1
|
Yang H, Shen X, Wang H, Shuai W. Tumour necrosis factor alpha-induced protein 3-interacting protein 3 overexpression protects against arrhythmogenic remodelling in the heart failure mice. Europace 2024; 27:euaf002. [PMID: 39800969 PMCID: PMC11757166 DOI: 10.1093/europace/euaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 12/11/2024] [Indexed: 01/25/2025] Open
Abstract
AIMS Ventricular arrhythmias (VAs), which can lead to sudden cardiac death, are the primary cause of mortality in patients with heart failure (HF). However, the precise mechanisms underlying these arrhythmias are not well understood. Recent studies have implicated tumour necrosis factor alpha-induced protein 3-interacting protein 3 (TNIP3) in pathological cardiac hypertrophy. Nevertheless, its role in isoproterenol (ISO)-associated VAs remains elusive. METHODS AND RESULTS We overexpressed TNIP3 in the myocardium using an adeno-associated virus 9 system, administered via tail vein injection. C57BL/6 mice received daily subcutaneous injections of ISO for two consecutive weeks to establish an HF model. We performed histopathology and electrophysiological studies to assess ventricular structural remodelling, electrical remodelling, and susceptibility to VAs. Additionally, RNA sequencing (RNA-Seq) and western blot analysis were conducted to elucidate the underlying mechanisms. The expression of TNIP3 was up-regulated following ISO treatment. TNIP3 overexpression significantly reversed ISO-induced cardiac dysfunction, fibrosis, electrical remodelling, and VAs susceptibility. Accordingly, RNA-Seq identifies that the inflammatory response takes an important role in ISO-induced Vas, and TNIP3 overexpression could alleviate ISO-induced cardiac proinflammatory response by promoting M1 to M2 macrophage polarization. Mechanistically, PI3K/Akt/NF-κB signalling is responsible for the protective effect of TNIP3 overexpression on ISO-induced HF. And PI3K/Akt signalling activation offset the protective effect of TNIP3 overexpression on ISO-induced cardiac inflammation and VAs. CONCLUSION The findings of this study highlight the critical role of TNIP3 in ISO-associated cardiac remodelling and VAs, which are induced by the inhibited activation of the PI3K/Akt/NF-κB signalling pathway.
Collapse
Affiliation(s)
- Hongjie Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan 430060, Hubei, P.R. of China
| | - Xiaoyan Shen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan 430060, Hubei, P.R. of China
| | - Huibo Wang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital; Institute of Cardiovascular Diseases, China Three Gorges University, Hu Bei Clinical Research Center for Ischemic Cardiovascular Disease, No. 183 Yiling Avenue, Yichang 443000, Hubei, China
| | - Wei Shuai
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan 430060, Hubei, P.R. of China
| |
Collapse
|
2
|
Alijanzadeh D, Moghim S, Zarand P, Akbarzadeh MA, Zarinfar Y, Khaheshi I. Reassessing Ivabradine: Potential Benefits and Risks in Atrial Fibrillation Therapy. Cardiovasc Drugs Ther 2024. [DOI: 10.1007/s10557-024-07652-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/12/2024] [Indexed: 01/03/2025]
|
3
|
Wong SL, Shih CL, Cho HY, Wu SN. Effective suppression of I h and I Na caused by capsazepine, known to be a blocker of TRPV1 receptor. Brain Res 2024; 1839:149008. [PMID: 38761846 DOI: 10.1016/j.brainres.2024.149008] [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: 02/06/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
A synthetic inhibitor of capsaicin-induced TRPV1 channel activation is called capsazepine (CPZ). In this study, we aimed to explore the effects of CPZ on hyperpolarization-activated cationic current (Ih) and voltage-gated Na + current (INa) in pituitary tumor (GH3) cells. Through patch-clamp recordings, we found that CPZ concentration-dependently inhibited Ih amplitude and slowed its activation time course. The IC50 and KD values were 3.1 and 3.16 μM, respectively. CPZ also shifted the steady-state activation curve of Ih towards a more hyperpolarized potential. However, there was no change in the gating charge of the curve. A modified Markovian model predicted the CPZ-induced decrease in the voltage-dependent hysteresis of Ih. CPZ suppressed INa in GH3 cells, without altering its activation or inactivation time course. Additionally, exposure to CPZ reduced spontaneous firing. These findings suggest that CPZ's inhibitory effects on Ih and INa are direct and not dependent on vanilloid receptor binding. This could provide light on an unidentified ionic mechanism influencing the membrane excitability of neurons and endocrine or neuroendocrine cells in vivo.
Collapse
Affiliation(s)
- Siew-Lee Wong
- Department of Pediatrics, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan
| | - Chia-Lung Shih
- Clinical Research Center, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City 60002, Taiwan.
| | - Hsin-Yen Cho
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan; Department of Research and Education, An Nan Hospital, China Medical University, Tainan 709040, Taiwan; School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, 804201 Taiwan.
| |
Collapse
|
4
|
Sripusanapan A, Yanpiset P, Sriwichaiin S, Siri-Angkul N, Chattipakorn SC, Chattipakorn N. Hyperpolarization-activated cyclic nucleotide-gated channel inhibitor in myocardial infarction: Potential benefits beyond heart rate modulation. Acta Physiol (Oxf) 2024; 240:e14085. [PMID: 38230890 DOI: 10.1111/apha.14085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/24/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024]
Abstract
Myocardial infarction (MI) and its associated complications including ventricular arrhythmias and heart failure are responsible for a significant incidence of morbidity and mortality worldwide. The ensuing cardiomyocyte loss results in neurohormone-driven cardiac remodeling, which leads to chronic heart failure in MI survivors. Ivabradine is a heart rate modulation agent currently used in treatment of chronic heart failure with reduced ejection fraction. The canonical target of ivabradine is the hyperpolarization-activated cyclic nucleotide-gated channels (HCN) in cardiac pacemaker cells. However, in post-MI hearts, HCN can also be expressed ectopically in non-pacemaker cardiomyocytes. There is an accumulation of intriguing evidence to suggest that ivabradine also possesses cardioprotective effects that are independent of heart rate reduction. This review aims to summarize and discuss the reported cardioprotective mechanisms of ivabradine beyond heart rate modulation in myocardial infarction through various molecular mechanisms including the prevention of reactive oxygen species-induced mitochondrial damage, improvement of autophagy system, modulation of intracellular calcium cycling, modification of ventricular electrophysiology, and regulation of matrix metalloproteinases.
Collapse
Affiliation(s)
- Adivitch Sripusanapan
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellent in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Panat Yanpiset
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellent in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sirawit Sriwichaiin
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellent in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Natthaphat Siri-Angkul
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellent in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellent in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellent in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
5
|
An X, Cho H. Increased GIRK channel activity prevents arrhythmia in mice with heart failure by enhancing ventricular repolarization. Sci Rep 2023; 13:22479. [PMID: 38110503 PMCID: PMC10728207 DOI: 10.1038/s41598-023-50088-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 12/15/2023] [Indexed: 12/20/2023] Open
Abstract
Ventricular arrhythmia causing sudden cardiac death is the leading mode of death in patients with heart failure. Yet, the mechanisms that prevent ventricular arrhythmias in heart failure are not well characterized. Using a mouse model of heart failure created by transverse aorta constriction, we show that GIRK channel, an important regulator of cardiac action potentials, is constitutively active in failing ventricles in contrast to normal cells. Evidence is presented indicating that the tonic activation of M2 muscarinic acetylcholine receptors by endogenously released acetylcholine contributes to the constitutive GIRK activity. This constitutive GIRK activity prevents the action potential prolongation in heart failure ventricles. Consistently, GIRK channel blockade with tertiapin-Q induces QT interval prolongation and increases the incidence of arrhythmia in heart failure, but not in control mice. These results suggest that constitutive GIRK channels comprise a key mechanism to protect against arrhythmia by providing repolarizing currents in heart failure ventricles.
Collapse
Affiliation(s)
- Xue An
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea.
| |
Collapse
|
6
|
Pay L, Yumurtaş AÇ, Tezen O, Çetin T, Keskin K, Eren S, Çinier G, Hayıroğlu Mİ, Çınar T, Tekkeşin Aİ. Effect of ivabradine on ventricular arrhythmias in heart failure patients with reduced ejection fraction. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20230703. [PMID: 37971125 PMCID: PMC10645178 DOI: 10.1590/1806-9282.20230703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/26/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND/INTRODUCTION Heart failure patients with reduced ejection fraction are at high risk for ventricular arrhythmias and sudden cardiac death. Ivabradine, a specific inhibitor of the If current in the sinoatrial node, provides heart rate reduction in sinus rhythm and angina control in chronic coronary syndromes. OBJECTIVE The effect of ivabradine on ventricular arrhythmias in heart failure patients with reduced ejection fraction patients has not been fully elucidated. The aim of this study was to investigate the effect of ivabradine use on life-threatening arrhythmias and long-term mortality in heart failure patients with reduced ejection fraction patients. METHODS In this retrospective study, 1,639 patients with heart failure patients with reduced ejection fraction were included. Patients were divided into two groups: ivabradine users and nonusers. Patients presenting with ventricular tachycardia, the presence of ventricular extrasystole, and ventricular tachycardia in 24-h rhythm monitoring, appropriate implantable cardioverter-defibrillator shocks, and long-term mortality outcomes were evaluated according to ivabradine use. RESULTS After adjustment for all possible variables, admission with ventricular tachycardia was three times higher in ivabradine nonusers (95% confidence interval 1.5-10.2). The presence of premature ventricular contractions and ventricular tachycardias in 24-h rhythm Holter monitoring was notably higher in ivabradine nonusers. According to the adjusted model for all variables, 4.1 times more appropriate implantable cardioverter-defibrillator shocks were observed in the ivabradine nonusers than the users (95%CI 1.8-9.6). Long-term mortality did not differ between these groups after adjustment for all covariates. CONCLUSION The use of ivabradine reduced the appropriate implantable cardioverter-defibrillator discharge in heart failure patients with reduced ejection fraction patients. Ivabradine has potential in the treatment of ventricular arrhythmias in heart failure patients with reduced ejection fraction patients.
Collapse
Affiliation(s)
- Levent Pay
- Ardahan State Hospital, Department of Cardiology – Ardahan, Turkey
| | - Ahmet Çağdaş Yumurtaş
- Dr. Siyami Ersek Thoracic and Cardiovascular Surgery Training Hospital, Department of Cardiology – İstanbul, Turkey
| | - Ozan Tezen
- Dr. Siyami Ersek Thoracic and Cardiovascular Surgery Training Hospital, Department of Cardiology – İstanbul, Turkey
| | - Tuğba Çetin
- Dr. Siyami Ersek Thoracic and Cardiovascular Surgery Training Hospital, Department of Cardiology – İstanbul, Turkey
| | - Kıvanç Keskin
- Dr. Siyami Ersek Thoracic and Cardiovascular Surgery Training Hospital, Department of Cardiology – İstanbul, Turkey
| | - Semih Eren
- Dr. Siyami Ersek Thoracic and Cardiovascular Surgery Training Hospital, Department of Cardiology – İstanbul, Turkey
| | - Göksel Çinier
- Başakşehir Çam and Sakura City Hospital, Department of Electrophysiology – İstanbul, Turkey
| | - Mert İlker Hayıroğlu
- Dr. Siyami Ersek Thoracic and Cardiovascular Surgery Training Hospital, Department of Cardiology – İstanbul, Turkey
| | - Tufan Çınar
- University of Maryland Medical Center Midtown Campus, Department of Medicine – Baltimore (MD), United States
| | - Ahmet İlker Tekkeşin
- Başakşehir Çam and Sakura City Hospital, Department of Electrophysiology – İstanbul, Turkey
| |
Collapse
|
7
|
Yang J, Lv T, Zhou J, Lin H, Zhao B, Lou H, Liu H, Zhang T, Guo H, Chi J. The effect of ivabradine therapy on dilated cardiomyopathy patients with congestive heart failure: a systematic review and meta-analysis. Front Cardiovasc Med 2023; 10:1149351. [PMID: 37915740 PMCID: PMC10616249 DOI: 10.3389/fcvm.2023.1149351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 10/03/2023] [Indexed: 11/03/2023] Open
Abstract
Background Ivabradine improves cardiac function in patients with heart failure, but its effect on dilated cardiomyopathy (DCM) remains unclear. We performed a systematic review and meta-analysis to study the efficacy and potential mechanisms of ivabradine's effect on cardiac function and prognosis in patients with DCM. Methods We searched PubMed, Cochrane Library, Embase, Web of Science, and four registers through September 28, 2022. All controlled trials of ivabradine for the treatment of DCM with congestive heart failure were included. Articles were limited to English, with the full text and necessary data available. We performed random- or fixed effects meta-analyses for all included outcome measures and compared the effect sizes for outcomes in patients treated with and without ivabradine. The quality of the studies was assessed using the Cochrane risk-of-bias tool for randomized trials (RoB2.0). Findings Five trials with 357 participants were included. The pooled risk ratio was 0.48 [95% confidence interval (CI) (0.18, 1.25)] for all-cause mortality and 0.38 [95% CI (0.12, 1.23)] for cardiac mortality. The pooled mean difference was -15.95 [95% CI (-19.97, -11.92)] for resting heart rate, 3.96 [95% CI (0.99, 6.93)] for systolic blood pressure, 2.93 [95% CI (2.09, 3.77)] for left ventricular ejection fraction, -5.90 [95% CI (-9.36, -2.44)] for left ventricular end-systolic diameter, -3.41 [95% CI (-5.24, -1.58)] for left ventricular end-diastolic diameter, -0.81 [95% CI (-1.00, -0.62)] for left ventricular end-systolic volume, -0.67 [95% CI (-0.86, -0.48)] for left ventricular end-diastolic volume, -11.01 [95% CI (-19.66, -2.35)] for Minnesota Living with Heart Failure score, and -0.52 [95% CI (-0.73, -0.31)] for New York Heart Association class. Interpretation Ivabradine reduces heart rate and ventricular volume, and improves cardiac function in patients with DCM, but showed no significant effect on the prognosis of patients.
