1
|
Berger V, Gabriel L, Lilliu E, Hackl B, Marksteiner J, Hilber K, Koenig X, Uhrin P, Todt H. Modulation of cardiac ventricular conduction: Impact on QRS duration, amplitude and dispersion. Eur J Pharmacol 2023; 941:175495. [PMID: 36621601 DOI: 10.1016/j.ejphar.2023.175495] [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: 08/02/2022] [Revised: 12/09/2022] [Accepted: 01/04/2023] [Indexed: 01/06/2023]
Abstract
Alterations in cardiac impulse conduction may exert both beneficial and detrimental effects. The assessment of ventricular conduction properties is of paramount importance both in clinical and in experimental settings. Currently the duration of the QRS complex is regarded as hallmark of in-vivo assessment of global ventricular conduction time. In addition, the amplitude of the QRS complex has been suggested to reflect ventricular conduction time in man and in rats. Here, for the first time, we systematically investigated the relationship between QRS duration ("QRS") and QRS amplitude ("RS-height"; RSh) in the murine ECG obtained during anesthesia. In mice harbouring a homozygous knockout of the transmembrane protein podoplanin (PDPN-/-; n = 10) we found both a shorter QRS and a greater RSh than in wild-type animals (n = 13). In both genotypes cumulative i.p. administration of 5 mg/kg and 10 mg/kg of the Na channel blocker flecainide resulted in dose-dependent QRS increase and RSh decrease, whereby the drug-induced changes in RSh were greater than in QRS. In both genotypes the flecainide-induced changes in QRS and in RSh were significantly correlated with each other (R = -0.56, P = 0.004). Whereas dispersion of QRS and RSh was similar between genotypes, dispersion of the ratio QRS/RSh was significantly smaller in PDPN-/- than in wild-types. We conclude that in the murine ECG QRS is inversely related to RSh. We suggest that both parameters should be considered in the analysis of ventricular conduction time in the murine ECG.
Collapse
Affiliation(s)
- Valerie Berger
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1190, Vienna, Austria.
| | - Ludwig Gabriel
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1190, Vienna, Austria.
| | - Elena Lilliu
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1190, Vienna, Austria.
| | - Benjamin Hackl
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1190, Vienna, Austria.
| | - Jessica Marksteiner
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1190, Vienna, Austria.
| | - Karlheinz Hilber
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1190, Vienna, Austria.
| | - Xaver Koenig
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1190, Vienna, Austria.
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1190, Vienna, Austria.
| | - Hannes Todt
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Waehringerstrasse 13a, 1190, Vienna, Austria.
| |
Collapse
|
2
|
Sykora M, Andelova K, Szeiffova Bacova B, Egan Benova T, Martiskova A, Knezl V, Tribulova N. Hypertension Induces Pro-arrhythmic Cardiac Connexome Disorders: Protective Effects of Treatment. Biomolecules 2023; 13:biom13020330. [PMID: 36830700 PMCID: PMC9953310 DOI: 10.3390/biom13020330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 02/11/2023] Open
Abstract
Prolonged population aging and unhealthy lifestyles contribute to the progressive prevalence of arterial hypertension. This is accompanied by low-grade inflammation and over time results in heart dysfunction and failure. Hypertension-induced myocardial structural and ion channel remodeling facilitates the development of both atrial and ventricular fibrillation, and these increase the risk of stroke and sudden death. Herein, we elucidate hypertension-induced impairment of "connexome" cardiomyocyte junctions. This complex ensures cell-to-cell adhesion and coupling for electrical and molecular signal propagation. Connexome dysfunction can be a key factor in promoting the occurrence of both cardiac arrhythmias and heart failure. However, the available literature indicates that arterial hypertension treatment can hamper myocardial structural remodeling, hypertrophy and/or fibrosis, and preserve connexome function. This suggests the pleiotropic effects of antihypertensive agents, including anti-inflammatory. Therefore, further research is required to identify specific molecular targets and pathways that will protect connexomes, and it is also necessary to develop new approaches to maintain heart function in patients suffering from primary or pulmonary arterial hypertension.
Collapse
|
3
|
He J, Xia G, Yang L, Jiang Z, Yang Y, Huo Z, Guo C. Crim1 inhibits angiotensin II-induced hypertrophy and preserves Kv4.2 expression in cardiomyocytes. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1201-1206. [PMID: 36311192 PMCID: PMC9588311 DOI: 10.22038/ijbms.2022.61459.13602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 07/20/2022] [Indexed: 11/17/2022]
Abstract
Objectives Angiotensin II (Ang II) plays a key role in the regulation of myocardial hypertrophy via downstream cysteine-rich transmembrane bone morphogenetic protein regulator 1 (Crim1). However, it is still unclear whether Crim1 is involved in ionic channel remodeling. The study aimed to explore the effects of Crim1 on transient outward potassium current (Ito) and Kv4.2 (the main subunit of Ito channel) expression in hypertrophic ventricular cardiomyocytes. Materials and Methods The ventricular cardiomyocytes were isolated from the neonatal rats. Hypertrophy was induced by Ang II. Crim1 expression was modulated by using adenovirus transfection. The expression of myosin heavy chain beta (β-MHC), Crim1, and Kv4.2 was determined by RT-qPCR and western blot. The cellular surface area was assessed using Image J software. Ito was recorded by the whole-cell patch clamp technique. Results Ang II-induced hypertrophy in cardiomyocytes was identified by their larger cellular surface area and higher mRNA expression of β-MHC. Ang II significantly decreased the expression of Crim1 and Kv4.2 and reduced Ito current density. However, Crim1 overexpression abolished the Ang II-induced hypertrophy and preserved the expression of Kv4.2 and Ito current density. Conclusion Crim1 overexpression inhibits Ang II-induced hypertrophy and preserves Ito current density via up-regulating Kv4.2 in ventricular cardiomyocytes from neonatal rats. Crim1 could have a role in the development of ventricular arrhythmia in hypertrophic hearts.
Collapse
Affiliation(s)
- Jionghong He
- Department of Cardiology, Guizhou Provincial People’s Hospital, Guiyang 550002 China, These authors contributed eqully to this work
| | - Guiling Xia
- Department of Cardiology, Guizhou Provincial People’s Hospital, Guiyang 550002 China, These authors contributed eqully to this work
| | - Long Yang
- Department of Cardiology, Guizhou Provincial People’s Hospital, Guiyang 550002 China,Corresponding author: Long Yang. Department of Cardiology, Guizhou Provincial People’s Hospital, Guiyang 550002 China, NO. 83 Zhongshandong Road, Guiyang 550002, China. Tel: +86-085185609229; Fax:+86-0851-85924943;
| | - Zhi Jiang
- Department of Cardiology, Guizhou Provincial People’s Hospital, Guiyang 550002 China
| | - Ying Yang
- Department of Cardiology, Guizhou Provincial People’s Hospital, Guiyang 550002 China
| | - Zhaomei Huo
- Department of Cardiology, Guizhou Provincial People’s Hospital, Guiyang 550002 China
| | - Chuxian Guo
- Department of Cardiology, Guizhou Provincial People’s Hospital, Guiyang 550002 China
| |
Collapse
|
4
|
Demers J, Ton A, Huynh F, Thibault S, Ducharme A, Paradis P, Nemer M, Fiset C. Atrial Electrical Remodeling in Mice With Cardiac‐Specific Overexpression of Angiotensin II Type 1 Receptor. J Am Heart Assoc 2022; 11:e023974. [PMID: 35435021 PMCID: PMC9238446 DOI: 10.1161/jaha.121.023974] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background
Elevated angiotensin II levels are thought to play an important role in atrial electrical and structural remodeling associated with atrial fibrillation. However, the mechanisms by which this remodeling occurs are still unclear. Accordingly, we explored the effects of angiotensin II on atrial remodeling using transgenic mice overexpressing angiotensin II type 1 receptor (AT1R) specifically in cardiomyocytes.
Methods and Results
Voltage‐clamp techniques, surface ECG, programmed electrical stimulations along with quantitative polymerase chain reaction, Western blot, and Picrosirius red staining were used to compare the atrial phenotype of AT1R mice and their controls at 50 days and 6 months. Atrial cell capacitance and fibrosis were increased only in AT1R mice at 6 months, indicating the presence of structural remodeling. Ca
2+
(
I
CaL
) and K
+
currents were not altered by AT1R overexpression (AT1R at 50 days). However,
I
CaL
density and Ca
V
1.2 messenger RNA expression were reduced by structural remodeling (AT1R at 6 months). Conversely, Na
+
current (
I
Na
) was reduced (−65%) by AT1R overexpression (AT1R at 50 days) and the presence of structural remodeling (AT1R at 6 months) yields no further effect. The reduced
I
Na
density was not explained by lower Na
V
1.5 expression but was rather associated with an increase in sarcolemmal protein kinase C alpha expression in the atria, suggesting that chronic AT1R activation reduced
I
Na
through protein kinase C alpha activation. Furthermore, connexin 40 expression was reduced in AT1R mice at 50 days and 6 months. These changes were associated with delayed atrial conduction time, as evidenced by prolonged P‐wave duration.
Conclusions
Chronic AT1R activation leads to slower atrial conduction caused by reduced
I
Na
density and connexin 40 expression.
Collapse
Affiliation(s)
- Julie Demers
- Research Center Montreal Heart Institute Montréal Québec Canada
- Faculty of Pharmacy Université de Montréal Montréal Québec Canada
| | - Anh‐Tuan Ton
- Research Center Montreal Heart Institute Montréal Québec Canada
- Faculty of Pharmacy Université de Montréal Montréal Québec Canada
| | - François Huynh
- Research Center Montreal Heart Institute Montréal Québec Canada
- Faculty of Pharmacy Université de Montréal Montréal Québec Canada
| | - Simon Thibault
- Research Center Montreal Heart Institute Montréal Québec Canada
- Faculty of Pharmacy Université de Montréal Montréal Québec Canada
| | - Anique Ducharme
- Research Center Montreal Heart Institute Montréal Québec Canada
- Faculty of Medicine Université de Montréal Montréal Québec Canada
| | | | | | - Céline Fiset
- Research Center Montreal Heart Institute Montréal Québec Canada
- Faculty of Pharmacy Université de Montréal Montréal Québec Canada
| |
Collapse
|
5
|
Shao X, Zhang X, Zhang R, Zhu R, Hou X, Yi W, Wu F, Hao L, Feng R. The atlas of ACE2 expression in fetal and adult human hearts reveals the potential mechanism of heart injured patients infected with SARS-CoV-2. Am J Physiol Cell Physiol 2022; 322:C723-C738. [PMID: 35138176 PMCID: PMC8977135 DOI: 10.1152/ajpcell.00169.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Numerous studies have shown that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can infect host cells through binding to angiotensin I converting enzyme 2 (ACE2) expressing in various tissues and organs. In this study, we deeply analyzed the single-cell expression profiles of ACE2 in fetal and adult human hearts to explore the potential mechanism of SARS-CoV-2 harming the heart. The molecular docking software was used to simulate the binding of SARS-CoV-2 and its variant spike protein with ACE2. The genes closely related to ACE2 in renin-angiotensin system (RAS) were identified by constructing a protein-protein interaction network. Through the analysis of single-cell transcription profiles at different stages of human embryos, we found that the expression level of ACE2 in ventricular myocytes was increased with embryonic development. The results of single-cell sequencing analysis showed that the expression of ACE2 in ventricular myocytes was upregulated in heart failure induced by dilated cardiomyopathy compared with normal hearts. The upregulation of ACE2 increases the risk of infection with SARS-CoV-2 in fetal and adult human hearts. We also further confirmed the expression of ACE2 and ACE2-related genes in normal and SARS-CoV-2-infected human pluripotent stem cell-derived cardiomyocytes. In addition, the pathway analysis revealed that ACE2 may regulate the differently expressed genes in heart failure through calcium signaling pathway and Wnt signaling pathway.
