. Int J Mol Sci 2020; 21:ijms21197215. [PMID: 33003641 PMCID: PMC7582575 DOI: 10.3390/ijms21197215] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 12/20/2022] Open
Abstract
The limited regenerative capacity of the injured myocardium leads to remodeling and often heart failure. Novel therapeutic approaches are essential. Induced pluripotent stem cells (iPSC) differentiated into cardiomyocytes are a potential future therapeutics. We hypothesized that organ-specific reprogramed fibroblasts may serve an advantageous source for future cardiomyocytes. Moreover, exosomes secreted from those cells may have a beneficial effect on cardiac differentiation and/or function. We compared RNA from different sources of human iPSC using chip gene expression. Protein expression was evaluated as well as exosome micro-RNA levels and their impact on embryoid bodies (EBs) differentiation. Statistical analysis identified 51 genes that were altered (p ≤ 0.05), and confirmed in the protein level, cardiac fibroblasts-iPSCs (CF-iPSCs) vs. dermal fibroblasts-iPSCs (DF-iPSCs). Several miRs were altered especially miR22, a key regulator of cardiac hypertrophy and remodeling. Lower expression of miR22 in CF-iPSCs vs. DF-iPSCs was observed. EBs treated with these exosomes exhibited more beating EBs p = 0.05. vs. control. We identify CF-iPSC and its exosomes as a potential source for cardiac recovery induction. The decrease in miR22 level points out that our CF-iPSC-exosomes are naïve of congestive heart cell memory, making them a potential biological source for future therapy for the injured heart.
Collapse
|
22
|
Torrente AG, Mesirca P, Bidaud I, Mangoni ME. Channelopathies of voltage-gated L-type Cav1.3/α 1D and T-type Cav3.1/α 1G Ca 2+ channels in dysfunction of heart automaticity. Pflugers Arch 2020; 472:817-830. [PMID: 32601767 DOI: 10.1007/s00424-020-02421-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/12/2020] [Accepted: 06/19/2020] [Indexed: 10/24/2022]
Abstract
The heart automaticity is a fundamental physiological function in vertebrates. The cardiac impulse is generated in the sinus node by a specialized population of spontaneously active myocytes known as "pacemaker cells." Failure in generating or conducting spontaneous activity induces dysfunction in cardiac automaticity. Several families of ion channels are involved in the generation and regulation of the heart automaticity. Among those, voltage-gated L-type Cav1.3 (α1D) and T-type Cav3.1 (α1G) Ca2+ channels play important roles in the spontaneous activity of pacemaker cells. Ca2+ channel channelopathies specifically affecting cardiac automaticity are considered rare. Recent research on familial disease has identified mutations in the Cav1.3-encoding CACNA1D gene that underlie congenital sinus node dysfunction and deafness (OMIM # 614896). In addition, both Cav1.3 and Cav3.1 channels have been identified as pathophysiological targets of sinus node dysfunction and heart block, caused by congenital autoimmune disease of the cardiac conduction system. The discovery of channelopathies linked to Cav1.3 and Cav3.1 channels underscores the importance of Ca2+ channels in the generation and regulation of heart's automaticity.
Collapse
Affiliation(s)
- Angelo G Torrente
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx Ion Channels Science and Therapeutics (ICST), Montpellier, France
| | - Pietro Mesirca
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx Ion Channels Science and Therapeutics (ICST), Montpellier, France
| | - Isabelle Bidaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 141, rue de la cardonille, 34094, Montpellier, France.,LabEx Ion Channels Science and Therapeutics (ICST), Montpellier, France
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 141, rue de la cardonille, 34094, Montpellier, France. .,LabEx Ion Channels Science and Therapeutics (ICST), Montpellier, France.