Collapse
Affiliation(s)
- Juntao Yang
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Tingting Lv
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Jiedong Zhou
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Hui Lin
- Ningbo Medical Center Lihuili Hospital (Lihuili Hospital Affiliated to Ningbo University), Ningbo, China
| | - Bingjie Zhao
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Haifei Lou
- Department of Cardiology, Shaoxing People’s Hospital, Shaoxing, China
| | - Hanxuan Liu
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Tao Zhang
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Hangyuan Guo
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Jufang Chi
- Department of Cardiology, Zhuji People’s Hospital, Zhuji, China
| |
Collapse
|
8
|
Thankam FG, La V, Agrawal DK. Single-cell genomics illustrates heterogeneous phenotypes of myocardial fibroblasts under ischemic insults. Biochem Cell Biol 2023; 101:12-51. [PMID: 36458696 DOI: 10.1139/bcb-2022-0229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Myocardial regenerative strategies are promising where the choice of ideal cell population is crucial for successful translational applications. Herein, we explored the regenerative/repair responses of infarct zone cardiac fibroblast(s) (CF) by unveiling their phenotype heterogeneity at single-cell resolution. CF were isolated from the infarct zone of Yucatan miniswine that suffered myocardial infarction, cultured under simulated ischemic and reperfusion, and grouped into control, ischemia, and ischemia/reperfusion. The single-cell RNA sequencing analysis revealed 19 unique cell clusters suggesting distinct subpopulations. The status of gene expression (log2 fold change (log2 FC) > 2 and log2 FC < -2) was used to define the characteristics of each cluster unveiling with diverse features, including the pro-survival/cardioprotective (Clusters 1, 3, 5, 9, and 18), vasculoprotective (Clusters 2 and 5), anti-inflammatory (Clusters 4 and 17), proliferative (Clusters 4 and 5), nonproliferative (Clusters 6, 8, 11, 16, 17, and 18), proinflammatory (Cluster 6), profibrotic/pathologic (Clusters 8 and 19), antihypertrophic (Clusters 8 and 10), extracellular matrix restorative (Clusters 9 and 12), angiogenic (Cluster 16), and normal (Clusters 7 and 15) phenotypes. Further understanding of these unique phenotypes of CF will provide significant translational opportunities for myocardial regeneration and cardiac management.
Collapse
Affiliation(s)
- Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Vy La
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
9
|
Sebastian S, Weinstein LS, Ludwig A, Munroe P, Tinker A. Slowing Heart Rate Protects Against Pathological Cardiac Hypertrophy. FUNCTION 2022; 4:zqac055. [PMID: 36540889 PMCID: PMC9761894 DOI: 10.1093/function/zqac055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/27/2022] [Indexed: 12/23/2022] Open
Abstract
We aimed to determine the pathophysiological impact of heart rate (HR) slowing on cardiac function. We have recently developed a murine model in which it is possible to conditionally delete the stimulatory heterotrimeric G-protein (Gαs) in the sinoatrial (SA) node after the addition of tamoxifen using cre-loxP technology. The addition of tamoxifen leads to bradycardia. We used this approach to examine the physiological and pathophysiological effects of HR slowing. We first looked at the impact on exercise performance by running the mice on a treadmill. After the addition of tamoxifen, mice with conditional deletion of Gαs in the SA node ran a shorter distance at a slower speed. Littermate controls preserved their exercise capacity after tamoxifen. Results consistent with impaired cardiac capacity in the mutants were also obtained with a dobutamine echocardiographic stress test. We then examined if HR reduction influenced pathological cardiac hypertrophy using two models: ligation of the left anterior descending coronary artery for myocardial infarction and abdominal aortic banding for hypertensive heart disease. In littermate controls, both procedures resulted in cardiac hypertrophy. However, induction of HR reduction prior to surgical intervention significantly ameliorated the hypertrophy. In order to assess potential protein kinase pathways that may be activated in the left ventricle by relative bradycardia, we used a phospho-antibody array and this revealed selective activation of phosphoinositide-3 kinase. In conclusion, HR reduction protects against pathological cardiac hypertrophy but limits physiological exercise capacity.
Collapse
Affiliation(s)
- Sonia Sebastian
- William Harvey Heart Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Lee S Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases/National Institutes of Health, Building 10, Room 8C101, Bethesda, MD 20892-1752, USA
| | - Andreas Ludwig
- Institut fuer Experimentelle und Klinische Pharmakologie und Toxikologie, Universitaet Erlangen-Nuernberg, Fahrstr. 17, 91054 Erlangen, Germany
| | - Patricia Munroe
- William Harvey Heart Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | | |
Collapse
|
10
|
Xue JB, Val-Blasco A, Davoodi M, Gómez S, Yaniv Y, Benitah JP, Gómez AM. Heart failure in mice induces a dysfunction of the sinus node associated with reduced CaMKII signaling. J Gen Physiol 2022; 154:213178. [PMID: 35452507 PMCID: PMC9040062 DOI: 10.1085/jgp.202112895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/18/2022] [Indexed: 11/20/2022] Open
Abstract
Dysfunction of the sinoatrial node (SAN), the natural heart pacemaker, is common in heart failure (HF) patients. SAN spontaneous activity relies on various ion currents in the plasma membrane (voltage clock), but intracellular Ca2+ ([Ca2+]i) release via ryanodine receptor 2 (RYR2; Ca2+ clock) plays an important synergetic role. Whereas remodeling of voltage-clock components has been revealed in HF, less is known about possible alterations to the Ca2+ clock. Here, we analyzed [Ca2+]i handling in SAN from a mouse HF model after transverse aortic constriction (TAC) and compared it with sham-operated animals. ECG data from awake animals showed slower heart rate in HF mice upon autonomic nervous system blockade, indicating intrinsic sinus node dysfunction. Confocal microscopy analyses of SAN cells within whole tissue showed slower and less frequent [Ca2+]i transients in HF. This correlated with fewer and smaller spontaneous Ca2+ sparks in HF SAN cells, which associated with lower RYR2 protein expression level and reduced phosphorylation at the CaMKII site. Moreover, PLB phosphorylation at the CaMKII site was also decreased in HF, which could lead to reduced sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) function and lower sarcoplasmic reticulum Ca2+ content, further depressing the Ca2+ clock. The inhibition of CaMKII with KN93 slowed [Ca2+]i transient rate in both groups, but this effect was smaller in HF SAN, consistent with less CaMKII activation. In conclusion, our data uncover that the mechanism of intrinsic pacemaker dysfunction in HF involves reduced CaMKII activation.
Collapse
Affiliation(s)
- Jian-Bin Xue
- Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Université Paris-Saclay, INSERM, Châtenay-Malabry, France
| | - Almudena Val-Blasco
- Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Université Paris-Saclay, INSERM, Châtenay-Malabry, France
| | - Moran Davoodi
- Biomedical Engineering, Technion Institute, Haifa, Israel
| | - Susana Gómez
- Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Université Paris-Saclay, INSERM, Châtenay-Malabry, France
| | - Yael Yaniv
- Biomedical Engineering, Technion Institute, Haifa, Israel
| | - Jean-Pierre Benitah
- Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Université Paris-Saclay, INSERM, Châtenay-Malabry, France
| | - Ana María Gómez
- Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Université Paris-Saclay, INSERM, Châtenay-Malabry, France
| |
Collapse
|
11
|
Inazumi H, Kuwahara K. NRSF/REST-Mediated Epigenomic Regulation in the Heart: Transcriptional Control of Natriuretic Peptides and Beyond. BIOLOGY 2022; 11:1197. [PMID: 36009824 PMCID: PMC9405064 DOI: 10.3390/biology11081197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022]
Abstract
Reactivation of fetal cardiac genes, including those encoding atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), is a key feature of pathological cardiac remodeling and heart failure. Intensive studies on the regulation of ANP and BNP have revealed the involvement of numerous transcriptional factors in the regulation of the fetal cardiac gene program. Among these, we identified that a transcriptional repressor, neuron-restrictive silencer factor (NRSF), also named repressor element-1-silencing transcription factor (REST), which was initially detected as a transcriptional repressor of neuron-specific genes in non-neuronal cells, plays a pivotal role in the transcriptional regulation of ANP, BNP and other fetal cardiac genes. Here we review the transcriptional regulation of ANP and BNP gene expression and the role of the NRSF repressor complex in the regulation of cardiac gene expression and the maintenance of cardiac homeostasis.
Collapse
Affiliation(s)
- Hideaki Inazumi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Koichiro Kuwahara
- Department of Cardiovascular Medicine, School of Medicine, Shinshu University, 3-1-1 Asahi, Nagano 390-8621, Japan
| |
Collapse
|
12
|
Piantoni C, Paina M, Molla D, Liu S, Bertoli G, Jiang H, Wang Y, Wang Y, Wang Y, DiFrancesco D, Barbuti A, Bucchi A, Baruscotti M. Chinese natural compound decreases pacemaking of rabbit cardiac sinoatrial cells by targeting second messenger regulation of f-channels. eLife 2022; 11:75119. [PMID: 35315774 PMCID: PMC8940175 DOI: 10.7554/elife.75119] [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: 10/29/2021] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Tongmai Yangxin (TMYX) is a complex compound of the Traditional Chinese Medicine (TCM) used to treat several cardiac rhythm disorders; however, no information regarding its mechanism of action is available. In this study we provide a detailed characterization of the effects of TMYX on the electrical activity of pacemaker cells and unravel its mechanism of action. Single-cell electrophysiology revealed that TMYX elicits a reversible and dose-dependent (2/6 mg/ml) slowing of spontaneous action potentials rate (−20.8/–50.2%) by a selective reduction of the diastolic phase (−50.1/–76.0%). This action is mediated by a negative shift of the If activation curve (−6.7/–11.9 mV) and is caused by a reduction of the cyclic adenosine monophosphate (cAMP)-induced stimulation of pacemaker channels. We provide evidence that TMYX acts by directly antagonizing the cAMP-induced allosteric modulation of the pacemaker channels. Noticeably, this mechanism functionally resembles the pharmacological actions of muscarinic stimulation or β-blockers, but it does not require generalized changes in cytoplasmic cAMP levels thus ensuring a selective action on rate. In agreement with a competitive inhibition mechanism, TMYX exerts its maximal antagonistic action at submaximal cAMP concentrations and then progressively becomes less effective thus ensuring a full contribution of If to pacemaker rate during high metabolic demand and sympathetic stimulation.