Collapse
Affiliation(s)
- Xiuli Shao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Xiaolin Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ruijia Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Rongli Zhu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Xiuyang Hou
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Weijue Yi
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Fengmin Wu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Liying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| | - Rui Feng
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, China
| |
Collapse
|
6
|
Emathinger JM, Nelson JW, Gurley SB. Advances in use of mouse models to study the renin-angiotensin system. Mol Cell Endocrinol 2021; 529:111255. [PMID: 33789143 PMCID: PMC9119406 DOI: 10.1016/j.mce.2021.111255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/19/2021] [Accepted: 03/20/2021] [Indexed: 12/28/2022]
Abstract
The renin-angiotensin system (RAS) is a highly complex hormonal cascade that spans multiple organs and cell types to regulate solute and fluid balance along with cardiovascular function. Much of our current understanding of the functions of the RAS has emerged from a series of key studies in genetically-modified animals. Here, we review key findings from ground-breaking transgenic models, spanning decades of research into the RAS, with a focus on their use in studying blood pressure. We review the physiological importance of this regulatory system as evident through the examination of mouse models for several major RAS components: angiotensinogen, renin, ACE, ACE2, and the type 1 A angiotensin receptor. Both whole-animal and cell-specific knockout models have permitted critical RAS functions to be defined and demonstrate how redundancy and multiplicity within the RAS allow for compensatory adjustments to maintain homeostasis. Moreover, these models present exciting opportunities for continued discovery surrounding the role of the RAS in disease pathogenesis and treatment for cardiovascular disease and beyond.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2/deficiency
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensinogen/deficiency
- Angiotensinogen/genetics
- Animals
- Blood Pressure/genetics
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Disease Models, Animal
- Gene Expression Regulation
- Humans
- Kidney/cytology
- Kidney/metabolism
- Mice
- Mice, Knockout
- Receptor, Angiotensin, Type 1/deficiency
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 2/deficiency
- Receptor, Angiotensin, Type 2/genetics
- Renin/deficiency
- Renin/genetics
- Renin-Angiotensin System/genetics
- Signal Transduction
- Water-Electrolyte Balance/genetics
Collapse
Affiliation(s)
- Jacqueline M Emathinger
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR, USA.
| | - Jonathan W Nelson
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR, USA.
| | - Susan B Gurley
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
7
|
Xiao S, Shimura D, Baum R, Hernandez DM, Agvanian S, Nagaoka Y, Katsumata M, Lampe PD, Kleber AG, Hong T, Shaw RM. Auxiliary trafficking subunit GJA1-20k protects connexin-43 from degradation and limits ventricular arrhythmias. J Clin Invest 2021; 130:4858-4870. [PMID: 32525845 DOI: 10.1172/jci134682] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 06/03/2020] [Indexed: 12/31/2022] Open
Abstract
Connexin-43 (Cx43) gap junctions provide intercellular coupling, which ensures rapid action potential propagation and synchronized heart contraction. Alterations in Cx43 localization and reductions in gap junction coupling occur in failing hearts, contributing to ventricular arrhythmias and sudden cardiac death. Recent reports have found that an internally translated Cx43 isoform, GJA1-20k, is an auxiliary subunit for the trafficking of Cx43 in heterologous expression systems. Here, we have created a mouse model by using CRISPR technology to mutate a single internal translation initiation site in Cx43 (M213L mutation), which generates full-length Cx43, but not GJA1-20k. We found that GJA1M213L/M213L mice had severely abnormal electrocardiograms despite preserved contractile function, reduced total Cx43, and reduced gap junctions, and they died suddenly at 2 to 4 weeks of age. Heterozygous GJA1M213L/WT mice survived to adulthood with increased ventricular ectopy. Biochemical experiments indicated that cytoplasmic Cx43 had a half-life that was 50% shorter than membrane-associated Cx43. Without GJA1-20k, poorly trafficked Cx43 was degraded. The data support that GJA1-20k, an endogenous entity translated independently of Cx43, is critical for Cx43 gap junction trafficking, maintenance of Cx43 protein, and normal electrical function of the mammalian heart.
Collapse
Affiliation(s)
- Shaohua Xiao
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA.,Department of Neurology, UCLA, Los Angeles, California, USA
| | - Daisuke Shimura
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| | - Rachel Baum
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Diana M Hernandez
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sosse Agvanian
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yoshiko Nagaoka
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Makoto Katsumata
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Paul D Lampe
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Andre G Kleber
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - TingTing Hong
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Robin M Shaw
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
8
|
Jin N, Wang Y, Liu L, Xue F, Jiang T, Xu M. Dysregulation of the Renin-Angiotensin System and Cardiometabolic Status in Mice Fed a Long-Term High-Fat Diet. Med Sci Monit 2019; 25:6605-6614. [PMID: 31523052 PMCID: PMC6738017 DOI: 10.12659/msm.914877] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background This study aimed to investigate the renin-angiotensin system (RAS) and cardiometabolic status in mice fed a long-term high-fat diet (HFD). Material/Methods C57BL/6J mice were randomly assigned to the control group on a normal diet (ND) (n=15) and the HFD group (n=15). Serum biomarkers were measured, including total cholesterol (TC), triglyceride (TG), insulin, glycated hemoglobin (HbA1c), brain natriuretic peptide (BNP), renin, angiotensin-converting enzyme (ACE), angiotensin II (Ang-II), Ang-II type 1 receptor (AT1R), and aldosterone. Cardiac histology was measured by the cross-sectional area (CSA) of cardiomyocytes and collagen deposition. Levels of myocardial intercalated disc (ICD) proteins and mRNA were analyzed by Western blot and real-time quantitative polymerase chain reaction (RT-qPCR), respectively. The localization of ICD proteins was evaluated by immunohistochemistry (IHC). Results Compared with ND, HFD resulted in increased blood glucose, body weight, TC, TG, HbA1c, insulin, and BNP and levels of serum ACE, Ang-II, aldosterone, AT1R, cardiomyocyte CSA, and interstitial collagen in the myocardium compared. Also, HFD significantly down-regulated connexin-43, and upregulated β-catenin, N-cadherin, and plakoglobin in the hearts of HFD mice compared with ND mice. However, the deposition of ICD proteins was not changed in the hearts of HFD mice compared with ND mice. Conclusions Long-term HFD in mice resulted in left ventricular hypertrophy, interstitial fibrosis, dysregulation of RAS, and abnormal expression of ICD proteins compared with ND mice, but did not affect the distribution of cardiomyocyte ICD proteins. Long-term HFD resulted in cardiac remodeling and altered expression of ICD proteins through RAS activation.
Collapse
Affiliation(s)
- Nana Jin
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China (mainland)
| | - Yu Wang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China (mainland)
| | - Lin Liu
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China (mainland)
| | - Feng Xue
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China (mainland)
| | - Tingbo Jiang
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China (mainland)
| | - Mingzhu Xu
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China (mainland)
| |
Collapse
|
9
|
Cao L, Chen Y, Lu L, Liu Y, Wang Y, Fan J, Yin Y. Angiotensin II upregulates fibroblast-myofibroblast transition through Cx43-dependent CaMKII and TGF-β1 signaling in neonatal rat cardiac fibroblasts. Acta Biochim Biophys Sin (Shanghai) 2018; 50:843-852. [PMID: 30060053 DOI: 10.1093/abbs/gmy090] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/05/2018] [Indexed: 02/07/2023] Open
Abstract
In cardiac fibroblasts, angiotensin II (Ang II) can increase connexin 43 (Cx43) expression and promote calmodulin-dependent protein kinase II (CaMKII) activation. Cx43 overexpression is crucial for the fibroblast-myofibroblast transition. The main purpose of the present study was to investigate the role of CaMKII in regulating Cx43 expression and to determine whether the CaMKII/Cx43 pathway is essential for controlling fibroblast activation and differentiation. In vivo, 4 weeks of Ang II infusion enhanced CaMKII activation but reduced Cx43 expression in hearts undergoing fibrosis remodeling, while in cultured neonatal rat fibroblasts, CaMKII activation upregulated Cx43 expression via transforming growth factor-beta1 (TGF-β1). CaMKII inhibition by Ang-(1-7) or autocamtide 2-related inhibitory peptide reversed the Ang II-induced changes in Cx43 expression and attenuated Ang II-induced upregulation of alpha smooth muscle actin and TGF-β1 in both Ang II-infused rats and cultured fibroblasts. Based on the in vivo and in vitro experimental results, CaMKII plays a pivotal role in the Ang II-mediated fibroblast-myofibroblast transition by modulating the expressions of TGF-β1 and Cx43. We conclude that Ang II mediates the fibroblast-myofibroblast transition partially via the Ang II/CaMKII/TGF-β1/Cx43 signaling pathway.
Collapse
Affiliation(s)
- Li Cao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunlin Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Lu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yihao Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaowen Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinqi Fan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuehui Yin
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Zankov DP, Salloum FN, Jiang M, Tseng GN. Chronic in vivo angiotensin II administration differentially modulates the slow delayed rectifier channels in atrial and ventricular myocytes. Heart Rhythm 2018; 16:108-116. [PMID: 30075281 DOI: 10.1016/j.hrthm.2018.07.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND In the heart, slow delayed rectifier channels provide outward currents (IKs) for action potential (AP) repolarization in a region- and context-dependent manner. In diseased hearts, chronic elevation of angiotensin II (Ang II) may remodel IKs in a region-dependent manner, contributing to atrial and ventricular arrhythmias of different mechanisms. OBJECTIVE The purpose of this study was to study whether/how chronic in vivo Ang II administration remodels IKs in atrial and ventricular myocytes. METHODS We used the guinea pig (GP) model whose myocytes express robust IKs. GPs were implanted with minipumps containing Ang II or vehicle. Treatment continued for 4-6 weeks. We used patch clamp, immunofluorescence/confocal microscopy, and immunoblots to evaluate changes in IKs function and to explore the underlying mechanisms. RESULTS We confirmed the pathologic state of the heart after chronic Ang II treatment. IKs density was increased in atrial myocytes but decreased in ventricular myocytes in Ang II- vs vehicle-treated animals. The former was correlated with an increase in KCNQ1/KCNE1 colocalization in myocyte periphery, whereas the latter was correlated with a decrease in KCNQ1 protein level. Interestingly, these changes in IKs were not translated into expected alterations in AP duration or plateau voltage, indicating that other currents were involved. In atrial myocytes from Ang II-treated animals, the L-type Ca channel current was increased, contributing to AP plateau elevation and AP duration prolongation. CONCLUSION IKs is differentially modulated by chronic in vivo Ang II administration between atrial and ventricular myocytes. Other currents remodeled by Ang II treatment also contribute to changes in action potentials.
Collapse
Affiliation(s)
- Dimitar P Zankov
- Department of Physiology & Biophysics, Virginia Commonwealth University, Richmond, Virginia
| | - Fadi N Salloum
- Department of Internal Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Min Jiang
- Department of Physiology & Biophysics, Virginia Commonwealth University, Richmond, Virginia
| | - Gea-Ny Tseng
- Department of Physiology & Biophysics, Virginia Commonwealth University, Richmond, Virginia; Department of Internal Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
11
|
Podzolkov МШ, Tarzimanova AI, Gataulin RG. CARDIOPROTECTIVE PROPERTIES OF LISINOPRIL: NEW POSSIBILITIES. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2018. [DOI: 10.20996/1819-6446-2018-14-3-319-323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aim. To study the changes in the stiffness of the arterial wall, vasomotor function of the endothelium, and appearance of new cases of atrial fibrillation (AF) in patients with arterial hypertension with long-term treatment with lisinopril.Material and method. 66 hypertensive patients with cardiac sinus rhythm at the age of 48-64 years (mean age 58.4±4.2 years) were included into the study. They were randomized into 2 groups: patients of group 1 (n=35) were prescribed lisinopril or a combination of lisinopril with hydrochlorothiazide over the 5-year follow-up; patients of group 2 (control) did not receive angiotensin converting enzyme inhibitors or angiotensin II receptor blockers. The follow-up duration was from September 2010 until June 2016. It included telephone calls once every 3 months and annual clinical, instrumental and laboratory examination. The new-onset AF was identified by the 24-hour Holter ECG monitoring results and by patient symptom diaries.Results. New-onset AF was registered in 2 patients (6%) in the lisinopril group and in 4 patients (13%) from the control group (p=0.001) over the 5-year follow-up. Lisinopril significantly reduced AF incidence in hypertensive patients. The patients on lisinopril were found to have no significant changes in the left ventricular mass index and left atrial size according to echocardiography done after the 5-year follow-up whereas in the patients of control group both parameters increased significantly. Lisinopril contributed to the maintenance of endothelial vasodilator function and prevented increase in arterial wall stiffness.Conclusion. Long term lisinopril treatment was found to significantly reduce the AF incidence in hypertensive patients over the 5-year follow-up. Lisinopril demonstrated organoprotective properties throughout the cardiovascular disease continuum and can be recommended for primary prevention of arrhythmia in hypertensive patients.