| |
Collapse
|
23
|
Yang M, Wang Y, Xiong X, Xie B, Liu J, Yin J, Zi L, Wang X, Tang Y, Huang C, Zhao Q. SK4 calcium-activated potassium channels activated by sympathetic nerves enhances atrial fibrillation vulnerability in a canine model of acute stroke. Heliyon 2020; 6:e03928. [PMID: 32420493 PMCID: PMC7215192 DOI: 10.1016/j.heliyon.2020.e03928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/24/2020] [Accepted: 05/01/2020] [Indexed: 12/02/2022] Open
Abstract
Background New-onset atrial fibrillation (AF) is common in patients with acute stroke (AS). Studies have shown that intermediate-conductance calcium-activated potassium channel channels (SK4) play an important role in cardiomyocyte automaticity. The aim of this study was to investigate the effects of SK4 on AF vulnerability in dogs with AS. Experimental Eighteen dogs were randomly divided into a control group, AS group and left stellate ganglion ablation (LSGA) group. In the control group, dogs received craniotomy without right middle cerebral artery occlusion (MCAO). AS dogs were established using a cerebral ischemic model with right MCAO. LSGA dogs underwent MCAO, and LSGA was performed. Results Three days later, the dispersion of the effective refractory period (dERP) and AF vulnerability in the AS group were significantly increased compared with those in the control group and LSGA group. However, no significant difference in dERP and AF vulnerability was found between the control group and the LSGA group. The SK4 inhibitor (TRAM-34) completely inhibited the inducibility of AF in AS dogs. SK4 expression and levels of noradrenaline (NE), β1-AR, p38 and c-Fos in the atrium were higher in the AS dogs than in the control group or LSGA group. However, no significant difference in SK4 expression or levels of NE, β1-AR, p38 and c-Fos in the left atrium was observed between the control group and LSGA group. Conclusion SK4 plays a key role in AF vulnerability in a canine model with AS. The effects of LSGA on AF vulnerability were associated with the p38 signaling pathways.
Collapse
Affiliation(s)
- Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Youcheng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan City, 430060, PR China
| | - Baojun Xie
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan City, 430060, PR China
| | - Jia Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan City, 430060, PR China
| | - Junkui Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Liuliu Zi
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Cardiovascular Research Institute of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuchang, Wuhan City, 430060, PR China
- Corresponding author.
| |
Collapse
|
24
|
Role of intermediate-conductance calcium-activated potassium channels in atrial fibrillation in canines with rapid atrial pacing. J Interv Card Electrophysiol 2020; 60:247-253. [PMID: 32248426 DOI: 10.1007/s10840-020-00736-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 03/24/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE The aim of the present study was to explore the role of intermediate-conductance Ca2+-activated K+ (SK4) in atrial fibrillation (AF) inducibility in canines with rapid atrial pacing. METHODS Eighteen dogs were divided into the control group, the pacing group and the stellate ganglion ablation (SGA) + pacing group. In the pacing group, dogs were subjected to rapid atrial pacing, and the atrial effective refractory period (AERP) and AF inducibility were measured. After cessation of 7-h pacing, SK4 inhibitor (TRAM-34) was administered. After SGA, the SGA + pacing group received the same procedure of pacing and electrophysiological measurement as the pacing group. The expression of SK4 was measured in the left atrium (LA) and the right atrium (RA) in the three groups. RESULTS The duration of the AERP decreased, while the number of AF episodes, the duration of induced AF, and the amplitude of stellate ganglion neural activity all increased after rapid atrial pacing. TRAM-34 completely inhibited AF induction in the pacing group. There was no significant difference in AERP shortening or AF vulnerability between the SGA + pacing group and the control group. The expression of SK4 in the LA and RA was higher in the pacing group than in the control and SGA + pacing groups. However, there was no significant difference in the expression of SK4 in the LA or the RA between the SGA + pacing group and the control group. CONCLUSION The higher expression of SK4 plays an important role in AF induction and the increased expression of SK4 in the atrium is related to SG activity during rapid atrial pacing.