Collapse
Affiliation(s)
- Chiara Piantoni
- Department of Biosciences, The Cell Physiology Lab and "Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata", Università degli Studi di Milano, Milano, Italy
| | - Manuel Paina
- Department of Biosciences, The Cell Physiology Lab and "Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata", Università degli Studi di Milano, Milano, Italy
| | - David Molla
- Department of Biosciences, The Cell Physiology Lab and "Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata", Università degli Studi di Milano, Milano, Italy
| | - Sheng Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Giorgia Bertoli
- Department of Biosciences, The Cell Physiology Lab and "Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata", Università degli Studi di Milano, Milano, Italy
| | - Hongmei Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Yanyan Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Institute of Traditional Chinese Medicine Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Dario DiFrancesco
- Department of Biosciences, The Cell Physiology Lab and "Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata", Università degli Studi di Milano, Milano, Italy
| | - Andrea Barbuti
- Department of Biosciences, The Cell Physiology Lab and "Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata", Università degli Studi di Milano, Milano, Italy
| | - Annalisa Bucchi
- Department of Biosciences, The Cell Physiology Lab and "Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata", Università degli Studi di Milano, Milano, Italy
| | - Mirko Baruscotti
- Department of Biosciences, The Cell Physiology Lab and "Centro Interuniversitario di Medicina Molecolare e Biofisica Applicata", Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
13
|
Caveolin-3 and Arrhythmias: Insights into the Molecular Mechanisms. J Clin Med 2022; 11:jcm11061595. [PMID: 35329921 PMCID: PMC8952412 DOI: 10.3390/jcm11061595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/02/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023] Open
Abstract
Caveolin-3 is a muscle-specific protein on the membrane of myocytes correlated with a variety of cardiovascular diseases. It is now clear that the caveolin-3 plays a critical role in the cardiovascular system and a significant role in cardiac protective signaling. Mutations in the gene encoding caveolin-3 cause a broad spectrum of clinical phenotypes, ranging from persistent elevations in the serum levels of creatine kinase in asymptomatic humans to cardiomyopathy. The influence of Caveolin-3(CAV-3) mutations on current density parallels the effect on channel trafficking. For example, mutations in the CAV-3 gene promote ventricular arrhythmogenesis in long QT syndrome 9 by a combined decrease in the loss of the inward rectifier current (IK1) and gain of the late sodium current (INa-L). The functional significance of the caveolin-3 has proved that caveolin-3 overexpression or knockdown contributes to the occurrence and development of arrhythmias. Caveolin-3 overexpression could lead to reduced diastolic spontaneous Ca2+ waves, thus leading to the abnormal L-Type calcium channel current-induced ventricular arrhythmias. Moreover, CAV-3 knockdown resulted in a shift to more negative values in the hyperpolarization-activated cyclic nucleotide channel 4 current (IHCN4) activation curve and a significant decrease in IHCN4 whole-cell current density. Recent evidence indicates that caveolin-3 plays a significant role in adipose tissue and is related to obesity development. The role of caveolin-3 in glucose homeostasis has attracted increasing attention. This review highlights the underlining mechanisms of caveolin-3 in arrhythmia. Progress in this field may contribute to novel therapeutic approaches for patients prone to developing arrhythmia.
Collapse
|
14
|
Depuydt AS, Peigneur S, Tytgat J. Review: HCN Channels in the Heart. Curr Cardiol Rev 2022; 18:e040222200836. [PMID: 35125083 PMCID: PMC9893134 DOI: 10.2174/1573403x18666220204142436] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/13/2021] [Accepted: 12/23/2021] [Indexed: 11/22/2022] Open
Abstract
Pacemaker cells are the basis of rhythm in the heart. Cardiovascular diseases, and in particular, arrhythmias are a leading cause of hospital admissions and have been implicated as a cause of sudden death. The prevalence of people with arrhythmias will increase in the next years due to an increase in the ageing population and risk factors. The current therapies are limited, have a lot of side effects, and thus, are not ideal. Pacemaker channels, also called hyperpolarizationactivated cyclic nucleotide-gated (HCN) channels, are the molecular correlate of the hyperpolarization- activated current, called Ih (from hyperpolarization) or If (from funny), that contribute crucially to the pacemaker activity in cardiac nodal cells and impulse generation and transmission in neurons. HCN channels have emerged as interesting targets for the development of drugs, in particular, to lower the heart rate. Nonetheless, their pharmacology is still rather poorly explored in comparison to many other voltage-gated ion channels or ligand-gated ion channels. Ivabradine is the first and currently the only clinically approved compound that specifically targets HCN channels. The therapeutic indication of ivabradine is the symptomatic treatment of chronic stable angina pectoris in patients with coronary artery disease with a normal sinus rhythm. Several other pharmacological agents have been shown to exert an effect on heart rate, although this effect is not always desired. This review is focused on the pacemaking process taking place in the heart and summarizes the current knowledge on HCN channels.
Collapse
Affiliation(s)
- Anne-Sophie Depuydt
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg, O&N2, PO Box 922, Herestraat 49, 3000Leuven, Belgium
| | - Steve Peigneur
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg, O&N2, PO Box 922, Herestraat 49, 3000Leuven, Belgium
| | - Jan Tytgat
- Toxicology and Pharmacology, University of Leuven (KU Leuven), Campus Gasthuisberg, O&N2, PO Box 922, Herestraat 49, 3000Leuven, Belgium
| |
Collapse
|
15
|
Ivabradine and Atrial Fibrillation: A Meta-analysis of Randomized Controlled Trials. J Cardiovasc Pharmacol 2021; 79:549-557. [PMID: 34983905 DOI: 10.1097/fjc.0000000000001209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/20/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT This was a meta-analysis of randomized control trials (RCTs) to evaluate the effect of ivabradine on the risk of atrial fibrillation (AF) as well as its effect on the ventricular rate in patients with AF. The PubMed, EMBASE, Cochrane Controlled Trials Register, and other databases were searched for RCTs of ivabradine. Thirteen trials with 37,533 patients met the inclusion criteria. The incidence of AF was significantly higher in the ivabradine treatment group than in the control group (odds ratio (OR), 1.23; 95% confidence interval (CI), 1.08-1.41), although it was reduced after cardiac surgery (OR, 0.70; 95% CI, 0.23-2.12). Regarding left ventricular ejection fraction (LVEF), ivabradine increased the risk of AF in both LVEF >40% (OR, 1.42; 95% CI, 1.24 to 1.63) and LVEF ≤40% subgroups (OR, 1.16; 95% CI, 0.98-1.37). The risk of AF was increased by both small and large cumulative doses of ivabradine (small cumulative dose: OR, 3.00; 95% CI, 0.48 to 18.93; large cumulative dose: OR, 1.05; 95% CI, 0.83-1.34). Furthermore, ivabradine may reduce the ventricular rate in patients with AF. In conclusion, we found that both large and small cumulative doses of ivabradine were associated with an increased incidence of AF, and the effect was more marked in the LVEF >40% subgroup. Nevertheless, ivabradine therapy is beneficial for the prevention of post-operative AF. Furthermore, ivabradine may be effective in controlling the ventricular rate in patients with AF, although more RCTs are needed to support this conclusion.
Collapse
|
16
|
Rodríguez-Angulo HO, Colombet-Naranjo D, Maza MC, Poveda C, Herreros-Cabello A, Mendoza I, Perera JC, Goyo JD, Gironès N, Fresno M. Molecular Remodeling of Cardiac Sinus Node Associated with Acute Chagas Disease Myocarditis. Microorganisms 2021; 9:microorganisms9112208. [PMID: 34835334 PMCID: PMC8620628 DOI: 10.3390/microorganisms9112208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 01/04/2023] Open
Abstract
Chagas disease principally affects Latin-American people, but it currently has worldwide distribution due to migration. Death among those with Chagas disease can occur suddenly and without warning, even in those who may not have evidence of clinical or structural cardiac disease and who are younger than 60 years old. HCN4 channels, one of the principal elements responsible for pacemaker currents, are associated with cardiac fetal reprogramming and supraventricular and ventricular arrhythmias, but their role in chagasic arrhythmias is not clear. We found that a single-dose administration of ivabradine, which blocks HCN4, caused QTc and QRS enlargement and an increase in P-wave amplitude and was associated with ventricular and supraventricular arrhythmias in mice challenged with isoproterenol, a chronotropic/ionotropic positive agent. Continuous treatment with ivabradine did not alter the QTc interval, but P-wave morphology was deeply modified, generating supraventricular arrhythmias. In addition, we found that repolarization parameters improved with ivabradine treatment. These effects could have been caused by the high HCN4 expression observed in auricular and ventricular tissue in infected mice. Thus, we suggest, for the first time, that molecular remodeling by overexpression of HCN4 channels may be related to supraventricular arrhythmias in acute Chagas disease, causing ivabradine over-response. Thus, ivabradine treatment should be administered with caution, while HCN4 overexpression may be an indicator of heart failure and/or sudden death risk.
Collapse
Affiliation(s)
- Héctor O. Rodríguez-Angulo
- Instituto Venezolano de Investigaciones Científicas, Caracas 1020A, Venezuela; (H.O.R.A.); (D.C.-N.)
- Unidad de Biología Celular, Departamento de Ciencias Morfológicas, Programa de Medicina, Facultad de Ciencias de la vida, Universidad Centroccidental Lisandro Alvarado, Barquisimeto 3001, Venezuela; (J.C.P.); (J.D.G.)
| | - Diana Colombet-Naranjo
- Instituto Venezolano de Investigaciones Científicas, Caracas 1020A, Venezuela; (H.O.R.A.); (D.C.-N.)
| | - María C. Maza
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.C.M.); (C.P.); (A.-H.C.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | - Cristina Poveda
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.C.M.); (C.P.); (A.-H.C.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | - Alfonso Herreros-Cabello
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.C.M.); (C.P.); (A.-H.C.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
| | - Iván Mendoza
- Instituto de Medicina Tropical, Universidad Central de Venezuela, Caracas 1060, Venezuela;
| | - Juan C. Perera
- Unidad de Biología Celular, Departamento de Ciencias Morfológicas, Programa de Medicina, Facultad de Ciencias de la vida, Universidad Centroccidental Lisandro Alvarado, Barquisimeto 3001, Venezuela; (J.C.P.); (J.D.G.)
| | - Juan D. Goyo
- Unidad de Biología Celular, Departamento de Ciencias Morfológicas, Programa de Medicina, Facultad de Ciencias de la vida, Universidad Centroccidental Lisandro Alvarado, Barquisimeto 3001, Venezuela; (J.C.P.); (J.D.G.)
| | - Núria Gironès
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.C.M.); (C.P.); (A.-H.C.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario de la Princesa, 28009 Madrid, Spain
- Correspondence: (N.G.); (M.F.)
| | - Manuel Fresno
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.C.M.); (C.P.); (A.-H.C.)
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, 28049 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario de la Princesa, 28009 Madrid, Spain
- Correspondence: (N.G.); (M.F.)
| |
Collapse
|
17
|
Oknińska M, Paterek A, Zambrowska Z, Mackiewicz U, Mączewski M. Effect of Ivabradine on Cardiac Ventricular Arrhythmias: Friend or Foe? J Clin Med 2021; 10:4732. [PMID: 34682854 PMCID: PMC8537674 DOI: 10.3390/jcm10204732] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/24/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Life-threatening ventricular arrhythmias, such as ventricular tachycardia and ventricular fibrillation remain an ongoing clinical problem and their prevention and treatment require optimization. Conventional antiarrhythmic drugs are associated with significant proarrhythmic effects that often outweigh their benefits. Another option, the implantable cardioverter defibrillator, though clearly the primary therapy for patients at high risk of ventricular arrhythmias, is costly, invasive, and requires regular monitoring. Thus there is a clear need for new antiarrhythmic treatment strategies. Ivabradine, a heartrate-reducing agent, an inhibitor of HCN channels, may be one of such options. In this review we discuss emerging data from experimental studies that indicate new mechanism of action of this drug and further areas of investigation and potential use of ivabradine as an antiarrhythmic agent. However, clinical evidence is limited, and the jury is still out on effects of ivabradine on cardiac ventricular arrhythmias in the clinical setting.