Collapse
|
12
|
Crassous PA, Shu P, Huang C, Gordan R, Brouckaert P, Lampe PD, Xie LH, Beuve A. Newly Identified NO-Sensor Guanylyl Cyclase/Connexin 43 Association Is Involved in Cardiac Electrical Function. J Am Heart Assoc 2017; 6:e006397. [PMID: 29269353 PMCID: PMC5778997 DOI: 10.1161/jaha.117.006397] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 10/05/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Guanylyl cyclase, a heme-containing α1β1 heterodimer (GC1), produces cGMP in response to Nitric oxide (NO) stimulation. The NO-GC1-cGMP pathway negatively regulates cardiomyocyte contractility and protects against cardiac hypertrophy-related remodeling. We recently reported that the β1 subunit of GC1 is detected at the intercalated disc with connexin 43 (Cx43). Cx43 forms gap junctions (GJs) at the intercalated disc that are responsible for electrical propagation. We sought to determine whether there is a functional association between GC1 and Cx43 and its role in cardiac homeostasis. METHODS AND RESULTS GC1 and Cx43 immunostaining at the intercalated disc and coimmunoprecipitation from membrane fraction indicate that GC1 and Cx43 are associated. Mice lacking the α subunit of GC1 (GCα1 knockout mice) displayed a significant decrease in GJ function (dye-spread assay) and Cx43 membrane lateralization. In a cardiac-hypertrophic model, angiotensin II treatment disrupted the GC1-Cx43 association and induced significant Cx43 membrane lateralization, which was exacerbated in GCα1 knockout mice. Cx43 lateralization correlated with decreased Cx43-containing GJs at the intercalated disc, predictors of electrical dysfunction. Accordingly, an ECG revealed that angiotensin II-treated GCα1 knockout mice had impaired ventricular electrical propagation. The phosphorylation level of Cx43 at serine 365, a protein-kinase A upregulated site involved in trafficking/assembly of GJs, was decreased in these models. CONCLUSIONS GC1 modulates ventricular Cx43 location, hence GJ function, and partially protects from electrical dysfunction in an angiotensin II hypertrophy model. Disruption of the NO-cGMP pathway is associated with cardiac electrical disturbance and abnormal Cx43 phosphorylation. This previously unknown NO/Cx43 signaling could be a protective mechanism against stress-induced arrhythmia.
Collapse
Affiliation(s)
- Pierre-Antoine Crassous
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School-Rutgers, Newark, NJ
| | - Ping Shu
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School-Rutgers, Newark, NJ
| | - Can Huang
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School-Rutgers, Newark, NJ
| | - Richard Gordan
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School-Rutgers, Newark, NJ
| | - Peter Brouckaert
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Paul D Lampe
- Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School-Rutgers, Newark, NJ
| | - Annie Beuve
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School-Rutgers, Newark, NJ
| |
Collapse
|
13
|
Abstract
Aims Angiotensin-converting enzyme (ACE), which metabolizes many peptides and plays a key role in blood pressure regulation and vascular remodeling, is expressed as a type-1 membrane glycoprotein on the surface of different cells, including endothelial cells of the heart. We hypothesized that the local conformation and, therefore, the properties of heart ACE could differ from lung ACE due to different microenvironment in these organs. Methods and results We performed ACE phenotyping (ACE levels, conformation and kinetic characteristics) in the human heart and compared it with that in the lung. ACE activity in heart tissues was 10–15 lower than that in lung. Various ACE effectors, LMW endogenous ACE inhibitors and HMW ACE-binding partners, were shown to be present in both heart and lung tissues. “Conformational fingerprint” of heart ACE (i.e., the pattern of 17 mAbs binding to different epitopes on the ACE surface) significantly differed from that of lung ACE, which reflects differences in the local conformations of these ACEs, likely controlled by different ACE glycosylation in these organs. Substrate specificity and pH-optima of the heart and lung ACEs also differed. Moreover, even within heart the apparent ACE activities, the local ACE conformations, and the content of ACE inhibitors differ in atria and ventricles. Conclusions Significant differences in the local conformations and kinetic properties of heart and lung ACEs demonstrate tissue specificity of ACE and provide a structural base for the development of mAbs able to distinguish heart and lung ACEs as a potential blood test for predicting atrial fibrillation risk.
Collapse
|
14
|
Becari C, Durand MT, Guimaraes AO, Lataro RM, Prado CM, de Oliveira M, Candido SCO, Pais P, Ribeiro MS, Bader M, Pesquero JB, Salgado MCO, Salgado HC. Elastase-2, a Tissue Alternative Pathway for Angiotensin II Generation, Plays a Role in Circulatory Sympathovagal Balance in Mice. Front Physiol 2017; 8:170. [PMID: 28386233 PMCID: PMC5363176 DOI: 10.3389/fphys.2017.00170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 03/07/2017] [Indexed: 12/26/2022] Open
Abstract
In vitro and ex vivo experiments indicate that elastase-2 (ELA-2), a chymotrypsin-serine protease elastase family member 2A, is an alternative pathway for angiotensin II (Ang II) generation. However, the role played by ELA-2 in vivo is unclear. We examined ELA-2 knockout (ELA-2KO) mice compared to wild-type (WT) mice and determined whether ELA-2 played a role in hemodynamics [arterial pressure (AP) and heart rate (HR)], cardiocirculatory sympathovagal balance and baroreflex sensitivity. The variability of systolic arterial pressure (SAP) and pulse interval (PI) for evaluating autonomic modulation was examined for time and frequency domains (spectral analysis), whereas a symbolic analysis was also used to evaluate PI variability. In addition, baroreflex sensitivity was examined using the sequence method. Cardiac function was evaluated echocardiographically under anesthesia. The AP was normal whereas the HR was reduced in ELA-2KO mice (425 ± 17 vs. 512 ± 13 bpm from WT). SAP variability and baroreflex sensitivity were similar in both strains. The LF power from the PI spectrum (33.6 ± 5 vs. 51.8 ± 4.8 nu from WT) and the LF/HF ratio (0.60 ± 0.1 vs. 1.45 ± 0.3 from WT) were reduced, whereas the HF power was increased (66.4 ± 5 vs. 48.2 ± 4.8 nu from WT) in ELA-2KO mice, indicating a shift toward parasympathetic modulation of HR. Echocardiographic examination showed normal fractional shortening and an ejection fraction in ELA-2KO mice; however, the cardiac output, stroke volume, and ventricular size were reduced. These findings provide the first evidence that ELA-2 acts on the sympathovagal balance of the heart, as expressed by the reduced sympathetic modulation of HR in ELA-2KO mice.
Collapse
Affiliation(s)
- Christiane Becari
- Department of Physiology, Ribeirão Preto Medical School, University of São PauloRibeirão Preto, Brazil; Department of Pharmacology, Ribeirão Preto Medical School, University of São PauloRibeirão Preto, Brazil; Department of Cardiovascular Diseases, Mayo ClinicRochester, MN, USA
| | - Marina T Durand
- Department of Physiology, Ribeirão Preto Medical School, University of São PauloRibeirão Preto, Brazil; Department of Medicine, University of Ribeirão PretoRibeirão Preto, Brazil
| | - Alessander O Guimaraes
- Max Delbruck Center for Molecular MedicineBerlin, Germany; Department of Biophysics, Federal University of São PauloSão Paulo, Brazil
| | - Renata M Lataro
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo Ribeirão Preto, Brazil
| | - Cibele M Prado
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo Ribeirão Preto, Brazil
| | - Mauro de Oliveira
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo Ribeirão Preto, Brazil
| | - Sarai C O Candido
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo Ribeirão Preto, Brazil
| | - Paloma Pais
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo Ribeirão Preto, Brazil
| | - Mauricio S Ribeiro
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo Ribeirão Preto, Brazil
| | - Michael Bader
- Max Delbruck Center for Molecular MedicineBerlin, Germany; Berlin Institute of Health, Charité-University Medicine BerlinBerlin, Germany; German Center for Cardiovascular Research, Partner Site BerlinBerlin, Germany
| | - Joao B Pesquero
- Department of Biophysics, Federal University of São Paulo São Paulo, Brazil
| | - Maria C O Salgado
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo Ribeirão Preto, Brazil
| | - Helio C Salgado
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo Ribeirão Preto, Brazil
| |
Collapse
|
15
|
Abstract
Cardiac arrhythmias can follow disruption of the normal cellular electrophysiological processes underlying excitable activity and their tissue propagation as coherent wavefronts from the primary sinoatrial node pacemaker, through the atria, conducting structures and ventricular myocardium. These physiological events are driven by interacting, voltage-dependent, processes of activation, inactivation, and recovery in the ion channels present in cardiomyocyte membranes. Generation and conduction of these events are further modulated by intracellular Ca2+ homeostasis, and metabolic and structural change. This review describes experimental studies on murine models for known clinical arrhythmic conditions in which these mechanisms were modified by genetic, physiological, or pharmacological manipulation. These exemplars yielded molecular, physiological, and structural phenotypes often directly translatable to their corresponding clinical conditions, which could be investigated at the molecular, cellular, tissue, organ, and whole animal levels. Arrhythmogenesis could be explored during normal pacing activity, regular stimulation, following imposed extra-stimuli, or during progressively incremented steady pacing frequencies. Arrhythmic substrate was identified with temporal and spatial functional heterogeneities predisposing to reentrant excitation phenomena. These could arise from abnormalities in cardiac pacing function, tissue electrical connectivity, and cellular excitation and recovery. Triggering events during or following recovery from action potential excitation could thereby lead to sustained arrhythmia. These surface membrane processes were modified by alterations in cellular Ca2+ homeostasis and energetics, as well as cellular and tissue structural change. Study of murine systems thus offers major insights into both our understanding of normal cardiac activity and its propagation, and their relationship to mechanisms generating clinical arrhythmias.
Collapse
Affiliation(s)
- Christopher L-H Huang
- Physiological Laboratory and the Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
16
|
O'Neal WT, Qureshi W, Zhang ZM, Soliman EZ. Bidirectional association between atrial fibrillation and congestive heart failure in the elderly. J Cardiovasc Med (Hagerstown) 2016; 17:181-6. [PMID: 26825329 DOI: 10.2459/jcm.0000000000000289] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AIMS The aim of this study was to examine the bidirectional association between atrial fibrillation and congestive heart failure (CHF) in older adults. METHODS We studied the association of atrial fibrillation at entry with incident CHF (N = 5281; 85% white, 42% male) and the association of CHF at entry with incident atrial fibrillation (N = 5233; 85% white, 42% male) in the Cardiovascular Health Study (CHS). Baseline atrial fibrillation was identified during the study electrocardiogram and by self-reported history, and incident cases were identified during subsequent study electrocardiograms and hospitalization data. Baseline CHF was identified by self-reported history and adjudication of medical records, and incident cases were identified using hospitalization data. Cox regression was used to compute hazard ratios and 95% confidence intervals (CIs) for the association between atrial fibrillation and incident CHF, and CHF and incident atrial fibrillation, separately. RESULTS Over a median follow-up of 12.6 years, 534 (10%) participants developed atrial fibrillation. CHF was associated with an increased risk of atrial fibrillation (hazard ratio 2.0, 95% CI 1.4, 3.0). A total of 1692 (32%) participants developed CHF over a median follow-up of 11.7 years and atrial fibrillation was associated with an increased risk of CHF (hazard ratio 1.9, 95% CI 1.5, 2.2). CONCLUSION Our results suggest that a bidirectional relationship exists between atrial fibrillation and CHF, with each condition influencing the development of the other.