Collapse
|
25
|
Nucleoside Diphosphate Kinase B Contributes to Arrhythmogenesis in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes from a Patient with Arrhythmogenic Right Ventricular Cardiomyopathy. J Clin Med 2020; 9:jcm9020486. [PMID: 32050722 PMCID: PMC7073527 DOI: 10.3390/jcm9020486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 12/29/2022] Open
Abstract
Background: Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a rare, inheritable cardiac disorder characterized by ventricular tachyarrhythmias, progressive loss of cardiomyocytes with fibrofatty replacement and sudden cardiac death. The exact underlying mechanisms are unclear. Methods: This study investigated the possible roles of nucleoside diphosphate kinase B (NDPK-B) and SK4 channels in the arrhythmogenesis of ARVC by using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Results: In hiPSC-CMs from a patient with ARVC, the expression levels of NDPK-B and SK4 channels were upregulated, the cell automaticity was increased and the occurrence rate of arrhythmic events was enhanced. Recombinant NDPK-B applied into hiPSC-CMs from either healthy donors or the patient enhanced SK4 channel current (ISK4), cell automaticity and the occurrence of arrhythmic events, whereas protein histidine phosphatase 1 (PHP-1), a counter actor of NDPK-B, prevented the NDPK-B effect. Application of PHP-1 alone or a SK4 channel blocker also reduced cell automaticity and arrhythmic events. Conclusion: This study demonstrated that the elevated NDPK-B expression, via activating SK4 channels, contributes to arrhythmogenesis in ARVC, and hence, NDPK-B may be a potential therapeutic target for treating arrhythmias in patients with ARVC.
Collapse
|
26
|
Yue X, Hazan A, Lotteau S, Zhang R, Torrente AG, Philipson KD, Ottolia M, Goldhaber JI. Na/Ca exchange in the atrium: Role in sinoatrial node pacemaking and excitation-contraction coupling. Cell Calcium 2020; 87:102167. [PMID: 32028091 DOI: 10.1016/j.ceca.2020.102167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/21/2020] [Indexed: 01/14/2023]
Abstract
Na/Ca exchange is the dominant calcium (Ca) efflux mechanism in cardiac myocytes. Although our knowledge of exchanger function (NCX1 in the heart) was originally established using biochemical and electrophysiological tools such as cardiac sarcolemmal vesicles and the giant patch technique [1-4], many advances in our understanding of the physiological/pathophysiological roles of NCX1 in the heart have been obtained using a suite of genetically modified mice. Early mouse studies focused on modification of expression levels of NCX1 in the ventricles, with transgenic overexpressors, global NCX1 knockout (KO) mice (which were embryonic lethal if homozygous), and finally ventricular-specific NCX1 KO [5-12]. We found, to our surprise, that ventricular cardiomyocytes lacking NCX1 can survive and function by engaging a clever set of adaptations to minimize Ca entry, while maintaining contractile function through an increase in excitation-contraction (EC) coupling gain [5,6,13]. Having studied ventricular NCX1 ablation in detail, we more recently focused on elucidating the role of NCX1 in the atria through altering NCX1 expression. Using a novel atrial-specific NCX1 KO mouse, we found unexpected changes in atrial cell morphology and calcium handling, together with dramatic alterations in the function of sinoatrial node (SAN) pacemaker activity. In this review, we will discuss these findings and their implications for cardiac disease.
