1
|
Zhao X, Liu Y, Han X, Wang X, Qu C, Liu X, Yang B. Dapagliflozin attenuates the vulnerability to atrial fibrillation in rats with lipopolysaccharide-induced myocardial injury. Int Immunopharmacol 2023; 125:111038. [PMID: 38149574 DOI: 10.1016/j.intimp.2023.111038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/24/2023] [Accepted: 10/07/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND Oxidative stress is an essential component participating in the development and maintenance of atrial fibrillation (AF). Dapagliflozin, a SGLT2 inhibitor, has been shown to exert cardioprotective effects by ameliorating oxidative stress in multiple heart disease models. However, its potential to attenuate lipopolysaccharide (LPS)-induced myocardial injury in rats remains unknown. AIM This study aims to investigate the role of dapagliflozin in LPS-induced myocardial injury and the potential mechanisms involved. METHODS Rats were intraperitoneally administered LPS to induce sepsis-like condition. The intervention was conducted with intraperitoneal injection of dapagliflozin or saline 1 h in advance. The effects of dapagliflozin were detected by electrophysiological recordings, western blot, qPCR, ELISA, HE staining, immunohistochemistry and fluorescence. We further validated the mechanism in vitro using HL-1 cells. RESULTS Dapagliflozin significantly improved LPS-induced myocardial injury, reduced susceptibility to AF, and mitigated atrial tissue inflammatory cell infiltration and atrial myocyte apoptosis. These were correlated with the Nrf2/HO-1 signaling pathway, which subsequently reduced oxidative stress. Subsequently, we used a specific inhibitor of the Nrf2/HO-1 pathway in vitro, reversed the anti-oxidative stress effects of dapagliflozin on HL-1 cells, further confirming the Nrf2/HO-1 pathway's pivotal role in dapagliflozin-mediated cardioprotection. CONCLUSION Dapagliflozin ameliorated myocardial injury and susceptibility to AF induced by LPS through anti-oxidative stress, which relied on upregulation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yating Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xiukun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| |
Collapse
|
2
|
Zong YJ, Liu XZ, Tu L, Sun Y. Cytomembrane Trafficking Pathways of Connexin 26, 30, and 43. Int J Mol Sci 2023; 24:10349. [PMID: 37373495 DOI: 10.3390/ijms241210349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The connexin gene family is the most prevalent gene that contributes to hearing loss. Connexins 26 and 30, encoded by GJB2 and GJB6, respectively, are the most abundantly expressed connexins in the inner ear. Connexin 43, which is encoded by GJA1, appears to be widely expressed in various organs, including the heart, skin, the brain, and the inner ear. The mutations that arise in GJB2, GJB6, and GJA1 can all result in comprehensive or non-comprehensive genetic deafness in newborns. As it is predicted that connexins include at least 20 isoforms in humans, the biosynthesis, structural composition, and degradation of connexins must be precisely regulated so that the gap junctions can properly operate. Certain mutations result in connexins possessing a faulty subcellular localization, failing to transport to the cell membrane and preventing gap junction formation, ultimately leading to connexin dysfunction and hearing loss. In this review, we provide a discussion of the transport models for connexin 43, connexins 30 and 26, mutations affecting trafficking pathways of these connexins, the existing controversies in the trafficking pathways of connexins, and the molecules involved in connexin trafficking and their functions. This review can contribute to a new way of understanding the etiological principles of connexin mutations and finding therapeutic strategies for hereditary deafness.
Collapse
Affiliation(s)
- Yan-Jun Zong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiao-Zhou Liu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Tu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| |
Collapse
|
3
|
Long non-coding RNA CCRR controls cardiac conduction via regulating intercellular coupling. Nat Commun 2018; 9:4176. [PMID: 30301979 PMCID: PMC6177441 DOI: 10.1038/s41467-018-06637-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 09/10/2018] [Indexed: 12/31/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as a new class of gene expression regulators playing key roles in many biological and pathophysiological processes. Here, we identify cardiac conduction regulatory RNA (CCRR) as an antiarrhythmic lncRNA. CCRR is downregulated in a mouse model of heart failure (HF) and in patients with HF, and this downregulation slows cardiac conduction and enhances arrhythmogenicity. Moreover, CCRR silencing induces arrhythmias in healthy mice. CCRR overexpression eliminates these detrimental alterations. HF or CCRR knockdown causes destruction of intercalated discs and gap junctions to slow longitudinal cardiac conduction. CCRR overexpression improves cardiac conduction by blocking endocytic trafficking of connexin43 (Cx43) to prevent its degradation via binding to Cx43-interacting protein CIP85, whereas CCRR silence does the opposite. We identified the functional domain of CCRR, which can reproduce the functional roles and pertinent molecular events of full-length CCRR. Our study suggests CCRR replacement a potential therapeutic approach for pathological arrhythmias.
Collapse
|
4
|
Shvedova M, Anfinogenova Y, Popov SV, Atochin DN. Connexins and Nitric Oxide Inside and Outside Mitochondria: Significance for Cardiac Protection and Adaptation. Front Physiol 2018; 9:479. [PMID: 29867537 PMCID: PMC5964197 DOI: 10.3389/fphys.2018.00479] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/16/2018] [Indexed: 12/27/2022] Open
Abstract
Irreversible myocardial damage happens in the presence of prolonged and severe ischemia. Several phenomena protect the heart against myocardial infarction and other adverse outcomes of ischemia and reperfusion (IR), namely: hibernation related to stunned myocardium, ischemic preconditioning (IPC), ischemic post-conditioning, and their pharmacological surrogates. Ischemic preconditioning consists in the induction of a brief IR to reduce damage of the tissue caused by prolonged and severe ischemia. Nitric oxide (NO) signaling plays an essential role in IPC. Nitric oxide-sensitive guanylate cyclase/cyclic guanosine-3′,5′-monophosphate (cGMP)-dependent protein kinase type I-signaling pathway protects against the IR injury during myocardial infarction. Mitochondrial ATP-sensitive and Ca2+-activated K+ channels are involved in NO-mediated signaling in IPC. Independently of the cGMP-mediated induction of NO production, S-nitrosation represents a regulatory molecular mechanism similar to phosphorylation and is essential for IPC. Unlike conditioning phenomena, the mechanistic basis of myocardial stunning and hibernation remains poorly understood. In this review article, we hypothesize that the disruption of electrical syncytium of the myocardium may underly myocardial stunning and hibernation. Considering that the connexins are the building blocks of gap junctions which represent primary structural basis of electrical syncytium, we discuss data on the involvement of connexins into myocardial conditioning, stunning, and hibernation. We also show how NO-mediated signaling is involved in myocardial stunning and hibernation. Connexins represent an essential element of adaptation phenomena of the heart at the level of both the cardio- myocytes and the mitochondria. Nitric oxide targets mitochondrial connexins which may affect electrical syncytium continuum in the heart. Mitochondrial connexins may play an essential role in NO-dependent mechanisms of myocardial adaptation to ischemia.
Collapse
Affiliation(s)
- Maria Shvedova
- Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Yana Anfinogenova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia.,RASA Center, National Research Tomsk Polytechnic University, Tomsk, Russia
| | - Sergey V Popov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Dmitriy N Atochin
- Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,RASA Center, National Research Tomsk Polytechnic University, Tomsk, Russia
| |
Collapse
|
5
|
Kohutova J, Elsnicova B, Holzerova K, Neckar J, Sebesta O, Jezkova J, Vecka M, Vebr P, Hornikova D, Szeiffova Bacova B, Egan Benova T, Hlavackova M, Tribulova N, Kolar F, Novakova O, Zurmanova JM. Anti-arrhythmic Cardiac Phenotype Elicited by Chronic Intermittent Hypoxia Is Associated With Alterations in Connexin-43 Expression, Phosphorylation, and Distribution. Front Endocrinol (Lausanne) 2018; 9:789. [PMID: 30740090 PMCID: PMC6357219 DOI: 10.3389/fendo.2018.00789] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/14/2018] [Indexed: 12/31/2022] Open
Abstract
Remodeling of the cellular distribution of gap junctions formed mainly by connexin-43 (Cx43) can be related to the increased incidence of cardiac arrhythmias. It has been shown that adaptation to chronic intermittent hypobaric hypoxia (IHH) attenuates the incidence and severity of ischemic and reperfusion ventricular arrhythmias and increases the proportion of anti-arrhythmic n-3 polyunsaturated fatty acids (n-3 PUFA) in heart phospholipids. Wistar rats were exposed to simulated IHH (7,000 m, 8-h/day, 35 exposures) and compared with normoxic controls (N). Cx43 expression, phosphorylation, localization and n-3 PUFA proportion were analyzed in left ventricular myocardium. Compared to N, IHH led to higher expression of total Cx43, its variant phosphorylated at Ser368 [p-Cx43(Ser368)], which maintains "end to end" communication, as well as p-Cx43(Ser364/365), which facilitates conductivity. By contrast, expression of non-phosphorylated Cx43 and p-Cx43(Ser278/289), attenuating intercellular communication, was lower in IHH than in N. IHH also resulted in increased expression of protein kinase A and protein kinase G while casein kinase 1 did not change compared to N. In IHH group, which exhibited reduced incidence of ischemic ventricular arrhythmias, Cx43 and p-Cx43(Ser368) were more abundant at "end to end" gap junctions than in N group and this difference was preserved after acute regional ischemia (10 min). We further confirmed higher n-3 PUFA proportion in heart phospholipids after adaptation to IHH, which was even further increased by ischemia. Our results suggest that adaptation to IHH alters expression, phosphorylation and distribution of Cx43 as well as cardioprotective n-3PUFA proportion suggesting that the anti-arrhythmic phenotype elicited by IHH can be at least partly related to the stabilization of the "end to end" conductivity between cardiomyocytes during brief ischemia.