Collapse
Affiliation(s)
| | | | | | | | - Michał Mączewski
- Centre of Postgraduate Medical Education, Department of Clinical Physiology, ul. Marymoncka 99/103, 01-813 Warsaw, Poland; (M.O.); (A.P.); (Z.Z.); (U.M.)
| |
Collapse
|
18
|
Wang J, Xia Y, Lu A, Wang H, Davis DR, Liu P, Beanlands RS, Liang W. Cardiomyocyte-specific deletion of β-catenin protects mouse hearts from ventricular arrhythmias after myocardial infarction. Sci Rep 2021; 11:17722. [PMID: 34489488 PMCID: PMC8421412 DOI: 10.1038/s41598-021-97176-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 08/23/2021] [Indexed: 11/09/2022] Open
Abstract
Wnt/β-catenin signaling is activated in the heart after myocardial infarction (MI). This study aims to investigate if β-catenin deletion affects post-MI ion channel gene alterations and ventricular tachycardias (VT). MI was induced by permanent ligation of left anterior descending artery in wild-type (WT) and cardiomyocyte-specific β-catenin knockout (KO) mice. KO mice showed reduced susceptibility to VT (18% vs. 77% in WT) at 8 weeks after MI, associated with reduced scar size and attenuated chamber dilation. qPCR analyses of both myocardial tissues and purified cardiomyocytes demonstrated upregulation of Wnt pathway genes in border and infarct regions after MI, including Wnt ligands (such as Wnt4) and receptors (such as Fzd1 and Fzd2). At 1 week after MI, cardiac sodium channel gene (Scn5a) transcript was reduced in WT but not in KO hearts, consistent with previous studies showing Scn5a inhibition by Wnt/β-catenin signaling. At 8 weeks after MI when Wnt genes have declined, Scn5a returned to near sham levels and K+ channel gene downregulations were not different between WT and KO mice. This study demonstrated that VT susceptibility in the chronic phase after MI is reduced in mice with cardiomyocyte-specific β-catenin deletion primarily through attenuated structural remodeling, but not ion channel gene alterations.
Collapse
Affiliation(s)
- Jerry Wang
- University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, ON, K1Y 4W7, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Ying Xia
- University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, ON, K1Y 4W7, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Aizhu Lu
- University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, ON, K1Y 4W7, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Hongwei Wang
- University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, ON, K1Y 4W7, Canada
| | - Darryl R Davis
- University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, ON, K1Y 4W7, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Peter Liu
- University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, ON, K1Y 4W7, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Rob S Beanlands
- University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, ON, K1Y 4W7, Canada
| | - Wenbin Liang
- University of Ottawa Heart Institute, 40 Ruskin St, Ottawa, ON, K1Y 4W7, Canada. .,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
19
|
Effect of ivabradine on cardiac arrhythmias: Antiarrhythmic or proarrhythmic? Heart Rhythm 2021; 18:1230-1238. [PMID: 33737235 DOI: 10.1016/j.hrthm.2021.03.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/28/2022]
Abstract
Cardiac arrhythmias are a major source of mortality and morbidity. Unfortunately, their treatment remains suboptimal. Major classes of antiarrhythmic drugs pose a significant risk of proarrhythmia, and their side effects often outweigh their benefits. Therefore, implantable devices remain the only truly effective antiarrhythmic therapy, and new strategies of antiarrhythmic treatment are required. Ivabradine is a selective heart rate-reducing agent, an inhibitor of hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels, currently approved for treatment of coronary artery disease and chronic heart failure. In this review, we focus on the clinical and basic science evidence for the antiarrhythmic and proarrhythmic effects of ivabradine. We attempt to dissect the mechanisms behind the effects of ivabradine and indicate the focus of future studies.
Collapse
|
20
|
Li Y, Wang K, Li Q, Hancox JC, Zhang H. Reciprocal interaction between IK1 and If in biological pacemakers: A simulation study. PLoS Comput Biol 2021; 17:e1008177. [PMID: 33690622 PMCID: PMC7984617 DOI: 10.1371/journal.pcbi.1008177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 03/22/2021] [Accepted: 02/17/2021] [Indexed: 11/19/2022] Open
Abstract
Pacemaking dysfunction (PD) may result in heart rhythm disorders, syncope or even death. Current treatment of PD using implanted electronic pacemakers has some limitations, such as finite battery life and the risk of repeated surgery. As such, the biological pacemaker has been proposed as a potential alternative to the electronic pacemaker for PD treatment. Experimentally and computationally, it has been shown that bio-engineered pacemaker cells can be generated from non-rhythmic ventricular myocytes (VMs) by knocking out genes related to the inward rectifier potassium channel current (IK1) or by overexpressing hyperpolarization-activated cyclic nucleotide gated channel genes responsible for the "funny" current (If). However, it is unclear if a bio-engineered pacemaker based on the modification of IK1- and If-related channels simultaneously would enhance the ability and stability of bio-engineered pacemaking action potentials. In this study, the possible mechanism(s) responsible for VMs to generate spontaneous pacemaking activity by regulating IK1 and If density were investigated by a computational approach. Our results showed that there was a reciprocal interaction between IK1 and If in ventricular pacemaker model. The effect of IK1 depression on generating ventricular pacemaker was mono-phasic while that of If augmentation was bi-phasic. A moderate increase of If promoted pacemaking activity but excessive increase of If resulted in a slowdown in the pacemaking rate and even an unstable pacemaking state. The dedicated interplay between IK1 and If in generating stable pacemaking and dysrhythmias was evaluated. Finally, a theoretical analysis in the IK1/If parameter space for generating pacemaking action potentials in different states was provided. In conclusion, to the best of our knowledge, this study provides a wide theoretical insight into understandings for generating stable and robust pacemaker cells from non-pacemaking VMs by the interplay of IK1 and If, which may be helpful in designing engineered biological pacemakers for application purposes.
Collapse
Affiliation(s)
- Yacong Li
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Kuanquan Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
- * E-mail: (KW); (HZ)
| | - Qince Li
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
- Peng Cheng Laboratory, Shenzhen, China
| | - Jules C. Hancox
- School of Physiology, Pharmacology and Neuroscience, Medical Sciences Building, University Walk, Bristol, United Kingdom
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
| | - Henggui Zhang
- Peng Cheng Laboratory, Shenzhen, China
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, United Kingdom
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- * E-mail: (KW); (HZ)
| |
Collapse
|
21
|
Hadova K, Kralova E, Doka G, Bies Pivackova L, Kmecova Z, Krenek P, Klimas J. Isolated downregulation of HCN2 in ventricles of rats with streptozotocin-induced diabetic cardiomyopathy. BMC Cardiovasc Disord 2021; 21:118. [PMID: 33653265 PMCID: PMC7927235 DOI: 10.1186/s12872-021-01929-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/11/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In spite of disrupted repolarization of diabetic heart, some studies report less tendency of diabetic heart to develop ventricular arrhythmias suggesting effective compensatory mechanism. We hypothesized that myocardial alterations in HCN2 and HCN4 channels occur under hyperglycaemia. METHODS Diabetes was induced in rats using a single injection of streptozotocin (STZ; 55 mg/kg body weight, i.p.). Basal ECG was measured. Expression of mRNA for HCN channels, potassium channels and microRNA 1 and 133a were measured in ventricular tissues. Protein expression of HCN2 channel isoform was assessed in five different regions of the heart by western blotting. Differentiated H9c2 cell line was used to examine HCN channels expression under hyperglycaemia in vitro. RESULTS Six weeks after STZ administration, heart rate was reduced, QRS complex duration, QT interval and T-wave were prolonged in diabetic rats compared to controls. mRNA and protein expressions of HCN2 decreased exclusively in the ventricles of diabetic rats. HCN2 expression levels in atria of STZ rats and H9c2 cells treated with excess of glucose were not changed. MicroRNA levels were stable in STZ rat hearts. We found significantly decreased mRNA levels of several potassium channels participating in repolarization, namely Kcnd2 (Ito1), Kcnh2 (IKr), Kcnq1 (IKs) and Kcnj11 (IKATP). CONCLUSIONS This result together with downregulated HCN2 channels suggest that HCN channels might be an integral part of ventricular electric remodelling and might play a role in cardiac repolarization projected in altered arrhythmogenic profile of diabetic heart.
Collapse
Affiliation(s)
- Katarina Hadova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, 832 32, Bratislava, Slovakia
| | - Eva Kralova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, 832 32, Bratislava, Slovakia
| | - Gabriel Doka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, 832 32, Bratislava, Slovakia
| | - Lenka Bies Pivackova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, 832 32, Bratislava, Slovakia
| | - Zuzana Kmecova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, 832 32, Bratislava, Slovakia
| | - Peter Krenek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, 832 32, Bratislava, Slovakia
| | - Jan Klimas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, Odbojarov 10, 832 32, Bratislava, Slovakia.
| |
Collapse
|
22
|
Nakagawa T, Yasaka T, Nakashima N, Takeya M, Oshita K, Tsuda M, Yamaura K, Takano M. Expression of the pacemaker channel HCN4 in excitatory interneurons in the dorsal horn of the murine spinal cord. Mol Brain 2020; 13:127. [PMID: 32948209 PMCID: PMC7501643 DOI: 10.1186/s13041-020-00666-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/07/2020] [Indexed: 11/17/2022] Open
Abstract
In the central nervous system, hyperpolarization-activated, cyclic nucleotide-gated (HCN1–4) channels have been implicated in neuronal excitability and synaptic transmission. It has been reported that HCN channels are expressed in the spinal cord, but knowledge about their physiological roles, as well as their distribution profiles, appear to be limited. We generated a transgenic mouse in which the expression of HCN4 can be reversibly knocked down using a genetic tetracycline-dependent switch and conducted genetically validated immunohistochemistry for HCN4. We found that the somata of HCN4-immunoreactive (IR) cells were largely restricted to the ventral part of the inner lamina II and lamina III. Many of these cells were either parvalbumin- or protein kinase Cγ (PKCγ)-IR. By using two different mouse strains in which reporters are expressed only in inhibitory neurons, we determined that the vast majority of HCN4-IR cells were excitatory neurons. Mechanical and thermal noxious stimulation did not induce c-Fos expression in HCN4-IR cells. PKCγ-neurons in this area are known to play a pivotal role in the polysynaptic pathway between tactile afferents and nociceptive projection cells that contributes to tactile allodynia. Therefore, pharmacological and/or genetic manipulations of HCN4-expressing neurons may provide a novel therapeutic strategy for the pain relief of tactile allodynia.
Collapse
Affiliation(s)
- Taku Nakagawa
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan.,Department of Anesthesiology, Graduate School of Medicine, Kyushu University, Fukuoka, 812-8582, Japan
| | - Toshiharu Yasaka
- Department of Health and Nutrition, Niigata University of Hearth and Welfare, Niigata, 950-3198, Japan
| | - Noriyuki Nakashima
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Mitsue Takeya
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Kensuke Oshita
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan.,Department of Anesthesiology, Kurume University School of Medicine, Kurume, 830-0011, Japan
| | - Makoto Tsuda
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Ken Yamaura
- Department of Anesthesiology, Graduate School of Medicine, Kyushu University, Fukuoka, 812-8582, Japan
| | - Makoto Takano
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan.
| |
Collapse
|
23
|
Chakraborty P, Rose RA, Nair K, Downar E, Nanthakumar K. The rationale for repurposing funny current inhibition for management of ventricular arrhythmia. Heart Rhythm 2020; 18:130-137. [PMID: 32738405 DOI: 10.1016/j.hrthm.2020.07.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/14/2020] [Accepted: 07/25/2020] [Indexed: 11/26/2022]
Abstract
Management of ventricular arrhythmia in structural heart disease is complicated by the toxicity of the limited antiarrhythmic options available. In others, proarrhythmia and deleterious hemodynamic and noncardiac effects prevent practical use. This necessitates new thinking in therapeutic agents for ventricular arrhythmia in structural heart disease. Ivabradine, a funny current (If) inhibitor, has proven safety in heart failure, angina, and inappropriate sinus tachycardia. Although it is commonly known that funny channels are primarily expressed in the sinoatrial node, atrioventricular node, and conducting system of the ventricle, ivabradine is known to exert effects on metabolism, ion homeostasis, and membrane electrophysiology of remodeled ventricular myocardium. This review considers novel concepts and evidence from clinical and experimental studies regarding this paradigm, with a potential role of ivabradine in ventricular arrhythmia.