Collapse
Affiliation(s)
- Xin Yue
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Adina Hazan
- Smidt Heart Institute, Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sabine Lotteau
- Smidt Heart Institute, Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Rui Zhang
- Smidt Heart Institute, Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Angelo G Torrente
- Institute for Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Michela Ottolia
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Joshua I Goldhaber
- Smidt Heart Institute, Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
27
|
Bueno-Levy H, Weisbrod D, Yadin D, Haron-Khun S, Peretz A, Hochhauser E, Arad M, Attali B. The Hyperpolarization-Activated Cyclic-Nucleotide-Gated Channel Blocker Ivabradine Does Not Prevent Arrhythmias in Catecholaminergic Polymorphic Ventricular Tachycardia. Front Pharmacol 2020; 10:1566. [PMID: 32009964 PMCID: PMC6978284 DOI: 10.3389/fphar.2019.01566] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/03/2019] [Indexed: 01/01/2023] Open
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is an inherited, stressed-provoked ventricular arrhythmia. CPVT is treated by β-adrenergic receptor blockers, Na+ channel inhibitors, sympathetic denervation, or by implanting a defibrillator. We showed recently that blockers of SK4 Ca2+-activated K+ channels depolarize the maximal diastolic potential, reduce the heart rate, and attenuate ventricular arrhythmias in CPVT. The aim of the present study was to examine whether the pacemaker channel inhibitor, ivabradine could demonstrate anti-arrhythmic properties in CPVT like other bradycardic agents used in this disease and to compare them with those of the SK4 channel blocker, TRAM-34. The effects of ivabradine were examined on the arrhythmic beating of human induced pluripotent stem cells derived cardiomyocytes (hiPSC-CMs) from CPVT patients, on sinoatrial node (SAN) calcium transients, and on ECG measurements obtained from transgenic mice model of CPVT. Ivabradine did neither prevent the arrhythmic pacing of hiPSC-CMs derived from CPVT patients, nor preclude the aberrant SAN calcium transients. In contrast to TRAM-34, ivabradine was unable to reduce in vivo the ventricular premature complexes and ventricular tachyarrhythmias in transgenic CPVT mice. In conclusion, ivabradine does not exhibit anti-arrhythmic properties in CPVT, which indicates that this blocker cannot be used as a plausible treatment for CPVT ventricular arrhythmias.
Collapse
Affiliation(s)
- Hanna Bueno-Levy
- Department of Physiology and Pharmacology, The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - David Weisbrod
- Department of Physiology and Pharmacology, The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dor Yadin
- Leviev Heart Center, Sheba Medical Center, Tel Aviv, Israel
| | - Shiraz Haron-Khun
- Department of Physiology and Pharmacology, The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Leviev Heart Center, Sheba Medical Center, Tel Aviv, Israel
| | - Asher Peretz
- Department of Physiology and Pharmacology, The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Edith Hochhauser
- The Cardiac Research Laboratory, Felsenstein Medical Research Center, Rabin Medical Center, Tel Aviv University, Petah Tikva, Israel
| | - Michael Arad
- Leviev Heart Center, Sheba Medical Center, Tel Aviv, Israel
| | - Bernard Attali
- Department of Physiology and Pharmacology, The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
28
|
Yang M, Zhao Q, Zhao H, Yang A, Wang F, Wang X, Tang Y, Huang C. Adipose‑derived stem cells overexpressing SK4 calcium‑activated potassium channel generate biological pacemakers. Int J Mol Med 2019; 44:2103-2112. [PMID: 31638180 PMCID: PMC6844603 DOI: 10.3892/ijmm.2019.4374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/11/2019] [Indexed: 01/14/2023] Open
Abstract
Recent studies have suggested that calcium-activated potassium channel (KCa) agonists increase the proportion of mouse embryonic stem cell-derived cardiomyocytes and promote the differentiation of pacemaker cells. In the present study, it was hypothesized that adipose-derived stem cells (ADSCs) can differentiate into pacemaker-like cells via over-expression of the SK4 gene. ADSCs were transduced with a recombinant adenovirus vector carrying the mouse SK4 gene, whereas the control group was transduced with GFP vector. ADSCs transduced with SK4 vector were implanted into the rat left ventricular free wall. Complete atrioventricular block (AVB) was established in isolated perfused rat hearts after 2 weeks. SK4 was successfully and stably expressed in ADSCs following transduction. The mRNA levels of the pluripotent markers Oct-4 and Sox-2 declined and that of the transcription factor Shox2 was upregulated following SK4 transduction. The expression of α-actinin and hyperpolarization-activated cyclic nucleotide-gated potassium channel 4 (HCN4) increased in the SK4 group. The hyperpolarizing activated pacemaker current If (8/20 cells) was detected in ADSCs transduced with SK4, but not in the GFP group. Furthermore, SK4 transduction induced the expression of p-ERK1/2 and p-p38 MAPK. In the ex vivo experiments, the heart rate of the SK4 group following AVB establishment was significantly higher compared with that in the GFP group. Immunofluorescence revealed that the transduced ADSCs were successfully implanted and expressed HCN4 in the SK4 group. In conclusion, SK4 induced ADSCs to differentiate into cardiomyocyte-like and pacemaker-like cells via activation of the extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase pathways. Therefore, ADSCs transduced with SK4 may be used to generate biological pacemakers in ex vivo rat hearts.
