1
|
Hu Y, Qu C, Zou Y, Liu X, Zhang C, Yang B. NBQX mediates ventricular fibrillation susceptibility in rat models of anxiety via the Nrf2/HO-1 pathway. Heliyon 2024; 10:e37358. [PMID: 39296140 PMCID: PMC11408043 DOI: 10.1016/j.heliyon.2024.e37358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/11/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
Objective Anxiety disorder (AD) is a common mental disorder related to cardiovascular disease morbidity. Oxidative stress plays a crucial role in the anxiety state and can lead to cardiac remodeling. Over-activation of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) in cardiomyocytes and neurons can cause oxidative stress. Additionally, the AMPAR inhibitor-2,3-dihydroxy-6-nitro-7-sulfamoyl-benzoquinoxaline-2,3-dione (NBQX) plays an important role in ameliorating oxidative stress. This study aimed to explore the anti-arrhythmic effects of NBQX in a rat model of anxiety. Methods The AD model was induced using empty bottle stimulation. Male Sprague Dawley rats were randomly divided into four groups: control + saline, control + NBQX, AD + saline, and AD + NBQX. Open field test was conducted to measure anxiety-like behavior. Electrophysiological experiments, histological analysis, biochemical detection and molecular biology were performed to verify the effects of NBQX on the amelioration of electrical remodeling and structural remodeling. Furthermore, the nuclear factor erythroid 2-related factor 2 (Nrf2) inhibitor (ML385) was used in vitro to demonstrate the signaling pathway. Results Oxidative stress levels increased with AMPAR over-activation in AD rats, leading to heightened vulnerability to ventricular fibrillation (VF). NBQX reverses anxiety and VF susceptibility. Our results showed that NBQX activated the Nrf2/heme oxygenase-1 (HO-1) pathway, leading to a decline in oxidative stress levels. Connexin 43 and ion channel expression was upregulated. NBQX treatment attenuated fibrosis and apoptosis. This effect was diminished by ML385 treatment in vitro. Conclusion NBQX can alleviate VF susceptibility in rat models of anxiety by alleviating electrical remodeling, structural remodeling via regulating the Nrf2/HO-1 pathway to some extent.
Collapse
Affiliation(s)
- Yiqian Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Ying Zou
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Cui Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, PR China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, PR China
- Hubei Key Laboratory of Cardiology, Wuhan, PR China
| |
Collapse
|
2
|
An X, Cho H. Increased GIRK channel activity prevents arrhythmia in mice with heart failure by enhancing ventricular repolarization. Sci Rep 2023; 13:22479. [PMID: 38110503 PMCID: PMC10728207 DOI: 10.1038/s41598-023-50088-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 12/15/2023] [Indexed: 12/20/2023] Open
Abstract
Ventricular arrhythmia causing sudden cardiac death is the leading mode of death in patients with heart failure. Yet, the mechanisms that prevent ventricular arrhythmias in heart failure are not well characterized. Using a mouse model of heart failure created by transverse aorta constriction, we show that GIRK channel, an important regulator of cardiac action potentials, is constitutively active in failing ventricles in contrast to normal cells. Evidence is presented indicating that the tonic activation of M2 muscarinic acetylcholine receptors by endogenously released acetylcholine contributes to the constitutive GIRK activity. This constitutive GIRK activity prevents the action potential prolongation in heart failure ventricles. Consistently, GIRK channel blockade with tertiapin-Q induces QT interval prolongation and increases the incidence of arrhythmia in heart failure, but not in control mice. These results suggest that constitutive GIRK channels comprise a key mechanism to protect against arrhythmia by providing repolarizing currents in heart failure ventricles.
Collapse
Affiliation(s)
- Xue An
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea.
| |
Collapse
|
3
|
Tibbs GR, Uprety R, Warren JD, Beyer NP, Joyce RL, Ferrer MA, Mellado W, Wong VSC, Goldberg DC, Cohen MW, Costa CJ, Li Z, Zhang G, Dephoure NE, Barman DN, Sun D, Ingólfsson HI, Sauve AA, Willis DE, Goldstein PA. An anchor-tether 'hindered' HCN1 inhibitor is antihyperalgesic in a rat spared nerve injury neuropathic pain model. Br J Anaesth 2023; 131:745-763. [PMID: 37567808 PMCID: PMC10541997 DOI: 10.1016/j.bja.2023.06.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Neuropathic pain impairs quality of life, is widely prevalent, and incurs significant costs. Current pharmacological therapies have poor/no efficacy and significant adverse effects; safe and effective alternatives are needed. Hyperpolarisation-activated cyclic nucleotide-regulated (HCN) channels are causally implicated in some forms of peripherally mediated neuropathic pain. Whilst 2,6-substituted phenols, such as 2,6-di-tert-butylphenol (26DTB-P), selectively inhibit HCN1 gating and are antihyperalgesic, the development of therapeutically tolerable, HCN-selective antihyperalgesics based on their inverse agonist activity requires that such drugs spare the cardiac isoforms and do not cross the blood-brain barrier. METHODS In silico molecular dynamics simulation, in vitro electrophysiology, and in vivo rat spared nerve injury methods were used to test whether 'hindered' variants of 26DTB-P (wherein a hydrophilic 'anchor' is attached in the para-position of 26DTB-P via an acyl chain 'tether') had the desired properties. RESULTS Molecular dynamics simulation showed that membrane penetration of hindered 26DTB-Ps is controlled by a tethered diol anchor without elimination of head group rotational freedom. In vitro and in vivo analysis showed that BP4L-18:1:1, a variant wherein a diol anchor is attached to 26DTB-P via an 18-carbon tether, is an HCN1 inverse agonist and an orally available antihyperalgesic. With a CNS multiparameter optimisation score of 2.25, a >100-fold lower drug load in the brain vs blood, and an absence of adverse cardiovascular or CNS effects, BP4L-18:1:1 was shown to be poorly CNS penetrant and cardiac sparing. CONCLUSIONS These findings provide a proof-of-concept demonstration that anchor-tethered drugs are a new chemotype for treatment of disorders involving membrane targets.
Collapse
Affiliation(s)
- Gareth R Tibbs
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Rajendra Uprety
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - J David Warren
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Nicole P Beyer
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Rebecca L Joyce
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Matthew A Ferrer
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | | | | | | | | | | | - Zhucui Li
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Guoan Zhang
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Noah E Dephoure
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Dipti N Barman
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Delin Sun
- Lawrence Livermore National Laboratory, Livermore, CA, USA
| | | | - Anthony A Sauve
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Dianna E Willis
- Burke Neurological Institute, White Plains, NY, USA; Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Peter A Goldstein
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA; Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Saleem F, Mansour H, Vichare R, Ayalasomayajula Y, Yassine J, Hesaraghatta A, Panguluri SK. Influence of Age on Hyperoxia-Induced Cardiac Pathophysiology in Type 1 Diabetes Mellitus (T1DM) Mouse Model. Cells 2023; 12:1457. [PMID: 37296578 PMCID: PMC10252211 DOI: 10.3390/cells12111457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/08/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Mechanical ventilation often results in hyperoxia, a condition characterized by excess SpO2 levels (>96%). Hyperoxia results in changes in the physiological parameters, severe cardiac remodeling, arrhythmia development, and alteration of cardiac ion channels, all of which can point toward a gradual increase in the risk of developing cardiovascular disease (CVD). This study extends the analysis of our prior work in young Akita mice, which demonstrated that exposure to hyperoxia worsens cardiac outcomes in a type 1 diabetic murine model as compared to wild-type (WT) mice. Age is an independent risk factor, and when present with a major comorbidity, such as type 1 diabetes (T1D), it can further exacerbate cardiac outcomes. Thus, this research subjected aged T1D Akita mice to clinical hyperoxia and analyzed the cardiac outcomes. Overall, aged Akita mice (60 to 68 weeks) had preexisting cardiac challenges compared to young Akita mice. Aged mice were overweight, had an increased cardiac cross-sectional area, and showed prolonged QTc and JT intervals, which are proposed as major risk factors for CVD like intraventricular arrhythmias. Additionally, exposure to hyperoxia resulted in severe cardiac remodeling and a decrease in Kv 4.2 and KChIP2 cardiac potassium channels in these rodents. Based on sex-specific differences, aged male Akita mice had a higher risk of poor cardiac outcomes than aged females. Aged male Akita mice had prolonged RR, QTc, and JT intervals even at baseline normoxic exposure. Moreover, they were not protected against hyperoxic stress through adaptive cardiac hypertrophy, which, at least to some extent, is due to reduced cardiac androgen receptors. This study in aged Akita mice aims to draw attention to the clinically important yet understudied subject of the effect of hyperoxia on cardiac parameters in the presence of preexisting comorbidities. The findings would help revise the provision of care for older T1D patients admitted to ICUs.
Collapse
Affiliation(s)
- Faizan Saleem
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Hussein Mansour
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Riddhi Vichare
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Yashwant Ayalasomayajula
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Jenna Yassine
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Anagha Hesaraghatta
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Siva Kumar Panguluri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
- Cell Biology, Microbiology and Molecular Biology, College of Arts and Sciences, University of South Florida, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| |
Collapse
|
5
|
Ismaili D, Schulz C, Horváth A, Koivumäki JT, Mika D, Hansen A, Eschenhagen T, Christ T. Human induced pluripotent stem cell-derived cardiomyocytes as an electrophysiological model: Opportunities and challenges-The Hamburg perspective. Front Physiol 2023; 14:1132165. [PMID: 36875015 PMCID: PMC9978010 DOI: 10.3389/fphys.2023.1132165] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Models based on human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) are proposed in almost any field of physiology and pharmacology. The development of human induced pluripotent stem cell-derived cardiomyocytes is expected to become a step forward to increase the translational power of cardiovascular research. Importantly they should allow to study genetic effects on an electrophysiological background close to the human situation. However, biological and methodological issues revealed when human induced pluripotent stem cell-derived cardiomyocytes were used in experimental electrophysiology. We will discuss some of the challenges that should be considered when human induced pluripotent stem cell-derived cardiomyocytes will be used as a physiological model.
Collapse
Affiliation(s)
- Djemail Ismaili
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Carl Schulz
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - András Horváth
- Translational Cardiology, Department of Cardiology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Jussi T. Koivumäki
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Delphine Mika
- Inserm, UMR-S 1180, Université Paris-Saclay, Orsay, France
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Torsten Christ
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
6
|
Jin X, Amoni M, Gilbert G, Dries E, Doñate Puertas R, Tomar A, Nagaraju CK, Pradhan A, Yule DI, Martens T, Menten R, Vanden Berghe P, Rega F, Sipido K, Roderick HL. InsP 3R-RyR Ca 2+ channel crosstalk facilitates arrhythmias in the failing human ventricle. Basic Res Cardiol 2022; 117:60. [PMID: 36378362 DOI: 10.1007/s00395-022-00967-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/13/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022]
Abstract
Dysregulated intracellular Ca2+ handling involving altered Ca2+ release from intracellular stores via RyR channels underlies both arrhythmias and reduced function in heart failure (HF). Mechanisms linking RyR dysregulation and disease are not fully established. Studies in animals support a role for InsP3 receptor Ca2+ channels (InsP3R) in pathological alterations in cardiomyocyte Ca2+ handling but whether these findings translate to the divergent physiology of human cardiomyocytes during heart failure is not determined. Using electrophysiological and Ca2+ recordings in human ventricular cardiomyocytes, we uncovered that Ca2+ release via InsP3Rs facilitated Ca2+ release from RyR and induced arrhythmogenic delayed after depolarisations and action potentials. InsP3R-RyR crosstalk was particularly increased in HF at RyR clusters isolated from the T-tubular network. Reduced SERCA activity in HF further facilitated the action of InsP3. Nanoscale imaging revealed co-localisation of InsP3Rs with RyRs in the dyad, which was increased in HF, providing a mechanism for augmented Ca2+ channel crosstalk. Notably, arrhythmogenic activity dependent on InsP3Rs was increased in tissue wedges from failing hearts perfused with AngII to promote InsP3 generation. These data indicate a central role for InsP3R-RyR Ca2+ signalling crosstalk in the pro-arrhythmic action of GPCR agonists elevated in HF and the potential for their therapeutic targeting.