Collapse
Affiliation(s)
- Wesley T O'Neal
- aDepartment of Internal Medicine bDepartment of Internal Medicine, Section on Cardiology cEpidemiological Cardiology Research Center (EPICARE), Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | | | | |
Collapse
|
17
|
Tse G, Yan BP, Chan YWF, Tian XY, Huang Y. Reactive Oxygen Species, Endoplasmic Reticulum Stress and Mitochondrial Dysfunction: The Link with Cardiac Arrhythmogenesis. Front Physiol 2016; 7:313. [PMID: 27536244 PMCID: PMC4971160 DOI: 10.3389/fphys.2016.00313] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/11/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Cardiac arrhythmias represent a significant problem globally, leading to cerebrovascular accidents, myocardial infarction, and sudden cardiac death. There is increasing evidence to suggest that increased oxidative stress from reactive oxygen species (ROS), which is elevated in conditions such as diabetes and hypertension, can lead to arrhythmogenesis. METHOD A literature review was undertaken to screen for articles that investigated the effects of ROS on cardiac ion channel function, remodeling and arrhythmogenesis. RESULTS Prolonged endoplasmic reticulum stress is observed in heart failure, leading to increased production of ROS. Mitochondrial ROS, which is elevated in diabetes and hypertension, can stimulate its own production in a positive feedback loop, termed ROS-induced ROS release. Together with activation of mitochondrial inner membrane anion channels, it leads to mitochondrial depolarization. Abnormal function of these organelles can then activate downstream signaling pathways, ultimately culminating in altered function or expression of cardiac ion channels responsible for generating the cardiac action potential (AP). Vascular and cardiac endothelial cells become dysfunctional, leading to altered paracrine signaling to influence the electrophysiology of adjacent cardiomyocytes. All of these changes can in turn produce abnormalities in AP repolarization or conduction, thereby increasing likelihood of triggered activity and reentry. CONCLUSION ROS plays a significant role in producing arrhythmic substrate. Therapeutic strategies targeting upstream events include production of a strong reducing environment or the use of pharmacological agents that target organelle-specific proteins and ion channels. These may relieve oxidative stress and in turn prevent arrhythmic complications in patients with diabetes, hypertension, and heart failure.
Collapse
Affiliation(s)
- Gary Tse
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong KongHong Kong, China
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong KongHong Kong, China
| | - Bryan P. Yan
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong KongHong Kong, China
- Department of Epidemiology and Preventive Medicine, Monash UniversityMelbourne, VIC, Australia
| | - Yin W. F. Chan
- Department of Psychology, School of Biological Sciences, University of CambridgeCambridge, UK
| | - Xiao Yu Tian
- Faculty of Medicine, School of Biomedical Sciences, Chinese University of Hong KongHong Kong, China
| | - Yu Huang
- Faculty of Medicine, School of Biomedical Sciences, Chinese University of Hong KongHong Kong, China
| |
Collapse
|
18
|
Chen JY, Liu JH, Wu HDI, Lin KH, Chang KC, Liou YM. Transforming Growth Factor-β1 T869C Gene Polymorphism Is Associated with Acquired Sick Sinus Syndrome via Linking a Higher Serum Protein Level. PLoS One 2016; 11:e0158676. [PMID: 27380173 PMCID: PMC4933337 DOI: 10.1371/journal.pone.0158676] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 06/20/2016] [Indexed: 12/19/2022] Open
Abstract
Background Familial sick sinus syndrome is associated with gene mutations and dysfunction of ion channels. In contrast, degenerative fibrosis of the sinus node tissue plays an important role in the pathogenesis of acquired sick sinus syndrome. There is a close relationship between transforming growth factor-β1 mediated cardiac fibrosis and acquired arrhythmia. It is of interest to examine whether transforming growth factor-β1 is involved in the pathogenesis of acquired sick sinus syndrome. Methods Overall, 110 patients with acquired SSS and 137 age/gender-matched controls were screened for transforming growth factor-β1 and cardiac sodium channel gene polymorphisms using gene sequencing or restriction fragment length polymorphism methods. An enzyme-linked immunosorbent assay was used to determine the serum level of transforming growth factor-β1. Results Two transforming growth factor-β1 gene polymorphisms (C-509T and T+869C) and one cardiac sodium channel gene polymorphism (H588R) have been identified. The C-dominant CC/CT genotype frequency of T869C was significantly higher in acquired sick sinus syndrome patients than in controls (OR 2.09, 95% CI 1.16–3.75, P = 0.01). Consistently, the level of serum transforming growth factor-β1 was also significantly greater in acquired sick sinus syndrome group than in controls (5.3±3.4 ng/ml vs. 3.7±2.4 ng/ml, P = 0.01). In addition, the CC/CT genotypes showed a higher transforming growth factor-β1 serum level than the TT genotype (4.25 ± 2.50 ng/ml vs. 2.71± 1.76 ng/ml, P = 0.028) in controls. Conclusion Transforming growth factor-β1 T869C polymorphism, correlated with high serum transforming growth factor-β1 levels, is associated with susceptibility to acquired sick sinus syndrome.
Collapse
Affiliation(s)
- Jan-Yow Chen
- Division of Cardiology, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Jiung-Hsiun Liu
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Hong-Dar Isaac Wu
- Department of Applied Mathematics and Institute of Statistics, National Chung Hsing University, Taichung, Taiwan
| | - Kuo-Hung Lin
- Division of Cardiology, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Kuan-Cheng Chang
- Division of Cardiology, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Ying-Ming Liou
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
19
|
Li H, Spagnol G, Zheng L, Stauch KL, Sorgen PL. Regulation of Connexin43 Function and Expression by Tyrosine Kinase 2. J Biol Chem 2016; 291:15867-80. [PMID: 27235399 DOI: 10.1074/jbc.m116.727008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Indexed: 11/06/2022] Open
Abstract
Connexin43 (Cx43) assembly and degradation, the regulation of electrical and metabolic coupling, as well as modulating the interaction with other proteins, involve phosphorylation. Here, we identified and characterized the biological significance of a novel tyrosine kinase that phosphorylates Cx43, tyrosine kinase 2 (Tyk2). Activation of Tyk2 led to a decrease in Cx43 gap junction communication by increasing the turnover rate of Cx43 from the plasma membrane. Tyk2 directly phosphorylated Cx43 residues Tyr-247 and Tyr-265, leading to indirect phosphorylation on residues Ser-279/Ser-282 (MAPK) and Ser-368 (PKC). Although this phosphorylation pattern is similar to what has been observed following Src activation, the response caused by Tyk2 occurred when Src was inactive in NRK cells. Knockdown of Tyk2 at the permissive temperature (active v-Src) in LA-25 cells decreased Cx43 phosphorylation, indicating that although activation of Tyk2 and v-Src leads to phosphorylation of the same Cx43CT residues, they are not identical in level at each site. Additionally, angiotensin II activation of Tyk2 increased the intracellular protein level of Cx43 via STAT3. These findings indicate that, like Src, Tyk2 can also inhibit gap junction communication by phosphorylating Cx43.
Collapse
Affiliation(s)
- Hanjun Li
- From the University of Nebraska Medical Center, Omaha, Nebraska 68105
| | - Gaelle Spagnol
- From the University of Nebraska Medical Center, Omaha, Nebraska 68105
| | - Li Zheng
- From the University of Nebraska Medical Center, Omaha, Nebraska 68105
| | - Kelly L Stauch
- From the University of Nebraska Medical Center, Omaha, Nebraska 68105
| | - Paul L Sorgen
- From the University of Nebraska Medical Center, Omaha, Nebraska 68105
| |
Collapse
|
20
|
Mathieu S, El Khoury N, Rivard K, Gélinas R, Goyette P, Paradis P, Nemer M, Fiset C. Reduction in Na(+) current by angiotensin II is mediated by PKCα in mouse and human-induced pluripotent stem cell-derived cardiomyocytes. Heart Rhythm 2016; 13:1346-54. [PMID: 26921763 DOI: 10.1016/j.hrthm.2016.02.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Ventricular arrhythmias and sudden cardiac deaths are among the leading causes of mortality in patients with heart failure, and the underlying mechanisms remain incompletely understood. Chronic elevation of angiotensin II (ANGII) is known to be one of the main contributors to heart failure. OBJECTIVE We tested whether ANGII can alter ventricular conduction and Na(+) current using transgenic mice with cardiomyocyte-restricted overexpression of ANGII type 1 receptor (AT1R). METHODS We used surface electrocardiograms along with current- and voltage-clamp techniques to characterize the electrophysiological properties of AT1R mice while the underlying regulatory mechanisms were explored using reverse transcription/quantitative polymerase chain reaction, Western blots, and immunofluorescence techniques. RESULTS Electrophysiological data indicated that chronic AT1R activation in ventricular myocytes caused a 60% reduction in Na(+) current density that slowed the maximal velocity of the action potential upstroke, leading to a prolongation of the QRS complex. These changes occur independently of cardiac hypertrophy, suggesting a direct role for ANGII/AT1R in slowing ventricular conduction. Western blots demonstrated a selective increase in sarcolemmal protein kinase Cα (PKCα) in AT1R mice, indicating PKCα activation. Furthermore, immunofluorescence analysis showed reorganization of PKCα expression to sarcolemma and colocalization with NaV1.5 in AT1R myocytes. The involvement of PKCα in regulating Na(+) current was subsequently demonstrated in human-induced pluripotent stem cell-derived cardiomyocytes where ANGII treatment reduced Na(+) current density. Concomitant treatment with αV5-3, a PKCα translocation inhibitor peptide, blocked the ANGII effect. CONCLUSION Overall, this study suggests that in mouse and human cardiomyocytes, PKCα is an important mediator of the ANGII-induced reduction in Na(+) current and may contribute to ventricular arrhythmias.
Collapse
Affiliation(s)
- Sophie Mathieu
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Nabil El Khoury
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Department of Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Katy Rivard
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Roselle Gélinas
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Philippe Goyette
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
| | - Pierre Paradis
- Lady Davis Institute, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Mona Nemer
- Ottawa University, Ottawa, Ontario, Canada
| | - Céline Fiset
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
21
|
Yasuno S, Kuwahara K, Kinoshita H, Yamada C, Nakagawa Y, Usami S, Kuwabara Y, Ueshima K, Harada M, Nishikimi T, Nakao K. Angiotensin II type 1a receptor signalling directly contributes to the increased arrhythmogenicity in cardiac hypertrophy. Br J Pharmacol 2014; 170:1384-95. [PMID: 23937445 DOI: 10.1111/bph.12328] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 07/11/2013] [Accepted: 07/21/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Angiotensin II has been implicated in the development of various cardiovascular ailments, including cardiac hypertrophy and heart failure. The fact that inhibiting its signalling reduced the incidences of both sudden cardiac death and heart failure in several large-scale clinical trials suggests that angiotensin II is involved in increased cardiac arrhythmogenicity during the development of heart failure. However, because angiotensin II also promotes structural remodelling, including cardiomyocyte hypertrophy and cardiac fibrosis, it has been difficult to assess its direct contribution to cardiac arrhythmogenicity independently of the structural effects. EXPERIMENTAL APPROACH We induced cardiac hypertrophy in wild-type (WT) and angiotensin II type 1a receptor knockout (AT1aR-KO) mice by transverse aortic constriction (TAC). The susceptibility to ventricular tachycardia (VT) assessed in an in vivo electrophysiological study was compared in the two genotypes. The effect of acute pharmacological blockade of AT1R on the incidences of arrhythmias was also assessed. KEY RESULTS As described previously, WT and AT1aR-KO mice with TAC developed cardiac hypertrophy to the same degree, but the incidence of VT was much lower in the latter. Moreover, although TAC induced an increase in tyrosine phosphorylation of connexin 43, a critical component of gap junctional channels, and a reduction in ventricular levels of connexin 43 protein in both genotypes, the effect was significantly ameliorated in AT1aR-KO mice. Acute pharmacological blockade of AT1R also reduced the incidence of arrhythmias. CONCLUSIONS AND IMPLICATIONS Our findings demonstrate that AT1aR-mediated signalling makes a direct contribution to the increase in arrhythmogenicity in hypertrophied hearts independently of structural remodelling.