Collapse
Affiliation(s)
- Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hongyi Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ankang Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fengyuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
29
|
Zhao H, Yang M, Wang F, Yang A, Zhao Q, Wang X, Tang Y, Wang T, Huang C. Overexpression of the medium‑conductance calcium‑activated potassium channel (SK4) and the HCN2 channel to generate a biological pacemaker. Mol Med Rep 2019; 20:3406-3414. [PMID: 31432175 DOI: 10.3892/mmr.2019.10591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/25/2019] [Indexed: 11/06/2022] Open
Abstract
Ion channels serve important roles in the excitation‑contraction coupling of cardiac myocytes. Previous studies have shown that the overexpression or activation of intermediate‑conductance calcium‑activated potassium channel (SK4, encoded by KCNN4) in embryonic stem cell‑derived cardiomyocytes can significantly increase their automaticity. The mechanism underlying this effect is hypothesized to be associated with the activation of hyperpolarization‑activated cyclic nucleotide‑gated channel 2 (HCN2). The aim of the present study was to explore whether a biological pacemaker could be constructed by overexpressing SK4 alone or in combination with HCN2 in a rat model. Ad‑green fluorescent protein (GFP), Ad‑KCNN4 and Ad‑HCN2 recombinant adenoviruses were injected into the left ventricle of Sprague‑Dawley rat hearts. The rats were divided into a GFP group (n=10), an SK4 group (n=10), a HCN2 group (n=10) and an SK4 + HCN2 (SK4/HCN2) group (n=10). The isolated hearts were perfused at 5‑7 days following injection, and a complete heart block model was established. Compared with the GFP group, overexpressing SK4 alone did not significantly increase the heart rate after establishment of a complete heart block model [98.1±8.9 vs. 96.7±7.6 beats per min (BPM)], The heart rates in the SK4/HCN2 (139.9±21.9 BPM) and HCN2 groups (111.7±5.5 BPM) were significantly increased compared with the GFP and SK4 groups, and the heart rates in the SK4/HCN2 group were significantly increased compared with the SK4 or HCN2 groups. In the HCN2 (n=8) and the SK4/HCN2 (n=7) groups, the shape of the spontaneous ventricular rhythm was the same as the pacing‑induced ectopic rhythm in the transgenically altered site. By contrast, these rhythms were different in the SK4 (n=10) and GFP (n=10) groups. There were no significant differences in action potential duration alternans or ventricular arrhythmia inducibility between the four groups (all P>0.05). Western blotting, reverse transcription‑quantitative PCR and immunohistochemistry analyses showed that the expression levels of SK4 and HCN2 were significantly increased at the transgene site. Biological pacemaker activity could be successfully generated by co‑overexpression of SK4 and HCN2 without increasing the risk of ventricular arrhythmias. The overexpression of SK4 alone is insufficient to generate biological pacemaker activity. The present study provided evidence that SK4 and HCN2 combined could construct an ectopic pacemaker, laying the groundwork for the development of improved biological pacing mechanisms in the future.