Collapse
Affiliation(s)
- Jana Kohutova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - Barbara Elsnicova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - Kristyna Holzerova
- Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jan Neckar
- Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Ondrej Sebesta
- Laboratory of Confocal and Fluorescence Microscopy, Faculty of Science, Charles University, Prague, Czechia
| | - Jana Jezkova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - Marek Vecka
- 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czechia
| | - Pavel Vebr
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - Daniela Hornikova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - Barbara Szeiffova Bacova
- Center of Experimental Medicine of the Slovak Academy of Sciences, Institute for Heart Research, Bratislava, Slovakia
| | - Tamara Egan Benova
- Center of Experimental Medicine of the Slovak Academy of Sciences, Institute for Heart Research, Bratislava, Slovakia
| | - Marketa Hlavackova
- Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Narcis Tribulova
- Center of Experimental Medicine of the Slovak Academy of Sciences, Institute for Heart Research, Bratislava, Slovakia
| | - Frantisek Kolar
- Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Olga Novakova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
- Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jitka M. Zurmanova
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
- *Correspondence: Jitka M. Zurmanova
| |
Collapse
|
6
|
Hibernating substrate of ventricular tachycardia: a three-dimensional metabolic and electro-anatomic assessment. J Interv Card Electrophysiol 2017; 48:247-254. [DOI: 10.1007/s10840-016-0219-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 12/14/2016] [Indexed: 10/20/2022]
|
7
|
Sukhacheva TV, Chudinovskikh YA, Eremeeva MV, Serov RA, Bockeria LA. Proliferative Potential of Cardiomyocytes in Hypertrophic Cardiomyopathy: Correlation with Myocardial Remodeling. Bull Exp Biol Med 2016; 162:160-169. [PMID: 27882462 DOI: 10.1007/s10517-016-3566-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Indexed: 01/07/2023]
Affiliation(s)
- T V Sukhacheva
- A. N. Bakulev Scientific Center for Cardiovascular Surgery, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - Yu A Chudinovskikh
- A. N. Bakulev Scientific Center for Cardiovascular Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - M V Eremeeva
- A. N. Bakulev Scientific Center for Cardiovascular Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - R A Serov
- A. N. Bakulev Scientific Center for Cardiovascular Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| | - L A Bockeria
- A. N. Bakulev Scientific Center for Cardiovascular Surgery, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
8
|
Chen Y, Xu J, Liao H, Ma Z, Zhang Y, Chen H, Huang Z, Hu J. Prostaglandin E2 and Connexin 43 crosstalk in the osteogenesis induced by extracorporeal shockwave. Med Hypotheses 2016; 94:123-5. [PMID: 27515217 DOI: 10.1016/j.mehy.2016.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 07/12/2016] [Accepted: 07/18/2016] [Indexed: 02/05/2023]
Abstract
As a type of mechanical stimulation, extracorporeal shockwave (ESW) has been widely used in the clinic to treat bone fracture delayed union and non-unions. A large number of studies have shown beneficial effects of ESW in promoting fracture healing by inducing bone regeneration; however, the underlying mechanisms remain unclear. ESW has been shown to induce the production of prostaglandin E2 (PGE2), which is essential for gap junction intercellular communication in response to mechanical stress. Among the 19 known gap junction subunits, connexin43 (Cx43) is the most prevalent for mediating the response of mechanical stress. However, to our knowledge, the effect of ESW on Cx43 expression has not been reported before. Herein, we propose that a crosstalk between PGE2 and Cx43 is involved in the enhancement of osteogenesis induced by ESW. We review the currently available data to propose an unrevealed, but important mechanism via which ESW treatment affects osteogenic differentiation of bone marrow stromal cells.
Collapse
Affiliation(s)
- Youbin Chen
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jiankun Xu
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China; Department of Orthopedics and Traumatology, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Haojie Liao
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zebin Ma
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yuantao Zhang
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Hongjiang Chen
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhonglian Huang
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jun Hu
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
9
|
Wasson S, Reddy HK, Dohrmann ML. Current Perspectives of Electrical Remodeling and Its Therapeutic Implications. J Cardiovasc Pharmacol Ther 2016; 9:129-44. [PMID: 15309249 DOI: 10.1177/107424840400900208] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Electrical remodeling involves alterations in the electrophysiologic milieu of myocardium in various disease states, such as ventricular hypertrophy, heart failure, atrial tachyarrhythmias, myocardial ischemia, and infarction that are associated with cardiac arrhythmias. Although research in this area dates back to early part of the 19th century, we still lack the exact knowledge of ionic remodeling, the role of various genes and channel proteins, and their relevance for the newer antiarrhythmic therapies. Structural remodeling may also have an impact on the electrical remodeling process, although differences in both structural and electrical remodeling are associated with different disease states. Various electrophysiologic, cellular, and structural alterations, including anisotropic conduction, increased intracellular calcium levels, and gap junction remodeling predispose to increased dispersion of action potential duration and refractoriness. This constitutes a favorable substrate for early and late afterdepolarizations and reentrant arrhythmias. Studying the role of ionic remodeling in the initiation and propagation of cardiac arrhythmias has significant relevance for developing newer antiarrhythmic therapies, for identifying patients at risk of developing fatal arrhythmias, and for implementing effective preventive measures. Further research is required to understand the specific effects of individual ion channel remodeling, to understand the signal transduction mechanisms, and to address whether detrimental effects of electrical remodeling can be altered.
Collapse
Affiliation(s)
- Sanjeev Wasson
- Division of Cardiology, University of Missouri Hospital, Columbia, Missouri 65212, USA
| | | | | |
Collapse
|
10
|
Sovari AA. Cellular and Molecular Mechanisms of Arrhythmia by Oxidative Stress. Cardiol Res Pract 2016; 2016:9656078. [PMID: 26981310 PMCID: PMC4770129 DOI: 10.1155/2016/9656078] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/10/2016] [Indexed: 12/25/2022] Open
Abstract
Current therapies for arrhythmia using ion channel blockade, catheter ablation, or an implantable cardioverter defibrillator have limitations, and it is important to search for new antiarrhythmic therapeutic targets. Both atrial fibrillation and heart failure, a condition with increased arrhythmic risk, are associated with excess amount of reactive oxygen species (ROS). There are several possible ways for ROS to induce arrhythmia. ROS can cause focal activity and reentry. ROS alter multiple cardiac ionic currents. ROS promote cardiac fibrosis and impair gap junction function, resulting in reduced myocyte coupling and facilitation of reentry. In order to design effective antioxidant drugs for treatment of arrhythmia, it is essential to explore the molecular mechanisms by which ROS exert these arrhythmic effects. Activation of Ca(2+)/CaM-dependent kinase II, c-Src tyrosine kinase, protein kinase C, and abnormal splicing of cardiac sodium channels are among the recently discovered molecular mechanisms of ROS-induced arrhythmia.
Collapse
Affiliation(s)
- Ali A. Sovari
- Cardiac Electrophysiology Section, Heart Institute, Cedars Sinai Medical Center, 127 S. San Vicente Boulevard, A3308, Los Angeles, CA 90048, USA
| |
Collapse
|
11
|
Reduced expression of adherens and gap junction proteins can have a fundamental role in the development of heart failure following cardiac hypertrophy in rats. Exp Mol Pathol 2015; 100:167-76. [PMID: 26708424 DOI: 10.1016/j.yexmp.2015.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 12/12/2015] [Accepted: 12/15/2015] [Indexed: 10/22/2022]
Abstract
Hypertension causes cardiac hypertrophy, cardiac dysfunction and heart failure (HF). The mechanisms implicated in the transition from compensated to decompensated cardiac hypertrophy are not fully understood. This study was aimed to investigate whether alterations in the expression of intercalated disk proteins could contribute to the transition of compensated cardiac hypertrophy to dilated heart development that culminates in HF. Male rats were submitted to abdominal aortic constriction and at 90 days post surgery (dps), three groups were observed: sham-operated animals (controls), animals with hypertrophic hearts (HH) and animals with hypertrophic + dilated hearts (HD). Blood pressure was evaluated. The hearts were collected and Western blot and immunofluorescence were performed to desmoglein-2, desmocollin-2, N-cadherin, plakoglobin, Bcatenin, and connexin-43. Cardiac systolic function was evaluated using the Vevo 2100 ultrasound system. Data were considered significant when p b 0.05. Seventy percent of the animals presented with HH and 30% were HD at 90 dps. The blood pressure increased in both groups. The amount of desmoglein-2 and desmocollin-2 expression was increased in both groups and no difference was observed in either group. The expression of N-cadherin, plakoglobin and B-catenin increased in the HHgroup and decreased in the HDgroup; and connexin-43 decreased only in theHDgroup. Therewas no difference between the ejection fraction and fractional shortening at 30 and 60 dps; however, they were decreased in the HD group at 90 dps. We found that while some proteins have increased expression accompanied by the increase in the cell volume associated with preserved systolic cardiac function in theHHgroup, these same proteins had decreased expression evenwithout significant reduction in the cell volume associated with decreased systolic cardiac function in HD group. The increased expression of desmoglein-2 and desmocollin-2 in both the HH and HD groups could work as a protective compensatory mechanism, helping tomaintain the dilated heart.We can hypothesize that inappropriate intercellular mechanical and electrical coupling associated with necrosis and/or apoptosis are important factors contributing to the transition to HF.
Collapse
|
12
|
Yang KC, Kyle JW, Makielski JC, Dudley SC. Mechanisms of sudden cardiac death: oxidants and metabolism. Circ Res 2015; 116:1937-55. [PMID: 26044249 PMCID: PMC4458707 DOI: 10.1161/circresaha.116.304691] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 02/09/2015] [Indexed: 02/07/2023]
Abstract
Ventricular arrhythmia is the leading cause of sudden cardiac death (SCD). Deranged cardiac metabolism and abnormal redox state during cardiac diseases foment arrhythmogenic substrates through direct or indirect modulation of cardiac ion channel/transporter function. This review presents current evidence on the mechanisms linking metabolic derangement and excessive oxidative stress to ion channel/transporter dysfunction that predisposes to ventricular arrhythmias and SCD. Because conventional antiarrhythmic agents aiming at ion channels have proven challenging to use, targeting arrhythmogenic metabolic changes and redox imbalance may provide novel therapeutics to treat or prevent life-threatening arrhythmias and SCD.
Collapse
Affiliation(s)
- Kai-Chien Yang
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.)
| | - John W Kyle
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.)
| | - Jonathan C Makielski
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.).
| | - Samuel C Dudley
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.).
| |
Collapse
|
13
|
Sialyltransferase7A, a Klf4-responsive gene, promotes cardiomyocyte apoptosis during myocardial infarction. Basic Res Cardiol 2015; 110:28. [PMID: 25860962 DOI: 10.1007/s00395-015-0484-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 03/15/2015] [Accepted: 03/31/2015] [Indexed: 01/01/2023]
Abstract
Myocardial infarction (MI) is one major cause of heart failure through its induction of cardiomyocyte death. However, the molecular mechanisms associated with MI-induced cardiomyocyte apoptosis in the context of sialylation of heart are not yet understood. In this study, we found that sialyltransferase7A (Siat7A), one of the members of sialyltransferase family, was significantly increased in the ischemic myocardium, as well as in the human cardiomyocyte cell line AC16 under hypoxic condition. The Sialyl-Tn antigen (Neu5Acα2-6GalNAc-O-Ser/Thr) synthesized by Siat7A also increased in the AC16 cardiomyocytes following hypoxic stimulus. Increased Siat7A promoted cardiomyocyte apoptosis. The knockdown of Siat7A expression reduced cardiomyocyte apoptosis in both of vivo and vitro. Furthermore, the decreased extracellular signal-regulated kinase ERK1 and ERK2 (ERK1/2) activity was involved in the Siat7A-induced cardiomyocyte apoptosis. Notably, we showed that Krüppel-like factor 4 (Klf4), one of the transcription factors, specifically bound to the Siat7A promoter by ChIP assays. Deletion and mutagenesis analysis identified that Klf4 could transactivate the Siat7A promoter region (nt -655 to -636 bp). The upregulated Siat7A expression, which was paralleled by the increased Klf4 in the ischemic myocardium, contributed to cardiomyocyte apoptosis. Our study suggests Siat7A could be a valuable target for developing treatments for MI patients.
