1
|
Marksteiner J, Dostal C, Ebner J, Szabó PL, Podesser BK, Baydar S, Goncalves AIA, Wagner A, Kratochwill K, Fichtinger P, Abraham D, Salzer I, Kubista H, Lilliu E, Hackl B, Sauer J, Todt H, Koenig X, Hilber K, Kiss A. Chronic administration of ivabradine improves cardiac Ca handling and function in a rat model of Duchenne muscular dystrophy. Sci Rep 2025; 15:8991. [PMID: 40089543 PMCID: PMC11910634 DOI: 10.1038/s41598-025-92927-4] [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: 09/16/2024] [Accepted: 03/04/2025] [Indexed: 03/17/2025] Open
Abstract
Duchenne muscular dystrophy (DMD), a severe muscle disease caused by mutations in the gene encoding for the intracellular protein dystrophin, is associated with impaired cardiac function and arrhythmias. A causative factor for complications in the dystrophic heart is abnormal calcium (Ca) handling in ventricular cardiomyocytes, and restoration of normal Ca homeostasis has emerged as therapeutic strategy. Here, we used a rodent model of DMD, the dystrophin-deficient DMDmdx rat, to test the following hypothesis: chronic administration of ivabradine (IVA), a drug clinically approved for the treatment of heart failure, improves Ca handling in dystrophic ventricular cardiomyocytes and thereby enhances contractile performance in the dystrophic heart. Intracellular Ca measurements revealed that 4-months administration of IVA to DMDmdx rats significantly improves Ca handling properties in dystrophic ventricular cardiomyocytes. In particular, IVA treatment increased electrically-evoked Ca transients and speeded their decay. This suggested enhanced sarcoplasmic reticulum Ca release and faster removal of Ca from the cytosol. Chronic IVA administration also enhanced the sarcoplasmic reticulum Ca load. Transthoracic echocardiography revealed a significant improvement of cardiac systolic function in IVA-treated DMDmdx rats. Thus, left ventricular ejection fraction and fractional shortening were enhanced, and end-systolic as well as end-diastolic diameters were diminished by the drug. Finally, chronic IVA administration neither significantly attenuated cardiac fibrosis and apoptosis, nor was vascular function improved by the drug. Collectively our findings suggest that long-term IVA administration enhances contractile function in the dystrophic heart by improvement of Ca handling in ventricular cardiomyocytes. Chronic IVA administration may be beneficial for DMD patients.
Collapse
Affiliation(s)
- Jessica Marksteiner
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Christopher Dostal
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Janine Ebner
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Petra Lujza Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Simge Baydar
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Ana I A Goncalves
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Anja Wagner
- Core Facility Proteomics, Medical University of Vienna, 1090, Vienna, Austria
| | - Klaus Kratochwill
- Core Facility Proteomics, Medical University of Vienna, 1090, Vienna, Austria
- Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University Vienna, 1090, Vienna, Austria
| | - Petra Fichtinger
- Center for Anatomy and Cell Biology, Medical University of Vienna, 1090, Vienna, Austria
| | - Dietmar Abraham
- Center for Anatomy and Cell Biology, Medical University of Vienna, 1090, Vienna, Austria
| | - Isabella Salzer
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Helmut Kubista
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Elena Lilliu
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Benjamin Hackl
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Jakob Sauer
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Hannes Todt
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Xaver Koenig
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Karlheinz Hilber
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria.
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090, Vienna, Austria
| |
Collapse
|
2
|
Sayers JR, Martinez-Navarro H, Sun X, de Villiers C, Sigal S, Weinberger M, Rodriguez CC, Riebel LL, Berg LA, Camps J, Herring N, Rodriguez B, Sauka-Spengler T, Riley PR. Cardiac conduction system regeneration prevents arrhythmias after myocardial infarction. NATURE CARDIOVASCULAR RESEARCH 2025; 4:163-179. [PMID: 39753976 PMCID: PMC11825367 DOI: 10.1038/s44161-024-00586-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 11/13/2024] [Indexed: 02/16/2025]
Abstract
Arrhythmias are a hallmark of myocardial infarction (MI) and increase patient mortality. How insult to the cardiac conduction system causes arrhythmias following MI is poorly understood. Here, we demonstrate conduction system restoration during neonatal mouse heart regeneration versus pathological remodeling at non-regenerative stages. Tissue-cleared whole-organ imaging identified disorganized bundling of conduction fibers after MI and global His-Purkinje disruption. Single-cell RNA sequencing (scRNA-seq) revealed specific molecular changes to regenerate the conduction network versus aberrant electrical alterations during fibrotic repair. This manifested functionally as a transition from normal rhythm to pathological conduction delay beyond the regenerative window. Modeling in the infarcted human heart implicated the non-regenerative phenotype as causative for heart block, as observed in patients. These findings elucidate the mechanisms underpinning conduction system regeneration and reveal how MI-induced damage elicits clinical arrhythmogenesis.
Collapse
Affiliation(s)
- Judy R Sayers
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Hector Martinez-Navarro
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Xin Sun
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Carla de Villiers
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Sarah Sigal
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Michael Weinberger
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Claudio Cortes Rodriguez
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Leto Luana Riebel
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Lucas Arantes Berg
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Julia Camps
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Neil Herring
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK
| | - Tatjana Sauka-Spengler
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Paul R Riley
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK.
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
3
|
Ziyatdinova NI, Kuptsova AM, Faskhutdinov LI, Shakirov RR, Zefirov TL. The Effect of α 2-Adrenergic Receptor Stimulation on the Isolated Hearts from Rats with Acute Myocardial Infarction against the Background of I f Blockade. Bull Exp Biol Med 2025; 178:295-300. [PMID: 39948175 DOI: 10.1007/s10517-025-06324-y] [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: 05/28/2024] [Indexed: 02/28/2025]
Abstract
We studied the effect of α2-adrenoreceptor stimulation after preliminary If blockade on the function of the Langendorff-isolated rat heart in the acute stage of experimental myocardial infarction. Stimulation of α2-adrenoreceptors with clonidine (10-6 M) against the background of If blockade with ZD7288 (10-9 M) reduced the contractility of the left ventricular myocardium, HR, coronary blood flow, and the duration of the left ventricular myocardium contraction in sham-operated rats and animals with acute myocardial infarction. The duration of relaxation and contraction cycle during α2-adrenoreceptor stimulation after preliminary If blockade (10-9 M) was increased in sham-operated rats and in animals with acute myocardial infarction. Stimulation of adrenoreceptors against the background of application of ZD7288 in a concentration of 10-5 M decreased the left ventricular pressure, HR, coronary blood flow, and the duration of left ventricular myocardial contraction in both groups. The duration of the relaxation and contraction cycle during α2-adrenoreceptor stimulation against the background of If blockade (10-5 M) increased in sham-operated rats and animals with acute myocardial infarction.
Collapse
Affiliation(s)
| | - A M Kuptsova
- Kazan (Volga region) State University, Kazan, Russia
| | | | - R R Shakirov
- Kazan (Volga region) State University, Kazan, Russia
| | - T L Zefirov
- Kazan (Volga region) State University, Kazan, Russia.
| |
Collapse
|
4
|
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]
|
5
|
Song R, Zhang L. MicroRNAs and therapeutic potentials in acute and chronic cardiac disease. Drug Discov Today 2024; 29:104179. [PMID: 39276921 DOI: 10.1016/j.drudis.2024.104179] [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: 06/18/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
microRNAs (miRNAs) are small regulatory RNAs implicated in various cardiac disorders. In this review, the role of miRNAs is discussed in relation to acute myocardial infarction and chronic heart failure. In both settings, miRNAs are altered, contributing to injury and adverse remodeling. Notably, miRNA profiles differ between acute ischemic injury and progressive heart failure. Owing to miRNA variabilities between disease stages and delivery difficulties, translation of animal studies to the clinic remains challenging. The identification of distinct miRNA signatures could lead to the development of miRNA therapies tailored to different disease stages. Here, we summarize the current understanding of miRNAs in acute and chronic cardiac diseases, identify knowledge gaps and discuss progress in developing miRNA-based treatment strategies.
Collapse
Affiliation(s)
- Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
6
|
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
|
7
|
Zhu M, Lu J, Li X, An Y, Li B, Liu W. Ivabradine Alleviates Experimental Autoimmune Myocarditis-Mediated Myocardial Injury. Int Heart J 2024; 65:109-118. [PMID: 38296563 DOI: 10.1536/ihj.23-330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Ivabradine (IVA) reduces heart rate by inhibiting hyperpolarization-activated cyclic nucleotide-gated channels (HCNs), which play a role in the promotion of pacemaker activity in cardiac sinoatrial node cells. HCNs are highly expressed in neural and myocardial tissues and are involved in the modulation of inflammatory neuropathic pain. However, whether IVA exerts any effect on myocardial inflammation in the pathogenesis of heart failure is unclear. We employed single-cell RNA sequencing (scRNA-seq) in porcine cardiac myosin-induced experimental autoimmune myocarditis rat model to determine the effects and mechanisms of IVA. Lewis rats (n = 32) were randomly divided into the normal, control, high-dose-IVA, and low-dose-IVA groups. Heart rate and blood pressure were measured on days 0 and 21, respectively. Echocardiography was performed on day 22, and inflammation of the myocardium was evaluated via histopathological examination. Western blot was employed to detect the expression of HCN1-4, MinK-related protein 1 (MiRP1), matrix metalloproteinase 2 (MMP-2), MMP-9, and transforming growth factor-β (TGF-β). Furthermore, enzyme-linked immunosorbent assay was performed to measure serum IL-1, IL-6, and TNF-α. The relative mRNA levels of collagen I, collagen III, and α-smooth muscle actin (α-SMA) were determined via qRT-PCR. We found that IVA reduced the total number of cells infiltrated into the myocardium, particularly in the subset of fibroblasts, endocardia, and monocytes. IVA administration ameliorated cardiac inflammation and reduced collagen production. Results of the echocardiography indicated that left ventricular internal diameter at end-systole LVIDs increased whereas left ventricular ejection fraction and left ventricular fractional shortening decreased in the control group. IVA improved cardiac performance. The expression of HCN4 and MiRP1 protein and the level of serum IL-1, IL-6, and TNF-α were decreased by IVA treatment. In conclusion, HCNs and the helper proteins were increased in the profile of myocardial inflammation. HCNs may be involved in the regulation of myocardial inflammation by inhibiting immune cell infiltration. Our findings can contribute to the development of IVA-based combination therapies for the future treatment of cardiac inflammation and heart failure.