Collapse
Affiliation(s)
- Hongyi Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Mei Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fengyuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ankang Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qingyan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Teng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
30
|
Fei YD, Wang Q, Hou JW, Li W, Cai XX, Yang YL, Zhang LH, Wei ZX, Chen TZ, Wang YP, Li YG. Macrophages facilitate post myocardial infarction arrhythmias: roles of gap junction and KCa3.1. Am J Cancer Res 2019; 9:6396-6411. [PMID: 31588225 PMCID: PMC6771231 DOI: 10.7150/thno.34801] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/10/2019] [Indexed: 12/24/2022] Open
Abstract
Effective therapeutic targets against post-myocardial infarction (MI) arrhythmias remain to be discovered. We aimed to investigate the role of macrophages in post-MI arrhythmias. Methods: Mononuclear cell accumulation, macrophage polarization from M0 to M1 subset, and gap junction formation were analyzed in MI patients and MI mice by flow cytometry, immunofluorescence and patch clamping. Differentially expressed genes were identified by RNA sequencing. Macrophages and cardiomyocytes were cocultured in vitro, and the effects of gap junction and KCa3.1 on electrophysiological properties were assessed by patch clamping. The effects of KCa3.1 inhibition on post-MI arrhythmias were assessed by intracardiac stimulation and ambulatory electrocardiograms in vivo. Results: Percentage of pro-inflammatory mononuclear cells were significantly elevated in patients with post-MI arrhythmias compared with MI patients without arrhythmias and healthy controls (p<0.001). Macrophages formed gap junction with cardiomyocytes in MI border zones of MI patient and mice, and pro-inflammatory macrophages were significantly increased 3 days post-MI (p<0.001). RNA sequencing identified Kcnn4 as the most differentially expressed gene encoding ion channel, and the upregulation is mainly attributed to macrophage accumulation and polarization into pro-inflammatory subset. In vitro coculture experiments demonstrated that connection with M0 macrophages via gap junction slightly shortened the action potential durations (APDs) of cardiomyocytes. However, the APD90 of cardiomyocytes connected with M1 macrophages were significantly prolonged (p<0.001), which were effectively attenuated by gap junction inhibition (p=0.002), KCa3.1 inhibition (p=0.008), KCa3.1 silencing (p<0.001) and store-operated Ca2+ channel inhibition (p=0.005). In vivo results demonstrated that KCa3.1 inhibition significantly decreased the QTc durations (p=0.031), intracardiac stimulation-induced ventricular arrhythmia durations (p=0.050) and incidence of premature ventricular contractions (p=0.030) in MI mice. Conclusion: Macrophage polarization leads to APD heterogeneity and post-MI arrhythmias via gap junction and KCa3.1 activation. The results provide evidences of a novel mechanism of post-MI heterogeneous repolarization and arrhythmias, rendering macrophages and KCa3.1 to be potential therapeutic targets.
Collapse
|
31
|
Positive Feedback Mechanisms among Local Ca Releases, NCX, and I CaL Ignite Pacemaker Action Potentials. Biophys J 2019. [PMID: 29539403 DOI: 10.1016/j.bpj.2017.12.043] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recent data suggest that cardiac pacemaker cell function is determined by numerous time-, voltage-, and Ca-dependent interactions of cell membrane electrogenic proteins (M-clock) and intracellular Ca cycling proteins (Ca-clock), forming a coupled-clock system. Many aspects of the coupled-clock system, however, remain underexplored. The key players of the system are Ca release channels (ryanodine receptors), generating local Ca releases (LCRs) from sarcoplasmic reticulum, electrogenic Na/Ca exchanger (NCX) current, and L-type Ca current (ICaL). We combined numerical model simulations with experimental simultaneous recordings of action potentials (APs) and Ca to gain further insight into the complex interactions within the system. Our simulations revealed a positive feedback mechanism, dubbed AP ignition, which accelerates the diastolic depolarization (DD) to reach AP threshold. The ignition phase begins when LCRs begin to occur and the magnitude of inward NCX current begins to increase. The NCX current, together with funny current and T-type Ca current accelerates DD, bringing the membrane potential to ICaL activation threshold. During the ignition phase, ICaL-mediated Ca influx generates more LCRs via Ca-induced Ca release that further activates inward NCX current, creating a positive feedback. Simultaneous recordings of membrane potential and confocal Ca images support the model prediction of the positive feedback among LCRs and ICaL, as diastolic LCRs begin to occur below and continue within the voltage range of ICaL activation. The ignition phase onset (identified within the fine DD structure) begins when DD starts to notably accelerate (∼0.15 V/s) above the recording noise. Moreover, the timing of the ignition onset closely predicted the duration of each AP cycle in the basal state, in the presence of autonomic receptor stimulation, and in response to specific inhibition of either the M-clock or Ca-clock, thus indicating general importance of the new coupling mechanism for regulation of the pacemaker cell cycle duration, and ultimately the heart rate.