Collapse
|
14
|
Yang KC, Bonini MG, Dudley SC. Mitochondria and arrhythmias. Free Radic Biol Med 2014; 71:351-361. [PMID: 24713422 PMCID: PMC4096785 DOI: 10.1016/j.freeradbiomed.2014.03.033] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 03/21/2014] [Accepted: 03/24/2014] [Indexed: 12/31/2022]
Abstract
Mitochondria are essential to providing ATP, thereby satisfying the energy demand of the incessant electrical activity and contractile action of cardiac muscle. Emerging evidence indicates that mitochondrial dysfunction can adversely affect cardiac electrical functioning by impairing the intracellular ion homeostasis and membrane excitability through reduced ATP production and excessive reactive oxygen species (ROS) generation, resulting in increased propensity to cardiac arrhythmias. In this review, the molecular mechanisms linking mitochondrial dysfunction to cardiac arrhythmias are discussed with an emphasis on the impact of increased mitochondrial ROS on the cardiac ion channels and transporters that are critical to maintaining normal electromechanical functioning of the cardiomyocytes. The potential of using mitochondria-targeted antioxidants as a novel antiarrhythmia therapy is highlighted.
Collapse
Affiliation(s)
- Kai-Chien Yang
- Lifespan Cardiovascular Institute, Providence VA Medical Center, and Brown University, Providence, RI 02903, USA
| | - Marcelo G Bonini
- Department of Medicine/Cardiology, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pathology, and University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Samuel C Dudley
- Lifespan Cardiovascular Institute, Providence VA Medical Center, and Brown University, Providence, RI 02903, USA.
| |
Collapse
|
15
|
Lionetti V, Matteucci M, Ribezzo M, Di Silvestre D, Brambilla F, Agostini S, Mauri P, Padeletti L, Pingitore A, Delsedime L, Rinaldi M, Recchia FA, Pucci A. Regional mapping of myocardial hibernation phenotype in idiopathic end-stage dilated cardiomyopathy. J Cell Mol Med 2014; 18:396-414. [PMID: 24444256 PMCID: PMC3955147 DOI: 10.1111/jcmm.12198] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 10/28/2013] [Indexed: 01/18/2023] Open
Abstract
Myocardial hibernation (MH) is a well-known feature of human ischaemic cardiomyopathy (ICM), whereas its presence in human idiopathic dilated cardiomyopathy (DCM) is still controversial. We investigated the histological and molecular features of MH in left ventricle (LV) regions of failing DCM or ICM hearts. We examined failing hearts from DCM (n = 11; 41.9 ± 5.45 years; left ventricle-ejection fraction (LV-EF), 18 ± 3.16%) and ICM patients (n = 12; 58.08 ± 1.7 years; LVEF, 21.5 ± 6.08%) undergoing cardiac transplantation, and normal donor hearts (N, n = 8). LV inter-ventricular septum (IVS) and antero-lateral free wall (FW) were transmurally (i.e. sub-epicardial, mesocardial and sub-endocardial layers) analysed. LV glycogen content was shown to be increased in both DCM and ICM as compared with N hearts (P < 0.001), with a U-shaped transmural distribution (lower values in mesocardium). Capillary density was homogenously reduced in both DCM and ICM as compared with N (P < 0.05 versus N), with a lower decrease independent of the extent of fibrosis in sub-endocardial and sub-epicardial layers of DCM as compared with ICM. HIF1-α and nestin, recognized ischaemic molecular hallmarks, were similarly expressed in DCM-LV and ICM-LV myocardium. The proteomic profile was overlapping by ˜50% in DCM and ICM groups. Morphological and molecular features of MH were detected in end-stage ICM as well as in end-stage DCM LV, despite epicardial coronary artery patency and lower fibrosis in DCM hearts. Unravelling the presence of MH in the absence of coronary stenosis may be helpful to design a novel approach in the clinical management of DCM.
Collapse
Affiliation(s)
- Vincenzo Lionetti
- Laboratory of Medical Science, Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy; Fondazione CNR-Regione Toscana "G. Monasterio", Pisa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Boulaksil M, Winckels SK, Engelen MA, Stein M, van Veen TA, Jansen JA, Linnenbank AC, Bierhuizen MF, Groenewegen WA, van Oosterhout MF, Kirkels JH, de Jonge N, Varró A, Vos MA, de Bakker JM, van Rijen HV. Heterogeneous Connexin43 distribution in heart failure is associated with dispersed conduction and enhanced susceptibility to ventricular arrhythmias. Eur J Heart Fail 2014; 12:913-21. [DOI: 10.1093/eurjhf/hfq092] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Mohamed Boulaksil
- Interuniversity Cardiology Institute of The Netherlands; Utrecht The Netherlands
- Division of Heart and Lungs, Department of Medical Physiology; University Medical Center Utrecht; Yalelaan 50 3584 CM Utrecht The Netherlands
| | - Stephan K.G. Winckels
- Division of Heart and Lungs, Department of Medical Physiology; University Medical Center Utrecht; Yalelaan 50 3584 CM Utrecht The Netherlands
- Division Laboratory, Department of Pathology; University Medical Center Utrecht; Utrecht The Netherlands
| | - Markus A. Engelen
- Division of Heart and Lungs, Department of Medical Physiology; University Medical Center Utrecht; Yalelaan 50 3584 CM Utrecht The Netherlands
- Department of Cardiology and Angiology; Hospital of the University of Muenster; Muenster Germany
| | - Mèra Stein
- Division of Heart and Lungs, Department of Medical Physiology; University Medical Center Utrecht; Yalelaan 50 3584 CM Utrecht The Netherlands
- Division of Heart and Lungs, Department of Cardiology; University Medical Center Utrecht; Utrecht The Netherlands
| | - Toon A.B. van Veen
- Division of Heart and Lungs, Department of Medical Physiology; University Medical Center Utrecht; Yalelaan 50 3584 CM Utrecht The Netherlands
| | - John A. Jansen
- Division of Heart and Lungs, Department of Medical Physiology; University Medical Center Utrecht; Yalelaan 50 3584 CM Utrecht The Netherlands
| | - André C. Linnenbank
- Interuniversity Cardiology Institute of The Netherlands; Utrecht The Netherlands
- Heart Failure Research Center; Academic Medical Center; Amsterdam The Netherlands
| | - Marti F.A. Bierhuizen
- Division of Heart and Lungs, Department of Medical Physiology; University Medical Center Utrecht; Yalelaan 50 3584 CM Utrecht The Netherlands
| | - W. Antoinette Groenewegen
- Division of Heart and Lungs, Department of Medical Physiology; University Medical Center Utrecht; Yalelaan 50 3584 CM Utrecht The Netherlands
| | | | - Johannes H. Kirkels
- Division of Heart and Lungs, Department of Cardiology; University Medical Center Utrecht; Utrecht The Netherlands
| | - Nicolaas de Jonge
- Division of Heart and Lungs, Department of Cardiology; University Medical Center Utrecht; Utrecht The Netherlands
| | - András Varró
- Department of Pharmacology and Pharmacotherapy; University of Szeged; Szeged Hungary
- Division for Cardiovascular Pharmacology; Hungarian Academy of Sciences; Szeged Hungary
| | - Marc A. Vos
- Division of Heart and Lungs, Department of Medical Physiology; University Medical Center Utrecht; Yalelaan 50 3584 CM Utrecht The Netherlands
| | - Jacques M.T. de Bakker
- Interuniversity Cardiology Institute of The Netherlands; Utrecht The Netherlands
- Division of Heart and Lungs, Department of Medical Physiology; University Medical Center Utrecht; Yalelaan 50 3584 CM Utrecht The Netherlands
- Heart Failure Research Center; Academic Medical Center; Amsterdam The Netherlands
| | - Harold V.M. van Rijen
- Division of Heart and Lungs, Department of Medical Physiology; University Medical Center Utrecht; Yalelaan 50 3584 CM Utrecht The Netherlands
| |
Collapse
|
17
|
Zhou J, Shu Y, Lü SH, Li JJ, Sun HY, Tang RY, Duan CM, Wang Y, Lin QX, Mou YC, Li X, Wang CY. The spatiotemporal development of intercalated disk in three-dimensional engineered heart tissues based on collagen/matrigel matrix. PLoS One 2013; 8:e81420. [PMID: 24260578 PMCID: PMC3829928 DOI: 10.1371/journal.pone.0081420] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 10/12/2013] [Indexed: 01/09/2023] Open
Abstract
Intercalated disk (ID), which electromechanically couples cardiomyocytes into a functional syncitium, is closely related to normal morphology and function of engineered heart tissues (EHTs), but the development mode of ID in the three-dimensional (3D) EHTs is still unclear. In this study, we focused on the spatiotemporal development of the ID in the EHTs constructed by mixing neonatal rat cardiomyocytes with collagen/Matrigel, and investigated the effect of 3D microenvironment provided by collagen/Matrigel matrix on the formation of ID. By histological and immmunofluorescent staining, the spatiotemporal distribution of ID-related junctions was detected. Furthermore, the ultra-structures of the ID in different developmental stages were observed under transmission electron microscope. In addition, the expression of the related proteins was quantitatively analyzed. The results indicate that accompanying the re-organization of cardiomyocytes in collagen/Matrigel matrix, the proteins of adherens junctions, desmosomes and gap junctions redistributed from diffused distribution to intercellular regions to form an integrated ID. The adherens junction and desmosome which are related with mechanical connection appeared earlier than gap junction which is essential for electrochemical coupling. These findings suggest that the 3D microenvironment based on collagen/Matrigel matrix could support the ordered assembly of the ID in EHTs and have implications for comprehending the ordered and coordinated development of ID during the functional organization of EHTs.