Collapse
Affiliation(s)
- Manlin Zhu
- Department of Cardiology, Fourth Affiliated Hospital, Harbin Medical University
| | - Jingjing Lu
- Department of Cardiology, Fourth Affiliated Hospital, Harbin Medical University
| | - Xiaomin Li
- Department of Cardiology, Fourth Affiliated Hospital, Harbin Medical University
| | - Yongqiang An
- Department of Cardiology, First Affiliated Hospital, Hebei Medical University
| | | | - Wei Liu
- Department of Cardiology, Fourth Affiliated Hospital, Harbin Medical University
| |
Collapse
|
8
|
Barbuti A, Baruscotti M, Bucchi A. The “Funny” Pacemaker Current. HEART RATE AND RHYTHM 2023:63-87. [DOI: 10.1007/978-3-031-33588-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Response of Isolated Rat Heart to α 2-Adrenergic Receptor Stimulation after I f Current Blockade in the Late Postinfarction Period. Bull Exp Biol Med 2022; 174:22-25. [PMID: 36437345 DOI: 10.1007/s10517-022-05640-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Indexed: 11/29/2022]
Abstract
We studied the effect of α2-adrenoreceptor activation after preliminary If-current blockade on the performance of the Langendorff-isolated rat heart 54 days after modeling myocardial infarction. Stimulation of α2-adrenoreceptors against the background of application of If blocker ZD7288 in concentrations of 10-9 and 10-5 M decreased myocardial inotropy in isolated rat hearts by 50 and 39% (p<0.05) and increased HR by 20 and 15% (p<0.05), respectively. Activation of α2-adrenoreceptors against the background of application of ZD7288 in a concentration of 10-9 and 10-5 M led to a decrease in the coronary flow in the isolated rat heart with the model of myocardial infarction by 21% (p<0.05) and 32% (p<0.05), respectively.
Collapse
|
10
|
Zhang S, Li R, Ma Y, Yan Y, Ma M, Zhang K, Zhou Y, Li L, Pan L, Ying H, Xue Y. Thyroid-stimulating hormone regulates cardiac function through modulating HCN2 via targeting microRNA-1a. FASEB J 2022; 36:e22561. [PMID: 36125044 DOI: 10.1096/fj.202200574r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/11/2022] [Accepted: 09/09/2022] [Indexed: 11/11/2022]
Abstract
Previous studies have found microRNA-1 (miR-1) and hyperpolarization-activated cyclic nucleotide-gated channel 2 (HCN2) may be involved in the pathogenesis of thyroid hormone (TH) induced cardiac hypertrophy. However, little is known about the role of miR-1 and HCN2 in thyroid stimulation hormone (TSH)-induced cardiac dysfunction. In order to investigate the molecular mechanisms of TSH induced cardiac dysfunction and the role of miR-1/HCN2 in that process, we evaluated the expression of miR-1a/HCN2 in the ventricular myocardium of hypothyroid mice and in TSH-stimulated H9c2 cardiomyocytes. Our data revealed that hypothyroidism mice had smaller hearts, ventricular muscle atrophy, and cardiac contractile dysfunction compared with euthyroid controls. The upregulation of miR-1a and downregulation of HCN2 were found in ventricular myocardium of hypothyroid mice and TSH-stimulated H9c2 cardiomyocytes, indicating that miR-1a and HCN2 may be involved in TSH-induced cardiac dysfunction. We also found that the regulation of miR-1a and HCN2 expression and HCN2 channel activity by TSH requires TSHR, while the regulation of HCN2 expression and HCN2 channel function by TSH requires miR-1a. Thus, our data revealed the potential mechanism of TSH-induced cardiac dysfunction and might shed new light on the pathological role of miR-1a/HCN2 in hypothyroid heart disease.
Collapse
Affiliation(s)
- Shengjie Zhang
- Department of Endocrinology and Metabolism, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.,CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ran Li
- Department of Endocrinology and Metabolism, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yiruo Ma
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ying Yan
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mei Ma
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Keqin Zhang
- Department of Endocrinology and Metabolism, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yun Zhou
- Department of Endocrinology and Metabolism, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ling Li
- Department of Endocrinology and Metabolism, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lingling Pan
- Department of Endocrinology and Metabolism, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Ying
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ying Xue
- Department of Endocrinology and Metabolism, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
11
|
New Insights into the Functions of MicroRNAs in Cardiac Fibrosis: From Mechanisms to Therapeutic Strategies. Genes (Basel) 2022; 13:genes13081390. [PMID: 36011301 PMCID: PMC9407613 DOI: 10.3390/genes13081390] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/16/2022] [Accepted: 08/03/2022] [Indexed: 02/06/2023] Open
Abstract
Cardiac fibrosis is a significant global health problem associated with almost all types of heart disease. Extensive cardiac fibrosis reduces tissue compliance and contributes to adverse outcomes, such as cardiomyocyte hypertrophy, cardiomyocyte apoptosis, and even heart failure. It is mainly associated with pathological myocardial remodeling, characterized by the excessive deposition of extracellular matrix (ECM) proteins in cardiac parenchymal tissues. In recent years, a growing body of evidence demonstrated that microRNAs (miRNAs) have a crucial role in the pathological development of cardiac fibrosis. More than sixty miRNAs have been associated with the progression of cardiac fibrosis. In this review, we summarized potential miRNAs and miRNAs-related regulatory mechanisms for cardiac fibrosis and discussed the potential clinical application of miRNAs in cardiac fibrosis.
Collapse
|
12
|
Hackl B, Lukacs P, Ebner J, Pesti K, Haechl N, Földi MC, Lilliu E, Schicker K, Kubista H, Stary-Weinzinger A, Hilber K, Mike A, Todt H, Koenig X. The Bradycardic Agent Ivabradine Acts as an Atypical Inhibitor of Voltage-Gated Sodium Channels. Front Pharmacol 2022; 13:809802. [PMID: 35586063 PMCID: PMC9108390 DOI: 10.3389/fphar.2022.809802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/24/2022] [Indexed: 12/19/2022] Open
Abstract
Background and purpose: Ivabradine is clinically administered to lower the heart rate, proposedly by inhibiting hyperpolarization-activated cyclic nucleotide-gated cation channels in the sinoatrial node. Recent evidence suggests that voltage-gated sodium channels (VGSC) are inhibited within the same concentration range. VGSCs are expressed within the sinoatrial node and throughout the conduction system of the heart. A block of these channels thus likely contributes to the established and newly raised clinical indications of ivabradine. We, therefore, investigated the pharmacological action of ivabradine on VGSCs in sufficient detail in order to gain a better understanding of the pro- and anti-arrhythmic effects associated with the administration of this drug. Experimental Approach: Ivabradine was tested on VGSCs in native cardiomyocytes isolated from mouse ventricles and the His-Purkinje system and on human Nav1.5 in a heterologous expression system. We investigated the mechanism of channel inhibition by determining its voltage-, frequency-, state-, and temperature-dependence, complemented by a molecular drug docking to the recent Nav1.5 cryoEM structure. Automated patch-clamp experiments were used to investigate ivabradine-mediated changes in Nav1.5 inactivation parameters and inhibition of different VGSC isoforms. Key results: Ivabradine inhibited VGSCs in a voltage- and frequency-dependent manner, but did not alter voltage-dependence of activation and fast inactivation, nor recovery from fast inactivation. Cardiac (Nav1.5), neuronal (Nav1.2), and skeletal muscle (Nav1.4) VGSC isoforms were inhibited by ivabradine within the same concentration range, as were sodium currents in native cardiomyocytes isolated from the ventricles and the His-Purkinje system. Molecular drug docking suggested an interaction of ivabradine with the classical local anesthetic binding site. Conclusion and Implications: Ivabradine acts as an atypical inhibitor of VGSCs. Inhibition of VGSCs likely contributes to the heart rate lowering effect of ivabradine, in particular at higher stimulation frequencies and depolarized membrane potentials, and to the observed slowing of intra-cardiac conduction. Inhibition of VGSCs in native cardiomyocytes and across channel isoforms may provide a potential basis for the anti-arrhythmic potential as observed upon administration of ivabradine.