Collapse
|
32
|
Human iPSC-Derived Cardiomyocytes for Investigation of Disease Mechanisms and Therapeutic Strategies in Inherited Arrhythmia Syndromes: Strengths and Limitations. Cardiovasc Drugs Ther 2018; 31:325-344. [PMID: 28721524 PMCID: PMC5550530 DOI: 10.1007/s10557-017-6735-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During the last two decades, significant progress has been made in the identification of genetic defects underlying inherited arrhythmia syndromes, which has provided some clinical benefit through elucidation of gene-specific arrhythmia triggers and treatment. However, for most arrhythmia syndromes, clinical management is hindered by insufficient knowledge of the functional consequences of the mutation in question, the pro-arrhythmic mechanisms involved, and hence the most optimal treatment strategy. Moreover, disease expressivity and sensitivity to therapeutic interventions often varies between mutations and/or patients, underlining the need for more individualized strategies. The development of the induced pluripotent stem cell (iPSC) technology now provides the opportunity for generating iPSC-derived cardiomyocytes (CMs) from human material (hiPSC-CMs), enabling patient- and/or mutation-specific investigations. These hiPSC-CMs may furthermore be employed for identification and assessment of novel therapeutic strategies for arrhythmia syndromes. However, due to their relative immaturity, hiPSC-CMs also display a number of essential differences as compared to adult human CMs, and hence there are certain limitations in their use. We here review the electrophysiological characteristics of hiPSC-CMs, their use for investigating inherited arrhythmia syndromes, and their applicability for identification and assessment of (novel) anti-arrhythmic treatment strategies.
Collapse
|
33
|
Haron-Khun S, Weisbrod D, Bueno H, Yadin D, Behar J, Peretz A, Binah O, Hochhauser E, Eldar M, Yaniv Y, Arad M, Attali B. SK4 K + channels are therapeutic targets for the treatment of cardiac arrhythmias. EMBO Mol Med 2017; 9:415-429. [PMID: 28219898 PMCID: PMC5376763 DOI: 10.15252/emmm.201606937] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a stress‐provoked ventricular arrhythmia, which also manifests sinoatrial node (SAN) dysfunction. We recently showed that SK4 calcium‐activated potassium channels are important for automaticity of cardiomyocytes derived from human embryonic stem cells. Here SK4 channels were identified in human induced pluripotent stem cell‐derived cardiomyocytes (hiPSC‐CMs) from healthy and CPVT2 patients bearing a mutation in calsequestrin 2 (CASQ2‐D307H) and in SAN cells from WT and CASQ2‐D307H knock‐in (KI) mice. TRAM‐34, a selective blocker of SK4 channels, prominently reduced delayed afterdepolarizations and arrhythmic Ca2+ transients observed following application of the β‐adrenergic agonist isoproterenol in CPVT2‐derived hiPSC‐CMs and in SAN cells from KI mice. Strikingly, in vivo ECG recording showed that intraperitoneal injection of the SK4 channel blockers, TRAM‐34 or clotrimazole, greatly reduced the arrhythmic features of CASQ2‐D307H KI and CASQ2 knockout mice at rest and following exercise. This work demonstrates the critical role of SK4 Ca2+‐activated K+ channels in adult pacemaker function, making them promising therapeutic targets for the treatment of cardiac ventricular arrhythmias such as CPVT.