Collapse
Affiliation(s)
- Shinji Yasuno
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto, Japan; EBM Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Dinov B, Kosiuk J, Kircher S, Bollmann A, Acou WJ, Arya A, Hindricks G, Rolf S. Impact of Metabolic Syndrome on Left Atrial Electroanatomical Remodeling and Outcomes After Radiofrequency Ablation of Nonvalvular Atrial Fibrillation. Circ Arrhythm Electrophysiol 2014; 7:483-9. [DOI: 10.1161/circep.113.001185] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Recent studies reported worse outcomes after atrial fibrillation (AF) ablation in patients with metabolic syndrome (MetS). However, mechanisms of AF recurrence in MetS remain unclear.
Method and Results—
We performed pulmonary vein isolation and voltage mapping in 236 patients with AF (age 61±9.6 years; persistent AF 64%; MetS 54%). Left atrial (LA) low voltage areas were semiquantitatively estimated and presented as low voltage index. MetS was defined according to National Cholesterol Education Program Adult Treatment Panel III. Follow-up for AF recurrence ≤12 months was performed. LA low voltage areas were observed in 46% of patients with MetS versus 8.2% patients without MetS ;
P
<0.0001. MetS was an independent predictor of LA low voltage areas: odds ratio, 11.64; 95% confidence interval, 4.381–30.903;
P
<0.0001. Observed AF recurrence at 12 months was 42.7% in MetS versus 36.1% in the non-MetS group (
P
=0.303). The presence of LA low voltage areas was a predictor of 12-month AF recurrence: odds ratio, 2.99; 95% confidence interval, 1.36–6.56;
P
=0.006. Probability of 12-month AF recurrence increased with 84.5% for every unit of low voltage Index.
Conclusions—
MetS was not associated with worse outcomes after radiofrequency catheter ablation of AF, but LA low voltage areas were more frequently observed in patients with MetS. The presence and extent of LA low voltage areas may influence the long-term outcomes after catheter ablation.
Collapse
Affiliation(s)
- Borislav Dinov
- From the Department of Electrophysiology, Heart Center, University of Leipzig, Leipzig, Germany
| | - Jedrzej Kosiuk
- From the Department of Electrophysiology, Heart Center, University of Leipzig, Leipzig, Germany
| | - Simon Kircher
- From the Department of Electrophysiology, Heart Center, University of Leipzig, Leipzig, Germany
| | - Andreas Bollmann
- From the Department of Electrophysiology, Heart Center, University of Leipzig, Leipzig, Germany
| | - Willem-Jan Acou
- From the Department of Electrophysiology, Heart Center, University of Leipzig, Leipzig, Germany
| | - Arash Arya
- From the Department of Electrophysiology, Heart Center, University of Leipzig, Leipzig, Germany
| | - Gerhard Hindricks
- From the Department of Electrophysiology, Heart Center, University of Leipzig, Leipzig, Germany
| | - Sascha Rolf
- From the Department of Electrophysiology, Heart Center, University of Leipzig, Leipzig, Germany
| |
Collapse
|
23
|
Herren AW, Bers DM, Grandi E. Post-translational modifications of the cardiac Na channel: contribution of CaMKII-dependent phosphorylation to acquired arrhythmias. Am J Physiol Heart Circ Physiol 2013; 305:H431-45. [PMID: 23771687 DOI: 10.1152/ajpheart.00306.2013] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The voltage-gated Na channel isoform 1.5 (NaV1.5) is the pore forming α-subunit of the voltage-gated cardiac Na channel, which is responsible for the initiation and propagation of cardiac action potentials. Mutations in the SCN5A gene encoding NaV1.5 have been linked to changes in the Na current leading to a variety of arrhythmogenic phenotypes, and alterations in the NaV1.5 expression level, Na current density, and/or gating have been observed in acquired cardiac disorders, including heart failure. The precise mechanisms underlying these abnormalities have not been fully elucidated. However, several recent studies have made it clear that NaV1.5 forms a macromolecular complex with a number of proteins that modulate its expression levels, localization, and gating and is the target of extensive post-translational modifications, which may also influence all these properties. We review here the molecular aspects of cardiac Na channel regulation and their functional consequences. In particular, we focus on the molecular and functional aspects of Na channel phosphorylation by the Ca/calmodulin-dependent protein kinase II, which is hyperactive in heart failure and has been causally linked to cardiac arrhythmia. Understanding the mechanisms of altered NaV1.5 expression and function is crucial for gaining insight into arrhythmogenesis and developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Anthony W Herren
- Department of Pharmacology, University of California Davis, Davis, California
| | | | | |
Collapse
|
24
|
Sovari AA, Rutledge CA, Jeong EM, Dolmatova E, Arasu D, Liu H, Vahdani N, Gu L, Zandieh S, Xiao L, Bonini MG, Duffy HS, Dudley SC. Mitochondria oxidative stress, connexin43 remodeling, and sudden arrhythmic death. Circ Arrhythm Electrophysiol 2013; 6:623-31. [PMID: 23559673 DOI: 10.1161/circep.112.976787] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Previously, we showed that a mouse model (ACE8/8) of cardiac renin-angiotensin system activation has a high rate of spontaneous ventricular tachycardia and sudden cardiac death secondary to a reduction in connexin43 level. Angiotensin-II activation increases reactive oxygen species (ROS) production, and ACE8/8 mice show increased cardiac ROS. We sought to determine the source of ROS and whether ROS played a role in the arrhythmogenesis. METHODS AND RESULTS Wild-type and ACE8/8 mice with and without 2 weeks of treatment with L-NIO (NO synthase inhibitor), sepiapterin (precursor of tetrahydrobiopterin), MitoTEMPO (mitochondria-targeted antioxidant), TEMPOL (a general antioxidant), apocynin (nicotinamide adenine dinucleotide phosphate oxidase inhibitor), allopurinol (xanthine oxidase inhibitor), and ACE8/8 crossed with P67 dominant negative mice to inhibit the nicotinamide adenine dinucleotide phosphate oxidase were studied. Western blotting, detection of mitochondrial ROS by MitoSOX Red, electron microscopy, immunohistochemistry, fluorescent dye diffusion technique for functional assessment of connexin43, telemetry monitoring, and in vivo electrophysiology studies were performed. Treatment with MitoTEMPO reduced sudden cardiac death in ACE8/8 mice (from 74% to 18%; P<0.005), decreased spontaneous ventricular premature beats, decreased ventricular tachycardia inducibility (from 90% to 17%; P<0.05), diminished elevated mitochondrial ROS to the control level, prevented structural damage to mitochondria, resulted in 2.6-fold increase in connexin43 level at the gap junctions, and corrected gap junction conduction. None of the other antioxidant therapies prevented ventricular tachycardia and sudden cardiac death in ACE8/8 mice. CONCLUSIONS Mitochondrial oxidative stress plays a central role in angiotensin II-induced gap junction remodeling and arrhythmia. Mitochondria-targeted antioxidants may be effective antiarrhythmic drugs in cases of renin-angiotensin system activation.
Collapse
Affiliation(s)
- Ali A Sovari
- Section of Cardiology and Center for Cardiovascular Research, University of Illinois at Chicago, IL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Riley G, Syeda F, Kirchhof P, Fabritz L. An introduction to murine models of atrial fibrillation. Front Physiol 2012; 3:296. [PMID: 22934047 PMCID: PMC3429067 DOI: 10.3389/fphys.2012.00296] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 07/08/2012] [Indexed: 01/28/2023] Open
Abstract
Understanding the mechanism of re-entrant arrhythmias in the past 30 years has allowed the development of almost curative therapies for many rhythm disturbances. The complex, polymorphic arrhythmias of atrial fibrillation (AF) and sudden death are, unfortunately, not yet well understood, and hence still in need of adequate therapy. AF contributes markedly to morbidity and mortality in aging Western populations. In the past decade, many genetically altered murine models have been described and characterized. Here, we review genetically altered murine models of AF; powerful tools that will enable a better understanding of the mechanisms of AF and the assessment of novel therapeutic interventions.
Collapse
Affiliation(s)
- Genna Riley
- Centre for Cardiovascular Sciences, School of Clinical and Experimental Medicine, University of Birmingham Birmingham, UK
| | | | | | | |
Collapse
|
26
|
Impact of QRS Duration and Morphology on the Risk of Sudden Cardiac Death in Asymptomatic Patients With Aortic Stenosis. J Am Coll Cardiol 2012; 59:1142-9. [DOI: 10.1016/j.jacc.2011.12.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 11/29/2011] [Accepted: 12/07/2011] [Indexed: 11/23/2022]
|
27
|
Rosenberg MA, Das S, Quintero Pinzon P, Knight AC, Sosnovik DE, Ellinor PT, Rosenzweig A. A Novel Transgenic Mouse Model of Cardiac Hypertrophy and Atrial Fibrillation. J Atr Fibrillation 2012; 4:415. [PMID: 28496713 PMCID: PMC3521534 DOI: 10.4022/jafib.415] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 10/11/2011] [Accepted: 12/12/2012] [Indexed: 12/18/2022]
Abstract
Cardiac hypertrophy is a major risk factor for the development of atrial fibrillation (AF). However, there are few animal models of AF associated with cardiac hypertrophy. In this study, we describe the in vivo electrophysiological characteristics and histopathology of a mouse model of cardiac hypertrophy that develops AF. Myostatin is a well-known negative regulator of skeletal muscle growth that was recently found to additionally regulate cardiac muscle growth. Using cardiac-specific expression of the inhibitory myostatin pro-peptide, we generated transgenic (TG) mice with dominant-negative regulation of MSTN (DN-MSTN). One line (DN-MSTN TG13) displayed ventricular hypertrophy, as well as spontaneous AF on the surface electrocardiogram (ECG), and was further evaluated. DN-MSTN TG13 had normal systolic function, but displayed atrial enlargement on cardiac MRI, as well as atrial fibrosis histologically. Baseline ECG revealed an increased P wave duration and QRS interval compared with wild-type littermate (WT) mice. Seven of 19 DN-MSTN TG13 mice had spontaneous or inducible AF, while none of the WT mice had atrial arrhythmias (p<0.05). Connexin40 (Cx40) was decreased in DN-MSTN TG13 mice, even in the absence of AF or significant atrial fibrosis, raising the possibility that MSTN signaling may play a role in Cx40 down-regulation and the development of AF in this mouse model. In conclusion, DN-MSTN TG13 mice represent a novel model of AF, in which molecular changes including an initial loss of Cx40 are noted prior to fibrosis and the development of atrial arrhythmias.