Collapse
Affiliation(s)
- Praloy Chakraborty
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; University Health Network, Toronto, Ontario, Canada
| | - Robert A Rose
- Libin Cardiovascular Institute of Alberta, An entity of the University of Calgary and Alberta Health Services, Calgary, Alberta, Canada
| | - Krishnakumar Nair
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; University Health Network, Toronto, Ontario, Canada
| | - Eugene Downar
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; University Health Network, Toronto, Ontario, Canada
| | - Kumaraswamy Nanthakumar
- The Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
24
|
Henry S, Szabó V, Sutus E, Pirity MK. RYBP is important for cardiac progenitor cell development and sarcomere formation. PLoS One 2020; 15:e0235922. [PMID: 32673370 PMCID: PMC7365410 DOI: 10.1371/journal.pone.0235922] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/24/2020] [Indexed: 12/28/2022] Open
Abstract
We have previously established that epigenetic regulator RING1 and YY1 binding protein (RYBP) is required for the contractility of embryonic stem (ES) cell derived cardiomyocytes (CMCs), suggesting its essential role in contractility. In order to investigate the underlying molecular events of this phenotype, we compared the transcriptomic profile of the wild type and Rybp null mutant ES cells and CMCs differentiated from these cell lines. We identified genes related to ion homeostasis, cell adhesion and sarcomeric organization affected in the Rybp null mutant CMCs, by using hierarchical gene clustering and Gene Ontology analysis. We have also demonstrated that the amount of RYBP is drastically reduced in the terminally differentiated wild type CMCs whilst it is broadly expressed in the early phase of differentiation when progenitors form. We also describe that RYBP is important for the proper expression of key cardiac transcription factors including Mesp1, Shh and Mef2c. These findings identify Rybp as a gene important for both early cardiac gene transcription and consequent sarcomere formation necessary for contractility. Since impairment of sarcomeric function and contractility plays a central role in reduced cardiac pump function leading to heart failures in human, current results might be relevant to the pathophysiology of cardiomyopathies.
Collapse
Affiliation(s)
- Surya Henry
- Biological Research Centre, Szeged, Hungary
- Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Viktória Szabó
- Biological Research Centre, Szeged, Hungary
- Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Enikő Sutus
- Biological Research Centre, Szeged, Hungary
- Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | | |
Collapse
|
25
|
Li Y, Wang K, Li Q, Zhang H. Biological pacemaker: from biological experiments to computational simulation. J Zhejiang Univ Sci B 2020; 21:524-536. [PMID: 32633107 DOI: 10.1631/jzus.b1900632] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Pacemaking dysfunction has become a significant disease that may contribute to heart rhythm disorders, syncope, and even death. Up to now, the best way to treat it is to implant electronic pacemakers. However, these have many disadvantages such as limited battery life, infection, and fixed pacing rate. There is an urgent need for a biological pacemaker (bio-pacemaker). This is expected to replace electronic devices because of its low risk of complications and the ability to respond to emotion. Here we survey the contemporary development of the bio-pacemaker by both experimental and computational approaches. The former mainly includes gene therapy and cell therapy, whilst the latter involves the use of multi-scale computer models of the heart, ranging from the single cell to the tissue slice. Up to now, a bio-pacemaker has been successfully applied in big mammals, but it still has a long way from clinical uses for the treatment of human heart diseases. It is hoped that the use of the computational model of a bio-pacemaker may accelerate this process. Finally, we propose potential research directions for generating a bio-pacemaker based on cardiac computational modeling.
Collapse
Affiliation(s)
- Yacong Li
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Kuanquan Wang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Qince Li
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China.,Peng Cheng Laboratory, Shenzhen 518052, China
| | - Henggui Zhang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin 150001, China.,School of Physics and Astronomy, The University of Manchester, Manchester, M13 9PL, UK.,Peng Cheng Laboratory, Shenzhen 518052, China
| |
Collapse
|
26
|
Notch1-mediated histone demethylation of HCN4 contributes to aconitine-induced ventricular myocardial dysrhythmia. Toxicol Lett 2020; 327:19-31. [DOI: 10.1016/j.toxlet.2020.03.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/12/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022]
|
27
|
Han Z, Wu X, Gao Y, Liu X, Bai J, Gu R, Lan R, Xu B, Xu W. PDK1-AKT signaling pathway regulates the expression and function of cardiac hyperpolarization-activated cyclic nucleotide-modulated channels. Life Sci 2020; 250:117546. [PMID: 32184125 DOI: 10.1016/j.lfs.2020.117546] [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: 01/03/2020] [Revised: 03/03/2020] [Accepted: 03/11/2020] [Indexed: 11/18/2022]
Abstract
AIM The enzyme 3-phosphoinositide-dependent protein kinase-1 (PDK1) is associated with cardiac and pathological remodeling and ion channel function regulation. However, whether it regulates hyperpolarization-activated cyclic nucleotide-modulated channels (HCNs) remains unclear. MAIN METHODS In the atrial myocytes of heart-specific PDK1 "knockout" mouse model and neonatal mice, protein kinase B (AKT)-related inhibitors or agonists as well as knockdown or overexpression plasmids were used to study the relationship between PDK1 and HCNs. KEY FINDINGS HCN1 expression and AKT phosphorylation at the Thr308 site were significantly decreased in atrial myocytes after PDK1 knockout or inhibition; in contrast, HCN2 and HCN4 levels were significantly increased. Also, a similar trend of HCNs expression has been observed in cultured atrial myocytes after PDK1 inhibition, as further demonstrated via immunofluorescence and patch-clamp experiments. Moreover, these results of PDK1 overexpression indicate an opposite trend compared with the previous experimental results. However, the results of PDK1 inhibition or overexpression could be reversed by activating or inhibiting AKT, respectively. SIGNIFICANCE These results indicate that the PDK1-AKT signaling pathway is involved in the regulation of HCN mRNA transcription, protein expression, HCN current density, and cell membrane location.
Collapse
Affiliation(s)
- Zhonglin Han
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Xiang Wu
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Yuan Gao
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Xuehua Liu
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Jian Bai
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Rong Gu
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - RongFang Lan
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Biao Xu
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China
| | - Wei Xu
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, China.
| |
Collapse
|
28
|
Rivolta I, Binda A, Masi A, DiFrancesco JC. Cardiac and neuronal HCN channelopathies. Pflugers Arch 2020; 472:931-951. [PMID: 32424620 DOI: 10.1007/s00424-020-02384-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are expressed as four different isoforms (HCN1-4) in the heart and in the central and peripheral nervous systems. In the voltage range of activation, HCN channels carry an inward current mediated by Na+ and K+, termed If in the heart and Ih in neurons. Altered function of HCN channels, mainly HCN4, is associated with sinus node dysfunction and other arrhythmias such as atrial fibrillation, ventricular tachycardia, and atrioventricular block. In recent years, several data have also shown that dysfunctional HCN channels, in particular HCN1, but also HCN2 and HCN4, can play a pathogenic role in epilepsy; these include experimental data from animal models, and data collected over genetic mutations of the channels identified and characterized in epileptic patients. In the central nervous system, alteration of the Ih current could predispose to the development of neurodegenerative diseases such as Parkinson's disease; since HCN channels are widely expressed in the peripheral nervous system, their dysfunctional behavior could also be associated with the pathogenesis of neuropathic pain. Given the fundamental role played by the HCN channels in the regulation of the discharge activity of cardiac and neuronal cells, the modulation of their function for therapeutic purposes is under study since it could be useful in various pathological conditions. Here we review the present knowledge of the HCN-related channelopathies in cardiac and neurological diseases, including clinical, genetic, therapeutic, and physiopathological aspects.
Collapse
Affiliation(s)
- Ilaria Rivolta
- School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Monza, Italy
| | - Anna Binda
- School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Monza, Italy
| | - Alessio Masi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Jacopo C DiFrancesco
- School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Monza, Italy. .,Department of Neurology, ASST San Gerardo Hospital, University of Milano-Bicocca, Via Pergolesi, 33, 20900, Monza, MB, Italy.
| |
Collapse
|
29
|
Li Y, Wang K, Li Q, Luo C, Zhang H. Role of I f Density on Electrical Action Potential of Bio-engineered Cardiac Pacemaker: A Simulation Study. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:3995-3998. [PMID: 31946747 DOI: 10.1109/embc.2019.8856350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Due to the inevitable drawbacks of the implantable electrical pacemaker, the biological pacemaker was believed to be an alternative therapy for heart failure. Previous experimental studies have shown that biological pacemaker could be produced by genetically manipulating non-pacemaking cardiac cells by suppressing the inward rectifier potassium current (IK1) and expressing the hyperpolarization- activated current (If). However, the role of If in such bio-engineered pacemaker is not clear. In this study, we simulated the action potential of biological pacemaker cells by manipulating If-IK1 parameters (i.e., inhibiting IK1 as well as incorporating If) to analyze possible mechanisms by which different If densities control pacemaking action potentials. Our simulation results showed different pacing mechanism between the bioengineered pacemaking cells with and without If. In addition, it was shown that a greater If density might result in a slower pacing frequency, and excessive of it might produce an early-afterdepolarizations-like action potential due to a sudden release of calcium from sarcoplasmic reticulum into the cytoplasm. This study indicated that when IK1 was significantly suppressed, incorporating If may not enhance the pacing ability of biological pacemaker, but lead to abnormal dynamics of intracellular ionic concentration, increasing risks of dysrhythmia in the heart.
Collapse
|
30
|
Lussier Y, Fürst O, Fortea E, Leclerc M, Priolo D, Moeller L, Bichet DG, Blunck R, D'Avanzo N. Disease-linked mutations alter the stoichiometries of HCN-KCNE2 complexes. Sci Rep 2019; 9:9113. [PMID: 31235733 PMCID: PMC6591248 DOI: 10.1038/s41598-019-45592-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 06/11/2019] [Indexed: 12/21/2022] Open
Abstract
The four hyperpolarization-activated cylic-nucleotide gated (HCN) channel isoforms and their auxiliary subunit KCNE2 are important in the regulation of peripheral and central neuronal firing and the heartbeat. Disruption of their normal function has been implicated in cardiac arrhythmias, peripheral pain, and epilepsy. However, molecular details of the HCN-KCNE2 complexes are unknown. Using single-molecule subunit counting, we determined that the number of KCNE2 subunits in complex with the pore-forming subunits of human HCN channels differs with each HCN isoform and is dynamic with respect to concentration. These interactions can be altered by KCNE2 gene-variants with functional implications. The results provide an additional consideration necessary to understand heart rhythm, pain, and epileptic disorders.
Collapse
Affiliation(s)
- Yoann Lussier
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Oliver Fürst
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Eva Fortea
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Marc Leclerc
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Dimitri Priolo
- Department of Physics, Université de Montréal, Montréal, Canada
| | - Lena Moeller
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Canada
| | - Daniel G Bichet
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Rikard Blunck
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada.,Department of Physics, Université de Montréal, Montréal, Canada
| | - Nazzareno D'Avanzo
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada. .,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
31
|
Oshita K, Kozasa Y, Nakagawa Y, Kuwabara Y, Kuwahara K, Nakagawa T, Nakashima N, Hiraki T, Takano M. Overexpression of the HCN2 channel increases the arrhythmogenicity induced by hypokalemia. J Physiol Sci 2019; 69:653-660. [PMID: 31087220 PMCID: PMC6583697 DOI: 10.1007/s12576-019-00684-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022]
Abstract
Hypokalemia, an abnormally low level of potassium (K+), is a electrolyte imbalance that commonly occurs in heart failure patients. Hypokalemia is well known to induce lethal ventricular arrhythmia. However, the effects of hypokalemia in failing hearts that have undergone electrophysiological remodeling, i.e., the reactivation of fetal-type ion channels, remain unexplored. We have examined the effect of hypokalemia in the myocytes of transgenic mice overexpressing the hyperpolarization-activated, cyclic nucleotide-sensitive (HCN) channel in the heart (HCN2-Tg mice). Perfusion with a mild hypokalemic solution containing 3 mM K+ induced ectopic ventricular automaticity in 55.0% of HCN2-Tg mouse myocytes. In the remaining HCN2-Tg mouse myocytes, the resting membrane potential (RMP) was more depolarized than that of wild-type myocytes subjected to the same treatment and could also be hyperpolarized by an HCN channel blocker. We conclude that in hypokalemia in our mice model, the HCN2 channel was constitutively activated at the hyperpolarized RMP, thereby destabilizing the electrophysiological activity of ventricular myocytes.