Collapse
Affiliation(s)
- Benjamin Hackl
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Peter Lukacs
- ELKH, Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary
| | - Janine Ebner
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Krisztina Pesti
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
- Semmelweis University, School of Ph.D. Studies, Budapest, Hungary
| | - Nicholas Haechl
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Mátyás C Földi
- ELKH, Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Elena Lilliu
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Klaus Schicker
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Helmut Kubista
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Karlheinz Hilber
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Arpad Mike
- ELKH, Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Hannes Todt
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Xaver Koenig
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Dai Y, Chen Y, Wei G, Zha L, Li X. Ivabradine protects rats against myocardial infarction through reinforcing autophagy via inhibiting PI3K/AKT/mTOR/p70S6K pathway. Bioengineered 2021; 12:1826-1837. [PMID: 33975512 PMCID: PMC8806854 DOI: 10.1080/21655979.2021.1925008] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/24/2021] [Indexed: 02/07/2023] Open
Abstract
Ivabradine (Iva), a heart rate reducing agent that specifically inhibits the pacemaker I(f) ionic current, has been demonstrated to be cardioprotective in many cardiovascular diseases. Autophagy is an evolutionarily conserved metabolic process that regulates cardiac homeostasis. This study is aimed to explore whether autophagy is functionally involved in the cardioprotective effect of Iva in a rat model of myocardial infarction (MI). We observed that Iva treatment (po, 10 mg/kg/day) showed significant recovery on the hemodynamics parameters in MI rats, including left ventricular systolic pressure, left ventricular end diastolic pressure, and maximal ascending/descending rate of left ventricular pressure. Also, Iva treatment dramatically decreased infarct size, inhibited myocardial apoptosis, and reduced the levels of pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6 in MI rats. Moreover, Iva treatment enhanced autophagy and inhibited PI3K/AKT/mTOR/p70S6K pathway in MI rats. Simultaneously, we observed that autophagy enhancer rapamycin (ip, 10 mg/kg/day) showed similar cardioprotective effects with Iva. Furthermore, we observed that addition of autophagy inhibitor 3-methyladenine (ip, 10 mg/kg/day) counteracted the therapeutic effect of Iva, addressing that Iva attenuated post-MI cardiac injury by enhancing autophagy. In summary, these findings demonstrated that Iva attenuated MI in rats by enhancing autophagy, and PI3K/AKT/mTOR/p70S6K pathway might be involved in the process. Autophagy activation by Iva may be a potential therapeutic strategy for the treatment of MI.
Collapse
Affiliation(s)
- Yingnan Dai
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| | - Yeping Chen
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| | - Guoqian Wei
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| | - Li Zha
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| | - Xueqi Li
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| |
Collapse
|
14
|
Martino A, Rebecchi M, Sette A, Cicogna F, Politano A, Sgueglia M, de Ruvo E, Volterrani M, Calo' L. Ivabradine versus bisoprolol in the treatment of inappropriate sinus tachycardia: a long-term follow-up study. J Cardiovasc Med (Hagerstown) 2021; 22:892-900. [PMID: 34747925 DOI: 10.2459/jcm.0000000000001203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AIM The aim of our study was to compare ivabradine versus bisoprolol in the short-term and long-term treatment of inappropriate sinus tachycardia. METHODS From this prospective, parallel-group, open-label study, consecutive patients affected by inappropriate sinus tachycardia received ivabradine or bisoprolol and were evaluated with Holter ECG, ECG stress test, European Heart Rhythm Association score and Minnesota Living With Heart Failure Questionnaire at baseline, after 3 and 24 months. RESULTS Overall, 40 patients were enrolled. Baseline parameters were comparable in the ivabradine and bisoprolol subgroups. Two patients had transient phosphenes with ivabradine and two others interrupted the drug after 3 months as they planned to become pregnant. Eight individuals treated with bisoprolol experienced hypotension and weakness, which caused drug discontinuation in five of them. Ivabradine was superior to bisoprolol in reducing Holter ECG mean heart rate (HR) and mean HR during daytime at short- and long-term follow-up. Moreover, ivabradine but not bisoprolol significantly reduced Holter ECG mean HR during night-time as well as maximal and minimal HR and significantly increased the time duration and maximal load reached at ECG stress test. The quality of life questionnaires significantly improved in both subgroups. CONCLUSION This study suggests that ivabradine is better tolerated than bisoprolol and seems to be superior in controlling the heart rate and improving exercise capacity in a small population of individuals affected by inappropriate sinus tachycardia during a short-term and long-term follow-up.
Collapse
|
15
|
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
|
16
|
Non-Coding RNAs in the Cardiac Action Potential and Their Impact on Arrhythmogenic Cardiac Diseases. HEARTS 2021. [DOI: 10.3390/hearts2030026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cardiac arrhythmias are prevalent among humans across all age ranges, affecting millions of people worldwide. While cardiac arrhythmias vary widely in their clinical presentation, they possess shared complex electrophysiologic properties at cellular level that have not been fully studied. Over the last decade, our current understanding of the functional roles of non-coding RNAs have progressively increased. microRNAs represent the most studied type of small ncRNAs and it has been demonstrated that miRNAs play essential roles in multiple biological contexts, including normal development and diseases. In this review, we provide a comprehensive analysis of the functional contribution of non-coding RNAs, primarily microRNAs, to the normal configuration of the cardiac action potential, as well as their association to distinct types of arrhythmogenic cardiac diseases.
Collapse
|
17
|
Scridon A, Halaţiu VB, Balan AI, Cozac DA, Moldovan V, Bănescu C, Perian M, Şerban RC. Long-Term Effects of Ivabradine on Cardiac Vagal Parasympathetic Function in Normal Rats. Front Pharmacol 2021; 12:596956. [PMID: 33897414 PMCID: PMC8061748 DOI: 10.3389/fphar.2021.596956] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/26/2021] [Indexed: 01/08/2023] Open
Abstract
Background: The complex interactions that exist between the pacemaker current, I f, and the parasympathetic nervous system could significantly influence the course of patients undergoing chronic therapy with the I f blocker ivabradine. We thus aimed to assess the effects of chronic ivabradine therapy on autonomic modulation and on the cardiovascular response to in situ and in vitro parasympathetic stimulation. The right atrial expression of HCN genes, encoding proteins for I f, was also evaluated. Methods: Sympathetic and parasympathetic heart rate variability parameters and right atrial HCN(1-4) RNA levels were analyzed in 6 Control and 10 ivabradine-treated male Wistar rats (IVA; 3 weeks, 10 mg/kg/day). The heart rate (HR) and systolic blood pressure (SBP) responses to in situ electrical stimulation of the vagus nerve (2-20 Hz) were assessed in 6 additional Control and 10 IVA rats. The spontaneous sinus node discharge rate (SNDR) response to in vitro cholinergic receptors stimulation using carbamylcholine (10-9-10-6 mol/L) was also assessed in these later rats. Results: Ivabradine significantly increased vagal modulation and shifted the sympatho-vagal balance toward vagal dominance. In Control, in situ vagus nerve stimulation induced progressive decrease in both the SBP (p = 0.0001) and the HR (p< 0.0001). Meanwhile, in IVA, vagal stimulation had no effect on the HR (p = 0.16) and induced a significantly lower drop in SBP (p< 0.05). IVA also displayed a significantly lower SNDR drop in response to carbamylcholine (p< 0.01) and significantly higher right atrial HCN4 expression (p = 0.02). Conclusion: Chronic ivabradine administration enhanced vagal modulation in healthy rats. In addition, ivabradine reduced the HR response to direct muscarinic receptors stimulation, canceled the cardioinhibitory response and blunted the hemodynamic response to in situ vagal stimulation. These data bring new insights into the mechanisms of ivabradine-related atrial proarrhythmia and suggest that long-term I f blockade may protect against excessive bradycardia induced by acute vagal activation.
Collapse
Affiliation(s)
- Alina Scridon
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania.,Center for Advanced Medical and Pharmaceutical Research, Târgu Mureş, Romania
| | - Vasile Bogdan Halaţiu
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania
| | - Alkora Ioana Balan
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania
| | - Dan Alexandru Cozac
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania.,Emergency Institute for Cardiovascular Diseases and Transplantation Târgu Mureş, Târgu Mureş, Romania
| | - Valeriu Moldovan
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania.,Center for Advanced Medical and Pharmaceutical Research, Târgu Mureş, Romania
| | - Claudia Bănescu
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania.,Center for Advanced Medical and Pharmaceutical Research, Târgu Mureş, Romania
| | - Marcel Perian
- University of Medicine, Pharmacy, Science and Technology "George Emil Palade" of Târgu Mureş, Târgu Mureş, Romania.,Center for Advanced Medical and Pharmaceutical Research, Târgu Mureş, Romania
| | - Răzvan Constantin Şerban
- Emergency Institute for Cardiovascular Diseases and Transplantation Târgu Mureş, Târgu Mureş, Romania
| |
Collapse
|
18
|
Paterek A, Sochanowicz B, Oknińska M, Śmigielski W, Kruszewski M, Mackiewicz U, Mączewski M, Leszek P. Ivabradine prevents deleterious effects of dopamine therapy in heart failure: No role for HCN4 overexpression. Biomed Pharmacother 2021; 136:111250. [PMID: 33450487 DOI: 10.1016/j.biopha.2021.111250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/27/2020] [Accepted: 01/03/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Exacerbations of chronic heart failure (CHF) are often treated with catecholamines to provide short term inotropic support, but this strategy is associated with long-term detrimental hemodynamic effects and increased ventricular arrhythmias (VA), possibly related to increased heart rate (HR). We hypothesized that ivabradine may prevent adverse effects of short-term dopamine treatment in CHF. METHODS Rats with post-myocardial infarction CHF received 2-week infusion of saline, dopamine(D), ivabradine(I) or D&I; cardiac function was assessed using echocardiography and pressure-volume loops while VA were assessed using telemetric ECG recording. Expression of HCN4, a potentially proarrhythmic channel blocked by ivabradine, was assessed in left ventricular (LV) myocardium. HCN4 expression was also assessed in human explanted normal and failing hearts and correlated with VA. FINDINGS Dopamine infusion had detrimental effects on hemodynamic parameters and LV remodeling and induced VA in CHF rats, while ivabradine completely prevented these effects. CHF rats demonstrated HCN4 overexpression in LV myocardium, and ivabradine and, unexpectedly, dopamine prevented this. Failing human hearts also exhibited HCN4 overexpression in LV myocardium that was unrelated to patient's sex, CHF etiology, VA severity or plasma NT-proBNP. INTERPRETATION HR reduction offered by ivabradine may be a feasible strategy to extract benefits of inotropic support in CHF exacerbations, avoiding detrimental effects on CHF biology or VA. Ivabradine may offer additional beneficial effects in this setting, going beyond pure HR reduction, however prevention of ventricular HCN4 overexpression is unlikely to play a major role.