Collapse
Affiliation(s)
- Xin Jin
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium.,Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Matthew Amoni
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Guillaume Gilbert
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Eef Dries
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Rosa Doñate Puertas
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Ashutosh Tomar
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Chandan K Nagaraju
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Ankit Pradhan
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - David I Yule
- Department of Pharmacology and Physiology, Medical Center School of Medicine and Dentistry, University of Rochester, 601 Elmwood Avenue, Box 711, Rochester, NY, 14642, USA
| | - Tobie Martens
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000, Leuven, Belgium.,Cell and Tissue Imaging Cluster (CIC), KU Leuven, 3000, Leuven, Belgium
| | - Roxane Menten
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, 3000, Leuven, Belgium.,Cell and Tissue Imaging Cluster (CIC), KU Leuven, 3000, Leuven, Belgium
| | - Filip Rega
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium.,Department of Cardiology and Department of Cardiac Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Karin Sipido
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium
| | - H Llewelyn Roderick
- Department of Cardiovascular Sciences, Laboratory of Experimental Cardiology, KU Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
7
|
Rong Z, Chen H, Zhang Z, Zhang Y, Ge L, Lv Z, Zou Y, Lv J, He Y, Li W, Chen L. Identification of cardiomyopathy-related core genes through human metabolic networks and expression data. BMC Genomics 2022; 23:47. [PMID: 35016605 PMCID: PMC8753885 DOI: 10.1186/s12864-021-08271-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 12/15/2021] [Indexed: 12/27/2022] Open
Abstract
Abstract
Background
Cardiomyopathy is a complex type of myocardial disease, and its incidence has increased significantly in recent years. Dilated cardiomyopathy (DCM) and ischemic cardiomyopathy (ICM) are two common and indistinguishable types of cardiomyopathy.
Results
Here, a systematic multi-omics integration approach was proposed to identify cardiomyopathy-related core genes that could distinguish normal, DCM and ICM samples using cardiomyopathy expression profile data based on a human metabolic network. First, according to the differentially expressed genes between different states (DCM/ICM and normal, or DCM and ICM) of samples, three sets of initial modules were obtained from the human metabolic network. Two permutation tests were used to evaluate the significance of the Pearson correlation coefficient difference score of the initial modules, and three candidate modules were screened out. Then, a cardiomyopathy risk module that was significantly related to DCM and ICM was determined according to the significance of the module score based on Markov random field. Finally, based on the shortest path between cardiomyopathy known genes, 13 core genes related to cardiomyopathy were identified. These core genes were enriched in pathways and functions significantly related to cardiomyopathy and could distinguish between samples of different states.
Conclusion
The identified core genes might serve as potential biomarkers of cardiomyopathy. This research will contribute to identifying potential biomarkers of cardiomyopathy and to distinguishing different types of cardiomyopathy.
Collapse
|
8
|
Inducing I to,f and phase 1 repolarization of the cardiac action potential with a Kv4.3/KChIP2.1 bicistronic transgene. J Mol Cell Cardiol 2021; 164:29-41. [PMID: 34823101 PMCID: PMC8884339 DOI: 10.1016/j.yjmcc.2021.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/22/2021] [Accepted: 11/11/2021] [Indexed: 11/22/2022]
Abstract
The fast transient outward potassium current (Ito,f) plays a key role in phase 1 repolarization of the human cardiac action potential (AP) and its reduction in heart failure (HF) contributes to the loss of contractility. Therefore, restoring Ito,f might be beneficial for treating HF. The coding sequence of a P2A peptide was cloned, in frame, between Kv4.3 and KChIP2.1 genes and ribosomal skipping was confirmed by Western blotting. Typical Ito,f properties with slowed inactivation and accelerated recovery from inactivation due to the association of KChIP2.1 with Kv4.3 was seen in transfected HEK293 cells. Both bicistronic components trafficked to the plasmamembrane and in adenovirus transduced rabbit cardiomyocytes both t-tubular and sarcolemmal construct labelling appeared. The resulting current was similar to Ito,f seen in human ventricular cardiomyocytes and was 50% blocked at ~0.8 mmol/l 4-aminopyridine and increased ~30% by 5 μmol/l NS5806 (an Ito,f agonist). Variation in the density of the expressed Ito,f, in rabbit cardiomyocytes recapitulated typical species-dependent variations in AP morphology. Simultaneous voltage recording and intracellular Ca2+ imaging showed that modification of phase 1 to a non-failing human phenotype improved the rate of rise and magnitude of the Ca2+ transient. Ito,f expression also reduced AP triangulation but did not affect ICa,L and INa magnitudes. This raises the possibility for a new gene-based therapeutic approach to HF based on selective phase 1 modification. Action potential phase 1 depends on fast transient outward current (Ito,f). Construction of a bicistronic transgene for Kv4.3 and KChIP2.1 with P2A separator Expressed bicistronic Kv4.3/KChIP2.1 proteins traffic to the cell surface membrane Viral transduction with Kv4.3/KChIP2.1 increases Ito,f in cardiomyocytes. Kv4.3/KChIP2.1 transgene expression increased AP phase 1 and EC coupling
Collapse
|
9
|
Peirlinck M, Sahli Costabal F, Kuhl E. Sex Differences in Drug-Induced Arrhythmogenesis. Front Physiol 2021; 12:708435. [PMID: 34489728 PMCID: PMC8417068 DOI: 10.3389/fphys.2021.708435] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/14/2021] [Indexed: 12/25/2022] Open
Abstract
The electrical activity in the heart varies significantly between men and women and results in a sex-specific response to drugs. Recent evidence suggests that women are more than twice as likely as men to develop drug-induced arrhythmia with potentially fatal consequences. Yet, the sex-specific differences in drug-induced arrhythmogenesis remain poorly understood. Here we integrate multiscale modeling and machine learning to gain mechanistic insight into the sex-specific origin of drug-induced cardiac arrhythmia at differing drug concentrations. To quantify critical drug concentrations in male and female hearts, we identify the most important ion channels that trigger male and female arrhythmogenesis, and create and train a sex-specific multi-fidelity arrhythmogenic risk classifier. Our study reveals that sex differences in ion channel activity, tissue conductivity, and heart dimensions trigger longer QT-intervals in women than in men. We quantify the critical drug concentration for dofetilide, a high risk drug, to be seven times lower for women than for men. Our results emphasize the importance of including sex as an independent biological variable in risk assessment during drug development. Acknowledging and understanding sex differences in drug safety evaluation is critical when developing novel therapeutic treatments on a personalized basis. The general trends of this study have significant implications on the development of safe and efficacious new drugs and the prescription of existing drugs in combination with other drugs.
Collapse
Affiliation(s)
- Mathias Peirlinck
- Department of Mechanical Engineering, Stanford University, Stanford, CA, United States
| | - Francisco Sahli Costabal
- Department of Mechanical and Metallurgical Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Nucleus for Cardiovascular Magnetic Resonance, Santiago, Chile
| | - Ellen Kuhl
- Department of Mechanical Engineering, Stanford University, Stanford, CA, United States
| |
Collapse
|
10
|
Zhu Y, Bai J, Lo A, Lu Y, Zhao J. Mechanisms underlying pro-arrhythmic abnormalities arising from Pitx2-induced electrical remodelling: an in silico intersubject variability study. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:106. [PMID: 33569408 PMCID: PMC7867875 DOI: 10.21037/atm-20-5660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Background Electrical remodelling as a result of the homeodomain transcription factor 2 (Pitx2)-dependent gene regulation induces atrial fibrillation (AF) with different mechanisms. The purpose of this study was to identify Pitx2-induced changes in ionic currents that cause action potential (AP) shortening and lead to triggered activity. Methods Populations of computational atrial AP models were developed based on AP recordings from sinus rhythm (SR) and AF patients. Models in the AF population were divided into triggered and untriggered AP groups to evaluate the relationship between each ion current regulated by Pitx2 and triggered APs. Untriggered AP models were then divided into shortened and unshortened AP groups to determine which Pitx2-dependent ion currents contribute to AP shortening. Results According to the physiological range of AP biomarkers measured experimentally, populations of 2,885 SR and 4,781 AF models out of the initial pool of 30,000 models were selected. Models in the AF population predicted AP shortening and triggered activity observed in experiments in Pitx2-induced remodelling conditions. The AF models included 925 triggered AP models, 1,412 shortened AP models and 2,444 unshortened AP models. Intersubject variability in IKs and ICaL primarily modulated variability in AP duration (APD) in all shortened and unshortened AP models, whereas intersubject variability in IK1 and SERCA mainly contributed to the variability in AP morphology in all triggered and untriggered AP models. The incidence of shortened AP was positively correlated with IKs and IK1 and was negatively correlated with INa , ICaL and SERCA, whereas the incidence of triggered AP was negatively correlated with IKs and IK1 and was positively correlated with INa , ICaL and SERCA. Conclusions Electrical remodelling due to Pitx2 upregulation may increase the incidence of shortened AP, whereas electrical remodelling arising from Pitx2 downregulation may favor to the genesis of triggered AP.
Collapse
Affiliation(s)
- Yijie Zhu
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou, China
| | - Jieyun Bai
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou, China
| | - Andy Lo
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Yaosheng Lu
- Department of Electronic Engineering, College of Information Science and Technology, Jinan University, Guangzhou, China
| | - Jichao Zhao
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
11
|
Lipovsky CE, Jimenez J, Guo Q, Li G, Yin T, Hicks SC, Bhatnagar S, Takahashi K, Zhang DM, Brumback BD, Goldsztejn U, Nadadur RD, Perez-Cervantez C, Moskowitz IP, Liu S, Zhang B, Rentschler SL. Chamber-specific transcriptional responses in atrial fibrillation. JCI Insight 2020; 5:135319. [PMID: 32841220 PMCID: PMC7526559 DOI: 10.1172/jci.insight.135319] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 08/19/2020] [Indexed: 12/30/2022] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia, yet the molecular signature of the vulnerable atrial substrate is not well understood. Here, we delineated a distinct transcriptional signature in right versus left atrial cardiomyocytes (CMs) at baseline and identified chamber-specific gene expression changes in patients with a history of AF in the setting of end-stage heart failure (AF+HF) that are not present in heart failure alone (HF). We observed that human left atrial (LA) CMs exhibited Notch pathway activation and increased ploidy in AF+HF but not in HF alone. Transient activation of Notch signaling within adult CMs in a murine genetic model is sufficient to increase ploidy in both atrial chambers. Notch activation within LA CMs generated a transcriptomic fingerprint resembling AF, with dysregulation of transcription factor and ion channel genes, including Pitx2, Tbx5, Kcnh2, Kcnq1, and Kcnip2. Notch activation also produced distinct cellular electrophysiologic responses in LA versus right atrial CMs, prolonging the action potential duration (APD) without altering the upstroke velocity in the left atrium and reducing the maximal upstroke velocity without altering the APD in the right atrium. Our results support a shared human/murine model of increased Notch pathway activity predisposing to AF. Distinct transcriptional changes occur in human left versus right atrial cardiomyocytes in atrial fibrillation, including Notch pathway activation, which alters electric properties and ploidy in murine models.