Collapse
Affiliation(s)
- Jin Zhou
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Yao Shu
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Shuang-Hong Lü
- Laboratory of Oncology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Jun-Jie Li
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Hong-Yu Sun
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Rong-Yu Tang
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Cui-Mi Duan
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Yan Wang
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Qiu-Xia Lin
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Yong-Chao Mou
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Xia Li
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Chang-Yong Wang
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
- * E-mail:
| |
Collapse
|
18
|
Zhang X, Wang Q, Gablaski B, Zhang X, Lucchesi P, Zhao Y. A microdevice for studying intercellular electromechanical transduction in adult cardiac myocytes. LAB ON A CHIP 2013; 13:3090-7. [PMID: 23753064 PMCID: PMC3770274 DOI: 10.1039/c3lc50414j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Intercellular electromechanical transduction in adult cardiac myocytes plays an important role in regulating heart function. The efficiency of intercellular electromechanical transduction has so far been investigated only to a limited extent, which is largely due to the lack of appropriate tools that can quantitatively assess the contractile performance of interconnected adult cardiac myocytes. In this paper we report a microengineered device that is capable of applying electrical stimulation to the selected adult cardiac myocyte in a longitudinally connected cell doublet and quantifying the intercellular electromechanical transduction by measuring the contractile performance of stimulated and un-stimulated cells in the same doublet. The capability of applying selective electrical stimulation on only one cell in the doublet is validated by examining cell contractile performance while blocking the intercellular communication. Quantitative assessment of cell contractile performance in isolated adult cardiac myocytes is performed by measuring the change in cell length. The proof-of-concept assessment of gap junction performance shows that the device is useful in studying the efficiency of gap junctions in adult cardiac myocytes, which is most relevant to the synchronized pumping performance of native myocardium. Collectively, this work provides a quantitative tool for studying intercellular electromechanical transduction and is expected to develop a comprehensive understanding of the role of intercellular communication in various heart diseases.
Collapse
Affiliation(s)
- Xu Zhang
- Laboratory for Biomedical Microsystems, Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210 USA
| | - Qian Wang
- Laboratory for Biomedical Microsystems, Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210 USA
| | - Brian Gablaski
- Center for Cardiovascular and Pulmonary Research, Nationwide Children’s Hospital, Columbus, OH, 43205 USA
| | - Xiaojin Zhang
- Center for Cardiovascular and Pulmonary Research, Nationwide Children’s Hospital, Columbus, OH, 43205 USA
| | - Pamela Lucchesi
- Center for Cardiovascular and Pulmonary Research, Nationwide Children’s Hospital, Columbus, OH, 43205 USA
| | - Yi Zhao
- Laboratory for Biomedical Microsystems, Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210 USA
| |
Collapse
|
19
|
King JH, Huang CLH, Fraser JA. Determinants of myocardial conduction velocity: implications for arrhythmogenesis. Front Physiol 2013; 4:154. [PMID: 23825462 PMCID: PMC3695374 DOI: 10.3389/fphys.2013.00154] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/10/2013] [Indexed: 12/19/2022] Open
Abstract
Slowed myocardial conduction velocity (θ) is associated with an increased risk of re-entrant excitation, predisposing to cardiac arrhythmia. θ is determined by the ion channel and physical properties of cardiac myocytes and by their interconnections. Thus, θ is closely related to the maximum rate of action potential (AP) depolarization [(dV/dt)max], as determined by the fast Na+ current (INa); the axial resistance (ra) to local circuit current flow between cells; their membrane capacitances (cm); and to the geometrical relationship between successive myocytes within cardiac tissue. These determinants are altered by a wide range of pathophysiological conditions. Firstly, INa is reduced by the impaired Na+ channel function that arises clinically during heart failure, ischemia, tachycardia, and following treatment with class I antiarrhythmic drugs. Such reductions also arise as a consequence of mutations in SCN5A such as those occurring in Lenègre disease, Brugada syndrome (BrS), sick sinus syndrome, and atrial fibrillation (AF). Secondly, ra, may be increased due to gap junction decoupling following ischemia, ventricular hypertrophy, and heart failure, or as a result of mutations in CJA5 found in idiopathic AF and atrial standstill. Finally, either ra or cm could potentially be altered by fibrotic change through the resultant decoupling of myocyte–myocyte connections and coupling of myocytes with fibroblasts. Such changes are observed in myocardial infarction and cardiomyopathy or following mutations in MHC403 and SCN5A resulting in hypertrophic cardiomyopathy (HCM) or Lenègre disease, respectively. This review defines and quantifies the determinants of θ and summarizes experimental evidence that links changes in these determinants with reduced myocardial θ and arrhythmogenesis. It thereby identifies the diverse pathophysiological conditions in which abnormal θ may contribute to arrhythmia.
Collapse
Affiliation(s)
- James H King
- Physiological Laboratory, Department of Physiology, Development and Neuroscience, University of Cambridge Cambridge, UK
| | | | | |
Collapse
|
20
|
Effects of hypokalemia on transmural dispersion of ventricular repolarization in left ventricular myocardium. ASIAN PAC J TROP MED 2013; 6:485-8. [PMID: 23711711 DOI: 10.1016/s1995-7645(13)60079-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Revised: 04/15/2013] [Accepted: 05/15/2013] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE To observe effects of hypokalemia on transmural heterogeneity of ventricular repolarization in left ventricular myocardium of rabbit, and explore the role of hypokalemia in malignant ventricular arrhythmia (MVA). METHODS A total of 20 rabbits were randomly divided into control group and hypokalemic group. Isolated hearts in the control group were simply perfused with modified Tyrode's solution, and were perfused with hypokalemic Tyrode's solution in hypokalemic group. Ventricular fibrillation threshold (VFT), 90% monophasic action potential repolarization duration (APD90) of subepicardial, midmyocardial and subendocardial myocardium, transmural dispersion of repolarization (TDR) and C×43 protein expression in three layers of myocardium were measured in both groups. RESULTS VFT in the control group and the hypokalemic group were (13.40 ± 2.95) V, and (7.00 ± 1.49) V, respectively. There was a significant difference between two groups (P<0.01). APD90 of three myocardial layers in the hypokalemic group were significantly prolonged than those in the control group (P<0.01). ΔAPD90 in the hypokalemic group and the control group were (38.10 ± 10.29) ms and (23.70 ± 5.68) ms, and TDR were (52.90 ± 14.55) ms and (36.10 ± 12.44) ms, respectively. ΔAPD90 and TDR in the hypokalemic group were significantly higher than those in the control group (P<0.05), and the increase in APD90 of midmyocardium was more significant in the hypokalemic group. Cx43 protein expression of all three myocardial layers were decreased significantly in the hypokalemic group (P<0.01), and ΔCx43 was significantly increased (P<0.05). Reduction of Cx43 protein expression was more significant in the midmyocardium. CONCLUSIONS Hypokalemic can increase transmural heterogeneity of C×43 expression and repolarization in left ventricular myocardium of rabbit, and decrease VFT and can induce MVA more easily.
Collapse
|
21
|
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
|
22
|
Hsu SJ, Byram BC, Bouchard RR, Dumont DM, Wolf PD, Trahey GE. Acoustic radiation force impulse imaging of mechanical stiffness propagation in myocardial tissue. ULTRASONIC IMAGING 2012; 34:142-58. [PMID: 22972912 PMCID: PMC3500656 DOI: 10.1177/0161734612456580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Acoustic radiation force impulse (ARFI) imaging has been shown to be capable of imaging local myocardial stiffness changes throughout the cardiac cycle. Expanding on these results, the authors present experiments using cardiac ARFI imaging to visualize and quantify the propagation of mechanical stiffness during ventricular systole. In vivo ARFI images of the left ventricular free wall of two exposed canine hearts were acquired. Images were formed while the heart was externally paced by one of two electrodes positioned on the epicardial surface and either side of the imaging plane. Two-line M-mode ARFI images were acquired at a sampling frequency of 120 Hz while the heart was paced from an external stimulating electrode. Two-dimensional ARFI images were also acquired, and an average propagation velocity across the lateral field of view was calculated. Directions and speeds of myocardial stiffness propagation were measured and compared with the propagations derived from the local electrocardiogram (ECG), strain, and tissue velocity measurements estimated during systole. In all ARFI images, the direction of myocardial stiffness propagation was seen to be away from the stimulating electrode and occurred with similar velocity magnitudes in either direction. When compared with the local epicardial ECG, the mechanical stiffness waves were observed to travel in the same direction as the propagating electrical wave and with similar propagation velocities. In a comparison between ARFI, strain, and tissue velocity imaging, the three methods also yielded similar propagation velocities.
Collapse
|
23
|
Wilders R. Arrhythmogenic right ventricular cardiomyopathy: considerations from in silico experiments. Front Physiol 2012; 3:168. [PMID: 22754532 PMCID: PMC3385583 DOI: 10.3389/fphys.2012.00168] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 05/09/2012] [Indexed: 11/22/2022] Open
Abstract
Objective: Arrhythmogenic right ventricular cardiomyopathy (ARVC) is associated with remodeling of gap junctions and also, although less well-defined, down-regulation of the fast sodium current. The gap junction remodeling and down-regulation of sodium current have been proposed as contributors to arrhythmogenesis in ARVC by slowing conduction. The objective of the present study was to assess the amount of conduction slowing due to the observed gap junction remodeling and down-regulation of sodium current. Methods: The effects of (changes in) gap junctional conductance, cell dimensions, and sodium current on both longitudinal and transversal conduction velocity were tested by simulating action potential propagation in linear strands of human ventricular cells that were either arranged end-to-end or side-by-side. Results: A 50% reduction in gap junction content, as commonly observed in ARVC, gives rise to an 11% decrease in longitudinal conduction velocity and a 29% decrease in transverse conduction velocity. A down-regulation of the sodium current through a 50% decrease in peak current density as well as a −15 mV shift in steady-state inactivation, as observed in an experimental model of ARVC, decreases conduction velocity in either direction by 32%. In combination, the gap junction remodeling and down-regulation of sodium current result in a 40% decrease in longitudinal conduction velocity and a 52% decrease in transverse conduction velocity. Conclusion: The gap junction remodeling and down-regulation of sodium current do result in conduction slowing, but heterogeneity of gap junction remodeling, in combination with down-regulation of sodium current, rather than gap junction remodeling per se may be a critical factor in arrhythmogenesis in ARVC.