Collapse
Affiliation(s)
- Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Barbara Sochanowicz
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Witold Śmigielski
- Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion, The Cardinal Stefan Wyszyński National Institute of Cardiology, Warsaw, Poland
| | - Marcin Kruszewski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland; Department of Molecular Biology and Translational Research, Institute of Rural Health, Lublin, Poland; Department of Medical Biology and Translational Research, Faculty of Medicine, University of Information Technology and Management, Rzeszów, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland.
| | - Przemysław Leszek
- Department of Heart Failure and Transplantology, The Cardinal Stefan Wyszyński National Institute of Cardiology, Warsaw, Poland
| |
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
|
Jiang LP, Lin JJ, Zhuang TP, Wang WW, Wu HZ, Zhang FL. Effects of ivabradine on ventricular electrophysiological remodeling after myocardial infarction in rats. Arch Med Sci 2020; 19:1497-1507. [PMID: 37732052 PMCID: PMC10507765 DOI: 10.5114/aoms.2020.101181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/22/2020] [Indexed: 09/22/2023] Open
Abstract
Introduction This study aims to investigate the effects of ivabradine (IVA) on ventricular electrophysiological remodeling after myocardial infarction (MI) in rats. Material and methods A total of 60 male Sprague-Dawley rats were randomly divided into five groups: an MI group, an IVA group, a metoprolol (MET) group, an IVA + MET group, and a sham group. After a four-week intervention, the ventricular electrophysiological parameters were detected by multichannel electrophysiological polygraph. Then, the morphological characteristics were evaluated using hematoxylin and eosin (H&E) and Masson's staining, and the expression of phosphorylated connexin 43 (p-Cx43) in the left ventricular wall was detected through immunohistochemistry and the Western blot test. Results The electrophysiological examination revealed that the induction rate and fatality rate of ventricular tachycardia (VT)/ventricular fibrillation (VF) were lower in both the IVA and the MET group, compared with the MI group (6/12, 6/12 vs. 10/11; and 1/12, 1/12 vs. 5/11; all p < 0.05), as well as the IVA + MET group (1/11 vs. 10/11, p < 0.01; and 1/11 vs. 5/11, p < 0.05). The induction rate of VT/VF was lower in the IVA + MET group, compared to the MET group (1/11 vs. 6/12, p < 0.05). H&E and Masson's staining revealed that compared with the MI group, the left ventricular infarction area was lower in the IVA, MET, and IVA + MET groups (p < 0.05, p < 0.05, and p < 0.01, respectively), while collagen volume fraction (CVF) also was lower in the other groups (all p < 0.01). The left ventricular infarction area and CVF both were lower in the IVA + MET group, compared to the MET group (p < 0.05 and p < 0.01, respectively). The immunohistochemistry and Western blot revealed that p-Cx43 expression was higher in the treatment groups, compared with the MI group (all p < 0.01). Conclusions IVA can reduce the incidence of ventricular arrhythmia after MI in male rats by improving both structural and electrical remodeling, and the combination of IVA and MET is even more effective.
Collapse
Affiliation(s)
- Li-ping Jiang
- Department of General Medicine, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Jing-jing Lin
- Department of Cardiology, Fujian Medical University Union Hospital and Fujian Provincial Institute of Coronary Disease, Fuzhou, China
| | - Ting-pei Zhuang
- Department of Cardiology, Fujian Medical University Union Hospital and Fujian Provincial Institute of Coronary Disease, Fuzhou, China
| | - Wei-wei Wang
- Department of Cardiology, Fujian Medical University Union Hospital and Fujian Provincial Institute of Coronary Disease, Fuzhou, China
| | - Hang-zhou Wu
- Medical Insurance Office, Fujian Medical University Union Hospital, Fuzhou, China
| | - Fei-long Zhang
- Department of Cardiology, Fujian Medical University Union Hospital and Fujian Provincial Institute of Coronary Disease, Fuzhou, China
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Šustr F, Stárek Z, Souček M, Novák J. Non-coding RNAs and Cardiac Arrhythmias. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:287-300. [PMID: 32285419 DOI: 10.1007/978-981-15-1671-9_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
Abstract
Cardiac arrhythmias represent wide and heterogenic group of disturbances in the cardiac rhythm. Pathophysiology of individual arrhythmias is highly complex and dysfunction in ion channels/currents involved in generation or spreading of action potential is usually documented. Non-coding RNAs (ncRNAs) represent highly variable group of molecules regulating the heart expression program, including regulation of the expression of individual ion channels and intercellular connection proteins, e.g. connexins.Within this chapter, we will describe basic electrophysiological properties of the myocardium. We will focus on action potential generation and spreading in pacemaker and non-pacemaker cells, including description of individual ion channels (natrium, potassium and calcium) and their ncRNA-mediated regulation. Most of the studies have so far focused on microRNAs, thus, their regulatory function will be described into greater detail. Clinical consequences of altered ncRNA regulatory function will also be described together with potential future directions of the research in the field.
Collapse
Affiliation(s)
- Filip Šustr
- Second Department of Internal Medicine of St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zdeněk Stárek
- First Department of Internal Medicine and Cardioangiology of St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Miroslav Souček
- Second Department of Internal Medicine of St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Novák
- Second Department of Internal Medicine of St. Anne's University Hospital in Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
- CEITEC - Central European Institute for Technology, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
24
|
Zhong LY, Fan XR, Shi ZJ, Fan ZC, Luo J, Lin N, Liu YC, Wu L, Zeng XR, Cao JM, Wei Y. Hyperpolarization-Activated Cyclic Nucleotide-Gated Ion (HCN) Channels Regulate PC12 Cell Differentiation Toward Sympathetic Neuron. Front Cell Neurosci 2019; 13:415. [PMID: 31616252 PMCID: PMC6763607 DOI: 10.3389/fncel.2019.00415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/28/2019] [Indexed: 12/15/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated ion channels (HCN channels) are widely expressed in the central and peripheral nervous systems and organs, while their functions are not well elucidated especially in the sympathetic nerve. The present study aimed to investigate the roles of HCN channel isoforms in the differentiation of sympathetic neurons using PC12 cell as a model. PC12 cells derived from rat pheochromocytoma were cultured and induced by nerve growth factor (NGF) (25 ng/ml) to differentiate to sympathetic neuron-like cells. Sympathetic directional differentiation of PC12 cells were evaluated by expressions of growth-associated protein 43 (GAP-43) (a growth cone marker), tyrosine hydroxylase (TH) (a sympathetic neuron marker) and neurite outgrowth. Results show that the HCN channel isoforms (HCN1-4) were all expressed in PC12 cells; blocking HCN channels with ivabradine suppressed NGF-induced GAP-43 expression and neurite outgrowth; silencing the expression of HCN2 and HCN4 using silenced using small interfering RNAs (siRNA), rather than HCN1 and HCN3, restrained GAP-43 expression and neurite outgrowth, while overexpression of HCN2 and HCN4 channels with gene transfer promoted GAP-43 expression and neurite outgrowth. Patch clamp experiments show that PC12 cells exhibited resting potentials (RP) of about −65 to −70 mV, and also presented inward HCN channel currents and outward (K+) currents, but no inward voltage-gated Na+ current was induced; NGF did not significantly affect the RP but promoted the establishment of excitability as indicated by the increased ability to depolarize and repolarize in the evoked suspicious action potentials (AP). We conclude that HCN2 and HCN4 channel isoforms, but not HCN1 and HCN3, promote the differentiation of PC12 cells toward sympathetic neurons. NGF potentiates the establishment of excitability during PC12 cell differentiation.
Collapse
Affiliation(s)
- Li-Ying Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xin-Rong Fan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhang-Jing Shi
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Zhong-Cai Fan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jian Luo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Na Lin
- Department of Respiratory Medicine, Rongcheng People's Hospital, Rongcheng, China
| | - Ying-Cai Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lin Wu
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.,Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Xiao-Rong Zeng
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Ji-Min Cao
- Key Laboratory of Cellular Physiology of Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Yan Wei
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
25
|
Li MCH, O'Brien TJ, Todaro M, Powell KL. Acquired cardiac channelopathies in epilepsy: Evidence, mechanisms, and clinical significance. Epilepsia 2019; 60:1753-1767. [PMID: 31353444 DOI: 10.1111/epi.16301] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 07/07/2019] [Accepted: 07/07/2019] [Indexed: 12/13/2022]
Abstract
There is growing evidence that cardiac dysfunction in patients with chronic epilepsy could play a pathogenic role in sudden unexpected death in epilepsy (SUDEP). Recent animal studies have revealed that epilepsy secondarily alters the expression of cardiac ion channels alongside abnormal cardiac electrophysiology and remodeling. These molecular findings represent novel evidence for an acquired cardiac channelopathy in epilepsy, distinct from inherited ion channels mutations associated with cardiocerebral phenotypes. Specifically, seizure activity has been shown to alter the messenger RNA (mRNA) and protein expression of voltage-gated sodium channels (Nav 1.1, Nav 1.5), voltage-gated potassium channels (Kv 4.2, Kv 4.3), sodium-calcium exchangers (NCX1), and nonspecific cation-conducting channels (HCN2, HCN4). The pathophysiology may involve autonomic dysfunction and structural cardiac disease, as both are independently associated with epilepsy and ion channel dysregulation. Indeed, in vivo and in vitro studies of cardiac pathology reveal a complex network of signaling pathways and transcription factors regulating ion channel expression in the setting of sympathetic overactivity, cardiac failure, and hypertrophy. Other mechanisms such as circulating inflammatory mediators or exogenous effects of antiepileptic medications lack evidence. Moreover, an acquired cardiac channelopathy may underlie the electrophysiologic cardiac abnormalities seen in chronic epilepsy, potentially contributing to the increased risk of malignant arrhythmias and sudden death. Therefore, further investigation is necessary to establish whether cardiac ion channel dysregulation similarly occurs in patients with epilepsy, and to characterize any pathogenic relationship with SUDEP.