Collapse
Affiliation(s)
- Michael A Rosenberg
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, CLS 9, Boston MA 02215
| | - Saumya Das
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, CLS 9, Boston MA 02215
| | - Pablo Quintero Pinzon
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, CLS 9, Boston MA 02215
| | - Ashley C Knight
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, CLS 9, Boston MA 02215
| | - David E Sosnovik
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 55 Fruit Street, Boston MA 02114
- Cardiology Division, Massachusetts General Hospital, 55 Fruit Street, Boston MA 02114
| | - Patrick T Ellinor
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 55 Fruit Street, Boston MA 02114
| | - Anthony Rosenzweig
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, CLS 9, Boston MA 02215
| |
Collapse
|
28
|
Direct action of angiotensin II on the conduction through papillary muscle preparations of rat heart immediately after reoxygenation. J Arrhythm 2012. [DOI: 10.1016/j.joa.2012.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
29
|
Sovari AA, Iravanian S, Dolmatova E, Jiao Z, Liu H, Zandieh S, Kumar V, Wang K, Bernstein KE, Bonini MG, Duffy HS, Dudley SC. Inhibition of c-Src tyrosine kinase prevents angiotensin II-mediated connexin-43 remodeling and sudden cardiac death. J Am Coll Cardiol 2012; 58:2332-9. [PMID: 22093512 DOI: 10.1016/j.jacc.2011.07.048] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 07/26/2011] [Indexed: 01/13/2023]
Abstract
OBJECTIVES The aim of this study was to test whether c-Src tyrosine kinase mediates connexin-43 (Cx43) reduction and sudden cardiac death in a transgenic mouse model of cardiac-restricted overexpression of angiotensin-converting enzyme (ACE8/8 mice). BACKGROUND Renin-angiotensin system activation is associated with an increased risk for arrhythmia and sudden cardiac death, but the mechanism is not well understood. The up-regulation of c-Src by angiotensin II may result in the reduction of Cx43, which impairs gap junction function and provides a substrate for arrhythmia. METHODS Wild-type and ACE8/8 mice with and without treatment with the c-Src inhibitor 1-(1,1-dimethylethyl)-1-(4-methylphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine (PP1) were studied. Telemetry monitoring, in vivo electrophysiologic studies, Western blot analyses for total and phosphorylated c-Src and Cx43, immunohistochemistry staining for Cx43, and functional assessment of Cx43 with fluorescent dye diffusion were performed. RESULTS The majority of the arrhythmic deaths resulted from ventricular tachycardia degenerating to ventricular fibrillation (83%). Levels of total and phosphorylated c-Src were increased and Cx43 reduced in ACE8/8 mice. PP1 reduced total and phosphorylated c-Src levels, increased Cx43 level by 2.1-fold (p < 0.005), increased Cx43 at the gap junctions (immunostaining), improved gap junctional communication (dye spread), and reduced ventricular tachycardia inducibility and sudden cardiac death. The survival rate increased from 11% to 86% with 4 weeks of PP1 treatment (p < 0.005). Treatment with an inactive analog did not change survival or Cx43 levels. CONCLUSIONS Renin-angiotensin system activation is associated with c-Src up-regulation, Cx43 loss, reduced myocyte coupling, and arrhythmic sudden death, which can be prevented by c-Src inhibition. This suggests that an increase in c-Src activity may help mediate renin-angiotensin system-induced arrhythmias and that c-Src inhibitors might exert antiarrhythmic activity.
Collapse
Affiliation(s)
- Ali A Sovari
- Section of Cardiology and Center for Cardiovascular Research, University of Illinois at Chicago, 840 S.Wood Street, Chicago, IL 60612, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Chen JY, Liou YM, Wu HDI, Lin KH, Chang KC. Promoter polymorphism G-6A, which modulates angiotensinogen gene expression, is associated with non-familial sick sinus syndrome. PLoS One 2012; 7:e29951. [PMID: 22242192 PMCID: PMC3252346 DOI: 10.1371/journal.pone.0029951] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 12/07/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND It is well known that familial sick sinus syndrome (SSS) is caused by functional alterations of ion channels and gap junction. Limited information is available on the mechanism of age-related non-familial SSS. Although evidence shows a close link between arrhythmia and the renin-angiotensin system (RAS), it remains to be determined whether the RAS is involved in the pathogenesis of non-familial SSS. METHODS In this study, 113 patients with documented non-familial SSS and 125 controls were screened for angiotensinogen (AGT) and gap junction protein-connexin 40 (Cx40) promoter polymorphisms by gene sequencing, followed by an association study. A luciferase assay was used to determine the transcriptional activity of the promoter polymorphism. The interaction between nuclear factors and the promoter polymorphism was characterized by an electrophoretic mobility shift assay (EMSA). RESULTS Association study showed the Cx40 -44/+71 polymorphisms are not associated with non-familial SSS; however, it indicated that four polymorphic sites at positions -6, -20, -152, and -217 in the AGT promoter are linked to non-familial SSS. Compared to controls, SSS patients had a lower frequency of the G-6A AA genotype (OR 2.88, 95% CI 1.58-5.22, P = 0.001) and a higher frequency of the G allele at -6 position (OR 2.65, 95% CI 1.54-4.57, P = 0.0003). EMSA and luciferase assays confirmed that nucleotide G at position -6 modulates the binding affinity with nuclear factors and yields a lower transcriptional activity than nucleotide A (P<0.01). CONCLUSION G-6A polymorphism, which modulates the transcriptional activity of the AGT promoter, may contribute to non-familial SSS susceptibility.
Collapse
Affiliation(s)
- Jan-Yow Chen
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
- Division of Cardiology, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Ying-Ming Liou
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- * E-mail:
| | - Hong-Dar Isaac Wu
- Department of Applied Mathematics and Institute of Statistics, National Chung Hsing University, Taichung, Taiwan
| | - Kuo-Hung Lin
- Division of Cardiology, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Kuan-Cheng Chang
- Division of Cardiology, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
31
|
Sovari AA, Bonini MG, Dudley SC. Effective antioxidant therapy for the management of arrhythmia. Expert Rev Cardiovasc Ther 2011; 9:797-800. [PMID: 21809958 DOI: 10.1586/erc.11.85] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
32
|
Rivard K, Grandy SA, Douillette A, Paradis P, Nemer M, Allen BG, Fiset C. Overexpression of type 1 angiotensin II receptors impairs excitation-contraction coupling in the mouse heart. Am J Physiol Heart Circ Physiol 2011; 301:H2018-27. [DOI: 10.1152/ajpheart.01092.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Transgenic mice that overexpress human type 1 angiotensin II receptor (AT1R) in the heart develop cardiac hypertrophy. Previously, we have shown that in 6-mo AT1R mice, which exhibit significant cardiac remodeling, fractional shortening is decreased. However, it is not clear whether altered contractility is attributable to AT1R overexpression or is secondary to cardiac hypertrophy/remodeling. Thus the present study characterized the effects of AT1R overexpression on ventricular L-type Ca2+ currents ( ICaL), cell shortening, and Ca2+ handling in 50-day and 6-mo-old male AT1R mice. Echocardiography showed there was no evidence of cardiac hypertrophy in 50-day AT1R mice but that fractional shortening was decreased. Cellular experiments showed that cell shortening, ICaL, and Cav1.2 mRNA expression were significantly reduced in 50-day and 6-mo-old AT1R mice compared with controls. In addition, Ca2+ transients and caffeine-induced Ca2+ transients were reduced whereas the time to 90% Ca2+ transient decay was prolonged in both age groups of AT1R mice. Western blot analysis revealed that sarcoplasmic reticulum Ca2+-ATPase and Na+/Ca2+ exchanger protein expression was significantly decreased in 50-day and 6-mo AT1R mice. Overall, the data show that cardiac contractility and the mechanisms that underlie excitation-contraction coupling are altered in AT1R mice. Furthermore, since the alterations in contractility occur before the development of cardiac hypertrophy, it is likely that these changes are attributable to the increased activity of the renin-angiotensin system brought about by AT1R overexpression. Thus it is possible that AT1R blockade may help maintain cardiac contractility in individuals with heart disease.
Collapse
Affiliation(s)
- Katy Rivard
- Research Centre, Montreal Heart Institute, Montreal,
- Faculty of Pharmacy, Université de Montréal, Montreal,
| | - Scott A. Grandy
- Research Centre, Montreal Heart Institute, Montreal,
- Faculty of Pharmacy, Université de Montréal, Montreal,
| | - Annie Douillette
- Research Centre, Montreal Heart Institute, Montreal,
- Faculty of Pharmacy, Université de Montréal, Montreal,
| | | | | | | | - Céline Fiset
- Research Centre, Montreal Heart Institute, Montreal,
- Faculty of Pharmacy, Université de Montréal, Montreal,
| |
Collapse
|
33
|
Luft FC. Connecting the renin-angiotensin-aldosterone system with sudden death. J Mol Med (Berl) 2011; 89:631-3. [PMID: 21607541 DOI: 10.1007/s00109-011-0772-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Friedrich C Luft
- Experimental and Clinical Research Center, Berlin, Lindenbergerweg, Germany.
| |
Collapse
|
34
|
Jeong EM, Liu M, Sturdy M, Gao G, Varghese ST, Sovari AA, Dudley SC. Metabolic stress, reactive oxygen species, and arrhythmia. J Mol Cell Cardiol 2011; 52:454-63. [PMID: 21978629 DOI: 10.1016/j.yjmcc.2011.09.018] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2011] [Revised: 08/20/2011] [Accepted: 09/19/2011] [Indexed: 02/07/2023]
Abstract
Cardiac arrhythmias can cause sudden cardiac death (SCD) and add to the current heart failure (HF) health crisis. Nevertheless, the pathological processes underlying arrhythmias are unclear. Arrhythmic conditions are associated with systemic and cardiac oxidative stress caused by reactive oxygen species (ROS). In excitable cardiac cells, ROS regulate both cellular metabolism and ion homeostasis. Increasing evidence suggests that elevated cellular ROS can cause alterations of the cardiac sodium channel (Na(v)1.5), abnormal Ca(2+) handling, changes of mitochondrial function, and gap junction remodeling, leading to arrhythmogenesis. This review summarizes our knowledge of the mechanisms by which ROS may cause arrhythmias and discusses potential therapeutic strategies to prevent arrhythmias by targeting ROS and its consequences. This article is part of a Special Issue entitled "Local Signaling in Myocytes".
Collapse
Affiliation(s)
- Euy-Myoung Jeong
- Section of Cardiology, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Palatinus JA, Rhett JM, Gourdie RG. The connexin43 carboxyl terminus and cardiac gap junction organization. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1818:1831-43. [PMID: 21856279 DOI: 10.1016/j.bbamem.2011.08.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Revised: 07/25/2011] [Accepted: 08/03/2011] [Indexed: 12/09/2022]
Abstract
The precise spatial order of gap junctions at intercalated disks in adult ventricular myocardium is thought vital for maintaining cardiac synchrony. Breakdown or remodeling of this order is a hallmark of arrhythmic disease of the heart. The principal component of gap junction channels between ventricular cardiomyocytes is connexin43 (Cx43). Protein-protein interactions and modifications of the carboxyl-terminus of Cx43 are key determinants of gap junction function, size, distribution and organization during normal development and in disease processes. Here, we review data on the role of proteins interacting with the Cx43 carboxyl-terminus in the regulation of cardiac gap junction organization, with particular emphasis on Zonula Occludens-1. The rapid progress in this area suggests that in coming years we are likely to develop a fuller understanding of the molecular mechanisms causing pathologic remodeling of gap junctions. With these advances come the promise of novel approach to the treatment of arrhythmia and the prevention of sudden cardiac death. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.