Collapse
Affiliation(s)
- Catherine E Lipovsky
- Department of Medicine, Cardiovascular Division.,Department of Developmental Biology, and
| | | | - Qiusha Guo
- Department of Medicine, Cardiovascular Division
| | - Gang Li
- Department of Medicine, Cardiovascular Division.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Tiankai Yin
- Department of Medicine, Cardiovascular Division
| | | | - Somya Bhatnagar
- Department of Medicine, Cardiovascular Division.,Department of Developmental Biology, and
| | | | | | - Brittany D Brumback
- Department of Medicine, Cardiovascular Division.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Uri Goldsztejn
- Department of Medicine, Cardiovascular Division.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Rangarajan D Nadadur
- Departments of Pediatrics, Pathology, and Human Genetics, Biological Sciences Division, University of Chicago, Chicago, Illinois, USA
| | - Carlos Perez-Cervantez
- Departments of Pediatrics, Pathology, and Human Genetics, Biological Sciences Division, University of Chicago, Chicago, Illinois, USA
| | - Ivan P Moskowitz
- Departments of Pediatrics, Pathology, and Human Genetics, Biological Sciences Division, University of Chicago, Chicago, Illinois, USA
| | | | - Bo Zhang
- Department of Developmental Biology, and
| | - Stacey L Rentschler
- Department of Medicine, Cardiovascular Division.,Department of Developmental Biology, and.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
12
|
Abstract
Changes of intracellular Ca2+ concentration regulate many aspects of cardiac myocyte function. About 99% of the cytoplasmic calcium in cardiac myocytes is bound to buffers, and their properties will therefore have a major influence on Ca2+ signaling. This article considers the fundamental properties and identities of the buffers and how to measure them. It reviews the effects of buffering on the systolic Ca2+ transient and how this may change physiologically, and in heart failure and both atrial and ventricular arrhythmias, as well. It is concluded that the consequences of this strong buffering may be more significant than currently appreciated, and a fuller understanding is needed for proper understanding of cardiac calcium cycling and contractility.
Collapse
Affiliation(s)
- Godfrey L Smith
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, UK (G.L.S.)
| | - David A Eisner
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, UK (D.A.E.)
| |
Collapse
|
13
|
Yuxiang L, Fujiu K. Frozen Heart and Arrhythmia. Int Heart J 2019; 60:1019-1021. [PMID: 31564707 DOI: 10.1536/ihj.19-407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Liu Yuxiang
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Katsuhito Fujiu
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo.,Department of Advanced Cardiology, Graduate School of Medicine, The University of Tokyo
| |
Collapse
|
14
|
Lee J, Kim S, Kim HM, Kim HJ, Yu FH. NaV1.6 and NaV1.7 channels are major endogenous voltage-gated sodium channels in ND7/23 cells. PLoS One 2019; 14:e0221156. [PMID: 31419255 PMCID: PMC6697327 DOI: 10.1371/journal.pone.0221156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/31/2019] [Indexed: 01/02/2023] Open
Abstract
ND7/23 cells are gaining traction as a host model to express peripheral sodium channels such as NaV1.8 and NaV1.9 that have been difficult to express in widely utilized heterologous cells, like CHO and HEK293. Use of ND7/23 as a model cell to characterize the properties of sodium channels requires clear understanding of the endogenous ion channels. To define the nature of the background sodium currents in ND7/23 cells, we aimed to comprehensively profile the voltage-gated sodium channel subunits by endpoint and quantitative reverse transcription-PCR and by whole-cell patch clamp electrophysiology. We found that untransfected ND7/23 cells express endogenous peak sodium currents that average -2.12nA (n = 15) and with kinetics typical of fast sodium currents having activation and inactivation completed within few milliseconds. Furthermore, sodium currents were reduced to virtually nil upon exposure to 100nM tetrodotoxin, indicating that ND7/23 cells have essentially null background for tetrodotoxin-resistant (TTX-R) currents. qRT-PCR profiling indicated a major expression of TTX-sensitive (TTX-S) NaV1.6 and NaV1.7 at similar levels and very low expression of TTX-R NaV1.9 transcripts. There was no expression of TTX-R NaV1.8 in ND7/23 cells. There was low expression of NaV1.1, NaV1.2, NaV1.3 and no expression of cardiac or skeletal muscle sodium channels. As for the sodium channel auxiliary subunits, β1 and β3 subunits were expressed, but not the β2 and β4 subunits that covalently associate with the α-subunits. In addition, our results also showed that only the mouse forms of NaV1.6, NaV1.7 and NaV1.9 sodium channels were expressed in ND7/23 cells that was originally generated as a hybridoma of rat embryonic DRG and mouse neuroblastoma cell-line. By molecular profiling of auxiliary β- and principal α-subunits of the voltage gated sodium channel complex, our results define the background sodium channels expressed in ND7/23 cells, and confirm their utility for detailed functional studies of emerging pain channelopathies ascribed to mutations of the TTX-R sodium channels of sensory neurons.
Collapse
Affiliation(s)
- Jisoo Lee
- Department of Pharmacology and Dental Therapeutics, Program in Neurobiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Shinae Kim
- Department of Pharmacology and Dental Therapeutics, Program in Neurobiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Hye-mi Kim
- Department of Pharmacology and Dental Therapeutics, Program in Neurobiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Hyun Jeong Kim
- Department of Dental Anesthesiology, Program in Neurobiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Frank H. Yu
- Department of Pharmacology and Dental Therapeutics, Program in Neurobiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
15
|
Calloe K. Doctoral Dissertation: The transient outward potassium current in healthy and diseased hearts. Acta Physiol (Oxf) 2019; 225 Suppl 717:e13225. [PMID: 30628199 DOI: 10.1111/apha.13225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Kirstine Calloe
- Section for Anatomy; Biochemistry and Physiology; Department for Veterinary and Animal Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Frederiksberg C Denmark
| |
Collapse
|
16
|
David JP, Lisewski U, Crump SM, Jepps TA, Bocksteins E, Wilck N, Lossie J, Roepke TK, Schmitt N, Abbott GW. Deletion in mice of X-linked, Brugada syndrome- and atrial fibrillation-associated Kcne5 augments ventricular K V currents and predisposes to ventricular arrhythmia. FASEB J 2018; 33:2537-2552. [PMID: 30289750 DOI: 10.1096/fj.201800502r] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
KCNE5 is an X-linked gene encoding KCNE5, an ancillary subunit to voltage-gated potassium (KV) channels. Human KCNE5 mutations are associated with atrial fibrillation (AF)- and Brugada syndrome (BrS)-induced cardiac arrhythmias that can arise from increased potassium current in cardiomyocytes. Seeking to establish underlying molecular mechanisms, we created and studied Kcne5 knockout ( Kcne5-/0) mice. Intracardiac ECG revealed that Kcne5 deletion caused ventricular premature beats, increased susceptibility to induction of polymorphic ventricular tachycardia (60 vs. 24% in Kcne5+/0 mice), and 10% shorter ventricular refractory period. Kcne5 deletion increased mean ventricular myocyte KV current density in the apex and also in the subpopulation of septal myocytes that lack fast transient outward current ( Ito,f). The current increases arose from an apex-specific increase in slow transient outward current-1 ( IKslow,1) (conducted by KV1.5) and Ito,f (conducted by KV4) and an increase in IKslow,2 (conducted by KV2.1) in both apex and septum. Kcne5 protein localized to the intercalated discs in ventricular myocytes, where KV2.1 was also detected in both Kcne5-/0 and Kcne5+/0 mice. In HL-1 cardiac cells and human embryonic kidney cells, KCNE5 and KV2.1 colocalized at the cell surface, but predominantly in intracellular vesicles, suggesting that Kcne5 deletion increases IK,slow2 by reducing KV2.1 intracellular sequestration. The human AF-associated mutation KCNE5-L65F negative shifted the voltage dependence of KV2.1-KCNE5 channels, increasing their maximum current density >2-fold, whereas BrS-associated KCNE5 mutations produced more subtle negative shifts in KV2.1 voltage dependence. The findings represent the first reported native role for Kcne5 and the first demonstrated Kcne regulation of KV2.1 in mouse heart. Increased KV current is a manifestation of KCNE5 disruption that is most likely common to both mouse and human hearts, providing a plausible mechanistic basis for human KCNE5-linked AF and BrS.-David, J.-P., Lisewski, U., Crump, S. M., Jepps, T. A., Bocksteins, E., Wilck, N., Lossie, J., Roepke, T. K., Schmitt, N., Abbott, G. W. Deletion in mice of X-linked, Brugada syndrome- and atrial fibrillation-associated Kcne5 augments ventricular KV currents and predisposes to ventricular arrhythmia.
Collapse
Affiliation(s)
- Jens-Peter David
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulrike Lisewski
- Medical Clinic and Polyclinic for Cardiology and Angiology, Charité Medical University of Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC), Charité Medical University of Berlin, Berlin, Germany
| | - Shawn M Crump
- Bioelectricity Laboratory, Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA; and
| | - Thomas A Jepps
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Elke Bocksteins
- Laboratory for Molecular Biophysics, Physiology, and Pharmacology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Nicola Wilck
- Medical Clinic and Polyclinic for Cardiology and Angiology, Charité Medical University of Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC), Charité Medical University of Berlin, Berlin, Germany
| | - Janine Lossie
- Medical Clinic and Polyclinic for Cardiology and Angiology, Charité Medical University of Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC), Charité Medical University of Berlin, Berlin, Germany
| | - Torsten K Roepke
- Medical Clinic and Polyclinic for Cardiology and Angiology, Charité Medical University of Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC), Charité Medical University of Berlin, Berlin, Germany
| | - Nicole Schmitt
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA; and
| |
Collapse
|
17
|
Okada JI, Yoshinaga T, Kurokawa J, Washio T, Furukawa T, Sawada K, Sugiura S, Hisada T. Arrhythmic hazard map for a 3D whole-ventricle model under multiple ion channel block. Br J Pharmacol 2018; 175:3435-3452. [PMID: 29745425 PMCID: PMC6086978 DOI: 10.1111/bph.14357] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 03/12/2018] [Accepted: 04/20/2018] [Indexed: 01/05/2023] Open
Abstract
Background and Purpose To date, proposed in silico models for preclinical cardiac safety testing are limited in their predictability and usability. We previously reported a multi‐scale heart simulation that accurately predicts arrhythmogenic risk for benchmark drugs. Experimental Approach We created a comprehensive hazard map of drug‐induced arrhythmia based on the electrocardiogram (ECG) waveforms simulated under wide range of drug effects using the multi‐scale heart simulator described here, implemented with cell models of human cardiac electrophysiology. Key Results A total of 9075 electrocardiograms constitute the five‐dimensional hazard map, with coordinates representing the extent of the block of each of the five ionic currents (rapid delayed rectifier potassium current (IKr), fast (INa) and late (INa,L) components of the sodium current, L‐type calcium current (ICa,L) and slow delayed rectifier current (IKs)), involved in arrhythmogenesis. Results of the evaluation of arrhythmogenic risk based on this hazard map agreed well with the risk assessments reported in the literature. ECG databases also suggested that the interval between the J‐point and the T‐wave peak is a superior index of arrhythmogenicity when compared to the QT interval due to its ability to characterize the multi‐channel effects compared with QT interval. Conclusion and Implications Because concentration‐dependent effects on electrocardiograms of any drug can be traced on this map based on in vitro current assay data, its arrhythmogenic risk can be evaluated without performing costly and potentially risky human electrophysiological assays. Hence, the map serves as a novel tool for use in pharmaceutical research and development.
Collapse
Affiliation(s)
- Jun-Ichi Okada
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan.,UT-Heart Inc., Tokyo, Japan
| | | | - Junko Kurokawa
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Takumi Washio
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan.,UT-Heart Inc., Tokyo, Japan
| | - Tetsushi Furukawa
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kohei Sawada
- Global CV Assessment, Eisai Co., Ltd., Ibaraki, Japan
| | - Seiryo Sugiura
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan.,UT-Heart Inc., Tokyo, Japan
| | | |
Collapse
|
18
|
Ortega A, Tarazón E, Gil-Cayuela C, García-Manzanares M, Martínez-Dolz L, Lago F, González-Juanatey JR, Cinca J, Jorge E, Portolés M, Roselló-Lletí E, Rivera M. Intercalated disc in failing hearts from patients with dilated cardiomyopathy: Its role in the depressed left ventricular function. PLoS One 2017; 12:e0185062. [PMID: 28934278 PMCID: PMC5608295 DOI: 10.1371/journal.pone.0185062] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 09/06/2017] [Indexed: 11/19/2022] Open
Abstract
Alterations in myocardial structure and reduced cardiomyocyte adhesions have been previously described in dilated cardiomyopathy (DCM). We studied the transcriptome of cell adhesion molecules in these patients and their relationships with left ventricular (LV) function decay. We also visualized the intercalated disc (ID) structure and organization. The transcriptomic profile of 23 explanted LV samples was analyzed using RNA-sequencing (13 DCM, 10 control [CNT]), focusing on cell adhesion genes. Electron microscopy analysis to visualize ID structural differences and immunohistochemistry experiments of ID proteins was also performed. RT-qPCR and western blot experiments were carried out on ID components. We found 29 differentially expressed genes, most of all, constituents of the ID structure. We found that the expression of GJA3, DSP and CTNNA3 was directly associated with LV ejection fraction (r = 0.741, P = 0.004; r = 0.674, P = 0.011 and r = 0.565, P = 0.044, respectively), LV systolic (P = 0.003, P = 0.003, P = 0.028, respectively) and diastolic dimensions (P = 0.006, P = 0.001, P = 0.025, respectively). Electron microscopy micrographs showed a reduced ID convolution index and immunogold labeling of connexin 46 (GJA gene), desmoplakin (DSP gene) and catenin α-3 (CTNNA3 gene) proteins in DCM patients. Moreover, we observed that protein and mRNA levels analyzed by RT-qPCR of these ID components were diminished in DCM group. In conclusion, we report significant gene and protein expression changes and found that the ID components GJA3, DSP and CTNNA3 were highly related to LV function. Microscopic observations indicated that ID is structurally compromised in these patients. These findings give new data for understanding the ventricular depression that characterizes DCM, opening new therapeutic perspectives for these critically diseased patients.