Collapse
Affiliation(s)
- Michael C H Li
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Marian Todaro
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia.,Department of Neurology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Kim L Powell
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Kura B, Parikh M, Slezak J, Pierce GN. The Influence of Diet on MicroRNAs that Impact Cardiovascular Disease. Molecules 2019; 24:molecules24081509. [PMID: 30999630 PMCID: PMC6514571 DOI: 10.3390/molecules24081509] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 12/19/2022] Open
Abstract
Food quality and nutritional habits strongly influence human health status. Extensive research has been conducted to confirm that foods rich in biologically active nutrients have a positive impact on the onset and development of different pathological processes, including cardiovascular diseases. However, the underlying mechanisms by which dietary compounds regulate cardiovascular function have not yet been fully clarified. A growing number of studies confirm that bioactive food components modulate various signaling pathways which are involved in heart physiology and pathology. Recent evidence indicates that microRNAs (miRNAs), small single-stranded RNA chains with a powerful ability to influence protein expression in the whole organism, have a significant role in the regulation of cardiovascular-related pathways. This review summarizes recent studies dealing with the impact of some biologically active nutrients like polyunsaturated fatty acids (PUFAs), vitamins E and D, dietary fiber, or selenium on the expression of many miRNAs, which are connected with cardiovascular diseases. Current research indicates that the expression levels of many cardiovascular-related miRNAs like miRNA-21, -30 family, -34, -155, or -199 can be altered by foods and dietary supplements in various animal and human disease models. Understanding the dietary modulation of miRNAs represents, therefore, an important field for further research. The acquired knowledge may be used in personalized nutritional prevention of cardiovascular disease or the treatment of cardiovascular disorders.
Collapse
Affiliation(s)
- Branislav Kura
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovak Republic.
| | - Mihir Parikh
- Institute of Cardiovascular Sciences and the Canadian Centre for Agri-food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St. Boniface Hospital, Winnipeg, MB R2H2A6, Canada.
- Department of Physiology and Pathophysiology, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E0W3, Canada.
| | - Jan Slezak
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 84104 Bratislava, Slovak Republic.
| | - Grant N Pierce
- Institute of Cardiovascular Sciences and the Canadian Centre for Agri-food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St. Boniface Hospital, Winnipeg, MB R2H2A6, Canada.
- Department of Physiology and Pathophysiology, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E0W3, Canada.
| |
Collapse
|
27
|
Dini L, Del Lungo M, Resta F, Melchiorre M, Spinelli V, Di Cesare Mannelli L, Ghelardini C, Laurino A, Sartiani L, Coppini R, Mannaioni G, Cerbai E, Romanelli MN. Selective Blockade of HCN1/HCN2 Channels as a Potential Pharmacological Strategy Against Pain. Front Pharmacol 2018; 9:1252. [PMID: 30467478 PMCID: PMC6237106 DOI: 10.3389/fphar.2018.01252] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 10/15/2018] [Indexed: 12/25/2022] Open
Abstract
A prominent role of hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels has been suggested based on their expression and (dys)function in dorsal root ganglion (DRG) neurons, being likely involved in peripheral nociception. Using HCN blockers as antinociceptive drugs is prevented by the widespread distribution of these channels. However, tissue-specific expression of HCN isoforms varies significantly, HCN1 and HCN2 being considered as major players in DRG excitability. We characterized the pharmacological effect of a novel compound, MEL55A, able to block selectively HCN1/HCN2 isoforms, on DRG neuron excitability in-vitro and for its antiallodynic properties in-vivo. HEK293 cells expressing HCN1, HCN2, or HCN4 isoforms were used to verify drug selectivity. The pharmacological profile of MEL55A was tested on mouse DRG neurons by patch-clamp recordings, and in-vivo in oxaliplatin-induced neuropathy by means of thermal hypersensitivity. Results were compared to the non-isoform-selective drug, ivabradine. MEL55A showed a marked preference toward HCN1 and HCN2 isoforms expressed in HEK293, with respect to HCN4. In cultured DRG, MEL55A reduced I h amplitude, both in basic conditions and after stimulation by forskolin, and cell excitability, its effect being quantitatively similar to that observed with ivabradine. MEL55A was able to relieve chemotherapy-induced neuropathic pain. In conclusion, selective blockade of HCN1/HCN2 channels, over HCN4 isoform, was able to modulate electrophysiological properties of DRG neurons similarly to that reported for classical I h blockers, ivabradine, resulting in a pain-relieving activity. The availability of small molecules with selectivity toward HCN channel isoforms involved in nociception might represent a safe and effective strategy against chronic pain.
Collapse
Affiliation(s)
- Leonardo Dini
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Martina Del Lungo
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Francesco Resta
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Michele Melchiorre
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Valentina Spinelli
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Carla Ghelardini
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Annunziatina Laurino
- 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
| | - Raffaele Coppini
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| | - Guido Mannaioni
- 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
| | - Maria Novella Romanelli
- Department of Neurosciences, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Florence, Italy
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Down-regulation of miR-133a/b in patients with myocardial infarction correlates with the presence of ventricular fibrillation. Biomed Pharmacother 2018; 99:65-71. [PMID: 29324314 DOI: 10.1016/j.biopha.2018.01.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 12/22/2017] [Accepted: 01/03/2018] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs (miRNAs) are important regulators of physiologic and pathologic conditions of the heart. Animal models of heart diseases have shown that miRNAs may contribute to the development of arrhythmias. However, little is known about the expression of muscle- and cardiac-specific miRNAs in patients with myocardial infarction (MI) who have developed ventricular fibrillation (VF). Our study included 47 patients who had died from myocardial infarction (MI), 23 with clinically proven VF and 24 without VF. Autopsy samples of infarcted tissue and remote myocardium were available (n = 94). Heart tissue from 8 healthy trauma victims was included as control. Expression of miR-1, miR-133a/b and miR-208 was analyzed using real-time PCR (qPCR). In patients with MI with VF, we observed down-regulation of miR-133a/b, and this down-regulation was even stronger 2-7 days after MI. miR-208 was up-regulated in remote myocardium irrespective of the presence of VF. Deregulation of miR-1 and miR-208 was not related to the presence of VF. Our results suggest that down-regulation of miR-133a/b might contribute to the development of VF in patients with MI. However, up-regulation of miR-1 and miR-208 in remote myocardium might play a role in cardiac remodeling after MI, at least to certain degree.
Collapse
|
31
|
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: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [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
|
32
|
Swan KW, Song BM, Chen AL, Chen TJ, Chan RA, Guidry BT, Katakam PVG, Kerut EK, Giles TD, Kadowitz PJ. Analysis of decreases in systemic arterial pressure and heart rate in response to the hydrogen sulfide donor sodium sulfide. Am J Physiol Heart Circ Physiol 2017; 313:H732-H743. [PMID: 28667054 PMCID: PMC5668608 DOI: 10.1152/ajpheart.00729.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 06/05/2017] [Accepted: 06/23/2017] [Indexed: 01/16/2023]
Abstract
The actions of hydrogen sulfide (H2S) on the heart and vasculature have been extensively reported. However, the mechanisms underlying the effects of H2S are unclear in the anesthetized rat. The objective of the present study was to investigate the effect of H2S on the electrocardiogram and examine the relationship between H2S-induced changes in heart rate (HR), mean arterial pressure (MAP), and respiratory function. Intravenous administration of the H2S donor Na2S in the anesthetized Sprague-Dawley rat decreased MAP and HR and produced changes in respiratory function. The administration of Na2S significantly increased the RR interval at some doses but had no effect on PR or corrected QT(n)-B intervals. In experiments where respiration was maintained with a mechanical ventilator, we observed that Na2S-induced decreases in MAP and HR were independent of respiration. In experiments where respiration was maintained by mechanical ventilation and HR was maintained by cardiac pacing, Na2S-induced changes in MAP were not significantly altered, whereas changes in HR were abolished. Coadministration of glybenclamide significantly increased MAP and HR responses at some doses, but methylene blue, diltiazem, and ivabradine had no significant effect compared with control. The decreases in MAP and HR in response to Na2S could be dissociated and were independent of changes in respiratory function, ATP-sensitive K+ channels, methylene blue-sensitive mechanism involving L-type voltage-sensitive Ca2+ channels, or hyperpolarization-activated cyclic nucleotide-gated channels. Cardiovascular responses observed in spontaneously hypertensive rats were more robust than those in Sprague-Dawley rats.NEW & NOTEWORTHY H2S is a gasotransmitter capable of producing a decrease in mean arterial pressure and heart rate. The hypotensive and bradycardic effects of H2S can be dissociated, as shown with cardiac pacing experiments. Responses were not blocked by diltiazem, ivabradine, methylene blue, or glybenclamide.