Collapse
Affiliation(s)
- Kensuke Oshita
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan.,Department of Anesthesiology, Kurume University School of Medicine, Kurume, Japan
| | - Yuko Kozasa
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan.,Department of Anesthesiology, Kurume University School of Medicine, Kurume, Japan
| | - Yasuaki Nakagawa
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshihiro Kuwabara
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koichiro Kuwahara
- Department of Cardiovascular Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Taku Nakagawa
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Noriyuki Nakashima
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
| | - Teruyuki Hiraki
- Department of Anesthesiology, Kurume University School of Medicine, Kurume, Japan
| | - Makoto Takano
- Department of Physiology, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan.
| |
Collapse
|
32
|
Tanguay J, Callahan KM, D'Avanzo N. Characterization of drug binding within the HCN1 channel pore. Sci Rep 2019; 9:465. [PMID: 30679654 PMCID: PMC6345760 DOI: 10.1038/s41598-018-37116-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/29/2018] [Indexed: 11/09/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels mediate rhythmic electrical activity of cardiac pacemaker cells, and in neurons play important roles in setting resting membrane potentials, dendritic integration, neuronal pacemaking, and establishing action potential threshold. Block of HCN channels slows the heart rate and is currently used to treat angina. However, HCN block also provides a promising approach to the treatment of neuronal disorders including epilepsy and neuropathic pain. While several molecules that block HCN channels have been identified, including clonidine and its derivative alinidine, lidocaine, mepivacaine, bupivacaine, ZD7288, ivabradine, zatebradine, and cilobradine, their low affinity and lack of specificity prevents wide-spread use. Different studies suggest that the binding sites of these inhibitors are located in the inner vestibule of HCN channels, but the molecular details of their binding remain unknown. We used computational docking experiments to assess the binding sites and mode of binding of these inhibitors against the recently solved atomic structure of human HCN1 channels, and a homology model of the open pore derived from a closely related CNG channel. We identify a possible hydrophobic groove in the pore cavity that plays an important role in conformationally restricting the location and orientation of drugs bound to the inner vestibule. Our results also help explain the molecular basis of the low-affinity binding of these inhibitors, paving the way for the development of higher affinity molecules.
Collapse
Affiliation(s)
- Jérémie Tanguay
- Department of Physics, Université de Montréal, Montréal, Canada
| | - Karen M Callahan
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Nazzareno D'Avanzo
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada.
| |
Collapse
|
33
|
Wang J, Yang Y, Li Y, Zhu J, Lian H, Shao X, Zhang H, Fu Y, Zhang L. Long‐term treatment with ivabradine in transgenic atrial fibrillation mice counteracts hyperpolarization‐activated cyclic nucleotide gated channel overexpression. J Cardiovasc Electrophysiol 2018; 30:242-252. [PMID: 30302853 DOI: 10.1111/jce.13772] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/24/2018] [Accepted: 10/08/2018] [Indexed: 01/20/2023]
Affiliation(s)
- Juan Wang
- Emergency and Intensive Care Center, State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing China
| | - Yan‐min Yang
- Emergency and Intensive Care Center, State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing China
| | - Yang Li
- Department of CardiologyChinese PLA General HospitalBeijing China
| | - Jun Zhu
- Emergency and Intensive Care Center, State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing China
| | - Hong Lian
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing China
| | - Xing‐hui Shao
- Emergency and Intensive Care Center, State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing China
| | - Han Zhang
- Emergency and Intensive Care Center, State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing China
| | - Yi‐cheng Fu
- Department of GeriatricsPeking University Third HospitalBeijing China
| | - Lian‐feng Zhang
- Key Laboratory of Human Disease Comparative MedicineMinistry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing China
| |
Collapse
|
34
|
Pirintr P, Limprasutr V, Saengklub N, Pavinadol P, Yapao N, Limvanicharat N, Kuecharoen H, Kijtawornrat A. Acute effect of ivabradine on heart rate and myocardial oxygen consumption in dogs with asymptomatic mitral valve degeneration. Exp Anim 2018; 67:441-449. [PMID: 29760343 PMCID: PMC6219887 DOI: 10.1538/expanim.18-0030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/17/2018] [Indexed: 01/30/2023] Open
Abstract
Degenerative mitral valve disease (DMVD) is a common cardiac disease in geriatric dogs characterized by the degeneration of the mitral valve, leading to decreased cardiac output and activation of the sympathetic and renin-angiotensin-aldosterone system. This disease results in an increased resting heart rate (HR) and myocardial oxygen consumption (MVO2). A recent publication demonstrated that dogs with asymptomatic DMVD had a significantly higher HR and systemic blood pressure (BP) than age-matched control dogs. This higher HR will eventually contribute to increased MVO2. This study aimed to determine the effects of a single oral dose of ivabradine on the HR, MVO2 as assessed by the rate-pressure product, and BP in dogs with asymptomatic DMVD. Seven beagles with naturally occurring DMVD were instrumented by the Holter recorder and an oscillometric device to measure electrocardiogram and BP for 24 and 12 h, respectively. Each dog was randomly subjected to receive either placebo or ivabradine (0.5, 1.0 and 2.0 mg/kg). The results revealed that oral administration of ivabradine significantly decreased the HR and rate-pressure product in a dose-dependent manner without adverse effects. The highest dose of 2.0 mg/kg significantly reduced systolic and mean BP. Therefore, the findings imply that a single oral ivabradine administration at a dose of 1.0 mg/kg is suitable for dogs with asymptomatic DMVD to reduce the HR and MVO2 without marked effects on BP. This may potentially make ivabradine promising for management of an elevated HR in DMVD dogs.
Collapse
Affiliation(s)
- Prapawadee Pirintr
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, 39 Henri Dunant Road, Pathumwan, Bangkok 10330, Thailand
| | - Vudhiporn Limprasutr
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, 254 Phayathai Road, Pathumwan, Bangkok 10330, Thailand
| | - Nakkawee Saengklub
- Department of Physiology, Faculty of Pharmacy, Mahidol University, 447 Sri Ayudhya Road, Rajathevi, Bangkok 10400, Thailand
| | - Parnpradub Pavinadol
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, 39 Henri Dunant Road, Pathumwan, Bangkok 10330, Thailand
| | - Napat Yapao
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, 39 Henri Dunant Road, Pathumwan, Bangkok 10330, Thailand
| | - Natthakarn Limvanicharat
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, 39 Henri Dunant Road, Pathumwan, Bangkok 10330, Thailand
| | - Hathaisiri Kuecharoen
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, 39 Henri Dunant Road, Pathumwan, Bangkok 10330, Thailand
| | - Anusak Kijtawornrat
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, 39 Henri Dunant Road, Pathumwan, Bangkok 10330, Thailand
- Research clusters: research study and testing of drug's effect related to cardiovascular system in laboratory animal, Chulalongkorn University, 39 Henri Dunant Road, Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
35
|
Badu-Boateng C, Jennings R, Hammersley D. The therapeutic role of ivabradine in heart failure. Ther Adv Chronic Dis 2018; 9:199-207. [PMID: 30364460 DOI: 10.1177/2040622318784556] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 05/30/2018] [Indexed: 12/18/2022] Open
Abstract
Heart failure represents a major global cause of morbidity and mortality. Ivabradine is a selective funny current (If) inhibitor, which acts on the sinoatrial node, resulting in a reduction in heart rate. Ivabradine is currently licensed for use in patients with symptomatic heart failure with reduced ejection fraction and a heart rate persistently at least 70 beats per minute in spite of otherwise optimal prognostic heart failure pharmacotherapy. In this review article, we examine the mechanism of action of ivabradine, evaluate the clinical trials underpinning its application in heart failure and discuss its current recommended clinical use in this capacity.
Collapse
Affiliation(s)
| | - Robert Jennings
- Department of Medicine, Frimley Park Hospital, Frimley, Camberley, UK
| | - Daniel Hammersley
- Department of Cardiology, Kings College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| |
Collapse
|
36
|
Spinelli V, Sartiani L, Mugelli A, Romanelli MN, Cerbai E. Hyperpolarization-activated cyclic-nucleotide-gated channels: pathophysiological, developmental, and pharmacological insights into their function in cellular excitability. Can J Physiol Pharmacol 2018; 96:977-984. [PMID: 29969572 DOI: 10.1139/cjpp-2018-0115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The hyperpolarization-activated cyclic-nucleotide-gated (HCN) proteins are voltage-dependent ion channels, conducting both Na+ and K+, blocked by millimolar concentrations of extracellular Cs+ and modulated by cyclic nucleotides (mainly cAMP) that contribute crucially to the pacemaker activity in cardiac nodal cells and subsidiary pacemakers. Over the last decades, much attention has focused on HCN current, If, in non-pacemaker cardiac cells and its potential role in triggering arrhythmias. In fact, in addition to pacemakers, HCN current is constitutively present in the human atria and has long been proposed to sustain atrial arrhythmias associated to different cardiac pathologies or triggered by various modulatory signals (catecholamines, serotonin, natriuretic peptides). An atypical If occurs in diseased ventricular cardiomyocytes, its amplitude being linearly related to the severity of cardiac hypertrophy. The properties of atrial and ventricular If and its modulation by pharmacological interventions has been object of intense study, including the synthesis and characterization of new compounds able to block preferentially HCN1, HCN2, or HCN4 isoforms. Altogether, clues emerge for opportunities of future pharmacological strategies exploiting the unique properties of this channel family: the prevalence of different HCN subtypes in organs and tissues, the possibility to target HCN gain- or loss-of-function associated with disease, the feasibility of novel isoform-selective drugs, as well as the discovery of HCN-mediated effects for old medicines.
Collapse
Affiliation(s)
- Valentina Spinelli
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Laura Sartiani
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Alessandro Mugelli
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Maria Novella Romanelli
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Elisabetta Cerbai
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| |
Collapse
|
37
|
Chow SL, Page RL, Depre C. Role of ivabradine and heart rate lowering in chronic heart failure: guideline update. Expert Rev Cardiovasc Ther 2018; 16:515-526. [PMID: 29902387 DOI: 10.1080/14779072.2018.1489235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION This review summarizes the current management of heart failure (HF) in patients with reduced ejection fraction and the potential role of heart rate lowering agents in select populations, as recommended in the updated guidelines. Areas covered: PubMed was searched for studies that evaluated the role of heart rate lowering or ivabradine in HF management. Expert commentary: Targeting heart rate may offer benefit when added to renin-angiotensin aldosterone antagonists, and beta-blockers. Ivabradine is a heart rate lowering agent that acts on the funny current (If) in the sinoatrial node, thereby reducing heart rate without directly affecting cardiac contraction and relaxation. Clinical data from a phase III trial demonstrated that ivabradine reduced the composite end point of cardiovascular death or hospital admission for worsening systolic HF, while maintaining an acceptable safety profile in patients receiving standard of care therapy. These data, in addition to more recently published guidelines, suggest ivabradine as a promising new treatment option for lowering heart rate after optimizing standard therapy in select patients with chronic HF.
Collapse
Affiliation(s)
- Sheryl L Chow
- a Department of Pharmacy , Western University of Health Sciences , Pomona , CA , USA
| | - Robert L Page
- b Department of Clinical Pharmacy , University of Colorado , Denver , CO , USA
| | - Christophe Depre
- c Clinical Research Medical Director , Amgen Inc ., Thousand Oaks , CA , USA
| |
Collapse
|
38
|
Sartiani L, Mannaioni G, Masi A, Novella Romanelli M, Cerbai E. The Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels: from Biophysics to Pharmacology of a Unique Family of Ion Channels. Pharmacol Rev 2017; 69:354-395. [PMID: 28878030 DOI: 10.1124/pr.117.014035] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/07/2017] [Indexed: 12/22/2022] Open
Abstract
Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels are important members of the voltage-gated pore loop channels family. They show unique features: they open at hyperpolarizing potential, carry a mixed Na/K current, and are regulated by cyclic nucleotides. Four different isoforms have been cloned (HCN1-4) that can assemble to form homo- or heterotetramers, characterized by different biophysical properties. These proteins are widely distributed throughout the body and involved in different physiologic processes, the most important being the generation of spontaneous electrical activity in the heart and the regulation of synaptic transmission in the brain. Their role in heart rate, neuronal pacemaking, dendritic integration, learning and memory, and visual and pain perceptions has been extensively studied; these channels have been found also in some peripheral tissues, where their functions still need to be fully elucidated. Genetic defects and altered expression of HCN channels are linked to several pathologies, which makes these proteins attractive targets for translational research; at the moment only one drug (ivabradine), which specifically blocks the hyperpolarization-activated current, is clinically available. This review discusses current knowledge about HCN channels, starting from their biophysical properties, origin, and developmental features, to (patho)physiologic role in different tissues and pharmacological modulation, ending with their present and future relevance as drug targets.