Collapse
Affiliation(s)
- Ronald Wilders
- Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam Amsterdam, Netherlands
| |
Collapse
|
24
|
Salameh A, Dhein S. Effects of mechanical forces and stretch on intercellular gap junction coupling. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:147-56. [PMID: 22245380 DOI: 10.1016/j.bbamem.2011.12.030] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 12/17/2011] [Accepted: 12/27/2011] [Indexed: 01/27/2023]
Abstract
Mechanical forces provide fundamental physiological stimulus in living organisms. Recent investigations demonstrated how various types of mechanical load, like strain, pressure, shear stress, or cyclic stretch can affect cell biology and gap junction intercellular communication (GJIC). Depending on the cell type, the type of mechanical load and on strength and duration of application, these forces can induce hypertrophic processes and modulate the expression and function of certain connexins such as Cx43, while others such as Cx37 or Cx40 are reported to be less mechanosensitive. In particular, not only expression but also subcellular localization of Cx43 is altered in cardiomyocytes submitted to cyclic mechanical stretch resulting in the typical elongated cell shape with an accentuation of Cx43 at the cell poles. In the heart both cardiomyocytes and fibroblasts can alter their GJIC in response to mechanical load. In the vasculature both endothelial cells and smooth muscle cells are subject to strain and cyclic stretch resulting from the pulsatile flow. In addition, vascular endothelial cells are mainly affected by shear stress resulting from the blood flow parallel to their surface. These mechanical forces lead to a regulation of GJIC in vascular tissue. In bones, osteocytes and osteoblasts are coupled via gap junctions, which also react to mechanical forces. Since gap junctions are involved in regulation of cell growth and differentiation, the mechanosensitivity of the regulation of these channels might open new perspectives to explain how cells can respond to mechanical load, and how stretch induces self-organization of a cell layer which might have implications for embryology and the development of organs. This article is part of a Special Issue entitled: The Communicating junctions, roles and dysfunctions.
Collapse
Affiliation(s)
- Aida Salameh
- Clinic for Pediatric Cardiology, University of Leipzig, Heart Centre, Germany
| | | |
Collapse
|
25
|
Dhein S, Rothe S, Busch A, Rojas Gomez DM, Boldt A, Reutemann A, Seidel T, Salameh A, Pfannmüller B, Rastan A, Kostelka M, Mohr FW. Effects of metoprolol therapy on cardiac gap junction remodelling and conduction in human chronic atrial fibrillation. Br J Pharmacol 2011; 164:607-16. [PMID: 21542828 PMCID: PMC3188900 DOI: 10.1111/j.1476-5381.2011.01460.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 03/16/2011] [Accepted: 04/06/2011] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE We investigated the influence of metoprolol on gap junction proteins connexin43 (Cx43) and connexin40 (Cx40) in atrial tissue from patients with/without atrial fibrillation (AF). EXPERIMENTAL APPROACH Left atrial tissue samples from 160 patients with AF or sinus rhythm (SR) with or without metoprolol (mean daily dose: 65.2 ± 9.1 mg·day⁻¹) were analysed for Cx43 and Cx40 by Western blot and immunohistology. Transverse and longitudinal conduction velocities were determined by 64 multi-electrode mapping. KEY RESULTS Both Cx43 and Cx40 expression were significantly increased in patients with AF versus SR. Cx43-expression in AF was significantly higher in patients receiving metoprolol, while Cx40 expression was unaffected by metoprolol treatment. In AF, the ratio of lateral/polar expression of Cx43 and Cx40 was enhanced due to increased expression at the sides of the cells (lateral) and a loss at the cell poles. This AF-induced increase in lateral/polar expression of Cx43, but not of Cx40, was significantly antagonized by metoprolol treatment. Functionally, in AF patients, transverse conduction velocity in atrial samples was significantly enhanced and this change was also significantly antagonized by metoprolol. CONCLUSIONS AND IMPLICATIONS AF induced enhanced lateral expression of Cx43 and Cx40 together with enhanced transverse conduction velocity in left atrial tissue. Alterations in localization of Cx43 and conduction changes were both antagonized by metoprolol, showing that pharmacological modulation of gap junction remodelling seems, in principle, possible. This finding may open new approaches to the development of anti-arrythmic drugs.
Collapse
Affiliation(s)
- S Dhein
- Clinic for Cardiac Surgery, University of Leipzig, Leipzig, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
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
|
27
|
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
|
28
|
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
|
29
|
Rhett JM, Jourdan J, Gourdie RG. Connexin 43 connexon to gap junction transition is regulated by zonula occludens-1. Mol Biol Cell 2011; 22:1516-28. [PMID: 21411628 PMCID: PMC3084674 DOI: 10.1091/mbc.e10-06-0548] [Citation(s) in RCA: 225] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cx43 gap junctions (GJs) are integral to the function of the mammalian heart. It is shown that ZO-1 dynamically regulates the transition between Cx43 connexons and GJ intercellular channels, determining the balance of connexon-mediated cell permeability to GJ communication. Importantly, a novel domain proximal to GJs is identified—the perinexus. Connexin 43 (Cx43) is a gap junction (GJ) protein widely expressed in mammalian tissues that mediates cell-to-cell coupling. Intercellular channels comprising GJ aggregates form from docking of paired connexons, with one each contributed by apposing cells. Zonula occludens-1 (ZO-1) binds the carboxy terminus of Cx43, and we have previously shown that inhibition of the Cx43/ZO-1 interaction increases GJ size by 48 h. Here we demonstrated that increases in GJ aggregation occur within 2 h (∼Cx43 half-life) following disruption of Cx43/ZO-1. Immunoprecipitation and Duolink protein–protein interaction assays indicated that inhibition targets ZO-1 binding with Cx43 in GJs as well as connexons in an adjacent domain that we term the “perinexus.” Consistent with GJ size increases being matched by decreases in connexons, inhibition of Cx43/ZO-1 reduced the extent of perinexal interaction, increased the proportion of connexons docked in GJs relative to undocked connexons in the plasma membrane, and increased GJ intercellular communication while concomitantly decreasing hemichannel-mediated membrane permeance in contacting, but not noncontacting, cells. ZO-1 small interfering RNA and overexpression experiments verified that loss and gain of ZO-1 function govern the transition of connexons into GJs. It is concluded that ZO-1 regulates the rate of undocked connexon aggregation into GJs, enabling dynamic partitioning of Cx43 channel function between junctional and proximal nonjunctional domains of plasma membrane.
Collapse
Affiliation(s)
- J Matthew Rhett
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | |
Collapse
|
30
|
Hussain W, Patel PM, Chowdhury RA, Cabo C, Ciaccio EJ, Lab MJ, Duffy HS, Wit AL, Peters NS. The Renin-Angiotensin system mediates the effects of stretch on conduction velocity, connexin43 expression, and redistribution in intact ventricle. J Cardiovasc Electrophysiol 2011; 21:1276-83. [PMID: 20487124 DOI: 10.1111/j.1540-8167.2010.01802.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
UNLABELLED Effect of Stretch on Conduction and Cx43. INTRODUCTION In disease states such as heart failure, myocardial infarction, and hypertrophy, changes in the expression and location of Connexin43 (Cx43) occur (Cx43 remodeling), and may predispose to arrhythmias. Stretch may be an important stimulus to Cx43 remodeling; however, it has only been investigated in neonatal cell cultures, which have different physiological properties than adult myocytes. We hypothesized that localized stretch in vivo causes Cx43 remodeling, with associated changes in conduction, mediated by the renin-angiotensin system (RAS). METHODS AND RESULTS In an open-chest canine model, a device was used to stretch part of the right ventricle (RV) by 22% for 6 hours. Activation mapping using a 312-electrode array was performed before and after stretch. Regional stretch did not change longitudinal conduction velocity (post-stretch vs baseline: 51.5 ± 5.2 vs 55.3 ± 8.1 cm/s, P = 0.24, n = 11), but significantly reduced transverse conduction velocity (28.7 ± 2.5 vs 35.4 ± 5.4 cm/s, P < 0.01). It also reduced total Cx43 expression, by Western blotting, compared with nonstretched RV of the same animal (86.1 ± 32.2 vs 100 ± 19.4%, P < 0.02, n = 11). Cx43 labeling redistributed to the lateral cell borders. Stretch caused a small but significant increase in the proportion of the dephosphorylated form of Cx43 (stretch 9.95 ± 1.4% vs control 8.74 ± 1.2%, P < 0.05). Olmesartan, an angiotensin II blocker, prevented the stretch-induced changes in Cx43 levels, localization, and conduction. CONCLUSION Myocardial stretch in vivo has opposite effects to that in neonatal myocytes in vitro. Stretch in vivo causes conduction changes associated with Cx43 remodeling that are likely caused by local stretch-induced activation of the RAS.
Collapse
Affiliation(s)
- Wajid Hussain
- Department of Cardiac Electrophysiology, Imperial College & St. Mary's Hospital, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Activated Met signalling in the developing mouse heart leads to cardiac disease. PLoS One 2011; 6:e14675. [PMID: 21347410 PMCID: PMC3036588 DOI: 10.1371/journal.pone.0014675] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 01/13/2011] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The Hepatocyte Growth Factor (HGF) is a pleiotropic cytokine involved in many physiological processes, including skeletal muscle, placenta and liver development. Little is known about its role and that of Met tyrosine kinase receptor in cardiac development. METHODOLOGY/PRINCIPAL FINDINGS In this study, we generated two transgenic mice with cardiac-specific, tetracycline-suppressible expression of either Hepatocyte Growth Factor (HGF) or the constitutively activated Tpr-Met kinase to explore: i) the effect of stimulation of the endogenous Met receptor by autocrine production of HGF and ii) the consequence of sustained activation of Met signalling in the heart. We first showed that Met is present in the neonatal cardiomyocytes and is responsive to exogenous HGF. Exogenous HGF starting from prenatal stage enhanced cardiac proliferation and reduced sarcomeric proteins and Connexin43 (Cx43) in newborn mice. As adults, these transgenics developed systolic contractile dysfunction. Conversely, prenatal Tpr-Met expression was lethal after birth. Inducing Tpr-Met expression during postnatal life caused early-onset heart failure, characterized by decreased Cx43, upregulation of fetal genes and hypertrophy. CONCLUSIONS/SIGNIFICANCE Taken together, our data show that excessive activation of the HGF/Met system in development may result in cardiac damage and suggest that Met signalling may be implicated in the pathogenesis of cardiac disease.