Collapse
Affiliation(s)
- Kevin W Swan
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Bryant M Song
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Allen L Chen
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Travis J Chen
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ryan A Chan
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Bradley T Guidry
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Prasad V G Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | | | - Thomas D Giles
- Division of Cardiology, Department of Internal Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Philip J Kadowitz
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana;
| |
Collapse
|
33
|
Frommeyer G, Sterneberg M, Dechering DG, Ellermann C, Bögeholz N, Kochhäuser S, Pott C, Fehr M, Eckardt L. Effective suppression of atrial fibrillation by ivabradine: Novel target for an established drug? Int J Cardiol 2017; 236:237-243. [DOI: 10.1016/j.ijcard.2017.02.055] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/25/2017] [Accepted: 02/15/2017] [Indexed: 12/28/2022]
|
34
|
Niccoli G, Borovac JA, Vetrugno V, Camici PG, Crea F. Ivabradine in acute coronary syndromes: Protection beyond heart rate lowering. Int J Cardiol 2017; 236:107-112. [DOI: 10.1016/j.ijcard.2017.02.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/07/2017] [Accepted: 02/13/2017] [Indexed: 12/26/2022]
|
35
|
Re A, Nanni S, Aiello A, Granata S, Colussi C, Campostrini G, Spallotta F, Mattiussi S, Pantisano V, D'Angelo C, Biroccio A, Rossini A, Barbuti A, DiFrancesco D, Trimarchi F, Pontecorvi A, Gaetano C, Farsetti A. Anacardic acid and thyroid hormone enhance cardiomyocytes production from undifferentiated mouse ES cells along functionally distinct pathways. Endocrine 2016; 53:681-8. [PMID: 26547215 DOI: 10.1007/s12020-015-0751-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/19/2015] [Indexed: 10/22/2022]
Abstract
The epigenetics of early commitment to embryonal cardiomyocyte is poorly understood. In this work, we compared the effect of thyroid hormone and that of anacardic acid, a naturally occurring histone acetylase inhibitor, or both in combination, on mouse embryonic stem cells (mES) differentiating into embryonal cardiomyocyte by embryoid bodies (EBs) formation. Although the results indicated that anacardic acid (AA) and thyroid hormone were both efficient in promoting cardiomyocyte differentiation, we noticed that a transient exposure of mES to AA alone was sufficient to enlarge the beating areas of EBs compared to those of untreated controls. This effect was associated with changes in the chromatin structure at the promoters of specific cardiomyogenic genes. Among them, a rapid induction of the transcription factor Castor 1 (CASZ1), important for cardiomyocytes differentiation and maturation during embryonic development, was observed in the presence of AA. In contrast, thyroid hormone (T 3) was more effective in stimulating spontaneous firing, thus suggesting a role in the production of a population of cardiomyocyte with pacemaker properties. In conclusion, AA and thyroid hormone both enhanced cardiomyocyte formation along in apparently distinct pathways.
Collapse
Affiliation(s)
- Agnese Re
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, 00143, Rome, Italy
| | - Simona Nanni
- Institute of Medical Pathology, Catholic University, Rome, Italy
| | - Aurora Aiello
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, 00143, Rome, Italy
| | - Serena Granata
- Institute of Medical Pathology, Catholic University, Rome, Italy
| | - Claudia Colussi
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, 00143, Rome, Italy
| | - Giulia Campostrini
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Francesco Spallotta
- Division of Cardiovascular Epigenetics, Internal Medicine Clinic III, Goethe University Frankfurt, 60590, Frankfurt, Germany
| | - Stefania Mattiussi
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, 00143, Rome, Italy
| | | | - Carmen D'Angelo
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Annamaria Biroccio
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Alessandra Rossini
- Bolzano Center for Biomedicine (Affiliated Institute of the University of Lübeck), European Academy Bozen/Bolzano (EURAC), Bolzano, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Andrea Barbuti
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Dario DiFrancesco
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Francesco Trimarchi
- Unit of Endocrinology, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | | | - Carlo Gaetano
- Division of Cardiovascular Epigenetics, Internal Medicine Clinic III, Goethe University Frankfurt, 60590, Frankfurt, Germany.
| | - Antonella Farsetti
- National Research Council (CNR), Institute of Cell Biology and Neurobiology, 00143, Rome, Italy.
- Internal Medicine Clinic III, Goethe University Frankfurt, Frankfurt, Germany.
| |
Collapse
|
36
|
|
37
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Cardiac-specific miRNA in cardiogenesis, heart function, and cardiac pathology (with focus on myocardial infarction). J Mol Cell Cardiol 2016; 94:107-121. [PMID: 27056419 DOI: 10.1016/j.yjmcc.2016.03.015] [Citation(s) in RCA: 217] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 02/09/2016] [Accepted: 03/24/2016] [Indexed: 12/21/2022]
Abstract
Cardiac miRNAs (miR-1, miR133a, miR-208a/b, and miR-499) are abundantly expressed in the myocardium. They play a central role in cardiogenesis, heart function and pathology. While miR-1 and miR-133a predominantly control early stages of cardiogenesis supporting commitment of cardiac-specific muscle lineage from embryonic stem cells and mesodermal precursors, miR-208 and miR-499 are involved in the late cardiogenic stages mediating differentiation of cardioblasts to cardiomyocytes and fast/slow muscle fiber specification. In the heart, miR-1/133a control cardiac conductance and automaticity by regulating all phases of the cardiac action potential. miR-208/499 located in introns of the heavy chain myosin genes regulate expression of sarcomeric contractile proteins. In cardiac pathology including myocardial infarction (MI), expression of cardiac miRNAs is markedly altered that leads to deleterious effects associated with heart wounding, arrhythmia, increased apoptosis, fibrosis, hypertrophy, and tissue remodeling. In acute MI, circulating levels of cardiac miRNAs are significantly elevated making them to be a promising diagnostic marker for early diagnosis of acute MI. Great cardiospecific capacity of these miRNAs is very helpful for enhancing regenerative properties and survival of stem cell and cardiac progenitor transplants and for reprogramming of mature non-cardiac cells to cardiomyocytes.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Molecular Genetic Diagnostics and Cell Biology, Division of Laboratory Medicine, Institute of Pediatrics, Research Center for Children's Health, 119991 Moscow, Russia
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia; Department of Biophysics, Biological Faculty, Moscow State University, Moscow 119991, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow 121609, Russia
| | - Yuri V Bobryshev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia; Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; School of Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia.
| |
Collapse
|
38
|
Rajagopalan V, Zhang Y, Ojamaa K, Chen YF, Pingitore A, Pol CJ, Saunders D, Balasubramanian K, Towner RA, Gerdes AM. Safe Oral Triiodo-L-Thyronine Therapy Protects from Post-Infarct Cardiac Dysfunction and Arrhythmias without Cardiovascular Adverse Effects. PLoS One 2016; 11:e0151413. [PMID: 26981865 PMCID: PMC4794221 DOI: 10.1371/journal.pone.0151413] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/26/2016] [Indexed: 12/11/2022] Open
Abstract
Background A large body of evidence suggests that thyroid hormones (THs) are beneficial for the treatment of cardiovascular disorders. We have shown that 3 days of triiodo-L-thyronine (T3) treatment in myocardial infarction (MI) rats increased left ventricular (LV) contractility and decreased myocyte apoptosis. However, no clinically translatable protocol is established for T3 treatment of ischemic heart disease. We hypothesized that low-dose oral T3 will offer safe therapeutic benefits in MI. Methods and Results Adult female rats underwent left coronary artery ligation or sham surgeries. T3 (~6 μg/kg/day) was available in drinking water ad libitum immediately following MI and continuing for 2 month(s) (mo). Compared to vehicle-treated MI, the oral T3-treated MI group at 2 mo had markedly improved anesthetized Magnetic Resonance Imaging-based LV ejection fraction and volumes without significant negative changes in heart rate, serum TH levels or heart weight, indicating safe therapy. Remarkably, T3 decreased the incidence of inducible atrial tachyarrhythmias by 88% and improved remodeling. These were accompanied by restoration of gene expression involving several key pathways including thyroid, ion channels, fibrosis, sympathetic, mitochondria and autophagy. Conclusions Low-dose oral T3 dramatically improved post-MI cardiac performance, decreased atrial arrhythmias and cardiac remodeling, and reversed many adverse changes in gene expression with no observable negative effects. This study also provides a safe and effective treatment/monitoring protocol that should readily translate to humans.