Collapse
Affiliation(s)
- Laura Sartiani
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Guido Mannaioni
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Alessio Masi
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Maria Novella Romanelli
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Elisabetta Cerbai
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| |
Collapse
|
39
|
Zhao X, Gu T. Dysfunctional Hyperpolarization-Activated Cyclic Nucleotide-gated Ion Channels in Cardiac Diseases. Braz J Cardiovasc Surg 2017; 31:203-6. [PMID: 27556324 PMCID: PMC5062718 DOI: 10.5935/1678-9741.20160030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are reverse
voltage-dependent, and their activation depends on the hyperpolarization of the
membrane and may be directly or indirectly regulated by the cyclic adenosine
monophosphate (cAMP) or other signal-transduction cascades. The distribution,
quantity and activation states of HCN channels differ in tissues throughout the
body. Evidence exhibits that HCN channels play critical roles in the generation
and conduction of the electrical impulse and the physiopathological process of
some cardiac diseases. They may constitute promising drug targets in the
treatment of these cardiac diseases. Pharmacological treatment targeting HCN
channels is of benefit to these cardiac conditions.
Collapse
Affiliation(s)
- Xiaoqi Zhao
- Department of Cardiac Surgery ICU, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Tianxiang Gu
- Department of Cardiac Surgery ICU, First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
40
|
Motloch LJ, Larbig R, Darabi T, Reda S, Motloch KA, Wernly B, Lichtenauer M, Gebing T, Schwaiger A, Zagidullin N, Wolny M, Hoppe UC. Long-QT syndrome-associated caveolin-3 mutations differentially regulate the hyperpolarization-activated cyclic nucleotide gated channel 4. Physiol Int 2017. [DOI: 10.1556/2060.104.2017.2.6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background
Caveolin-3 (cav-3) mutations are linked to the long-QT syndrome (LQTS) causing distinct clinical symptoms. Hyperpolarization-activated cyclic nucleotide channel 4 (HCN4) underlies the pacemaker current If. It associates with cav-3 and both form a macromolecular complex.
Methods
To examine the effects of human LQTS-associated cav-3 mutations on HCN4-channel function, HEK293-cells were cotransfected with HCN4 and wild-type (WT) cav-3 or a LQTS-associated cav-3 mutant (T78M, A85T, S141R, or F97C). HCN4 currents were recorded using the whole-cell patch-clamp technique.
Results
WT cav-3 significantly decreased HCN4 current density and shifted midpoint of activation into negative direction. HCN4 current properties were differentially modulated by LQTS-associated cav-3 mutations. When compared with WT cav-3, A85T, F97C, and T78M did not alter the specific effect of cav-3, but S141R significantly increased HCN4 current density. Compared with WT cav-3, no significant modifications of voltage dependence of steady-state activation curves were observed. However, while WT cav-3 alone had no significant effect on HCN4 current activation, all LQTS-associated cav-3 mutations significantly accelerated HCN4 activation kinetics.
Conclusions
Our results indicate that HCN4 channel function is modulated by cav-3. LQTS-associated mutations of cav-3 differentially influence pacemaker current properties indicating a pathophysiological role in clinical manifestations.
Collapse
Affiliation(s)
- LJ Motloch
- 1Department of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - R Larbig
- 2Division of Electrophysiology, Department of Cardiovascular Medicine, University Hospital Muenster, Muenster, Germany
| | - T Darabi
- 1Department of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - S Reda
- 1Department of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - KA Motloch
- 3Research Program for Ophthalmology and Glaucoma Research, University Clinic of Ophthalmology and Optometry, Paracelsus Medical University/SALK, Salzburg, Austria
| | - B Wernly
- 1Department of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - M Lichtenauer
- 1Department of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - T Gebing
- 1Department of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - A Schwaiger
- 1Department of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - N Zagidullin
- 4Department of Internal Diseases, Bashkir State Medical University, Ufa, Russia
| | - M Wolny
- 1Department of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| | - UC Hoppe
- 1Department of Internal Medicine II, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
41
|
Foster DB, Liu T, Kammers K, O'Meally R, Yang N, Papanicolaou KN, Talbot CC, Cole RN, O'Rourke B. Integrated Omic Analysis of a Guinea Pig Model of Heart Failure and Sudden Cardiac Death. J Proteome Res 2016; 15:3009-28. [PMID: 27399916 DOI: 10.1021/acs.jproteome.6b00149] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Here, we examine key regulatory pathways underlying the transition from compensated hypertrophy (HYP) to decompensated heart failure (HF) and sudden cardiac death (SCD) in a guinea pig pressure-overload model by integrated multiome analysis. Relative protein abundances from sham-operated HYP and HF hearts were assessed by iTRAQ LC-MS/MS. Metabolites were quantified by LC-MS/MS or GC-MS. Transcriptome profiles were obtained using mRNA microarrays. The guinea pig HF proteome exhibited classic biosignatures of cardiac HYP, left ventricular dysfunction, fibrosis, inflammation, and extravasation. Fatty acid metabolism, mitochondrial transcription/translation factors, antioxidant enzymes, and other mitochondrial procsses, were downregulated in HF but not HYP. Proteins upregulated in HF implicate extracellular matrix remodeling, cytoskeletal remodeling, and acute phase inflammation markers. Among metabolites, acylcarnitines were downregulated in HYP and fatty acids accumulated in HF. The correlation of transcript and protein changes in HF was weak (R(2) = 0.23), suggesting post-transcriptional gene regulation in HF. Proteome/metabolome integration indicated metabolic bottlenecks in fatty acyl-CoA processing by carnitine palmitoyl transferase (CPT1B) as well as TCA cycle inhibition. On the basis of these findings, we present a model of cardiac decompensation involving impaired nuclear integration of Ca(2+) and cyclic nucleotide signals that are coupled to mitochondrial metabolic and antioxidant defects through the CREB/PGC1α transcriptional axis.
Collapse
Affiliation(s)
- D Brian Foster
- Division of Cardiology, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Ting Liu
- Division of Cardiology, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Kai Kammers
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland 21205, United States
| | - Robert O'Meally
- Proteomics Core Facility, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Ni Yang
- Division of Cardiology, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Kyriakos N Papanicolaou
- Division of Cardiology, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - C Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Robert N Cole
- Proteomics Core Facility, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Brian O'Rourke
- Division of Cardiology, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| |
Collapse
|
42
|
|
43
|
Yamada C, Kuwahara K, Yamazaki M, Nakagawa Y, Nishikimi T, Kinoshita H, Kuwabara Y, Minami T, Yamada Y, Shibata J, Nakao K, Cho K, Arai Y, Honjo H, Kamiya K, Nakao K, Kimura T. The renin-angiotensin system promotes arrhythmogenic substrates and lethal arrhythmias in mice with non-ischaemic cardiomyopathy. Cardiovasc Res 2015; 109:162-73. [PMID: 26531129 DOI: 10.1093/cvr/cvv248] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 10/29/2015] [Indexed: 11/13/2022] Open
Abstract
AIMS The progression of pathological left ventricular remodelling leads to cardiac dysfunction and contributes to the occurrence of malignant arrhythmias and sudden cardiac death. The underlying molecular mechanisms remain unclear, however. Our aim was to examine the role of the renin-angiotensin system (RAS) in the mechanism underlying arrhythmogenic cardiac remodelling using a transgenic mouse expressing a cardiac-specific dominant-negative form of neuron-restrictive silencer factor (dnNRSF-Tg). This mouse model exhibits progressive cardiac dysfunction leading to lethal arrhythmias. METHODS AND RESULTS Subcutaneous administration of aliskiren, a direct renin inhibitor, significantly suppressed the progression of pathological cardiac remodelling and improved survival among dnNRSF-Tg mice while reducing arrhythmogenicity. Genetic deletion of the angiotensin type 1a receptor (AT1aR) similarly suppressed cardiac remodelling and sudden death. In optical mapping analyses, spontaneous ventricular tachycardia (VT) and fibrillation (VF) initiated by breakthrough-type excitations originating from focal activation sites and maintained by functional re-entry were observed in dnNRSF-Tg hearts. Under constant pacing, dnNRSF-Tg hearts exhibited markedly slowed conduction velocity, which likely contributes to the arrhythmogenic substrate. Aliskiren treatment increased conduction velocity and reduced the incidence of sustained VT. These effects were associated with suppression of cardiac fibrosis and restoration of connexin 43 expression in dnNRSF-Tg ventricles. CONCLUSION Renin inhibition or genetic deletion of AT1aR suppresses pathological cardiac remodelling that leads to the generation of substrates maintaining VT/VF and reduces the occurrence of sudden death in dnNRSF-Tg mice. These findings demonstrate the significant contribution of RAS activation to the progression of arrhythmogenic substrates.
Collapse
Affiliation(s)
- Chinatsu Yamada
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Koichiro Kuwahara
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masatoshi Yamazaki
- Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Yasuaki Nakagawa
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Toshio Nishikimi
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hideyuki Kinoshita
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoshihiro Kuwabara
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takeya Minami
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yuko Yamada
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan Department of Peptide Research, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Junko Shibata
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kazuhiro Nakao
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan Department of Peptide Research, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kosai Cho
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yuji Arai
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Haruo Honjo
- Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Kaichiro Kamiya
- Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Kazuwa Nakao
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
44
|
Kashyap M, Yoshimura N, Smith PP, Chancellor M, Tyagi P. Characterization of the role of HCN channels in β3-adrenoceptor mediated rat bladder relaxation. Bladder (San Franc) 2015; 2. [PMID: 26709376 PMCID: PMC4690542 DOI: 10.14440/bladder.2015.44] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Objective The second messenger cAMP is involved in both β3 adrenoceptor (β3-AR) mediated detrusor relaxation and the kinetics of Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. Here we characterized the effect HCN channel activation and possible interaction with β3-AR in bladder. Materials and Methods Bladder tissues from Sprague-Dawley rats and Human organ donors were obtained for studying species-specific expression of HCN channels by real-time qPCR and Western Blot. Effect of β3-agonist on rat bladder strips (0.5 × 0.5 × 7 mm in size) was studied during activation and blockade of HCN channels by Lamotrigine and ZD7288, respectively. Results Expression of all four genes encoding for HCN channels (HCN1-4) was detected separately in bladder mucosa and detrusor from human and rat bladders. Species based differences were evident from relatively higher expression of HCN4 isoform in human bladder and that of HCN1 in rat bladder. Western blot confirmed the findings at mRNA level. Cumulative application β3-AR agonist CL316,243 produced a concentration dependent decrease in resting tension of rat bladder strips expressed as integral of mechanical activity. Pre-incubation of HCN channel blocker ZD 7288 opposed the relaxant effect of CL316,243, whereas co-administration of lamotrigine with CL316,243 at equal molar concentrations caused an additive decrease in resting tension. Cumulative addition of ZD7288 and lamotrigine in absence of CL316,243 showed opposing effects on detrusor contractility. Conclusions Species-specific differences were noted in expression of HCN channels in bladder. Opposing effects ZD7288 and Lamotrigine in the action of β3-AR agonist demonstrate possible functional interaction of HCN channels and β3-AR in detrusor contractility.