Collapse
Affiliation(s)
- Shiraz Haron-Khun
- Department of Physiology and Pharmacology, The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Tel Aviv, Israel
| | - David Weisbrod
- Department of Physiology and Pharmacology, The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hanna Bueno
- Department of Physiology and Pharmacology, The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dor Yadin
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Tel Aviv, Israel
| | - Joachim Behar
- Laboratory of Bioenergetic and Bioelectric Systems, Biomedical Engineering Faculty, Technion-Israel Institute of Technology, Haifa, Israel
| | - Asher Peretz
- Department of Physiology and Pharmacology, The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ofer Binah
- Department of Physiology, Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Edith Hochhauser
- The Cardiac Research Laboratory of the Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel Aviv University, Petah Tikva, Israel
| | - Michael Eldar
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Tel Aviv, Israel
| | - Yael Yaniv
- Laboratory of Bioenergetic and Bioelectric Systems, Biomedical Engineering Faculty, Technion-Israel Institute of Technology, Haifa, Israel
| | - Michael Arad
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Tel Aviv, Israel
| | - Bernard Attali
- Department of Physiology and Pharmacology, The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
34
|
Chai S, Wan X, Nassal DM, Liu H, Moravec CS, Ramirez-Navarro A, Deschênes I. Contribution of two-pore K + channels to cardiac ventricular action potential revealed using human iPSC-derived cardiomyocytes. Am J Physiol Heart Circ Physiol 2017; 312:H1144-H1153. [PMID: 28341634 DOI: 10.1152/ajpheart.00107.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/16/2017] [Accepted: 03/22/2017] [Indexed: 01/12/2023]
Abstract
Two-pore K+ (K2p) channels have been described in modulating background conductance as leak channels in different physiological systems. In the heart, the expression of K2p channels is heterogeneous with equivocation regarding their functional role. Our objective was to determine the K2p expression profile and their physiological and pathophysiological contribution to cardiac electrophysiology. Induced pluripotent stem cells (iPSCs) generated from humans were differentiated into cardiomyocytes (iPSC-CMs). mRNA was isolated from these cells, commercial iPSC-CM (iCells), control human heart ventricular tissue (cHVT), and ischemic (iHF) and nonischemic heart failure tissues (niHF). We detected 10 K2p channels in the heart. Comparing quantitative PCR expression of K2p channels between human heart tissue and iPSC-CMs revealed K2p1.1, K2p2.1, K2p5.1, and K2p17.1 to be higher expressed in cHVT, whereas K2p3.1 and K2p13.1 were higher in iPSC-CMs. Notably, K2p17.1 was significantly lower in niHF tissues compared with cHVT. Action potential recordings in iCells after K2p small interfering RNA knockdown revealed prolongations in action potential depolarization at 90% repolarization for K2p2.1, K2p3.1, K2p6.1, and K2p17.1. Here, we report the expression level of 10 human K2p channels in iPSC-CMs and how they compared with cHVT. Importantly, our functional electrophysiological data in human iPSC-CMs revealed a prominent role in cardiac ventricular repolarization for four of these channels. Finally, we also identified K2p17.1 as significantly reduced in niHF tissues and K2p4.1 as reduced in niHF compared with iHF. Thus, we advance the notion that K2p channels are emerging as novel players in cardiac ventricular electrophysiology that could also be remodeled in cardiac pathology and therefore contribute to arrhythmias.NEW & NOTEWORTHY Two-pore K+ (K2p) channels are traditionally regarded as merely background leak channels in myriad physiological systems. Here, we describe the expression profile of K2p channels in human-induced pluripotent stem cell-derived cardiomyocytes and outline a salient role in cardiac repolarization and pathology for multiple K2p channels.
Collapse
Affiliation(s)
- Sam Chai
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio.,Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | - Xiaoping Wan
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | - Drew M Nassal
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio.,Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | - Haiyan Liu
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | | | - Angelina Ramirez-Navarro
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | - Isabelle Deschênes
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio; .,Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| |
Collapse
|