Collapse
Affiliation(s)
- Viswanathan Rajagopalan
- Department of Biomedical Sciences, New York Institute of Technology-College of Osteopathic Medicine, Old Westbury, New York, United States of America
- * E-mail: (AMG); (VR)
| | - Youhua Zhang
- Department of Biomedical Sciences, New York Institute of Technology-College of Osteopathic Medicine, Old Westbury, New York, United States of America
| | - Kaie Ojamaa
- Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Yue-feng Chen
- Department of Biomedical Sciences, New York Institute of Technology-College of Osteopathic Medicine, Old Westbury, New York, United States of America
| | | | - Christine J. Pol
- Department of Biomedical Sciences, New York Institute of Technology-College of Osteopathic Medicine, Old Westbury, New York, United States of America
| | - Debra Saunders
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | | | - Rheal A. Towner
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - A. Martin Gerdes
- Department of Biomedical Sciences, New York Institute of Technology-College of Osteopathic Medicine, Old Westbury, New York, United States of America
- * E-mail: (AMG); (VR)
| |
Collapse
|
39
|
Ivabradine Prevents Low Shear Stress Induced Endothelial Inflammation and Oxidative Stress via mTOR/eNOS Pathway. PLoS One 2016; 11:e0149694. [PMID: 26890696 PMCID: PMC4758626 DOI: 10.1371/journal.pone.0149694] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 02/02/2016] [Indexed: 12/01/2022] Open
Abstract
Ivabradine not only reduces heart rate but has other cardiac and vascular protective effects including anti-inflammation and anti-oxidation. Since endothelial nitric oxide synthase (eNOS) is a crucial enzyme in maintaining endothelial activity, we aimed to investigate the impact of ivabradine in low shear stress (LSS) induced inflammation and endothelial injury and the role of eNOS played in it. Endothelial cells (ECs) were subjected to LSS at 2dyne/cm2, with 1 hour of ivabradine (0.04μM) or LY294002 (10μM) pre-treatment. The mRNA expression of IL-6, VCAM-1 along with eNOS were measured by QPCR. Reactive oxygen species (ROS) was detected by dihydroethidium (DHE) and DCF, and protein phosphorylation was detected by western blot. It demonstrated that ivabradine decreased LSS induced inflammation and oxidative stress in endothelial cells. Western blot showed reduced rictor and Akt-Ser473 as well as increased eNOS-Thr495 phosphorylation. However, mTORC1 pathway was only increased when LSS applied within 30 minutes. These effects were reversed by ivabradine. It would appear that ivabradine diminish ROS generation by provoking mTORC2/Akt phosphorylation and repressing mTORC1 induced eNOS-Thr495 activation. These results together suggest that LSS induced endothelial inflammation and oxidative stress are suppressed by ivabradine via mTORC2/Akt activation and mTORC1/eNOS reduction.
Collapse
|
40
|
Novella Romanelli M, Sartiani L, Masi A, Mannaioni G, Manetti D, Mugelli A, Cerbai E. HCN Channels Modulators: The Need for Selectivity. Curr Top Med Chem 2016; 16:1764-91. [PMID: 26975509 PMCID: PMC5374843 DOI: 10.2174/1568026616999160315130832] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 12/27/2022]
Abstract
Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels, the molecular correlate of the hyperpolarization-activated current (If/Ih), are membrane proteins which play an important role in several physiological processes and various pathological conditions. In the Sino Atrial Node (SAN) HCN4 is the target of ivabradine, a bradycardic agent that is, at the moment, the only drug which specifically blocks If. Nevertheless, several other pharmacological agents have been shown to modulate HCN channels, a property that may contribute to their therapeutic activity and/or to their side effects. HCN channels are considered potential targets for developing drugs to treat several important pathologies, but a major issue in this field is the discovery of isoform-selective compounds, owing to the wide distribution of these proteins into the central and peripheral nervous systems, heart and other peripheral tissues. This survey is focused on the compounds that have been shown, or have been designed, to interact with HCN channels and on their binding sites, with the aim to summarize current knowledge and possibly to unveil useful information to design new potent and selective modulators.
Collapse
Affiliation(s)
- Maria Novella Romanelli
- University of Florence, Department of Neurosciences, Psychology, Drug Research and Child's Health, Section of Pharmaceutical and Nutraceutical Sciences, via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy.
| | | | | | | | | | | | | |
Collapse
|
41
|
Annamaria M, Lupo PP, Foresti S, De Ambroggi G, de Ruvo E, Sciarra L, Cappato R, Calo L. Treatment of inappropriate sinus tachycardia with ivabradine. J Interv Card Electrophysiol 2015; 46:47-53. [PMID: 26467151 DOI: 10.1007/s10840-015-0066-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/05/2015] [Indexed: 11/26/2022]
Abstract
BACKGROUND Inappropriate sinus tachycardia (IST) often causes palpitations, dyspnea, and exercise intolerance, that are generally treated with beta blockers and non-dihydropyridine calcium-channel antagonists. Ivabradine, a selective inhibitor of cardiac pacemaker If current, has recently emerged as an effective and safe alternative to conventional drugs for IST. METHODS We performed a systematic overview of clinical studies on the therapeutic yield of ivabradine in patients with inappropriate sinus tachycardia, published in MEDLINE database from January 2000 to March 2015. RESULTS Overall, five case reports were found, all showing efficacy of ivabradine in subjects affected by IST. Eight non-randomized clinical studies demonstrated short- and medium-term safety and efficacy of ivabradine administration in IST, also in adjunction to or in comparison with metoprolol. One double-blind randomized crossover study also showed that ivabradine is superior to placebo for heart rate (HR) reduction and symptoms control in patients affected by IST. CONCLUSIONS Ivabradine is effective and safe in short- and medium-term treatment of IST. However, long-term follow-up studies and randomized studies comparing ivabradine with beta blockers are still lacking.
Collapse
Affiliation(s)
- Martino Annamaria
- Division of Cardiology, Policlinic Casilino, ASL RMB, Via Casilina 1049, Rome, Italy
- Cardiovascular, Respiratory, Nephrologic and Geriatrics Sciences Department, Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Pier Paolo Lupo
- Arrhythmia and Electrophysiology II Center, Humanitas Gavazzeni Clinics, Bergamo, Italy
| | - Sara Foresti
- Arrhythmia and Electrophysiology II Center, Humanitas Gavazzeni Clinics, Bergamo, Italy
| | - Guido De Ambroggi
- Arrhythmia and Electrophysiology II Center, Humanitas Gavazzeni Clinics, Bergamo, Italy
| | - Ermenegildo de Ruvo
- Division of Cardiology, Policlinic Casilino, ASL RMB, Via Casilina 1049, Rome, Italy
| | - Luigi Sciarra
- Division of Cardiology, Policlinic Casilino, ASL RMB, Via Casilina 1049, Rome, Italy
| | - Riccardo Cappato
- Arrhythmia and Electrophysiology II Center, Humanitas Gavazzeni Clinics, Bergamo, Italy
- Arrhythmia and Electrophysiology Research Center, IRCCS Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Leonardo Calo
- Division of Cardiology, Policlinic Casilino, ASL RMB, Via Casilina 1049, Rome, Italy.
| |
Collapse
|
42
|
Pereira-Barretto AC. Cardiac and Hemodynamic Benefits: Mode of Action of Ivabradine in Heart Failure. Adv Ther 2015; 32:906-19. [PMID: 26521191 DOI: 10.1007/s12325-015-0257-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Indexed: 01/19/2023]
Abstract
Heart failure has seen a number of therapeutic advances in recent years. Despite this, heart failure is still related to increasing rates of morbidity, repeated hospitalizations, and mortality. Ivabradine is a recent treatment option for heart failure. It has a mode of action that includes reduction in heart rate, and leads to improvement in outcomes related to heart failure mortality and morbidity, as demonstrated by the results of the SHIFT trial in patients with systolic heart failure, functional classes II and III on the New York Heart Association classification, and left ventricular ejection fraction ≤ 35%. These results are intriguing since many heart failure drugs reduce heart rate without such benefits, or with quite different effects, making it more difficult to understand the novelty of ivabradine in this setting. Many of the drugs used in heart failure modify heart rate, but most have other pathophysiological effects beyond their chronotropic action, which affect their efficacy in preventing morbidity and mortality outcomes. For instance, heart rate reduction at rest or exercise with ivabradine prolongs diastolic perfusion time, improves coronary blood flow, and increases exercise capacity. Another major difference is the increase in stroke volume observed with ivabradine, which may underlie its beneficial cardiac effects. Finally, there is mounting evidence from both preclinical and clinical studies that ivabradine has an anti-remodeling effect, improving left ventricular structures and functions. All together, these mechanisms have a positive impact on the prognosis of ivabradine-treated patients with heart failure, making a compelling argument for use of ivabradine in combination with other treatments.
Collapse
|
43
|
Tao H, Shi KH, Yang JJ, Li J. Epigenetic mechanisms in atrial fibrillation: New insights and future directions. Trends Cardiovasc Med 2015; 26:306-18. [PMID: 26475117 DOI: 10.1016/j.tcm.2015.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 08/23/2015] [Accepted: 08/28/2015] [Indexed: 11/28/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia. AF is a complex disease that results from genetic and environmental factors and their interactions. In recent years, numerous studies have shown that epigenetic mechanisms significantly participate in AF pathogenesis. Even though a poor understanding of the molecular and electrophysiologic mechanisms of AF, accumulated evidence has suggested that the relevance of epigenetic changes in the development of AF. The aim of this review is to describe the present knowledge about the epigenetic regulatory features significantly participates in AF, and look ahead on new perspectives of epigenetic mechanisms research. Epigenetic regulatory features such as DNA methylation, histone modification, and microRNA influence gene expression by epigenetic mechanisms and by directly binding to various factor response elements in the target gene promoters. Given the role of epigenetic alterations in regulating genes, there is potential for the integration of factors-induced epigenetic alterations as informative factors in the risk assessment process. In this review, new insight into the epigenetic mechanisms in AF pathogenesis is discussed, with special emphasis on DNA methylation, histone modification, and microRNA. Further studies are needed to reveal the potential targets of epigenetic mechanisms, and it can be developed as a therapeutic target for AF.
Collapse
Affiliation(s)
- Hui Tao
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, China; Cardiovascular Research Center, Anhui Medical University, Hefei, China
| | - Kai-Hu Shi
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, China; Cardiovascular Research Center, Anhui Medical University, Hefei, China.
| | - Jing-Jing Yang
- Department of Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China.