Collapse
Affiliation(s)
- Mahendra Kashyap
- Department of Urology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Naoki Yoshimura
- Department of Urology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Phillip P Smith
- Department of Surgery, University of Connecticut Health Center, Farmington, CT, USA
| | - Michael Chancellor
- Department of Urology, William Beaumont School of Medicine, Rochester, MI, USA
| | - Pradeep Tyagi
- Department of Urology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
45
|
Hara M, Takahashi T, Mitsumasu C, Igata S, Takano M, Minami T, Yasukawa H, Okayama S, Nakamura K, Okabe Y, Tanaka E, Takemura G, Kosai KI, Yamashita Y, Matsuishi T. Disturbance of cardiac gene expression and cardiomyocyte structure predisposes Mecp2-null mice to arrhythmias. Sci Rep 2015; 5:11204. [PMID: 26073556 PMCID: PMC4466896 DOI: 10.1038/srep11204] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 05/08/2015] [Indexed: 01/13/2023] Open
Abstract
Methyl-CpG-binding protein 2 (MeCP2) is an epigenetic regulator of gene expression that is essential for normal brain development. Mutations in MeCP2 lead to disrupted neuronal function and can cause Rett syndrome (RTT), a neurodevelopmental disorder. Previous studies reported cardiac dysfunction, including arrhythmias in both RTT patients and animal models of RTT. In addition, recent studies indicate that MeCP2 may be involved in cardiac development and dysfunction, but its role in the developing and adult heart remains unknown. In this study, we found that Mecp2-null ESCs could differentiate into cardiomyocytes, but the development and further differentiation of cardiovascular progenitors were significantly affected in MeCP2 deficiency. In addition, we revealed that loss of MeCP2 led to dysregulation of endogenous cardiac genes and myocardial structural alterations, although Mecp2-null mice did not exhibit obvious cardiac functional abnormalities. Furthermore, we detected methylation of the CpG islands in the Tbx5 locus, and showed that MeCP2 could target these sequences. Taken together, these results suggest that MeCP2 is an important regulator of the gene-expression program responsible for maintaining normal cardiac development and cardiomyocyte structure.
Collapse
Affiliation(s)
- Munetsugu Hara
- 1] Division of Gene Therapy and Regenerative Medicine, Cognitive and Molecular Research Institute of Brain Diseases [2] Department of Pediatrics, Kurume University School of Medicine, Kurume, Japan
| | - Tomoyuki Takahashi
- 1] Division of Gene Therapy and Regenerative Medicine, Cognitive and Molecular Research Institute of Brain Diseases [2] Department of Pediatrics, Kurume University School of Medicine, Kurume, Japan
| | - Chiaki Mitsumasu
- 1] Division of Gene Therapy and Regenerative Medicine, Cognitive and Molecular Research Institute of Brain Diseases [2] Department of Pediatrics, Kurume University School of Medicine, Kurume, Japan
| | - Sachiyo Igata
- Department of Physiology, Kurume University School of Medicine, Kurume, Japan
| | - Makoto Takano
- Department of Physiology, Kurume University School of Medicine, Kurume, Japan
| | - Tomoko Minami
- Cardiovascular Research Institute, Kurume University, Kurume, Japan
| | - Hideo Yasukawa
- Cardiovascular Research Institute, Kurume University, Kurume, Japan
| | - Satoko Okayama
- Department of Anatomy, Kurume University School of Medicine, Kurume, Japan
| | - Keiichiro Nakamura
- Department of Anatomy, Kurume University School of Medicine, Kurume, Japan
| | - Yasunori Okabe
- 1] Division of Gene Therapy and Regenerative Medicine, Cognitive and Molecular Research Institute of Brain Diseases [2] Department of Physiology, Kurume University School of Medicine, Kurume, Japan
| | - Eiichiro Tanaka
- Department of Physiology, Kurume University School of Medicine, Kurume, Japan
| | - Genzou Takemura
- Department of Internal Medicine, Asahi University School of Dentistry, Gifu, Japan
| | - Ken-ichiro Kosai
- 1] Division of Gene Therapy and Regenerative Medicine, Cognitive and Molecular Research Institute of Brain Diseases [2] Department of Pediatrics, Kurume University School of Medicine, Kurume, Japan [3] Department of Gene Therapy and Regenerative Medicine, Advanced Therapeutics Course, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yushiro Yamashita
- Department of Pediatrics, Kurume University School of Medicine, Kurume, Japan
| | - Toyojiro Matsuishi
- 1] Division of Gene Therapy and Regenerative Medicine, Cognitive and Molecular Research Institute of Brain Diseases [2] Department of Pediatrics, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
46
|
Li YD, Hong YF, Yusufuaji Y, Tang BP, Zhou XH, Xu GJ, Li JX, Sun L, Zhang JH, Xin Q, Xiong J, Ji YT, Zhang Y. Altered expression of hyperpolarization-activated cyclic nucleotide-gated channels and microRNA-1 and -133 in patients with age-associated atrial fibrillation. Mol Med Rep 2015; 12:3243-3248. [PMID: 26005035 PMCID: PMC4526032 DOI: 10.3892/mmr.2015.3831] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 12/09/2014] [Indexed: 01/08/2023] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) cation channels mediate pacemaker currents in the atrium. The microRNA (miR) families miR-1 and miR-133 regulate the expression of multiple genes involved in myocardial function, including HCN channels. It was hypothesized that age‑dependent changes in HCN2, HCN4, miR‑1 and miR‑133 expression may contribute to age‑associated atrial fibrillation, and therefore the correlation between expression levels, among adult (≤65 years) and aged patients (≥65 years), and sinus rhythm was determined. Right atrial appendage samples were collected from 60 patients undergoing coronary artery bypass grafting. Reverse transcription-quantitative polymerase chain reaction (PCR) and western blot analyses were performed in order to determine target RNA and protein expression levels. Compared with aged patients with sinus rhythm, aged patients with atrial fibrillation exhibited significantly higher HCN2 and HCN4 channel mRNA and protein expression levels (P<0.05), but significantly lower expression levels of miR‑1 and miR‑133 (P<0.05). In addition, aged patients with sinus rhythm exhibited significantly higher expression levels of HCN2 and HCN4 channel mRNA and protein (P<0.05), but significantly lower expression levels of miR‑1 and ‑133 (P<0.05), compared with those of adult patients with sinus rhythm. Expression levels of HCN2 and HCN4 increased with age, and a greater increase was identified in patients with age‑associated atrial fibrillation compared with that in those with aged sinus rhythm. These electrophysiological changes may contribute to the induction of ectopic premature beats that trigger atrial fibrillation.
Collapse
Affiliation(s)
- Yao-Dong Li
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| | - Yi-Fan Hong
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| | - Yueerguli Yusufuaji
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| | - Bao-Peng Tang
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| | - Xian-Hui Zhou
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| | - Guo-Jun Xu
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| | - Jin-Xin Li
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| | - Lin Sun
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| | - Jiang-Hua Zhang
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| | - Qiang Xin
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| | - Jian Xiong
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| | - Yu-Tong Ji
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| | - Yu Zhang
- Department of Cardiology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang Uyghur 830011, P.R. China
| |
Collapse
|
47
|
Li YD, Ji YT, Zhou XH, Jiang T, Hong YF, Li JX, Xing Q, Xiong J, Yusufuaji Y, Tang BP. Effects of ivabradine on cardiac electrophysiology in dogs with age-related atrial fibrillation. Med Sci Monit 2015; 21:1414-20. [PMID: 25982136 PMCID: PMC4444168 DOI: 10.12659/msm.894320] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Ivabradine is an inhibitor of mixed Na+-K+ current that could combine with HCN channels to reduce the transmembrane velocity of funny current (If), heart rate, and cardiac efficiency, and thus be used for the treatment of cardiovascular diseases such as chronic heart failure. As an ion channel blocker, Ivabradine is also a potential antiarrhythmic agent. Material/Methods Twelve aging dogs (8–10 years old) underwent rapid atrial pacing for 2 months to induce age-related AF in this study. The dogs were randomly divided into the Ivabradine group and aging-AF group. The effects of Ivabradine on the electrophysiological parameters, including the effective refractory period (ERP) of the pulmonary veins and atrium, duration of AF, and inducing rate of AF, were investigated. Results As compared to the aging-AF group, the ERPs of the left superior pulmonary vein (139.00±4.18 ms vs. 129.00±4.08 ms, P=0.005) and left auricle (135.00±3.53 ms vs. 122.00±4.47 ms, P=0.001) were significantly increased, while the duration of AF (46.60±5.07 s vs. 205.40±1.14 s, P=0.001) and inducing rate of AF (25% vs. 60%, P=0.001) were significantly decreased. Conclusions Ivabradine could effectively reduce the inducing rate of AF, and thus be used as an upstream drug for the prevention of age-related AF.
Collapse
Affiliation(s)
- Yao-Dong Li
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Yu-Tong Ji
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Xian-Hui Zhou
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Tao Jiang
- Clinical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Yi-fan Hong
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Jin-Xin Li
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Qiang Xing
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Jian Xiong
- Pacing and Electrophysiology Department, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China, Urumqi, China (mainland)
| | - Yueerguli Yusufuaji
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Bao-Peng Tang
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| |
Collapse
|
48
|
Ectopic automaticity induced in ventricular myocytes by transgenic overexpression of HCN2. J Mol Cell Cardiol 2015; 80:81-9. [DOI: 10.1016/j.yjmcc.2014.12.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 12/05/2014] [Accepted: 12/22/2014] [Indexed: 11/22/2022]
|
49
|
Herrmann S, Schnorr S, Ludwig A. HCN channels--modulators of cardiac and neuronal excitability. Int J Mol Sci 2015; 16:1429-47. [PMID: 25580535 PMCID: PMC4307311 DOI: 10.3390/ijms16011429] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 12/31/2014] [Indexed: 01/06/2023] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels comprise a family of cation channels activated by hyperpolarized membrane potentials and stimulated by intracellular cyclic nucleotides. The four members of this family, HCN1-4, show distinct biophysical properties which are most evident in the kinetics of activation and deactivation, the sensitivity towards cyclic nucleotides and the modulation by tyrosine phosphorylation. The four isoforms are differentially expressed in various excitable tissues. This review will mainly focus on recent insights into the functional role of the channels apart from their classic role as pacemakers. The importance of HCN channels in the cardiac ventricle and ventricular hypertrophy will be discussed. In addition, their functional significance in the peripheral nervous system and nociception will be examined. The data, which are mainly derived from studies using transgenic mice, suggest that HCN channels contribute significantly to cellular excitability in these tissues. Remarkably, the impact of the channels is clearly more pronounced in pathophysiological states including ventricular hypertrophy as well as neural inflammation and neuropathy suggesting that HCN channels may constitute promising drug targets in the treatment of these conditions. This perspective as well as the current therapeutic use of HCN blockers will also be addressed.
Collapse
Affiliation(s)
- Stefan Herrmann
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Sabine Schnorr
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Andreas Ludwig
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
50
|
Li YD, Hong YF, Zhang Y, Zhou XH, Ji YT, Li HL, Hu GJ, Li JX, Sun L, Zhang JH, Xin Q, Yusufuaji Y, Xiong J, Tang BP. Association between reversal in the expression of hyperpolarization-activated cyclic nucleotide-gated (HCN) channel and age-related atrial fibrillation. Med Sci Monit 2014; 20:2292-7. [PMID: 25404650 PMCID: PMC4242900 DOI: 10.12659/msm.892505] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Background We compared cardiac electrophysiological indicators and regional expression levels of cardiac hyperpolarization-activated cyclic nucleotide-gated (HCN) channels between adult and aged dogs to identify possible mechanisms of age-related atrial fibrillation. Material/Methods Corrected sinus node recovery time (SNRTc) and effective refractory period (ERP) of the atrium and pulmonary veins were measured in 10 adult (3–6 years old) and 10 aged dogs (>9 years old). Expression levels of HCN2 and HCN4 channel mRNAs and proteins were measured in the sinoatrial node, atrium, and pulmonary veins by real-time PCR and Western blotting. Results Aged dogs exhibited a higher induction rate of atrial fibrillation (AF) in response to electrical stimulation, longer AF duration after induction, longer SNRTc, longer right atrial effective refractory period (AERP), shorter left AERP, and increased AERP dispersion compared to adults. Expression levels of HCN2 and HCN4 channel mRNAs and proteins were lower in the sinoatrial node but higher in the atrium and pulmonary veins of aged dogs. Conclusions Changes in atrial electrophysiological indicators in aged dogs revealed sinoatrial node dysfunction. There was a reversal in the local tissue distribution of HCN2 and HCN4 channel mRNA and protein, a decrease in sinoatrial node expression, and increase in atrial and pulmonary vein expression with age. Changes in atrial electrophysiological characteristics and regional HCN channel expression patterns were associated with the onset and maintenance of age-related atrial fibrillation.
Collapse
Affiliation(s)
- Yao-Dong Li
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Yi-Fan Hong
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Yu Zhang
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Xian-Hui Zhou
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Yu-Tong Ji
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Hong-Liang Li
- Heart Rhythm Institute and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Guo-Jun Hu
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Jin-Xin Li
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Lin Sun
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Jiang-Hua Zhang
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Qiang Xin
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Yueerguli Yusufuaji
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Jian Xiong
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Bao-Peng Tang
- Department of Cardiology, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| |
Collapse
|