Collapse
|
32
|
Spray DC, Suadicani SO, Srinivas M, Gutstein DE, Fishman GI. Gap Junctions in the Cardiovascular System. Compr Physiol 2011. [DOI: 10.1002/cphy.cp020104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
33
|
Wang B, Wen Q, Xie X, Liu S, Liu M, Tao Y, Li Z, Suo P, Shen A, Wang J, Ma X. Mutation analysis of Connexon43 gene in Chinese patients with congenital heart defects. Int J Cardiol 2010; 145:487-9. [DOI: 10.1016/j.ijcard.2009.06.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Accepted: 06/06/2009] [Indexed: 10/20/2022]
|
34
|
Chao MW, Kozlosky J, Po IP, Strickland PO, Svoboda KKH, Cooper K, Laumbach RJ, Gordon MK. Diesel exhaust particle exposure causes redistribution of endothelial tube VE-cadherin. Toxicology 2010; 279:73-84. [PMID: 20887764 DOI: 10.1016/j.tox.2010.09.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 09/07/2010] [Accepted: 09/21/2010] [Indexed: 11/18/2022]
Abstract
Whether diesel exhaust particles (DEPs) potentially have a direct effect on capillary endothelia was examined by following the adherens junction component, vascular endothelial cell cadherin (VE-cadherin). This molecule is incorporated into endothelial adherens junctions at the cell surface, where it forms homodimeric associations with adjacent cells and contributes to the barrier function of the vasculature (Dejana et al., 2008; Venkiteswaran et al., 2002; Villasante et al., 2007). Human umbilical vein endothelial cells (HUVECs) that were pre-formed into capillary-like tube networks in vitro were exposed to DEPs for 24h. After exposure, the integrity of VE-cadherin in adherens junctions was assessed by immunofluorescence analysis, and demonstrated that increasing concentrations of DEPs caused increasing redistribution of VE-cadherin away from the cell-cell junctions toward intracellular locations. Since HUVEC tube networks are three-dimensional structures, whether particles entered the endothelial cells or tubular lumens was also examined. The data indicate that translocation of the particles does occur. The results, obtained in a setting that removes the confounding effects of inflammatory cells or blood components, suggest that if DEPs encounter alveolar capillaries in vivo, they may be able to directly affect the endothelial cell-cell junctions.
Collapse
Affiliation(s)
- Ming-Wei Chao
- Pharmacology and Toxicology, Rutgers University, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Salameh A, Krautblatter S, Karl S, Blanke K, Gomez DR, Dhein S, Pfeiffer D, Janousek J. The signal transduction cascade regulating the expression of the gap junction protein connexin43 by beta-adrenoceptors. Br J Pharmacol 2010; 158:198-208. [PMID: 19719782 DOI: 10.1111/j.1476-5381.2009.00344.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE In mammalian heart, connexin43 (Cx43) represents the predominant connexin in the working myocardium. As the beta-adrenoceptor is involved in many cardiac diseases, we wanted to clarify the pathway by which beta-adrenoceptor stimulation may control Cx43 expression. EXPERIMENTAL APPROACH Cultured neonatal rat cardiomyocytes were stimulated with isoprenaline. Cx43 expression as well as activation of p38 mitogen-activated protein kinase (MAPK), p42/44 MAPK, JUN NH(2)-terminal kinase (JNK) and nuclear translocation of the transcription factors activator protein 1 (AP1) and CRE-binding protein (CREB) were investigated. Additionally, we assessed Cx43 expression and distribution in left ventricular biopsies from patients without any significant heart disease, and from patients with either congestive heart failure [dilated cardiomyopathy (DCM)] or hypertrophic cardiomyopathy (HCM). KEY RESULTS Isoprenaline exposure caused about twofold up-regulation of Cx43 protein with a pEC(50) of 7.92 +/- 0.11, which was inhibited by propranolol, SB203580 (4-(4-fluorophenyl)-2-(4-methylsulphinylphenyl)-5-(4-pyridyl)-1H-imidazole) (p38 inhibitor), PD98059 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one) (MAPK 1 kinase inhibitor) (Alexis Biochemicals, San Diego, CA, USA) or cyclosporin A. Similar findings were obtained for Cx43 mRNA. Furthermore, Cx43 up-regulation was accompanied by phosphorylation of p38, p42/44 and JNK, and by translocation of AP1 and CREB to the nucleus. Analysis of Cx43 protein and mRNA in ventricular biopsies revealed that in patients with DCM, Cx43 content was significantly lower, and in patients with HCM, Cx43 content was significantly higher, relative to patients without any cardiomyopathy. More importantly, Cx43 distribution also changed with more Cx43 being localized at the lateral border of the cardiomyocytes. CONCLUSION AND IMPLICATION Beta-adrenoceptor stimulation up-regulated cardiac Cx43 expression via a protein kinase A and MAPK-regulated pathway, possibly involving AP1 and CREB. Cardiomyopathy altered Cx43 expression and distribution.
Collapse
Affiliation(s)
- A Salameh
- Department of Paediatric Cardiology, University of Leipzig, Heart Centre, Leipzig, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Ordered assembly of the adhesive and electrochemical connections within newly formed intercalated disks in primary cultures of adult rat cardiomyocytes. J Biomed Biotechnol 2010; 2010:624719. [PMID: 20467587 PMCID: PMC2868981 DOI: 10.1155/2010/624719] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2009] [Accepted: 02/17/2010] [Indexed: 02/06/2023] Open
Abstract
The intercalated disk (ID) is a complex structure that electromechanically couples adjoining cardiac myocytes into a functional syncitium. The integrity of the disk is essential for normal cardiac function, but how the diverse elements are assembled into a fully integrated structure is not well understood. In this study, we examined the assembly of new IDs in primary cultures of adult rat cardiac myocytes. From 2 to 5 days after dissociation, the cells flatten and spread, establishing new cell-cell contacts in a manner that recapitulates the in vivo processes that occur during heart development and myocardial remodeling. As cells make contact with their neighbors, transmembrane adhesion proteins localize along the line of apposition, concentrating at the sites of membrane attachment of the terminal sarcomeres. Cx43 gap junctions and ankyrin-G, an essential cytoskeletal component of voltage gated sodium channel complexes, were secondarily recruited to membrane domains involved in cell-cell contacts. The consistent order of the assembly process suggests that there are specific scaffolding requirements for integration of the mechanical and electrochemical elements of the disk. Defining the relationships that are the foundation of disk assembly has important implications for understanding the mechanical dysfunction and cardiac arrhythmias that accompany alterations of ID architecture.
Collapse
|
37
|
Strom M, Wan X, Poelzing S, Ficker E, Rosenbaum DS. Gap junction heterogeneity as mechanism for electrophysiologically distinct properties across the ventricular wall. Am J Physiol Heart Circ Physiol 2009; 298:H787-94. [PMID: 20035026 DOI: 10.1152/ajpheart.00887.2009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gap junctions are critical to maintaining synchronized impulse propagation and repolarization. Heterogeneous expression of the principal ventricular gap junction protein connexin43 (Cx43) is associated with action potential duration (APD) dispersion across the anterior ventricular wall. Little is known about Cx43 expression patterns and their disparate impact on regional electrophysiology throughout the heart. We aimed to determine whether the anterior and posterior regions of the heart are electrophysiologically distinct. Multisegment, high-resolution optical mapping was performed in canine wedge preparations harvested separately from the anterior left ventricle (aLV; n = 8) and posterior left ventricle (pLV; n = 8). Transmural APD dispersion was significantly greater on the aLV than the pLV (45 +/- 13 vs. 26 +/- 8.0 ms; P < 0.05). Conduction velocity dispersion was also significantly higher (P < 0.05) across the aLV (39 +/- 7%) than the pLV (16 +/- 3%). Carbenoxolone perfusion significantly enhanced APD and conduction velocity dispersion on the aLV (by 1.53-fold and 1.36-fold, respectively), but not the pLV (by 1.27-fold and 1.2-fold, respectively), and produced a 4.2-fold increase in susceptibility to inducible arrhythmias in the aLV. Confocal immunofluorescence microscopy revealed significantly (P < 0.05) greater transmural dispersion of Cx43 expression on the aLV (44 +/- 10%) compared with the pLV wall (8.3 +/- 0.7%), suggesting that regional expression of Cx43 expression patterns may account for regional electrophysiological differences. Computer simulations affirmed that localized uncoupling at the epicardial-midmyocardial interface is sufficient to produce APD gradients observed on the aLV. These data demonstrate that the aLV and pLV differ importantly with respect to their electrophysiological properties and Cx43 expression patterns. Furthermore, local underexpression of Cx43 is closely associated with transmural electrophysiological heterogeneity on the aLV. Therefore, regional and transmural heterogeneous Cx43 expression patterns may be an important mechanism underlying arrhythmia susceptibility, particularly in disease states where gap junction expression is altered.
Collapse
Affiliation(s)
- Maria Strom
- The Heart and Vascular Research Center, and The Department Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44109-1998, USA
| | | | | | | | | |
Collapse
|
38
|
Chen CC, Lin CC, Lee TM. 17beta-Estradiol decreases vulnerability to ventricular arrhythmias by preserving connexin43 protein in infarcted rats. Eur J Pharmacol 2009; 629:73-81. [PMID: 20004189 DOI: 10.1016/j.ejphar.2009.11.050] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Revised: 11/11/2009] [Accepted: 11/23/2009] [Indexed: 11/19/2022]
Abstract
Epidemiological studies showed that a lower mortality rate of sudden cardiac death among women than among men may depend on the action of female sex hormones. This study assessed whether 17beta-estradiol exerts anti-arrhythmic effects through enhanced Connexin43 (Cx43) expression after infarction. Two weeks after ovariectomy, female Wistar rats were randomly assigned to coronary artery ligation or sham-operation. Twenty-four hours after coronary ligation, ovariectomized rats were randomized into vehicle, subcutaneous estradiol treatment, tamoxifen, or subcutaneous estradiol treatment+tamoxifen and followed for 4weeks. To verify the role of estradiol-related nitric oxide in modulating the expression of Cx43, N-nitro-L-arginine methyl ester was also assessed in an in vitro study. Myocardial Cx43 expression revealed a significant decrease in vehicle-treated infarcted rats compared with sham-operated rats at 24h and 4weeks after infarction. Attenuated Cx43 expression was blunted after administering estradiol, assessed by immunofluorescent analysis, Western blotting, and real-time quantitative RT-PCR of Cx43. The vulnerability for ventricular arrhythmia during programmed stimulation in estradiol-treated infarcted rats was significantly lower than in vehicle-treated infarcted rats. The beneficial effect of estradiol on Cx43 was abolished by tamoxifen. In addition, the invitro study demonstrated that the amount of Cx43 showed significant reduction after adding N-nitro-L-arginine methyl ester. Chronic administration of estradiol after infarction is associated with attenuated reduction of gap junction proteins probably through a nitric oxide-dependent pathway via the estrogen receptor and thus plays a critical role in the beneficial effect on arrhythmic vulnerability response to programmed electrical stimulation.