Collapse
Affiliation(s)
- Ana Ortega
- Cardiocirculatory Unit, Health Research Institute La Fe, Valencia, Spain
- Center for Biomedical Research Network in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Estefanía Tarazón
- Cardiocirculatory Unit, Health Research Institute La Fe, Valencia, Spain
- Center for Biomedical Research Network in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Carolina Gil-Cayuela
- Cardiocirculatory Unit, Health Research Institute La Fe, Valencia, Spain
- Center for Biomedical Research Network in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - María García-Manzanares
- Cardiocirculatory Unit, Health Research Institute La Fe, Valencia, Spain
- Center for Biomedical Research Network in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Luis Martínez-Dolz
- Center for Biomedical Research Network in Cardiovascular Diseases (CIBERCV), Madrid, Spain
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, Valencia, Spain
| | - Francisca Lago
- Center for Biomedical Research Network in Cardiovascular Diseases (CIBERCV), Madrid, Spain
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - José Ramón González-Juanatey
- Center for Biomedical Research Network in Cardiovascular Diseases (CIBERCV), Madrid, Spain
- Cellular and Molecular Cardiology Research Unit, Department of Cardiology and Institute of Biomedical Research, University Clinical Hospital, Santiago de Compostela, Spain
| | - Juan Cinca
- Center for Biomedical Research Network in Cardiovascular Diseases (CIBERCV), Madrid, Spain
- Cardiology Service of Santa Creu i Sant Pau Hospital, Barcelona, Spain
| | - Esther Jorge
- Center for Biomedical Research Network in Cardiovascular Diseases (CIBERCV), Madrid, Spain
- Cardiology Service of Santa Creu i Sant Pau Hospital, Barcelona, Spain
| | - Manuel Portolés
- Cardiocirculatory Unit, Health Research Institute La Fe, Valencia, Spain
- Center for Biomedical Research Network in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Esther Roselló-Lletí
- Cardiocirculatory Unit, Health Research Institute La Fe, Valencia, Spain
- Center for Biomedical Research Network in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Miguel Rivera
- Cardiocirculatory Unit, Health Research Institute La Fe, Valencia, Spain
- Center for Biomedical Research Network in Cardiovascular Diseases (CIBERCV), Madrid, Spain
- * E-mail:
| |
Collapse
|
19
|
Significance of integrated in silico transmural ventricular wedge preparation models of human non-failing and failing hearts for safety evaluation of drug candidates. J Pharmacol Toxicol Methods 2017; 83:30-41. [DOI: 10.1016/j.vascn.2016.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/08/2016] [Accepted: 08/17/2016] [Indexed: 01/20/2023]
|
20
|
Transmural electrophysiological heterogeneity, the T-wave and ventricular arrhythmias. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 122:202-214. [DOI: 10.1016/j.pbiomolbio.2016.05.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/21/2016] [Accepted: 05/03/2016] [Indexed: 01/05/2023]
|
21
|
McKinnon D, Rosati B. Transmural gradients in ion channel and auxiliary subunit expression. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 122:165-186. [PMID: 27702655 DOI: 10.1016/j.pbiomolbio.2016.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 09/30/2016] [Indexed: 12/11/2022]
Abstract
Evolution has acted to shape the action potential in different regions of the heart in order to produce a maximally stable and efficient pump. This has been achieved by creating regional differences in ion channel expression levels within the heart as well as differences between equivalent cardiac tissues in different species. These region- and species-dependent differences in channel expression are established by regulatory evolution, evolution of the regulatory mechanisms that control channel expression levels. Ion channel auxiliary subunits are obvious targets for regulatory evolution, in order to change channel expression levels and/or modify channel function. This review focuses on the transmural gradients of ion channel expression in the heart and the role that regulation of auxiliary subunit expression plays in generating and shaping these gradients.
Collapse
Affiliation(s)
- David McKinnon
- Department of Veterans Affairs Medical Center, Northport, NY, USA; Institute of Molecular Cardiology, Stony Brook University, Stony Brook, NY, USA; Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Barbara Rosati
- Department of Veterans Affairs Medical Center, Northport, NY, USA; Institute of Molecular Cardiology, Stony Brook University, Stony Brook, NY, USA; Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
22
|
Ortega A, Gil-Cayuela C, Tarazón E, García-Manzanares M, Montero JA, Cinca J, Portolés M, Rivera M, Roselló-Lletí E. New Cell Adhesion Molecules in Human Ischemic Cardiomyopathy. PCDHGA3 Implications in Decreased Stroke Volume and Ventricular Dysfunction. PLoS One 2016; 11:e0160168. [PMID: 27472518 PMCID: PMC4966940 DOI: 10.1371/journal.pone.0160168] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/14/2016] [Indexed: 11/25/2022] Open
Abstract
Background Intercalated disks are unique structures in cardiac tissue, in which adherens junctions, desmosomes, and GAP junctions co-localize, thereby facilitating cardiac muscle contraction and function. Protocadherins are involved in these junctions; however, their role in heart physiology is poorly understood. We aimed to analyze the transcriptomic profile of adhesion molecules in patients with ischemic cardiomyopathy (ICM) and relate the changes uncovered with the hemodynamic alterations and functional depression observed in these patients. Methods and Results Twenty-three left ventricular tissue samples from patients diagnosed with ICM (n = 13) undergoing heart transplantation and control donors (CNT, n = 10) were analyzed using RNA sequencing. Forty-two cell adhesion genes involved in cellular junctions were differentially expressed in ICM myocardium. Notably, the levels of protocadherin PCDHGA3 were related with the stroke volume (r = –0.826, P = 0.003), ejection fraction (r = –0.793, P = 0.004) and left ventricular end systolic and diastolic diameters (r = 0.867, P = 0.001; r = 0.781, P = 0.005, respectively). Conclusions Our results support the importance of intercalated disks molecular alterations, closely involved in the contractile function, highlighting its crucial significance and showing gene expression changes not previously described. Specifically, altered PCDHGA3 gene expression was strongly associated with reduced stroke volume and ventricular dysfunction in ICM, suggesting a relevant role in hemodynamic perturbations and cardiac performance for this unexplored protocadherin.
Collapse
Affiliation(s)
- Ana Ortega
- Cardiocirculatory Unit, The Health Research Institute La Fe, Valencia, Spain
| | | | - Estefanía Tarazón
- Cardiocirculatory Unit, The Health Research Institute La Fe, Valencia, Spain
| | | | - José Anastasio Montero
- Cardiovascular Surgery Service, University and Polytechnic La Fe Hospital, Valencia, Spain
| | - Juan Cinca
- Cardiology Service of Santa Creu i Sant Pau Hospital, Barcelona, Spain
| | - Manuel Portolés
- Cardiocirculatory Unit, The Health Research Institute La Fe, Valencia, Spain
| | - Miguel Rivera
- Cardiocirculatory Unit, The Health Research Institute La Fe, Valencia, Spain
| | - Esther Roselló-Lletí
- Cardiocirculatory Unit, The Health Research Institute La Fe, Valencia, Spain
- * E-mail:
| |
Collapse
|
23
|
Holzem KM, Gomez JF, Glukhov AV, Madden EJ, Koppel AC, Ewald GA, Trenor B, Efimov IR. Reduced response to IKr blockade and altered hERG1a/1b stoichiometry in human heart failure. J Mol Cell Cardiol 2016; 96:82-92. [PMID: 26093152 PMCID: PMC4683114 DOI: 10.1016/j.yjmcc.2015.06.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 05/27/2015] [Accepted: 06/01/2015] [Indexed: 01/27/2023]
Abstract
Heart failure (HF) claims 250,000 lives per year in the US, and nearly half of these deaths are sudden and presumably due to ventricular tachyarrhythmias. QT interval and action potential (AP) prolongation are hallmark proarrhythmic changes in the failing myocardium, which potentially result from alterations in repolarizing potassium currents. Thus, we aimed to examine whether decreased expression of the rapid delayed rectifier potassium current, IKr, contributes to repolarization abnormalities in human HF. To map functional IKr expression across the left ventricle (LV), we optically imaged coronary-perfused LV free wall from donor and end-stage failing human hearts. The LV wedge preparation was used to examine transmural AP durations at 80% repolarization (APD80), and treatment with the IKr-blocking drug, E-4031, was utilized to interrogate functional expression. We assessed the percent change in APD80 post-IKr blockade relative to baseline APD80 (∆APD80) and found that ∆APD80s are reduced in failing versus donor hearts in each transmural region, with 0.35-, 0.43-, and 0.41-fold reductions in endo-, mid-, and epicardium, respectively (p=0.008, 0.037, and 0.022). We then assessed hERG1 isoform gene and protein expression levels using qPCR and Western blot. While we did not observe differences in hERG1a or hERG1b gene expression between donor and failing hearts, we found a shift in the hERG1a:hERG1b isoform stoichiometry at the protein level. Computer simulations were then conducted to assess IKr block under E-4031 influence in failing and nonfailing conditions. Our results confirmed the experimental observations and E-4031-induced relative APD80 prolongation was greater in normal conditions than in failing conditions, provided that the cellular model of HF included a significant downregulation of IKr. In human HF, the response to IKr blockade is reduced, suggesting decreased functional IKr expression. This attenuated functional response is associated with altered hERG1a:hERG1b protein stoichiometry in the failing human LV, and failing cardiomyoctye simulations support the experimental findings. Thus, of IKr protein and functional expression may be important determinants of repolarization remodeling in the failing human LV.
Collapse
Affiliation(s)
- Katherine M Holzem
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Juan F Gomez
- Polytechnic University of Valencia, Valencia, Spain
| | - Alexey V Glukhov
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Eli J Madden
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Aaron C Koppel
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Gregory A Ewald
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | | | - Igor R Efimov
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA; Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia.
| |
Collapse
|
24
|
Potassium Channel Interacting Protein 2 (KChIP2) is not a transcriptional regulator of cardiac electrical remodeling. Sci Rep 2016; 6:28760. [PMID: 27349185 PMCID: PMC4923891 DOI: 10.1038/srep28760] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/08/2016] [Indexed: 12/20/2022] Open
Abstract
The heart-failure relevant Potassium Channel Interacting Protein 2 (KChIP2) augments CaV1.2 and KV4.3. KChIP3 represses CaV1.2 transcription in cardiomyocytes via interaction with regulatory DNA elements. Hence, we tested nuclear presence of KChIP2 and if KChIP2 translocates into the nucleus in a Ca2+ dependent manner. Cardiac biopsies from human heart-failure patients and healthy donor controls showed that nuclear KChIP2 abundance was significantly increased in heart failure; however, this was secondary to a large variation of total KChIP2 content. Administration of ouabain did not increase KChIP2 content in nuclear protein fractions in anesthetized mice. KChIP2 was expressed in cell lines, and Ca2+ ionophores were applied in a concentration- and time-dependent manner. The cell lines had KChIP2-immunoreactive protein in the nucleus in the absence of treatments to modulate intracellular Ca2+ concentration. Neither increasing nor decreasing intracellular Ca2+ concentrations caused translocation of KChIP2. Microarray analysis did not identify relief of transcriptional repression in murine KChIP2−/− heart samples. We conclude that although there is a baseline presence of KChIP2 in the nucleus both in vivo and in vitro, KChIP2 does not directly regulate transcriptional activity. Moreover, the nuclear transport of KChIP2 is not dependent on Ca2+. Thus, KChIP2 does not function as a conventional transcription factor in the heart.