Collapse
Affiliation(s)
- Joseph A Palatinus
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | | | | |
Collapse
|
36
|
Yoshida M, Ohkusa T, Nakashima T, Takanari H, Yano M, Takemura G, Honjo H, Kodama I, Mizukami Y, Matsuzaki M. Alterations in adhesion junction precede gap junction remodelling during the development of heart failure in cardiomyopathic hamsters. Cardiovasc Res 2011; 92:95-105. [DOI: 10.1093/cvr/cvr182] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Masaaki Yoshida
- Division of Cardiology, Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1, Minami-kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Tomoko Ohkusa
- Division of Cardiology, Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1, Minami-kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Tadamitsu Nakashima
- Division of Cardiology, Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1, Minami-kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Hiroki Takanari
- Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University, Japan
| | - Masafumi Yano
- Division of Cardiology, Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1, Minami-kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Genzou Takemura
- Department of Cardiology, Gifu University Graduate School of Medicine, Japan
| | - Haruo Honjo
- Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University, Japan
| | | | | | - Masunori Matsuzaki
- Division of Cardiology, Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1, Minami-kogushi, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
37
|
Iravanian S, Sovari AA, Lardin HA, Liu H, Xiao HD, Dolmatova E, Jiao Z, Harris BS, Witham EA, Gourdie RG, Duffy HS, Bernstein KE, Dudley SC. Inhibition of renin-angiotensin system (RAS) reduces ventricular tachycardia risk by altering connexin43. J Mol Med (Berl) 2011; 89:677-87. [PMID: 21553032 DOI: 10.1007/s00109-011-0761-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2010] [Revised: 03/19/2011] [Accepted: 04/04/2011] [Indexed: 01/13/2023]
Abstract
Renin-angiotensin system (RAS) activation is associated with arrhythmias. We investigated the effects of RAS inhibition in cardiac-specific angiotensin-converting enzyme (ACE) overexpression (ACE 8/8) mice, which exhibit proclivity to ventricular tachycardia (VT) and sudden death because of reduced connexin43 (Cx43). ACE 8/8 mice were treated with an ACE inhibitor (captopril) or an angiotensin receptor type-1 blocker (losartan). Subsequently, electrophysiological studies were performed, and the hearts were extracted for Cx43 quantification using immunoblotting, immunohistochemistry, fluorescent dye spread method, and sodium current quantification using whole cell patch clamping. VT was induced in 12.5% of captopril-treated ACE 8/8 and in 28.6% of losartan-treated mice compared to 87.5% of untreated mice (P < 0.01). Losartan and captopril treatment increased total Cx43 2.4-fold (P = 0.01) and the Cx43 phosphorylation ratio 2.3-fold (P = 0.005). Treatment was associated with a recovery of gap junctional conductance. Survival in treated mice improved to 0.78 at 10 weeks (95% confidence interval 0.64 to 0.92), compared to the expected survival of less than 0.50. In a model of RAS activation, arrhythmic risk was correlated with reduced Cx43 amount and phosphorylation. RAS inhibition resulted in increased total and phosphorylated Cx43, decreased VT inducibility, and improved survival.
Collapse
Affiliation(s)
- Shahriar Iravanian
- Division of Cardiology, Atlanta Veterans Affairs Medical Center and Emory University, GA, 30033, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Mueller EE, Momen A, Massé S, Zhou YQ, Liu J, Backx PH, Henkelman RM, Nanthakumar K, Stewart DJ, Husain M. Electrical remodelling precedes heart failure in an endothelin-1-induced model of cardiomyopathy. Cardiovasc Res 2011; 89:623-33. [PMID: 21062919 DOI: 10.1093/cvr/cvq351] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
AIMS Binary transgenic (BT) mice with doxycycline (DOX)-suppressible cardiac-specific overexpression of endothelin-1 (ET-1) exhibit progressive heart failure (HF), QRS prolongation, and death following DOX withdrawal. However, the molecular basis and reversibility of the electrophysiological abnormalities in this model were not known. Here, we assess the mechanisms underlying ET-1-mediated electrical remodelling, and its role in HF. METHODS AND RESULTS BT vs. non-BT littermate controls were withdrawn from DOX and serially studied with ultrasound biomicroscopy, octapolar catheters, multielectrode epicardial mapping, histopathology, western blot, immunohistochemistry, and qRT-PCR. Abnormalities in ventricular activation and -dV/dt were detected as early as 4 weeks after transgene activation, when the structure and function of the heart remained unaffected. By 8 weeks of ET-1 overexpression, biventricular systolic and diastolic dysfunction, myocardial fibrosis, and cardiomyocyte hypertrophy were observed. Intracardiac and epicardial electrograms revealed prolonged conduction and ventricular activation, reduced -dV/dt, and abnormal atrioventricular nodal function. Within 4 weeks of ET-1 induction, connexin 40 (Cx40) protein and Cx43 mRNA, protein, and phosphorylation levels were reduced by 36, 64, 93, and 69%, respectively; Na(v)1.5 mRNA and protein levels were reduced by 30 and 50%, respectively, as was Na(+) channel conductance. Importantly, the associated electrophysiological abnormalities at this time point were reversible upon suppression of ET-1 overexpression and completely prevented the development of structural and functional remodelling. CONCLUSION ET-1-mediated electrical remodelling correlates with reduced Cx40, Cx43, and Na(v)1.5 expression and decreased Na(+) channel conductance and precedes HF. The sequence and reversibility of this phenotype suggest that a primary abnormality in electrical remodelling may contribute to the pathogenesis of HF.
Collapse
Affiliation(s)
- Erin E Mueller
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zografos T, Katritsis DG. Inhibition of the renin-angiotensin system for prevention of atrial fibrillation. Pacing Clin Electrophysiol 2011; 33:1270-85. [PMID: 20636314 DOI: 10.1111/j.1540-8159.2010.02832.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Atrial fibrillation (AF) is a source of considerable morbidity and mortality. There has been compelling evidence supporting the role of renin-angiotensin system (RAS) in the genesis and perpetuation of AF through atrial remodeling, and experimental studies have validated the utilization of RAS inhibition for AF prevention. This article reviews clinical trials on the use of angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) for the prevention of AF. Results have been variable, depending on the clinical background of treated patients. ACEIs and ARBs appear beneficial for primary prevention of AF in patients with heart failure, whereas they are not equally effective in hypertensive patients with normal left ventricular function. Furthermore, the use of ACEIs or ARBs for secondary prevention of AF has been found beneficial only after electrical cardioversion. Additional data are needed to establish the potential clinical role of renin-angiotensin inhibition for prevention of AF.
Collapse
Affiliation(s)
- Theodoros Zografos
- Department of Cardiology, Athens Euroclinic, 9 Athanassiadou Street, Athens, Greece
| | | |
Collapse
|
40
|
O'Quinn MP, Palatinus JA, Harris BS, Hewett KW, Gourdie RG. A peptide mimetic of the connexin43 carboxyl terminus reduces gap junction remodeling and induced arrhythmia following ventricular injury. Circ Res 2011; 108:704-15. [PMID: 21273554 DOI: 10.1161/circresaha.110.235747] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Remodeling of connexin (Cx)43 gap junctions (GJs) is linked to ventricular arrhythmia. OBJECTIVES A peptide mimetic of the carboxyl terminal (CT) of Cx43, incorporating a postsynaptic density-95/disks-large/ZO-1 (PDZ)-binding domain, reduces Cx43/ZO-1 interaction and GJ size remodeling in vitro. Here, we determined: (1) whether the Cx43-CT mimetic αCT1 altered GJ remodeling following left ventricular (LV) injury in vivo; (2) whether αCT1 affected arrhythmic propensity; and (3) the mechanism of αCT1 effects on arrhythmogenicity and GJ remodeling. METHODS AND RESULTS A cryoinjury model generating a reproducible wound and injury border zone (IBZ) in the LV was used. Adherent methylcellulose patches formulated to locally release αCT1 (< 48 hours) were placed on cryoinjuries. Relative to controls, Cx43/ZO-1 colocalization in the IBZ was reduced by αCT1 by 24 hours after injury. Programmed electric stimulation ex vivo and optical mapping of voltage transients indicated that peptide-treated hearts showed reduced inducible arrhythmias and increased ventricular depolarization rates 7 to 9 days after injury. At 24 hours and 1 week after injury, αCT1-treated hearts maintained Cx43 in intercalated disks (IDs) in the IBZ, whereas by 1 week after injury, controls demonstrated Cx43 remodeling from IDs to lateralized distributions. Over a postinjury time course of 1 week, αCT1-treated IBZs showed increased Cx43 phosphorylation at serine368 (Cx43-pS368) relative to control tissues. In biochemical assays, αCT1 promoted phosphorylation of serine368 by protein kinase (PK)C-ε in a dose-dependent manner that was modulated by, but did not require ZO-1 PDZ2. CONCLUSIONS αCT1 increases Cx43-pS368 in vitro in a PKC-ε-dependent manner and in the IBZ in vivo acutely following ventricular injury. αCT1-mediated increase in Cx43-pS368 phosphorylation may contribute to reductions in inducible-arrhythmia following injury.
Collapse
Affiliation(s)
- Michael P O'Quinn
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, USA
| | | | | | | | | |
Collapse
|
41
|
Stein M, Boulaksil M, Jansen JA, Herold E, Noorman M, Joles JA, van Veen TAB, Houtman MJC, Engelen MA, Hauer RNW, de Bakker JMT, van Rijen HVM. Reduction of fibrosis-related arrhythmias by chronic renin-angiotensin-aldosterone system inhibitors in an aged mouse model. Am J Physiol Heart Circ Physiol 2010; 299:H310-21. [PMID: 20435847 DOI: 10.1152/ajpheart.01137.2009] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Myocardial fibrosis increases arrhythmia vulnerability of the diseased heart. The renin-angiotensin-aldosterone system (RAAS) governs myocardial collagen synthesis. We hypothesized that reducing cardiac fibrosis by chronic RAAS inhibition would result in reduced arrhythmia vulnerability of the senescent mouse heart. Wild-type mice (52 wk old) were treated for 36 wk: 1) untreated control (C); 2) eplerenone (E); 3) losartan (L); and 4) cotreatment with eplerenone and losartan (EL). Ventricular epicardial activation mapping was performed on Langendorff-perfused hearts. Arrhythmia inducibility was tested by one to three premature stimuli and burst pacing. Longitudinal and transverse conduction velocity and dispersion of conduction were determined during pacing at a basic cycle length of 150 ms. Sirius red staining (collagen) was performed. As a result, in the RV of mice in the E, L, and EL groups, transverse conduction velocity was significantly increased and anisotropic ratio was significantly decreased compared with those values of mice in the C group. Anisotropic reentrant arrhythmias were induced in 52% of untreated mice and significantly reduced to 22%, 26%, and 16% in the E, L, and EL groups, respectively. Interstitial fibrosis was significantly decreased in both the RV and LV of all treated groups. Scattered patches of replacement fibrosis were found in 90% of untreated hearts, which were significantly reduced in the E, L, and EL groups. A strong correlation between the abundance of patchy fibrosis and arrhythmia inducibility was found. In conclusion, chronic RAAS inhibition limited aging-related interstitial fibrosis. The lower arrhythmogeneity of treated mice was directly correlated to the reduced amount of patchy fibrosis.
Collapse
Affiliation(s)
- Mera Stein
- Div. of Heart & Lungs, Dept. of Medical, Physiology, Univ. Medical Ctr. Utrecht, Yalelaan 50, 3584 CM Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Activation of the AT1 angiotensin II (Ang II) receptors has various effects including vasoconstriction, hypertrophy, and possibly hyperplasia of vascular smooth muscle cells and cardiomyocytes and increase in extracellular collagen matrix synthesis. These actions lead to the development of cardiovascular hypertrophy and fibrosis, as well as arterial stiffness, which are some key factors in the development of the cardiovascular and renal complications. In clinical studies, it has been shown that renin-angiotensin blockade has direct and specific implications in the evolution of heart failure, coronary disease, stroke, and hypertensive and diabetic renal disease. The beneficial cardiovascular and renal effects of blocking the renin-angiotensin-aldosterone system reported in numerous clinical trials may be at least partially related to the actions of these drugs on cardiovascular and renal fibrosis, and arterial stiffness. These effects are now well-established and lead the international medical societies to propose the use of the renin-angiotensin system (RAS) blockers as initial treatment (both angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers) in several cardiovascular, metabolic, and renal disorders such as hypertension, heart failure, and proteinuria.