Collapse
Affiliation(s)
- Chien-Chang Chen
- Cardiology Section, Department of Surgery, Chi-Mei Medical Center, Tainan, Taiwan
| | | | | |
Collapse
|
39
|
Waghabi MC, Coutinho-Silva R, Feige JJ, Higuchi MDL, Becker D, Burnstock G, Araújo-Jorge TCD. Gap junction reduction in cardiomyocytes following transforming growth factor-β treatment and Trypanosoma cruzi infection. Mem Inst Oswaldo Cruz 2009; 104:1083-90. [DOI: 10.1590/s0074-02762009000800004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Accepted: 10/19/2009] [Indexed: 11/21/2022] Open
Affiliation(s)
- Mariana C Waghabi
- Instituto Oswaldo Cruz-Fiocruz, Brasil; University College Medical School, UK
| | - Robson Coutinho-Silva
- University College Medical School, UK; Universidade Federal do Rio de Janeiro, Brasil
| | - Jean-Jacques Feige
- Institut National de la Santé et de la Recherche Médicale; Institut de Recherches en Technologies et Sciences pour le Vivant, France
| | | | | | | | | |
Collapse
|
40
|
Huang T, Wan Y, Zhu Y, Fang X, Hiramatsu N, Hayakawa K, Paton AW, Paton JC, Kitamura M, Yao J. Downregulation of gap junction expression and function by endoplasmic reticulum stress. J Cell Biochem 2009; 107:973-83. [PMID: 19492336 DOI: 10.1002/jcb.22202] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Gap junctional intercellular communication (GJIC) plays a critical role in the control of multiple cell behavior as well as in the maintenance of tissue and organ homeostasis. However, mechanisms involved in the regulation of gap junctions (GJs) have not been fully understood. Given endoplasmic reticulum (ER) stress and dysfunction of GJs coexist in several pathological situations, we asked whether GJs could be regulated by ER stress. Incubation of mesangial cells with ER stress-inducing agents (thapsigargin, tunicamycin, and AB(5) subtilase cytotoxin) resulted in a decrease in connexin 43 (Cx43) expression at both protein and mRNA levels. This was accompanied by a loss of GJIC, as evidenced by the reduced numbers of dye-coupled cells after single cell microinjection or scrape loading dye transfer. Further studies demonstrated that ER stress significantly inhibited the promoter activity of the Cx43 gene, reduced [(35)S]-methionine incorporation into Cx43 protein and accelerated degradation of Cx43. ER stress also decreased the Cx43 protein levels in several different cell types, including human umbilical vein endothelial cells, mouse-derived renin-secreting cells and human hepatoma cells. Furthermore, induction of ER stress by hypoxic chemicals thenoyltrifluoroacetone and cobalt chloride was found to be associated with a reduction in Cx43. Our findings thus reveal a close link between ER stress and GJs. ER stress may represent a novel mechanism underlying the altered GJs in a variety of pathological situations.
Collapse
Affiliation(s)
- Tao Huang
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Jansen JA, van Veen TAB, de Bakker JMT, van Rijen HVM. Cardiac connexins and impulse propagation. J Mol Cell Cardiol 2009; 48:76-82. [PMID: 19729017 DOI: 10.1016/j.yjmcc.2009.08.018] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 07/29/2009] [Accepted: 08/21/2009] [Indexed: 12/15/2022]
Abstract
Gap junctions form the intercellular pathway for cell-to-cell transmission of the cardiac impulse from its site of origin, the sinoatrial node, along the atria, the atrioventricular conduction system to the ventricular myocardium. The component parts of gap junctions are proteins called connexins (Cx), of which three main isoforms are found in the conductive and working myocardial cells: Cx40, Cx43, and Cx45. These isoforms are regionally expressed in the heart, which suggests a specific role or function of a specific connexin in a certain part of the heart. Using genetically modified mice, the function of these connexins in the different parts of the heart have been assessed in the past years. This review will follow the cardiac impulse on its path through the heart and recapitulate the role of the different connexins in the different cardiac compartments.
Collapse
Affiliation(s)
- John A Jansen
- Department of Medical Physiology, Division Heart and Lungs, University Medical Center Utrecht, The Netherlands
| | | | | | | |
Collapse
|
42
|
Callis TE, Pandya K, Seok HY, Tang RH, Tatsuguchi M, Huang ZP, Chen JF, Deng Z, Gunn B, Shumate J, Willis MS, Selzman CH, Wang DZ. MicroRNA-208a is a regulator of cardiac hypertrophy and conduction in mice. J Clin Invest 2009; 119:2772-86. [PMID: 19726871 DOI: 10.1172/jci36154] [Citation(s) in RCA: 623] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 06/10/2009] [Indexed: 12/25/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small noncoding RNAs that have gained status as important regulators of gene expression. Here, we investigated the function and molecular mechanisms of the miR-208 family of miRNAs in adult mouse heart physiology. We found that miR-208a, which is encoded within an intron of alpha-cardiac muscle myosin heavy chain gene (Myh6), was actually a member of a miRNA family that also included miR-208b, which was determined to be encoded within an intron of beta-cardiac muscle myosin heavy chain gene (Myh7). These miRNAs were differentially expressed in the mouse heart, paralleling the expression of their host genes. Transgenic overexpression of miR-208a in the heart was sufficient to induce hypertrophic growth in mice, which resulted in pronounced repression of the miR-208 regulatory targets thyroid hormone-associated protein 1 and myostatin, 2 negative regulators of muscle growth and hypertrophy. Studies of the miR-208a Tg mice indicated that miR-208a expression was sufficient to induce arrhythmias. Furthermore, analysis of mice lacking miR-208a indicated that miR-208a was required for proper cardiac conduction and expression of the cardiac transcription factors homeodomain-only protein and GATA4 and the gap junction protein connexin 40. Together, our studies uncover what we believe are novel miRNA-dependent mechanisms that modulate cardiac hypertrophy and electrical conduction.
Collapse
Affiliation(s)
- Thomas E Callis
- Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Slezak J, Tribulova N, Okruhlicova L, Dhingra R, Bajaj A, Freed D, Singal P. Hibernating myocardium: pathophysiology, diagnosis, and treatment. Can J Physiol Pharmacol 2009; 87:252-65. [PMID: 19370079 DOI: 10.1139/y09-011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Comprehensive management of patients with chronic ischemic disease is a critically important component of clinical practice. Cardiac myocytes have the potential to adapt to limited flow conditions by adjusting contractile function, reducing metabolism, conserving resources, and preserving myocardial integrity to cope with an oxygen and (or) nutrition shortage. A prime metabolic feature of cardiac myocytes affected by chronic ischemia is the return to a fetal gene pattern with predominance of carbohydrates as the substrate for energy. Structural adaptation with multiple intracellular changes is part of the remodeling process in hibernating myocardium. Transmural heterogeneity, which defines the pattern of injury in ventricular cardiomyocytes and the response to chronic ischemia, is a multifactorial process originating from functional, metabolic, and flow differences in subendocardial and subepicardial regions. Autophagy is typically activated in hibernating myocardium and has been identified as a prosurvival mechanism. Chronic ischemia is associated with changes in the number, size, and distribution of gap junctions and may give rise to conduction disturbances and arrhythmogenesis. Differentiation between viable and nonviable myocardium by assessing sensitivity of inotropic reserve is a crucial diagnostic tool that is correlated with the prognosis and outcome for improved contractility after restoration of blood perfusion in afflicted myocardium.Reliable and accurate diagnosis of ischemic, scar, and viable tissues is critical for recover strategies. Although early surgical reinstitution of blood flow is most effective in restoring physiologic function of the hibernating myocardium, several new approaches offer promising alternatives. Among others, vascular endothelial growth factor and fibroblast growth factor-2 (FGF-2), especially its lo-FGF-2 isoform, have been shown to be effective in rapid neovascularization. Substances such as statins, resveratrol, some hormones, and omega-3 fatty acids can improve recovery effect in chronically underperfused hearts. For patients with drug-refractory ischemia, intramyocardial transplantation of stem cells into predefined areas of the heart can enhance vascularization and have beneficial effects on cardiac function. This review of ischemic injury, its heterogeneity, accurate diagnosis, and newer methods of treatment, shows there is much information and tremendous hope for better management of patients with coronary heart disease.
Collapse
Affiliation(s)
- Jan Slezak
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | | | | | | | | | | | | |
Collapse
|
44
|
Stein M, van Veen TAB, Remme CA, Boulaksil M, Noorman M, van Stuijvenberg L, van der Nagel R, Bezzina CR, Hauer RNW, de Bakker JMT, van Rijen HVM. Combined reduction of intercellular coupling and membrane excitability differentially affects transverse and longitudinal cardiac conduction. Cardiovasc Res 2009; 83:52-60. [PMID: 19389723 DOI: 10.1093/cvr/cvp124] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Reduced excitability and gap junction expression are commonly found in electrically remodelled diseased hearts, but their contribution to slow conduction and arrhythmias is unclear. In this study, we have investigated the effect of isolated and combined reductions in membrane excitability and intercellular coupling on impulse propagation and arrhythmogeneity in genetically modified mice. METHODS AND RESULTS Cx43 and Scn5a(1798insD/+) heterozygous (HZ) mice were crossbred to create a mixed offspring: wild-type (WT, n = 15), Cx43 HZ (n = 14), Scn5a(1798insD/+) (Scn5a) HZ (n = 17), and Cx43/Scn5a(1798insD/+) (Cx43/Scn5a) HZ (n = 15) mice. After ECG recording, epicardial activation mapping (208 recording sites) was performed on Langendorff-perfused hearts. Arrhythmia inducibility was tested by one to three premature stimuli and burst pacing. Conduction velocity longitudinal (CV(L)) and transverse (CV(T)) to fibre orientation and dispersion of conduction were determined during S1-S1 pacing (150 ms). Connexin43 (Cx43) and sodium channel Nav1.5 protein expression and myocardial tissue collagen content were determined by immunohistology. Compared with WT animals, P, QRS, and QTc intervals were prolonged in Scn5a HZ and Cx43/Scn5a HZ, but not in Cx43 HZ animals. Scn5a HZ mice showed decreased CV(L) in right ventricle (RV) but not in left ventricle compared with WT. In the RV of Cx43/Scn5a HZ, CV(T) was reduced, but CV(L) was not different from WT. Arrhythmia inducibility was low and not increased in either single- or double-mutant mice. CONCLUSION Reduction of both electrical coupling and excitability results in normal conduction velocity parallel to fibre orientation but in pronounced conduction slowing transverse to fibre orientation in RV only, although this does not affect arrhythmogeneity.