Collapse
|
25
|
Elshrif MM, Cherry EM. Electrophysiological properties under heart failure conditions in a human ventricular cell: a modeling study. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2015; 2014:4324-9. [PMID: 25570949 DOI: 10.1109/embc.2014.6944581] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Heart failure (HF) is one of the major diseases across the world. During HF the electrophysiology of the failing heart is remodeled, which renders the heart more susceptible to ventricular arrhythmias. In this study, we quantitatively analyze the effects of electrophysiological remodeling of the major currents of human ventricular myocytes on the dynamics of the failing heart. We develop a HF model using a modified version of a recently published model of the human ventricular action potential, the O'Hara-Virag-Varro-Rudy (OVVR) model. The proposed HF model incorporates recently available HF clinical data. It can reproduce most of the action potential (AP) properties of failing myocytes, including action potential duration (APD), amplitude (APA), notch (APN), plateau (APP), resting membrane potential (RMP), and maximum upstroke velocity (dV/dtmax). In addition, the model reproduces the behavior of the [Na+], concentration and [Ca(2)+]i dynamics. Moreover, the HF model exhibits alternans with a fast pacing frequency and can induce early afterdepolarizations (EADs). Additionally, blocking the late sodium current shortens the APD and suppresses EADs, in agreement with experimental findings. The dynamics of the proposed model are assessed through investigating the rate dependence of the AP and the dynamics of the major currents. The steady-state (S-S) and S1-S2 restitution curves along with accommodation to an abrupt change in cycle length were evaluated. Our study should help to elucidate the roles of alterations in electrophysiological properties during HF. Also, this HF cellular model could be used to study HF in a realistic geometry and could be embedded into a model of HF electromechanics to investigate electrical and mechanical properties simultaneously during HF.
Collapse
|
26
|
Abstract
Optimal cardiac function depends on proper timing of excitation and contraction in various regions of the heart, as well as on appropriate heart rate. This is accomplished via specialized electrical properties of various components of the system, including the sinoatrial node, atria, atrioventricular node, His-Purkinje system, and ventricles. Here we review the major regionally determined electrical properties of these cardiac regions and present the available data regarding the molecular and ionic bases of regional cardiac function and dysfunction. Understanding these differences is of fundamental importance for the investigation of arrhythmia mechanisms and pharmacotherapy.
Collapse
Affiliation(s)
- Daniel C Bartos
- Department of Pharmacology, University of California Davis, Davis, California, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, California, USA
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California Davis, Davis, California, USA
| |
Collapse
|
27
|
Poulet C, Wettwer E, Grunnet M, Jespersen T, Fabritz L, Matschke K, Knaut M, Ravens U. Late Sodium Current in Human Atrial Cardiomyocytes from Patients in Sinus Rhythm and Atrial Fibrillation. PLoS One 2015; 10:e0131432. [PMID: 26121051 PMCID: PMC4485891 DOI: 10.1371/journal.pone.0131432] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/01/2015] [Indexed: 12/19/2022] Open
Abstract
Slowly inactivating Na+ channels conducting “late” Na+ current (INa,late) contribute to ventricular arrhythmogenesis under pathological conditions. INa,late was also reported to play a role in chronic atrial fibrillation (AF). The objective of this study was to investigate INa,late in human right atrial cardiomyocytes as a putative drug target for treatment of AF. To activate Na+ channels, cardiomyocytes from transgenic mice which exhibit INa,late (ΔKPQ), and right atrial cardiomyocytes from patients in sinus rhythm (SR) and AF were voltage clamped at room temperature by 250-ms long test pulses to -30 mV from a holding potential of -80 mV with a 100-ms pre-pulse to -110 mV (protocol I). INa,late at -30 mV was not discernible as deviation from the extrapolated straight line IV-curve between -110 mV and -80 mV in human atrial cells. Therefore, tetrodotoxin (TTX, 10 μM) was used to define persistent inward current after 250 ms at -30 mV as INa,late. TTX-sensitive current was 0.27±0.06 pA/pF in ventricular cardiomyocytes from ΔKPQ mice, and amounted to 0.04±0.01 pA/pF and 0.09±0.02 pA/pF in SR and AF human atrial cardiomyocytes, respectively. With protocol II (holding potential -120 mV, pre-pulse to -80 mV) TTX-sensitive INa,late was always larger than with protocol I. Ranolazine (30 μM) reduced INa,late by 0.02±0.02 pA/pF in SR and 0.09±0.02 pA/pF in AF cells. At physiological temperature (37°C), however, INa,late became insignificant. Plateau phase and upstroke velocity of action potentials (APs) recorded with sharp microelectrodes in intact human trabeculae were more sensitive to ranolazine in AF than in SR preparations. Sodium channel subunits expression measured with qPCR was high for SCN5A with no difference between SR and AF. Expression of SCN8A and SCN10A was low in general, and lower in AF than in SR. In conclusion, We confirm for the first time a TTX-sensitive current (INa,late) in right atrial cardiomyocytes from SR and AF patients at room temperature, but not at physiological temperature. While our study provides evidence for the presence of INa,late in human atria, the potential of such current as a target for the treatment of AF remains to be demonstrated.
Collapse
Affiliation(s)
- Claire Poulet
- Department of Pharmacology and Toxicology, Medical Faculty, TU Dresden, Dresden, Germany
| | - Erich Wettwer
- Department of Pharmacology and Toxicology, Medical Faculty, TU Dresden, Dresden, Germany
| | - Morten Grunnet
- Danish Arrhythmia Research Centre, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Jespersen
- Danish Arrhythmia Research Centre, University of Copenhagen, Copenhagen, Denmark
| | - Larissa Fabritz
- Centre for Cardiovascular Sciences, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Klaus Matschke
- Clinic for Cardiac Surgery, Heart Center Dresden, Dresden, Germanymailto
| | - Michael Knaut
- Clinic for Cardiac Surgery, Heart Center Dresden, Dresden, Germanymailto
| | - Ursula Ravens
- Department of Pharmacology and Toxicology, Medical Faculty, TU Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
28
|
Elshrif MM, Shi P, Cherry EM. Representing variability and transmural differences in a model of human heart failure. IEEE J Biomed Health Inform 2015; 19:1308-20. [PMID: 26068919 DOI: 10.1109/jbhi.2015.2442833] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
During heart failure (HF) at the cellular level, the electrophysiological properties of single myocytes get remodeled, which can trigger the occurrence of ventricular arrhythmias that could be manifested in many forms such as early afterdepolarizations (EADs) and alternans (ALTs). In this paper, based on experimentally observed human HF data, specific ionic and exchanger current strengths are modified from a recently developed human ventricular cell model: the O'Hara-Virág-Varró-Rudy (OVVR) model. A new transmural HF-OVVR model is developed that incorporates HF changes and variability of the observed remodeling. This new heterogeneous HF-OVVR model is able to replicate many of the failing action potential (AP) properties and the dynamics of both [Ca(2+)]i and [Na(+)]i in accordance with experimental data. Moreover, it is able to generate EADs for different cell types and exhibits ALTs at modest pacing rate for transmural cell types. We have assessed the HF-OVVR model through the examination of the AP duration and the major ionic currents' rate dependence in single myocytes. The evaluation of the model comes from utilizing the steady-state (S-S) and S1-S2 restitution curves and from probing the accommodation of the HF-OVVR model to an abrupt change in cycle length. In addition, we have investigated the effect of chosen currents on the AP properties, such as blocking the slow sodium current to shorten the AP duration and suppress the EADs, and have found good agreement with experimental observations. This study should help elucidate arrhythmogenic mechanisms at the cellular level and predict unseen properties under HF conditions. In addition, this AP cell model might be useful for modeling and simulating HF at the tissue and organ levels.
Collapse
|
29
|
Grubb S, Aistrup GL, Koivumäki JT, Speerschneider T, Gottlieb LA, Mutsaers NAM, Olesen SP, Calloe K, Thomsen MB. Preservation of cardiac function by prolonged action potentials in mice deficient of KChIP2. Am J Physiol Heart Circ Physiol 2015; 309:H481-9. [PMID: 26055791 DOI: 10.1152/ajpheart.00166.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/29/2015] [Indexed: 11/22/2022]
Abstract
Inherited ion channelopathies and electrical remodeling in heart disease alter the cardiac action potential with important consequences for excitation-contraction coupling. Potassium channel-interacting protein 2 (KChIP2) is reduced in heart failure and interacts under physiological conditions with both Kv4 to conduct the fast-recovering transient outward K(+) current (Ito,f) and with CaV1.2 to mediate the inward L-type Ca(2+) current (ICa,L). Anesthetized KChIP2(-/-) mice have normal cardiac contraction despite the lower ICa,L, and we hypothesized that the delayed repolarization could contribute to the preservation of contractile function. Detailed analysis of current kinetics shows that only ICa,L density is reduced, and immunoblots demonstrate unaltered CaV1.2 and CaVβ₂ protein levels. Computer modeling suggests that delayed repolarization would prolong the period of Ca(2+) entry into the cell, thereby augmenting Ca(2+)-induced Ca(2+) release. Ca(2+) transients in disaggregated KChIP2(-/-) cardiomyocytes are indeed comparable to wild-type transients, corroborating the preserved contractile function and suggesting that the compensatory mechanism lies in the Ca(2+)-induced Ca(2+) release event. We next functionally probed dyad structure, ryanodine receptor Ca(2+) sensitivity, and sarcoplasmic reticulum Ca(2+) load and found that increased temporal synchronicity of the Ca(2+) release in KChIP2(-/-) cardiomyocytes may reflect improved dyad structure aiding the compensatory mechanisms in preserving cardiac contractile force. Thus the bimodal effect of KChIP2 on Ito,f and ICa,L constitutes an important regulatory effect of KChIP2 on cardiac contractility, and we conclude that delayed repolarization and improved dyad structure function together to preserve cardiac contraction in KChIP2(-/-) mice.