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
44
|
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
|
45
|
MicroRNAs: New players in pharmacotherapy for ischemic heart diseases? Int J Cardiol 2015; 185:117-8. [PMID: 25791107 DOI: 10.1016/j.ijcard.2015.03.139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/11/2015] [Indexed: 11/20/2022]
|
46
|
Dedkov EI, Bogatyryov Y, McCooey DS, Christensen LP, Weiss RM, Tomanek RJ. Effect of Chronic Heart Rate Reduction by If Current Inhibitor Ivabradine on Left Ventricular Remodeling and Systolic Performance in Middle-Aged Rats With Postmyocardial Infarction Heart Failure. J Cardiovasc Pharmacol Ther 2014; 20:299-312. [DOI: 10.1177/1074248414553231] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 08/14/2014] [Indexed: 11/15/2022]
Abstract
Background: A large myocardial infarction (MI) initiates progressive cardiac remodeling that leads to systolic heart failure (HF). Long-term heart rate reduction (HRR) induced by the I f current inhibitor ivabradine (IVA) ameliorates left ventricular (LV) remodeling and improves systolic performance in young post-MI rats. However, the beneficial effects of chronic IVA treatment in middle-aged rats remain to be determined. Methods: A large MI was induced in 12-month-old rats by left coronary artery ligation. Rats were treated with IVA via osmotic pumps intraperitoneal in a dose of 10.5 mg/kg/d (MI + IVA) and compared with MI and sham-operated animals 12 weeks after MI. Results: Heart rate in MI + IVA rats was on average 29% lower than that of rats in the MI group. Left ventricular remodeling was comparable between post-MI groups, although MI + IVA rats did not show the compensatory thickening of the noninfarcted myocardium. Chronic HRR had no effect on transverse cardiac myocyte size and capillary growth, but it reduced the collagen content in noninfarcted myocardium. Left ventricular systolic performance remained similarly impaired in MI and MI + IVA rats. Moreover, abrupt IVA withdrawal led to worsening HF and reduction of coronary reserve. Conclusion: Our data reveal that chronic IVA-induced HRR does not provide sustainable benefits for LV systolic performance in middle-aged rats with post-MI HF.
Collapse
Affiliation(s)
- Eduard I. Dedkov
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Yevgen Bogatyryov
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Daniela Scaldaferri McCooey
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, USA
| | - Lance P. Christensen
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Robert M. Weiss
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Cardiovascular Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Robert J. Tomanek
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Cardiovascular Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
47
|
Boštjančič E, Glavač D. miRNome in myocardial infarction: Future directions and perspective. World J Cardiol 2014; 6:939-958. [PMID: 25276296 PMCID: PMC4176804 DOI: 10.4330/wjc.v6.i9.939] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 03/28/2014] [Accepted: 06/27/2014] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs), which are small and non-coding RNAs, are genome encoded from viruses to humans. They contribute to various developmental, physiological and pathological processes in living organisms. A huge amount of research results revealed that miRNAs regulate these processes also in the heart. miRNAs may have cell-type-specific or tissue-specific expression patterns or may be expressed ubiquitously. Primary studies of miRNA involvement in hypertrophy, heart failure and myocardial infarction analyzed miRNAs that are enriched in or specific for cardiomyocytes; however, growing evidence suggest that other miRNAs, not cardiac or muscle-specific, play a significant role in cardiovascular disease. Abnormal miRNA regulation has been shown to be involved in cardiac diseases, suggesting that miRNAs might affect cardiac structure and function. In this review, we focus on miRNAs that have been found to contribute to the pathogenesis of myocardial infarction (MI) and the response post-MI and characterized as diagnostic, prognostic and therapeutic targets. The majority of these studies were performed using mouse and rat models of MI, with a focus on the identification of basic cellular and molecular pathways involved in MI and in the response post-MI. Much research has also been performed on animal and human plasma samples from MI individuals to identify miRNAs that are possible prognostic and/or diagnostic targets of MI and other MI-related diseases. A large proportion of research is focused on miRNAs as promising therapeutic targets and biomarkers of drug responses and/or stem cell treatment approaches. However, only a few studies have described miRNA expression in human heart tissue following MI.
Collapse
|
48
|
D'Souza A, Bucchi A, Johnsen AB, Logantha SJRJ, Monfredi O, Yanni J, Prehar S, Hart G, Cartwright E, Wisloff U, Dobryznski H, DiFrancesco D, Morris GM, Boyett MR. Exercise training reduces resting heart rate via downregulation of the funny channel HCN4. Nat Commun 2014; 5:3775. [PMID: 24825544 PMCID: PMC4024745 DOI: 10.1038/ncomms4775] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/01/2014] [Indexed: 12/14/2022] Open
Abstract
Endurance athletes exhibit sinus bradycardia, that is a slow resting heart rate, associated with a higher incidence of sinus node (pacemaker) disease and electronic pacemaker implantation. Here we show that training-induced bradycardia is not a consequence of changes in the activity of the autonomic nervous system but is caused by intrinsic electrophysiological changes in the sinus node. We demonstrate that training-induced bradycardia persists after blockade of the autonomous nervous system in vivo in mice and in vitro in the denervated sinus node. We also show that a widespread remodelling of pacemaker ion channels, notably a downregulation of HCN4 and the corresponding ionic current, If. Block of If abolishes the difference in heart rate between trained and sedentary animals in vivo and in vitro. We further observe training-induced downregulation of Tbx3 and upregulation of NRSF and miR-1 (transcriptional regulators) that explains the downregulation of HCN4. Our findings provide a molecular explanation for the potentially pathological heart rate adaptation to exercise training. Endurance athletes are known to have a low resting heart rate. Here, D'Souza et al. propose that training-induced bradycardia is the result of electrophysiological changes in the sinus node, challenging the classical view that training-induced bradycardia is caused by increased activity of the autonomic nervous system.
Collapse
Affiliation(s)
- Alicia D'Souza
- 1] Institute of Cardiovascular Sciences, University of Manchester, Manchester M13 9NT, UK [2]
| | - Annalisa Bucchi
- 1] Department of Biosciences, University of Milano, Milano 20133, Italy [2]
| | - Anne Berit Johnsen
- 1] Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim 7491, Norway [2]
| | - Sunil Jit R J Logantha
- 1] Institute of Cardiovascular Sciences, University of Manchester, Manchester M13 9NT, UK [2]
| | - Oliver Monfredi
- Institute of Cardiovascular Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Joseph Yanni
- Institute of Cardiovascular Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Sukhpal Prehar
- Institute of Cardiovascular Sciences, University of Manchester, Manchester M13 9NT, UK
| | - George Hart
- Institute of Cardiovascular Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Elizabeth Cartwright
- Institute of Cardiovascular Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Ulrik Wisloff
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Halina Dobryznski
- Institute of Cardiovascular Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Dario DiFrancesco
- Department of Biosciences, University of Milano, Milano 20133, Italy
| | - Gwilym M Morris
- Institute of Cardiovascular Sciences, University of Manchester, Manchester M13 9NT, UK
| | - Mark R Boyett
- Institute of Cardiovascular Sciences, University of Manchester, Manchester M13 9NT, UK
| |
Collapse
|
49
|
Selective and specific inhibition of If with ivabradine for the treatment of coronary artery disease or heart failure. Drugs 2014; 73:1569-86. [PMID: 24065301 PMCID: PMC3786091 DOI: 10.1007/s40265-013-0117-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Heart rate is an important contributor in the pathophysiology of both coronary artery disease (CAD) and heart failure (HF). Ivabradine is an anti-anginal and anti-ischaemic agent, which selectively and specifically inhibits the If current in the sino-atrial node and provides pure heart rate reduction without altering other cardiac parameters, including conduction, and without directly affecting other haemodynamic parameters. It is approved for the treatment of CAD and HF. This article summarises the pharmacological properties, pharmacokinetics, clinical efficacy and tolerability of ivabradine in the treatment of CAD and HF, and presents evidence demonstrating that the pharmacological and clinical properties and clinical efficacy of ivabradine make it an important therapeutic choice for patients with stable CAD or HF. The positive effect of ivabradine on angina pectoris symptoms and its ability to reduce myocardial ischemia make it an important agent in the management of patients with stable CAD or chronic HF. Further studies are underway to add to the already robust evidence of ivabradine for the prevention of cardiovascular events in patients with CAD but without clinical HF. The SIGNIFY (Study assessInG the morbidity–mortality beNefits of the If inhibitor ivabradine in patients with coronarY artery disease) trial includes patients with stable CAD and an LVEF above 40 %, with no clinical sign of HF, and is investigating the long-term effects (over a period of 48 months) of ivabradine in a large study population. So far, this study has included more than 19,000 patients from 51 countries.
Collapse
|
50
|
Mackiewicz U, Gerges JY, Chu S, Duda M, Dobrzynski H, Lewartowski B, Mączewski M. Ivabradine Protects Against Ventricular Arrhythmias in Acute Myocardial Infarction in the Rat. J Cell Physiol 2014; 229:813-23. [DOI: 10.1002/jcp.24507] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 10/31/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Urszula Mackiewicz
- Department of Clinical Physiology; Medical Center of Postgraduate Education; Warsaw Poland
| | - Joseph Y. Gerges
- School of Biomedical Sciences; University of Manchester; Manchester UK
| | - Sandy Chu
- School of Biomedical Sciences; University of Manchester; Manchester UK
| | - Monika Duda
- Department of Clinical Physiology; Medical Center of Postgraduate Education; Warsaw Poland
| | - Halina Dobrzynski
- School of Biomedical Sciences; University of Manchester; Manchester UK
| | - Bohdan Lewartowski
- Department of Clinical Physiology; Medical Center of Postgraduate Education; Warsaw Poland
| | - Michał Mączewski
- Department of Clinical Physiology; Medical Center of Postgraduate Education; Warsaw Poland
| |
Collapse
|