Collapse
Affiliation(s)
- Mèra Stein
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Yalelaan 50, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Human connexin31.9, unlike its orthologous protein connexin30.2 in the mouse, is not detectable in the human cardiac conduction system. J Mol Cell Cardiol 2009; 46:553-9. [DOI: 10.1016/j.yjmcc.2008.12.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Revised: 12/04/2008] [Accepted: 12/09/2008] [Indexed: 12/11/2022]
|
46
|
Lin LC, Wu CC, Lin MS, Lin SF, Liu YB. Reducing the cyclic variations of ultrasonic integrated backscatters and myocardial electrical synchronism by reversibly blocking intercellular communications with heptanol. ULTRASOUND IN MEDICINE & BIOLOGY 2009; 35:209-218. [PMID: 18977069 DOI: 10.1016/j.ultrasmedbio.2008.08.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Revised: 07/27/2008] [Accepted: 08/23/2008] [Indexed: 05/27/2023]
Abstract
The purpose of this study is to provide direct evidence for the role of intercellular communications in electrical synchronization and mechanical function of myocardium. We used heptanol, a reversible inhibitor of gap junctions, at low (0.16 mM) and high (0.5 mM) concentration as perfusate for 18 Langendorff-perfused rabbit hearts to study its effects on myocardial electrical and mechanical functions. Optical mapping was performed to measure conduction velocity (CV) and action potential duration (APD). Ultrasonic integrated backscatter and Doppler tissue imaging (DTI) were used to evaluate the intrinsic and global myocardial contractile performance. The CV decreased during low-dose heptanol infusion and became much slower at high dose (high dose vs. baseline, 50.8 +/- 10.2 cm/s vs. 69.3 +/- 8.8 cm/s, p < 0.001). After washout of heptanol, CV completely recovered. The alterations of APD by heptanol infusion were similar to CV. The APD dispersion, standard deviation of APD(80), was increased after heptanol infusion (low dose vs. baseline, 5.9 +/- 1.1 ms vs. 4.3 +/- 1.1 ms, p = 0.004; high dose, 6.0 +/- 1.3 ms, vs. baseline, p = 0.035). However, washout did not restore the APD dispersion which became even larger after washout (13.6 +/- 1.9 ms vs. high dose and baseline, both p < 0.001). Regarding contractile function, heptanol treatment resulted in a progressive decrease of cardiac cycle-dependent variations of integrated backscatter (CVIBS; low dose vs. baseline, 6.1 +/- 1.7 dB vs. 7.2 +/- 1.8 dB, p = 0.007; high dose 1.7 +/- 0.3 dB vs. baseline, p < 0.001) and peak systolic strain rate (low dose vs. baseline, -1.5 +/- 0.6 1/s vs. -1.9 +/- 0.6 1/s, p = 0.014; high dose -0.4 +/- 0.2 1/s; vs. baseline, p < 0.001). That both CVIBS and strain rate incompletely recovered after heptanol washout may be attributed to the increased APD dispersion. In conclusion, uncoupling of gap junctions resulted in slowing CV, increased repolarization heterogeneity, reduced CVIBS and impaired myocardial contractility. There was a reversible dose-response relationship between the myocardial electromechanical functions and gap junction coupling.
Collapse
Affiliation(s)
- Lung-Chun Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
47
|
Grikscheit K, Thomas N, Bruce AF, Rothery S, Chan J, Severs NJ, Dupont E. Coexpression of connexin 45 with connexin 43 decreases gap junction size. ACTA ACUST UNITED AC 2008; 15:185-93. [PMID: 18649189 DOI: 10.1080/15419060802013943] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In the human heart, ventricular myocytes express connexin 43 (Cx43) and traces of Cx45. In congestive heart failure, Cx43 levels decrease, Cx45 levels increase and gap junction size decreases. To determine whether alterations of connexin coexpression ratio influence gap junction size, we engineered a rat liver epithelial cell line that endogenously expresses Cx43 to coexpress inducible levels of Cx45 under stimulation of the insect hormone, ponasterone A. In cells induced to express Cx45, gap junction sizes are significantly reduced (by 15% to 20%; p < 0.001), an effect that occurs despite increased levels of junctional connexons made from both connexins. In contrast, coexpression of Cx40 with Cx43 does not lead to any change in gap junction size. These results are consistent with the idea that increased Cx45 expression in the failing ventricle contributes to decreased gap junction size.
Collapse
Affiliation(s)
- Katharina Grikscheit
- Department of Cardiac Medicine, National Heart and Lung Institute, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
48
|
Jackson PEM, Feng QP, Jones DL. Nitric oxide depresses connexin 43 after myocardial infarction in mice. Acta Physiol (Oxf) 2008; 194:23-33. [PMID: 18394025 DOI: 10.1111/j.1748-1716.2008.01858.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIMS Heart failure (HF) is a major cause of death and morbidity. Connexin 43 (Cx43) content is reduced in the failing myocardium, but regulating factors have not been identified. In HF, inducible nitric oxide synthase (iNOS)-induced high levels of nitric oxide (NO) cause apoptosis and cardiac dysfunction. However, a direct iNOS-Cx43 link has not been demonstrated. We investigated this relationship in mice after myocardial infarction. METHODS Effects of myocardial infarction were evaluated 2 weeks after coronary artery ligation in wild-type C57BL/6 (WT) and iNOS(-/-) knockout mice. Myocardial Cx43 and Cx45 content were assessed by immunofluorescence confocal imaging and western blotting. Cardiac function was evaluated in anaesthetized mice using a micro pressure-tipped catheter inserted into the left ventricle. RESULTS Despite similar infarct size, deficiency in iNOS resulted in significantly lower plasma nitrate/nitrite levels, better haemodynamic performance and lower mortality 2 weeks after coronary ligation. Myocardial Cx43, but not Cx45, content was lower in WT mice following ligation. The reduction in Cx43 was less in iNOS(-/-) compared with WT mice. To assess the direct effect of NO on Cx43 expression, cultured neonatal mouse cardiomyocytes were employed. Incubation with the NO donor, S-nitroso-N-acetylpenicillamine, elicited a dose-dependent decrease in Cx43 content in cultured neonatal cardiomyocytes. CONCLUSIONS Increased NO production from iNOS depressed cardiac performance and contributed to the decreased myocardial Cx43 content 2 weeks after myocardial infarction.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Blotting, Western/methods
- Cells, Cultured
- Connexin 43/analysis
- Connexin 43/metabolism
- Depression, Chemical
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Confocal
- Models, Animal
- Myocardial Infarction/metabolism
- Myocardium/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type II/antagonists & inhibitors
- Nitric Oxide Synthase Type II/genetics
- Nitric Oxide Synthase Type II/metabolism
- Random Allocation
- S-Nitroso-N-Acetylpenicillamine/pharmacology
Collapse
Affiliation(s)
- P E M Jackson
- Department of Physiology, University of Western Ontario, London, ON, Canada
| | | | | |
Collapse
|
49
|
Kontogeorgis A, Li X, Kang EY, Feig JE, Ponzio M, Kang G, Kaba RA, Wit AL, Fisher EA, Morley GE, Peters NS, Coetzee WA, Gutstein DE. Decreased connexin43 expression in the mouse heart potentiates pacing-induced remodeling of repolarizing currents. Am J Physiol Heart Circ Physiol 2008; 295:H1905-16. [PMID: 18757477 PMCID: PMC2614590 DOI: 10.1152/ajpheart.590.2008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Gap junction redistribution and reduced expression, a phenomenon termed gap junction remodeling (GJR), is often seen in diseased hearts and may predispose toward arrhythmias. We have recently shown that short-term pacing in the mouse is associated with changes in connexin43 (Cx43) expression and localization but not with increased inducibility into sustained arrhythmias. We hypothesized that short-term pacing, if imposed on murine hearts with decreased Cx43 abundance, could serve as a model for evaluating the electrophysiological effects of GJR. We paced wild-type (normal Cx43 abundance) and heterozygous Cx43 knockout (Cx43+/−; 66% mean reduction in Cx43) mice for 6 h at 10–15% above their average sinus rate. We investigated the electrophysiological effects of pacing on the whole animal using programmed electrical stimulation and in isolated ventricular myocytes with patch-clamp studies. Cx43+/− myocytes had significantly shorter action potential durations (APD) and increased steady-state (Iss) and inward rectifier (IK1) potassium currents compared with those of wild-type littermate cells. In Cx43+/− hearts, pacing resulted in a significant prolongation of ventricular effective refractory period and APD and significant diminution of Iss compared with unpaced Cx43+/− hearts. However, these changes were not seen in paced wild-type mice. These data suggest that Cx43 abundance plays a critical role in regulating currents involved in myocardial repolarization and their response to pacing. Our study may aid in understanding how dyssynchronous activation of diseased, Cx43-deficient myocardial tissue can lead to electrophysiological changes, which may contribute to the worsened prognosis often associated with pacing in the failing heart.
Collapse
Affiliation(s)
- Andrianos Kontogeorgis
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
McLachlan CS, Almsherqi ZA, Mossop P, Suzuki J, Leong ST, Deng Y. Down regulation of immuno-detectable cardiac connexin-43 in BALB/c mice following acute fasting. Int J Cardiol 2008; 136:99-102. [PMID: 18606469 DOI: 10.1016/j.ijcard.2008.03.098] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2008] [Accepted: 03/30/2008] [Indexed: 11/18/2022]
Abstract
Acute starvation effects for connexin-43 protein expression, in the heart, had not been previously explored. Hence we examined acute fasting on the myocardial immuno-histochemical expression of connexin-43 in 3 groups of 8-week old female BALB/c mice. Groups consisted of control mice (n=5), fasting for 24 h (N=5) and 48 h (N=3). Under light microscopy all control fed cases revealed the presence of some immuno-detectable staining for connexin-43 that is either present or weakly observed in some or all of the regions of interest, that include the cross-sectional left ventricular sub-endocardium, mid-myocardium and papillary muscle. Whereas mice that underwent 24 or 48 h of acute starvation, connexin-43 expression was either difficult to detect visually (N=3) or was completely absent (N=5) at 40x magnification using a light microscope. In starved mice with no membrane staining for connexin-43 we observed an increase in the intracellular accumulation of cytoplasmic connexin-43 expression.
Collapse
|