Collapse
Affiliation(s)
- Søren Grubb
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gary L Aistrup
- Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, Illinois
| | - Jussi T Koivumäki
- Simula Research Laboratory, Center for Biomedical Computing and Center for Cardiological Innovation, Oslo, Norway
| | - Tobias Speerschneider
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lisa A Gottlieb
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nancy A M Mutsaers
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren-Peter Olesen
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kirstine Calloe
- Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten B Thomsen
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| |
Collapse
|
30
|
Bohnen MS, Iyer V, Sampson KJ, Kass RS. Novel mechanism of transient outward potassium channel current regulation in the heart: implications for cardiac electrophysiology in health and disease. Circ Res 2015; 116:1633-5. [PMID: 25953919 DOI: 10.1161/circresaha.115.306438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Michael S Bohnen
- From the Department of Pharmacology (M.S.B., K.J.S., R.S.K.), and Department of Medicine (V.I.), College of Physicians & Surgeons, Columbia University, NY
| | - Vivek Iyer
- From the Department of Pharmacology (M.S.B., K.J.S., R.S.K.), and Department of Medicine (V.I.), College of Physicians & Surgeons, Columbia University, NY
| | - Kevin J Sampson
- From the Department of Pharmacology (M.S.B., K.J.S., R.S.K.), and Department of Medicine (V.I.), College of Physicians & Surgeons, Columbia University, NY
| | - Robert S Kass
- From the Department of Pharmacology (M.S.B., K.J.S., R.S.K.), and Department of Medicine (V.I.), College of Physicians & Surgeons, Columbia University, NY.
| |
Collapse
|
31
|
Long VP, Bonilla IM, Vargas-Pinto P, Nishijima Y, Sridhar A, Li C, Mowrey K, Wright P, Velayutham M, Kumar S, Lee NY, Zweier JL, Mohler PJ, Györke S, Carnes CA. Heart failure duration progressively modulates the arrhythmia substrate through structural and electrical remodeling. Life Sci 2015; 123:61-71. [PMID: 25596015 PMCID: PMC4763601 DOI: 10.1016/j.lfs.2014.12.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/15/2014] [Accepted: 12/19/2014] [Indexed: 01/10/2023]
Abstract
AIMS Ventricular arrhythmias are a common cause of death in patients with heart failure (HF). Structural and electrical abnormalities in the heart provide a substrate for such arrhythmias. Canine tachypacing-induced HF models of 4-6 weeks duration are often used to study pathophysiology and therapies for HF. We hypothesized that a chronic canine model of HF would result in greater electrical and structural remodeling than a short term model, leading to a more arrhythmogenic substrate. MAIN METHODS HF was induced by ventricular tachypacing for one (short-term) or four (chronic) months to study remodeling. KEY FINDINGS Left ventricular contractility was progressively reduced, while ventricular hypertrophy and interstitial fibrosis were evident at 4 month but not 1 month of HF. Left ventricular myocyte action potentials were prolonged after 4 (p<0.05) but not 1 month of HF. Repolarization instability and early afterdepolarizations were evident only after 4 months of HF (p<0.05), coinciding with a prolonged QTc interval (p<0.05). The transient outward potassium current was reduced in both HF groups (p<0.05). The outward component of the inward rectifier potassium current was reduced only in the 4 month HF group (p<0.05). The delayed rectifier potassium currents were reduced in 4 (p<0.05) but not 1 month of HF. Reactive oxygen species were increased at both 1 and 4 months of HF (p<0.05). SIGNIFICANCE Reduced Ito, outward IK1, IKs, and IKr in HF contribute to EAD formation. Chronic, but not short term canine HF, results in the altered electrophysiology and repolarization instability characteristic of end-stage human HF.
Collapse
Affiliation(s)
- Victor P Long
- College of Pharmacy, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Ingrid M Bonilla
- College of Pharmacy, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Pedro Vargas-Pinto
- College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Arun Sridhar
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Chun Li
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | | | - Patrick Wright
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Murugesan Velayutham
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Sanjay Kumar
- College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Nam Y Lee
- College of Pharmacy, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Jay L Zweier
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Peter J Mohler
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Sandor Györke
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Cynthia A Carnes
- College of Pharmacy, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
32
|
Haugaard MM, Hesselkilde EZ, Pehrson S, Carstensen H, Flethøj M, Præstegaard KF, Sørensen US, Diness JG, Grunnet M, Buhl R, Jespersen T. Pharmacologic inhibition of small-conductance calcium-activated potassium (SK) channels by NS8593 reveals atrial antiarrhythmic potential in horses. Heart Rhythm 2014; 12:825-35. [PMID: 25542425 DOI: 10.1016/j.hrthm.2014.12.028] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND Small-conductance calcium-activated potassium (SK) channels have been found to play an important role in atrial repolarization and atrial fibrillation (AF). OBJECTIVE The purpose of this study was to investigate the existence and functional role of SK channels in the equine heart. METHODS Cardiac biopsies were analyzed to investigate the expression level of the most prominent cardiac ion channels, with special focus on SK channels, in the equine heart. Subcellular distribution of SK isoform 2 (SK2) was assessed by immunohistochemistry and confocal microscopy. The electrophysiologic and anti-AF effects of the relative selective SK channel inhibitor NS8593 (5 mg/kg IV) were evaluated in anesthetized horses, focusing on the potential of NS8593 to terminate acute pacing-induced AF, drug-induced changes in atrial effective refractory period, AF duration and vulnerability, and ventricular depolarization and repolarization times. RESULTS Analysis revealed equivalent mRNA transcript levels of the 3 SK channel isoforms in atria compared to ventricles. Immunohistochemistry and confocal microscopy displayed a widespread distribution of SK2 in both atrial and ventricular cardiomyocytes. NS8593 terminated all induced AF episodes (duration ≥15 minutes), caused pronounced prolongation of atrial effective refractory period, and reduced AF duration and vulnerability. QRS duration and QTc interval were not affected by treatment. CONCLUSION SK channels are widely distributed in atrial and ventricular cardiomyocytes and contribute to atrial repolarization. Inhibition by NS8593 terminates pacing-induced AF of short duration and decreases AF duration and vulnerability without affecting ventricular conduction and repolarization. Thus, inhibition by NS8593 demonstrates clear atrial antiarrhythmic properties in healthy horses.
Collapse
Affiliation(s)
- Maria Mathilde Haugaard
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Eva Zander Hesselkilde
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steen Pehrson
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Taastrup, Denmark
| | - Helena Carstensen
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Flethøj
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kirstine Færgemand Præstegaard
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Rikke Buhl
- Department of Large Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Jespersen
- Danish National Foundation Research Centre in Arrhythmias (DARC) and Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Wang DY, Abbasi C, El-Rass S, Li JY, Dawood F, Naito K, Sharma P, Bousette N, Singh S, Backx PH, Cox B, Wen XY, Liu PP, Gramolini AO. Endoplasmic reticulum resident protein 44 (ERp44) deficiency in mice and zebrafish leads to cardiac developmental and functional defects. J Am Heart Assoc 2014; 3:e001018. [PMID: 25332179 PMCID: PMC4323785 DOI: 10.1161/jaha.114.001018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Endoplasmic reticulum (ER) resident protein 44 (ERp44) is a member of the protein disulfide isomerase family, is induced during ER stress, and may be involved in regulating Ca(2+) homeostasis. However, the role of ERp44 in cardiac development and function is unknown. The aim of this study was to investigate the role of ERp44 in cardiac development and function in mice, zebrafish, and embryonic stem cell (ESC)-derived cardiomyocytes to determine the underlying role of ERp44. METHODS AND RESULTS We generated and characterized ERp44(-/-) mice, ERp44 morphant zebrafish embryos, and ERp44(-/-) ESC-derived cardiomyocytes. Deletion of ERp44 in mouse and zebrafish caused significant embryonic lethality, abnormal heart development, altered Ca(2+) dynamics, reactive oxygen species generation, activated ER stress gene profiles, and apoptotic cell death. We also determined the cardiac phenotype in pressure overloaded, aortic-banded ERp44(+/-) mice: enhanced ER stress activation and increased mortality, as well as diastolic cardiac dysfunction with a significantly lower fractional shortening. Confocal and LacZ histochemical staining showed a significant transmural gradient for ERp44 in the adult heart, in which high expression of ERp44 was observed in the outer subepicardial region of the myocardium. CONCLUSIONS ERp44 plays a critical role in embryonic heart development and is crucial in regulating cardiac cell Ca(2+) signaling, ER stress, ROS-induced oxidative stress, and activation of the intrinsic mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Ding-Yan Wang
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Cynthia Abbasi
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Suzan El-Rass
- Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.) Keenan Research Center for Biomedical Science and Zebrafish Center for Advanced Drug Discovery, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada (S.E.R., X.Y.W.)
| | - Jamie Yuanjun Li
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Fayez Dawood
- Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.)
| | - Kotaro Naito
- Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.)
| | - Parveen Sharma
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Nicolas Bousette
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Shalini Singh
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Peter H Backx
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.) Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.)
| | - Brian Cox
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.)
| | - Xiao-Yan Wen
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.) Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.) Keenan Research Center for Biomedical Science and Zebrafish Center for Advanced Drug Discovery, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada (S.E.R., X.Y.W.)
| | - Peter P Liu
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.) Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.)
| | - Anthony O Gramolini
- Department of Physiology, University of Toronto, Ontario, Canada (D.Y.W., C.A., J.Y.L., P.S., N.B., S.S., P.H.B., B.C., X.Y.W., P.P.L., A.O.G.) Faculty of Medicine and Institute of Medical Science, University of Toronto, Ontario, Canada (S.E.R., F.D., K.N., P.H.B., X.Y.W., P.P.L., A.O.G.)
| |
Collapse
|
34
|
Gomez JF, Cardona K, Romero L, Ferrero JM, Trenor B. Electrophysiological and structural remodeling in heart failure modulate arrhythmogenesis. 1D simulation study. PLoS One 2014; 9:e106602. [PMID: 25191998 PMCID: PMC4156355 DOI: 10.1371/journal.pone.0106602] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 08/05/2014] [Indexed: 01/24/2023] Open
Abstract
Background Heart failure is a final common pathway or descriptor for various cardiac pathologies. It is associated with sudden cardiac death, which is frequently caused by ventricular arrhythmias. Electrophysiological remodeling, intercellular uncoupling, fibrosis and autonomic imbalance have been identified as major arrhythmogenic factors in heart failure etiology and progression. Objective In this study we investigate in silico the role of electrophysiological and structural heart failure remodeling on the modulation of key elements of the arrhythmogenic substrate, i.e., electrophysiological gradients and abnormal impulse propagation. Methods Two different mathematical models of the human ventricular action potential were used to formulate models of the failing ventricular myocyte. This provided the basis for simulations of the electrical activity within a transmural ventricular strand. Our main goal was to elucidate the roles of electrophysiological and structural remodeling in setting the stage for malignant life-threatening arrhythmias. Results Simulation results illustrate how the presence of M cells and heterogeneous electrophysiological remodeling in the human failing ventricle modulate the dispersion of action potential duration and repolarization time. Specifically, selective heterogeneous remodeling of expression levels for the Na+/Ca2+ exchanger and SERCA pump decrease these heterogeneities. In contrast, fibroblast proliferation and cellular uncoupling both strongly increase repolarization heterogeneities. Conduction velocity and the safety factor for conduction are also reduced by the progressive structural remodeling during heart failure. Conclusion An extensive literature now establishes that in human ventricle, as heart failure progresses, gradients for repolarization are changed significantly by protein specific electrophysiological remodeling (either homogeneous or heterogeneous). Our simulations illustrate and provide new insights into this. Furthermore, enhanced fibrosis in failing hearts, as well as reduced intercellular coupling, combine to increase electrophysiological gradients and reduce electrical propagation. In combination these changes set the stage for arrhythmias.
Collapse
Affiliation(s)
- Juan F. Gomez
- Instituto de Investigación en Ingeniería Biomédica, Universitat Politècnica de València, Valencia, Spain
| | - Karen Cardona
- Instituto de Investigación en Ingeniería Biomédica, Universitat Politècnica de València, Valencia, Spain
| | - Lucia Romero
- Instituto de Investigación en Ingeniería Biomédica, Universitat Politècnica de València, Valencia, Spain
| | - Jose M. Ferrero
- Instituto de Investigación en Ingeniería Biomédica, Universitat Politècnica de València, Valencia, Spain
| | - Beatriz Trenor
- Instituto de Investigación en Ingeniería Biomédica, Universitat Politècnica de València, Valencia, Spain
- * E-mail:
| |
Collapse
|
35
|
Schmitt N, Grunnet M, Olesen SP. Cardiac potassium channel subtypes: new roles in repolarization and arrhythmia. Physiol Rev 2014; 94:609-53. [PMID: 24692356 DOI: 10.1152/physrev.00022.2013] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
About 10 distinct potassium channels in the heart are involved in shaping the action potential. Some of the K+ channels are primarily responsible for early repolarization, whereas others drive late repolarization and still others are open throughout the cardiac cycle. Three main K+ channels drive the late repolarization of the ventricle with some redundancy, and in atria this repolarization reserve is supplemented by the fairly atrial-specific KV1.5, Kir3, KCa, and K2P channels. The role of the latter two subtypes in atria is currently being clarified, and several findings indicate that they could constitute targets for new pharmacological treatment of atrial fibrillation. The interplay between the different K+ channel subtypes in both atria and ventricle is dynamic, and a significant up- and downregulation occurs in disease states such as atrial fibrillation or heart failure. The underlying posttranscriptional and posttranslational remodeling of the individual K+ channels changes their activity and significance relative to each other, and they must be viewed together to understand their role in keeping a stable heart rhythm, also under menacing conditions like attacks of reentry arrhythmia.