Collapse
|
43
|
Abstract
Atrial fibrillation (AF) is the most common clinically encountered abnormal heart beat. It is associated with an increased risk of stroke and symptoms of heart failure. Current therapies are directed toward controlling the rate of ventricular activation and preventing strokes through anticoagulation. Attempts at suppressing the arrhythmia are often ineffective, in part because the underlying pathogenesis is poorly understood. Recently, structural and electrical remodeling has been shown to occur during AF. These changes involve alterations in gene regulation and help perpetuate the arrhythmia. Some signals for remodeling are have been identified. Moreover, AF is associated with oxidative stress, and this redox imbalance may contribute to the altered gene regulation. One likely mediator of this change in transcriptional regulation is the redox sensitive transcription factor, nuclear factor-kappaB (NF-kappaB). Recently, NF-kappaB has been shown to downregulate transcription of the cardiac sodium channel in response to oxidative stress. NF-kappaB may contribute to the regulation of other ion channels, transcription factors, or splicing factors altered in AF and may represent a therapeutic target in AF management.
Collapse
Affiliation(s)
- Ge Gao
- Section of Cardiology, University of Illinois at Chicago, and the Jesse Brown VA Medical Center, Chicago, Illinois 60612, USA
| | | |
Collapse
|
44
|
Genetic variation in angiotensin-converting enzyme-related pathways associated with sudden cardiac arrest risk. Heart Rhythm 2009; 6:1306-14. [PMID: 19716087 DOI: 10.1016/j.hrthm.2009.06.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Accepted: 06/03/2009] [Indexed: 01/13/2023]
Abstract
BACKGROUND Angiotensin-converting enzyme (ACE)-related pathways influence arrhythmias and sudden cardiac arrest (SCA) risk. OBJECTIVE The purpose of this study was to investigate whether genetic variations in ACE-related pathways are associated with SCA risk. Because these pathways are sex dependent and are influenced by estrogen, we examined these genotype-SCA associations in the full study population and tested for interaction with gender. METHODS In a population-based case-control study set in King County, Washington, 211 SCA cases (80% male; mean age 59 years,) and 730 age- and gender-matched controls of European descent were genotyped for 47 single nucleotide polymorphisms (SNPs) in eight genes (ACE, AGT, REN, AGTR1, AGTR2, ACE2, KNG1, BDKRB2). The association of SNPs and haplotypes with SCA risk was examined using logistic regression. RESULTS AGTR1 SNP rs1492099 (allele frequency 15%) was associated with decreased SCA risk (odds ratio [OR] 0.62, 95% confidence interval [CI] 0.4-0.9). Haplotype variation in AGTR2 was associated with SCA risk (global haplotype test P = .001), with haplotype 2 (allele frequency 27%) associated with increased risk (OR 1.26, 95% CI 1.1-1.5). There was interaction with gender on SCA risk for variation in KNG1 (interaction P value range 0.0004-0.017 for 6/8 SNPs). KNG1 SNP rs710448 (allele frequency 42%) was associated with decreased risk (OR 0.44, 95% CI 0.3-0.8) among women but not men. Other SNPs and haplotypes in the eight genes examined were not associated with SCA risk after multiple testing correction. CONCLUSION Variations in AGTR1 and AGTR2 are associated with SCA risk in a population-based case-control study. There was evidence of interaction with gender on SCA risk for variation in KNG1. These study findings, if replicated, suggest that variation in genes in ACE-related pathways influence SCA risk.
Collapse
|
45
|
Watanabe H, Kaiser DW, Makino S, MacRae CA, Ellinor PT, Wasserman BS, Kannankeril PJ, Donahue BS, Roden DM, Darbar D. ACE I/D polymorphism associated with abnormal atrial and atrioventricular conduction in lone atrial fibrillation and structural heart disease: implications for electrical remodeling. Heart Rhythm 2009; 6:1327-32. [PMID: 19648063 DOI: 10.1016/j.hrthm.2009.05.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2008] [Accepted: 05/11/2009] [Indexed: 12/19/2022]
Abstract
BACKGROUND The angiotensin-converting enzyme (ACE) gene contains a common polymorphism based on the insertion (I) or deletion (D) of a 287-bp intronic DNA fragment. The D allele is associated with higher ACE activity and thus higher angiotensin II levels. Angiotensin II stimulates cardiac fibrosis and conduction heterogeneity. OBJECTIVE The purpose of this study was to determine whether the ACE I/D polymorphism modulates cardiac electrophysiology. METHODS Three different cohorts of patients were studied: 69 patients with paroxysmal lone atrial fibrillation (AF), 151 patients with structural heart disease and no history of AF, and 161 healthy subjects without cardiovascular disease or AF. Patients taking drugs that affect cardiac conduction were excluded from the study. ECG parameters during sinus rhythm were compared among the ACE I/D genotypes. RESULTS The ACE I/D polymorphism was associated with the PR interval and heart block in the lone AF cohort. In multivariable linear regression models, the D allele was associated with longer PR interval in the lone AF and heart disease cohorts (12.0-ms and 7.1-ms increase per D allele, respectively). P-wave duration showed a similar trend, with increase in PR interval across ACE I/D genotypes in the lone AF and heart disease cohorts. CONCLUSION The ACE D allele is associated with electrical remodeling in patients with lone AF and in those with heart disease, but not in control subjects. ACE activity may play a role in cardiac remodeling after the development of AF and heart disease.
Collapse
Affiliation(s)
- Hiroshi Watanabe
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Shimoni Y, Emmett T, Schmidt R, Nygren A, Kargacin G. Sex-dependent impairment of cardiac action potential conduction in type 1 diabetic rats. Am J Physiol Heart Circ Physiol 2009; 296:H1442-50. [DOI: 10.1152/ajpheart.01150.2008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The incidence of diabetes mellitus is increasing. Cardiac dysfunction often develops, resulting in diverse arrhythmias. These arise from ion channel remodeling or from altered speed and pattern of impulse propagation. Few studies have investigated impulse propagation in the diabetic heart. We previously showed a reduced conduction reserve in the diabetic heart, with associated changes in intercellular gap junctions. The present study investigated whether these effects are sex specific. Hearts from control and streptozotocin-diabetic male and female rats were used. Optical mapping was performed with the voltage-sensitive dye di-4-ANEPPS, using Langendorff-perfused hearts. Isolated ventricular cells and tissue sections were used for immunofluorescent labeling of the gap junction protein connexin43 (Cx43). The gap junction uncoupler heptanol (0.75 mM) or elevated K+ (9 mM, to reduce cell excitability) produced significantly greater slowing of propagation in diabetic males than females. In ovariectomized diabetic females, 9 mM K+ slowed conduction significantly more than in nonovariectomized females. The subcellular redistribution (lateralization) of the gap junction protein Cx43 was smaller in diabetic females. Pretreatment of diabetic males with the angiotensin-converting enzyme inhibitor quinapril reduced Cx43 lateralization and the effects of 9 mM K+ on propagation. In conclusion, the slowing of cardiac impulse propagation in type 1 diabetes is smaller in female rats, partly due to the presence of female sex hormones. This difference is (partly) mediated by sex differences in activation of the cardiac renin-angiotensin system.
Collapse
|
47
|
Johnson LN, Koval M. Cross-talk between pulmonary injury, oxidant stress, and gap junctional communication. Antioxid Redox Signal 2009; 11:355-67. [PMID: 18816185 PMCID: PMC2933150 DOI: 10.1089/ars.2008.2183] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Gap junction channels interconnect several different types of cells in the lung, ranging from the alveolar epithelium to the pulmonary vasculature, each of which expresses a unique subset of gap junction proteins (connexins). Major lung functions regulated by gap junctional communication include coordination of ciliary beat frequency and inflammation. Gap junctions help enable the alveolus to regulate surfactant secretion as an integrated system, in which type I cells act as mechanical sensors that transmit calcium transients to type II cells. Thus, disruption of epithelial gap junctional communication, particularly during acute lung injury, can interfere with these processes and increase the severity of injury. Consistent with this, connexin expression is altered during lung injury, and connexin-deficiency has a negative impact on the injury response and lung-growth control. It has recently been shown that alcohol abuse is a significant risk factor associated with acute respiratory distress syndrome. Oxidant stress and hormone-signaling cascades in the lung induced by prolonged alcohol ingestion are discussed, as well as the effects of these pathways on connexin expression and function.
Collapse
Affiliation(s)
- Latoya N Johnson
- Division of Pulmonary, Allergy and Critical Care Medicine, and Emory Alcohol and Lung Biology Center, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
48
|
Li N, Dai DZ, Dai Y. CPU86017 and its isomers improve hypoxic pulmonary hypertension by attenuating increased ETA receptor expression and extracellular matrix accumulation. Naunyn Schmiedebergs Arch Pharmacol 2008; 378:541-52. [DOI: 10.1007/s00210-008-0309-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Accepted: 05/02/2008] [Indexed: 12/17/2022]
|
49
|
Shen XZ, Xiao HD, Li P, Lin CX, Billet S, Okwan-Duodu D, Adams JW, Bernstein EA, Xu Y, Fuchs S, Bernstein KE. New insights into the role of angiotensin-converting enzyme obtained from the analysis of genetically modified mice. J Mol Med (Berl) 2008; 86:679-84. [PMID: 18443752 DOI: 10.1007/s00109-008-0325-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 02/08/2008] [Accepted: 02/11/2008] [Indexed: 11/29/2022]
Abstract
Angiotensin-converting enzyme (ACE) has been well-recognized for its role in blood pressure regulation. ACE is made by many tissues, though it is most abundantly expressed on the luminal surface of vascular endothelium. ACE knockout mice show a profound phenotype with low blood pressure, but also with hemopoietic and developmental defects, which complicates understanding the biological functions of ACE in individual tissue types. Using a promoter-swapping strategy, several mouse lines with unique ACE tissue expression patterns were studied. These include mice with ACE expression in the liver (ACE 3/3), the heart (ACE 8/8), and macrophages (ACE 10/10). We also investigated mice with a selective inactivation of either the N- or C-terminal ACE catalytic domain. Our studies indicate that ACE plays a role in many other physiologic processes beyond simple blood pressure control.
Collapse
Affiliation(s)
- Xiao Z Shen
- Department of Pathology, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tissue specific expression of angiotensin converting enzyme: a new way to study an old friend. Int Immunopharmacol 2008; 8:171-6. [PMID: 18182222 DOI: 10.1016/j.intimp.2007.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2007] [Revised: 08/09/2007] [Accepted: 08/12/2007] [Indexed: 11/20/2022]
Abstract
Angiotensin-converting enzyme (ACE) plays a central role in blood pressure regulation by producing the vasoconstrictor angiotensin II. When ACE knockout mice were studied, they presented with a complicated phenotype, including cardiovascular, reproductive, hematologic and developmental defects. The complexity of an ACE knockout mouse emphasizes the advantages and disadvantages of the classic knockout strategy. An animal lacking all ACE is very different from a wild type animal, and can be modeled as representing an extreme phenotype. To understand the role of ACE in a tissue and organ specific fashion, our group used targeted homologous recombination to create mouse models in which a promoter swapping strategy results in very restricted tissue patterns of ACE expression. Mice with ACE expression only in the heart, termed ACE 8/8 mice, present with atria enlargement and electrical conduction defects, but normal ventricular function. Mice with ACE expression only in monocytes and macrophages, termed ACE 10/10 mice, have a marked resistance to the growth of melanoma due to an enhanced immune response characterized by increased tumor specific CD8+ T cells and increased proinflammatory cytokines. These mice may define a new means of augmenting the immune response, potentially useful in human clinical situations. The promoter swapping strategy permits scientific investigation of questions unapproachable by other experimental approaches.
Collapse
|