Collapse
|
36
|
Grubb S, Speerschneider T, Occhipinti D, Fiset C, Olesen SP, Thomsen MB, Calloe K. Loss of K+ currents in heart failure is accentuated in KChIP2 deficient mice. J Cardiovasc Electrophysiol 2014; 25:896-904. [PMID: 24678923 DOI: 10.1111/jce.12422] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 03/06/2014] [Accepted: 03/12/2014] [Indexed: 10/25/2022]
Abstract
INTRODUCTION KV 4 together with KV Channel-Interacting Protein 2 (KChIP2) mediate the fast recovering transient outward potassium current (I(to,f)) in the heart. KChIP2 is downregulated in human heart failure (HF), potentially underlying the loss of I(to,f). We investigated remodeling associated with HF hypothesizing that KChIP2 plays a central role in the modulation of outward K(+) currents in HF. METHODS AND RESULTS HF was induced by aortic banding in wild-type (WT) and KChIP2 deficient (KChIP2(-/-)) mice, evaluated by echocardiography. Action potentials were measured by floating microelectrodes in intact hearts. Ventricular cardiomyocytes were isolated and whole-cell currents were recorded by patch clamp. Left ventricular action potentials in KChIP2(-/-) mice were prolonged in a rate dependent manner, consistent with patch-clamp data showing loss of a fast recovering outward K(+) current and upregulation of the slow recovering I(to,s) and I(Kur). HF decreased all outward K(+) currents in WT mice and did not change the relative contribution of I(to,f) in WT mice. Compared to WT HF, KChIP2(-/-) HF had a larger reduction of K(+) -current density. However, the relative APD prolongation caused by HF was shorter for KChIP2(-/-) compared with WT, and the APs of the 2 HF mouse types were indistinguishable. CONCLUSION I(to,f) is just one of many K(+) currents being downregulated in murine HF. The downregulation of repolarizing currents in HF is accentuated in KChIP2(-/-) mice. However, the prolongation of APs associated with HF is less in KChIP2(-/-) compared to WT, suggesting other compensatory mechanism(s) in the KChIP2(-/-) mouse.
Collapse
Affiliation(s)
- Søren Grubb
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tobias Speerschneider
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dona Occhipinti
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Céline Fiset
- Faculty of Pharmacy, Research Center of the Montreal Heart Institute, University of Montreal, Montreal, Quebec, Canada
| | - Søren-Peter Olesen
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten B Thomsen
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kirstine Calloe
- The Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Veterinary Clinical and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
37
|
Sato T, Kobayashi T, Kuno A, Miki T, Tanno M, Kouzu H, Itoh T, Ishikawa S, Kojima T, Miura T, Tohse N. Type 2 diabetes induces subendocardium-predominant reduction in transient outward K+ current with downregulation of Kv4.2 and KChIP2. Am J Physiol Heart Circ Physiol 2014; 306:H1054-65. [DOI: 10.1152/ajpheart.00414.2013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the present study, we examined if and how cardiac ion channels are modified by type 2 diabetes mellitus (T2DM). Subendocardial (Endo) myocytes and subepicardial (Epi) myocytes were isolated from left ventricles of Otsuka-Long-Evans-Tokushima Fatty rats (OLETF) rats, a rat model of T2DM, and Otsuka-Long-Evans-Tokushima (LETO) rats (nondiabetic control rats). Endo and Epi myocytes were used for whole cell patch-clamp recordings and for protein and mRNA analyses. Action potential durations in Endo and Epi myocytes were longer in OLETF rats than in LETO rats, and the difference was larger in Endo myocytes. Steady-state transient outward K+ current ( Ito) density was reduced in Endo but not Epi myocytes of OLETF rats compared with LETO rats, although the contribution of the fast component of Ito recovery from inactivation was smaller in both Endo and Epi myocytes of OLETF rats than in LETO rats. Kv4.2 protein was reduced only in Endo myocytes in OLETF rats, although voltage-gated K+ channel-interacting protein 2 (KChIP2) protein levels in both Endo and Epi myocytes were lower in OLETF rats than in LETO rats. Corresponding regional differences in mRNA levels of KChIP2 and Kv4.2 were observed between OLETF and LETO rats. mRNA levels of Iroquois homeobox 5 in Endo myocytes were 53% higher in OLETF rats than in LETO rats. Densities of inward rectifier K+ current and L-type Ca2+ current and mRNA levels of Kv4.3 and Kv1.4 were similar in OLETF and LETO rats. In conclusion, T2DM induces Endo-predominant prolongation of the action potential duration via a reduction of the fast component of Ito recovery from inactivation and reduced steady-state Ito, in which downregulation of Kv4.2 and KChIP2 may be involved. Increased Iroquois homeobox 5 expression may underlie Kv4.2 downregulation in T2DM.
Collapse
Affiliation(s)
- Tatsuya Sato
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takeshi Kobayashi
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Kuno
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan; and
| | - Takayuki Miki
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masaya Tanno
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hidemichi Kouzu
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takahito Itoh
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Satoko Ishikawa
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute of Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Noritsugu Tohse
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
38
|
Yuan L, Koivumäki JT, Liang B, Lorentzen LG, Tang C, Andersen MN, Svendsen JH, Tfelt-Hansen J, Maleckar M, Schmitt N, Olesen MS, Jespersen T. Investigations of the Navβ1b sodium channel subunit in human ventricle; functional characterization of the H162P Brugada syndrome mutant. Am J Physiol Heart Circ Physiol 2014; 306:H1204-12. [PMID: 24561865 DOI: 10.1152/ajpheart.00405.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Brugada syndrome (BrS) is a rare inherited disease that can give rise to ventricular arrhythmia and ultimately sudden cardiac death. Numerous loss-of-function mutations in the cardiac sodium channel Nav1.5 have been associated with BrS. However, few mutations in the auxiliary Navβ1-4 subunits have been linked to this disease. Here we investigated differences in expression and function between Navβ1 and Navβ1b and whether the H162P/Navβ1b mutation found in a BrS patient is likely to be the underlying cause of disease. The impact of Navβ subunits was investigated by patch-clamp electrophysiology, and the obtained in vitro values were used for subsequent in silico modeling. We found that Navβ1b transcripts were expressed at higher levels than Navβ1 transcripts in the human heart. Navβ1 and Navβ1b coexpressed with Nav1.5 induced a negative shift on steady state of activation and inactivation compared with Nav1.5 alone. Furthermore, Navβ1b was found to increase the current level when coexpressed with Nav1.5, Navβ1b/H162P mutated subunit peak current density was reduced by 48% (-645 ± 151 vs. -334 ± 71 pA/pF), V1/2 steady-state inactivation shifted by -6.7 mV (-70.3 ± 1.5 vs. -77.0 ± 2.8 mV), and time-dependent recovery from inactivation slowed by >50% compared with coexpression with Navβ1b wild type. Computer simulations revealed that these electrophysiological changes resulted in a reduction in both action potential amplitude and maximum upstroke velocity. The experimental data thereby indicate that Navβ1b/H162P results in reduced sodium channel activity functionally affecting the ventricular action potential. This result is an important replication to support the notion that BrS can be linked to the function of Navβ1b and is associated with loss-of-function of the cardiac sodium channel.
Collapse
Affiliation(s)
- Lei Yuan
- Danish National Research Foundation Centre for Cardiac Arrhythmia (DARC), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Haynes P, Nava KE, Lawson BA, Chung CS, Mitov MI, Campbell SG, Stromberg AJ, Sadayappan S, Bonnell MR, Hoopes CW, Campbell KS. Transmural heterogeneity of cellular level power output is reduced in human heart failure. J Mol Cell Cardiol 2014; 72:1-8. [PMID: 24560668 DOI: 10.1016/j.yjmcc.2014.02.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 01/22/2014] [Accepted: 02/11/2014] [Indexed: 01/04/2023]
Abstract
Heart failure is associated with pump dysfunction and remodeling but it is not yet known if the condition affects different transmural regions of the heart in the same way. We tested the hypotheses that the left ventricles of non-failing human hearts exhibit transmural heterogeneity of cellular level contractile properties, and that heart failure produces transmural region-specific changes in contractile function. Permeabilized samples were prepared from the sub-epicardial, mid-myocardial, and sub-endocardial regions of the left ventricular free wall of non-failing (n=6) and failing (n=10) human hearts. Power, an in vitro index of systolic function, was higher in non-failing mid-myocardial samples (0.59±0.06μWmg(-1)) than in samples from the sub-epicardium (p=0.021) and the sub-endocardium (p=0.015). Non-failing mid-myocardial samples also produced more isometric force (14.3±1.33kNm(-2)) than samples from the sub-epicardium (p=0.008) and the sub-endocardium (p=0.026). Heart failure reduced power (p=0.009) and force (p=0.042) but affected the mid-myocardium more than the other transmural regions. Fibrosis increased with heart failure (p=0.021) and mid-myocardial tissue from failing hearts contained more collagen than matched sub-epicardial (p<0.001) and sub-endocardial (p=0.043) samples. Power output was correlated with the relative content of actin and troponin I, and was also statistically linked to the relative content and phosphorylation of desmin and myosin light chain-1. Non-failing human hearts exhibit transmural heterogeneity of contractile properties. In failing organs, region-specific fibrosis produces the greatest contractile deficits in the mid-myocardium. Targeting fibrosis and sarcomeric proteins in the mid-myocardium may be particularly effective therapies for heart failure.
Collapse
Affiliation(s)
- Premi Haynes
- Department of Physiology, University of Kentucky, Lexington, KY, USA; Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Kristofer E Nava
- Department of Physiology, University of Kentucky, Lexington, KY, USA; Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Benjamin A Lawson
- Department of Physiology, University of Kentucky, Lexington, KY, USA; Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Charles S Chung
- Department of Physiology, University of Kentucky, Lexington, KY, USA; Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Mihail I Mitov
- Department of Physiology, University of Kentucky, Lexington, KY, USA; Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Stuart G Campbell
- Department of Physiology, University of Kentucky, Lexington, KY, USA; Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | | | - Sakthivel Sadayappan
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University, Chicago, USA
| | - Mark R Bonnell
- Division of Cardiothoracic Surgery, University of Kentucky, Lexington, KY, USA
| | - Charles W Hoopes
- Division of Cardiothoracic Surgery, University of Kentucky, Lexington, KY, USA
| | - Kenneth S Campbell
- Department of Physiology, University of Kentucky, Lexington, KY, USA; Center for Muscle Biology, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
40
|
Changes in Cx43 and NaV1.5 expression precede the occurrence of substantial fibrosis in calcineurin-induced murine cardiac hypertrophy. PLoS One 2014; 9:e87226. [PMID: 24498049 PMCID: PMC3909068 DOI: 10.1371/journal.pone.0087226] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 12/20/2013] [Indexed: 11/19/2022] Open
Abstract
In mice, the calcium-dependent phosphatase calcineurin A (CnA) induces a transcriptional pathway leading to pathological cardiac hypertrophy. Interestingly, induction of CnA has been frequently noticed in human hypertrophic and failing hearts. Independently, the arrhythmia vulnerability of such hearts has been regularly associated with remodeling of parameters determining electrical conduction (expression level of connexin43 (Cx43) and NaV1.5, connective tissue architecture), for which the precise molecular basis and sequence of events is still unknown. Recently, we observed reduced Cx43 and NaV1.5 expression in 4-week old mouse hearts, overexpressing a constitutively active form of CnA (MHC-CnA model), but the order of events is still unknown. Therefore, three key parameters of conduction (Cx43, NaV1.5 and connective tissue expression) were characterized in MHC-CnA ventricles versus wild-type (WT) during postnatal development on a weekly basis. At postnatal week 1, CnA overexpression induced cardiac hypertrophy in MHC-CnA. Moreover, protein and RNA levels of both Cx43 and NaV1.5 were reduced by at least 50% as compared to WT. Cx43 immunoreactive signal was reduced at week 2 in MHC-CnA. At postnatal week 3, Cx43 was less phosphorylated and RNA level of Cx43 normalized to WT values, although the protein level was still reduced. Additionally, MHC-CnA hearts displayed substantial fibrosis relative to WT, which was accompanied by increased RNA levels for genes previously associated with fibrosis such as Col1a1, Col1a2, Col3a1, Tgfb1, Ctgf, Timp1 and microRNA miR-21. In MHC-CnA, reduction in Cx43 and NaV1.5 expression thus coincided with overexpression of CnA and hypertrophy development and preceded significant presence of fibrosis. At postnatal week 4 the alterations in conductional parameters observed in the MHC-CnA model lead to abnormal conduction and arrhythmias, similar to those observed in cardiac remodeling in heart failure patients. The MHC-CnA model, therefore, provides for a unique model to resolve the molecular origin of conductional remodeling in detail.
Collapse
|
41
|
Chung CS, Campbell KS. Temperature and transmural region influence functional measurements in unloaded left ventricular cardiomyocytes. Physiol Rep 2013; 1:e00158. [PMID: 24400159 PMCID: PMC3871472 DOI: 10.1002/phy2.158] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 10/18/2013] [Accepted: 10/23/2013] [Indexed: 12/21/2022] Open
Abstract
Intact cardiomyocytes are increasingly being used to investigate the molecular mechanisms of contraction and to screen new therapeutic compounds. The function of the cardiomyocytes is often measured from the calcium transients and sarcomere length profiles. We studied the role of experimental temperature and transmural region on indices of function in freshly isolated, unloaded cardiomyocytes. Intact cardiomyocytes were isolated from the subendocardium, midmyocardium, and subepicardium of 3-month-old Sprague-Dawley rats. Myocytes from each region were studied at 25°C, 31°C, and 37°C. Cytosolic calcium transients were measured using Fura-2 fluorescence, whereas sarcomere length shortening and relengthening profiles were measured using high-speed video capture. For both the calcium transients and sarcomere length profiles, the time to peak and the time to half relaxation decreased significantly with increasing temperature. Increasing temperature also raised the minimum and maximum calcium levels of all cells. Of note, there was a reduced coefficient of variation (standard deviation divided by the mean) at higher temperatures for calcium fluorescence amplitudes, time to peak calcium, and rates of sarcomeric shortening and relengthening. The amplitudes and minimum of the calcium transients were significantly dependent on transmural region, and several sarcomere length parameters exhibited statistical interactions between temperature and transmural region. Together, these results show that biological variability can be reduced by performing experiments at 37°C rather than at room temperature, and by isolating cells from a specific transmural region. Adopting these procedures will improve the statistical power of subsequent analyses and increase the efficiency of future experiments.
Collapse
Affiliation(s)
- Charles S Chung
- Department of Physiology and Center for Muscle Biology, University of Kentucky Lexington, Kentucky
| | - Kenneth S Campbell
- Department of Physiology and Center for Muscle Biology, University of Kentucky Lexington, Kentucky
| |
Collapse
|
42
|
Speerschneider T, Grubb S, Metoska A, Olesen SP, Calloe K, Thomsen MB. Development of heart failure is independent of K+ channel-interacting protein 2 expression. J Physiol 2013; 591:5923-37. [PMID: 24099801 DOI: 10.1113/jphysiol.2013.263483] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Abnormal ventricular repolarization in ion channelopathies and heart disease is a major cause of ventricular arrhythmias and sudden cardiac death. K(+) channel-interacting protein 2 (KChIP2) expression is significantly reduced in human heart failure (HF), contributing to a loss of the transient outward K(+) current (Ito). We aim to investigate the possible significance of a changed KChIP2 expression on the development of HF and proarrhythmia. Transverse aortic constrictions (TAC) and sham operations were performed in wild-type (WT) and KChIP2(-/-) mice. Echocardiography was performed before and every 2 weeks after the operation. Ten weeks post-surgery, surface ECG was recorded and we paced the heart in vivo to induce arrhythmias. Afterwards, tissue from the left ventricle was used for immunoblotting. Time courses of HF development were comparable in TAC-operated WT and KChIP2(-/-) mice. Ventricular protein expression of KChIP2 was reduced by 70% after 10 weeks TAC in WT mice. The amplitudes of the J and T waves were enlarged in KChIP2(-/-) control mice. Ventricular effective refractory period, RR, QRS and QT intervals were longer in mice with HF compared to sham-operated mice of either genotype. Pacing-induced ventricular tachycardia (VT) was observed in 5/10 sham-operated WT mice compared with 2/10 HF WT mice with HF. Interestingly, and contrary to previously published data, sham-operated KChIP2(-/-) mice were resistant to pacing-induced VT resulting in only 1/10 inducible mice. KChIP2(-/-) with HF mice had similar low vulnerability to inducible VT (1/9). Our results suggest that although KChIP2 is downregulated in HF, it is not orchestrating the development of HF. Moreover, KChIP2 affects ventricular repolarization and lowers arrhythmia susceptibility. Hence, downregulation of KChIP2 expression in HF may be antiarrhythmic in mice via reduction of the fast transient outward K(+) current.
Collapse
Affiliation(s)
- Tobias Speerschneider
- M. B. Thomsen: Danish National Research Foundation Centre for Cardiac Arrhythmia, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 3b Blegdamsvej, building 12.5.36, Copenhagen N, Denmark.
| | | | | | | | | | | |
Collapse
|
43
|
McGahon MK, Yarham JM, Daly A, Guduric-Fuchs J, Ferguson LJ, Simpson DA, Collins A. Distinctive profile of IsomiR expression and novel microRNAs in rat heart left ventricle. PLoS One 2013; 8:e65809. [PMID: 23799049 PMCID: PMC3683050 DOI: 10.1371/journal.pone.0065809] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 05/03/2013] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs (miRNAs) are single-stranded non-coding RNAs that negatively regulate target gene expression through mRNA cleavage or translational repression. There is mounting evidence that they play critical roles in heart disease. The expression of known miRNAs in the heart has been studied at length by microarray and quantitative PCR but it is becoming evident that microRNA isoforms (isomiRs) are potentially physiologically important. It is well known that left ventricular (patho)physiology is influenced by transmural heterogeneity of cardiomyocyte phenotype, and this likely reflects underlying heterogeneity of gene expression. Given the significant role of miRNAs in regulating gene expression, knowledge of how the miRNA profile varies across the ventricular wall will be crucial to better understand the mechanisms governing transmural physiological heterogeneity. To determinine miRNA/isomiR expression profiles in the rat heart we investigated tissue from different locations across the left ventricular wall using deep sequencing. We detected significant quantities of 145 known rat miRNAs and 68 potential novel orthologs of known miRNAs, in mature, mature* and isomiR formation. Many isomiRs were detected at a higher frequency than their canonical sequence in miRBase and have different predicted targets. The most common miR-133a isomiR was more effective at targeting a construct containing a sequence from the gelsolin gene than was canonical miR-133a, as determined by dual-fluorescence assay. We identified a novel rat miR-1 homolog from a second miR-1 gene; and a novel rat miRNA similar to miR-676. We also cloned and sequenced the rat miR-486 gene which is not in miRBase (v18). Signalling pathways predicted to be targeted by the most highly detected miRNAs include Ubiquitin-mediated Proteolysis, Mitogen-Activated Protein Kinase, Regulation of Actin Cytoskeleton, Wnt signalling, Calcium Signalling, Gap junctions and Arrhythmogenic Right Ventricular Cardiomyopathy. Most miRNAs are not expressed in a gradient across the ventricular wall, with exceptions including miR-10b, miR-21, miR-99b and miR-486.
Collapse
Affiliation(s)
- Mary K. McGahon
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| | - Janet M. Yarham
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| | - Aideen Daly
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| | - Jasenka Guduric-Fuchs
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| | - Lyndsey J. Ferguson
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| | - David A. Simpson
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| | - Anthony Collins
- Centre for Vision and Vascular Science, Queen’s University Belfast, Belfast, County Antrim, United Kingdom
| |
Collapse
|
44
|
Kelly A, Ghouri IA, Kemi OJ, Bishop MJ, Bernus O, Fenton FH, Myles RC, Burton FL, Smith GL. Subepicardial action potential characteristics are a function of depth and activation sequence in isolated rabbit hearts. Circ Arrhythm Electrophysiol 2013; 6:809-17. [PMID: 23733913 DOI: 10.1161/circep.113.000334] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Electric excitability in the ventricular wall is influenced by cellular electrophysiology and passive electric properties of the myocardium. Action potential (AP) rise time, an indicator of myocardial excitability, is influenced by conduction pattern and distance from the epicardial surface. This study examined AP rise times and conduction velocity as the depolarizing wavefront approaches the epicardial surface. METHODS AND RESULTS Two-photon excitation of di-4-aminonaphthenyl-pyridinum-propylsulfonate was used to measure electric activity at discrete epicardial layers of isolated Langendorff-perfused rabbit hearts to a depth of 500 μm. Endo-to-epicardial wavefronts were studied during right atrial or ventricular endocardial pacing. Similar measurements were made with epi-to-endocardial, transverse, and longitudinal pacing protocols. Results were compared with data from a bidomain model of 3-dimensional (3D) electric propagation within ventricular myocardium. During right atrial and endocardial pacing, AP rise time (10%-90% of upstroke) decreased by ≈50% between 500 and 50 μm from the epicardial surface, whereas conduction velocity increased and AP duration was only slightly shorter (≈4%). These differences were not observed with other conduction patterns. The depth-dependent changes in rise time were larger at higher pacing rates. Modeling data qualitatively reproduced the behavior seen experimentally and demonstrated a parallel reduction in peak I(Na) and electrotonic load as the wavefront approaches the epicardial surface. CONCLUSIONS Decreased electrotonic load at the epicardial surface results in more rapid AP upstrokes and higher conduction velocities compared with the bulk myocardium. Combined effects of tissue depth and pacing rate on AP rise time reduce conduction safety and myocardial excitability within the ventricular wall.
Collapse
Affiliation(s)
- Allen Kelly
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Takanari H, Nalos L, Stary-Weinzinger A, de Git KCG, Varkevisser R, Linder T, Houtman MJC, Peschar M, de Boer TP, Tidwell RR, Rook MB, Vos MA, van der Heyden MAG. Efficient and specific cardiac IK1 inhibition by a new pentamidine analogue. Cardiovasc Res 2013; 99:203-14. [DOI: 10.1093/cvr/cvt103] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
46
|
Cardiac ion channels and mechanisms for protection against atrial fibrillation. Rev Physiol Biochem Pharmacol 2013; 162:1-58. [PMID: 21987061 DOI: 10.1007/112_2011_3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Atrial fibrillation (AF) is recognised as the most common sustained cardiac arrhythmia in clinical practice. Ongoing drug development is aiming at obtaining atrial specific effects in order to prevent pro-arrhythmic, devastating ventricular effects. In principle, this is possible due to a different ion channel composition in the atria and ventricles. The present text will review the aetiology of arrhythmias with focus on AF and include a description of cardiac ion channels. Channels that constitute potentially atria-selective targets will be described in details. Specific focus is addressed to the recent discovery that Ca(2+)-activated small conductance K(+) channels (SK channels) are important for the repolarisation of atrial action potentials. Finally, an overview of current pharmacological treatment of AF is included.
Collapse
|
47
|
Szuts V, Ménesi D, Varga-Orvos Z, Zvara Á, Houshmand N, Bitay M, Bogáts G, Virág L, Baczkó I, Szalontai B, Geramipoor A, Cotella D, Wettwer E, Ravens U, Deák F, Puskás LG, Papp JG, Kiss I, Varró A, Jost N. Altered expression of genes for Kir ion channels in dilated cardiomyopathy. Can J Physiol Pharmacol 2013; 91:648-56. [PMID: 23889090 DOI: 10.1139/cjpp-2012-0413] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Dilated cardiomyopathy (DCM) is a multifactorial disease characterized by left ventricular dilation that is associated with systolic dysfunction and increased action potential duration. The Kir2.x K⁺ channels (encoded by KCNJ genes) regulate the inward rectifier current (IK1) contributing to the final repolarization in cardiac muscle. Here, we describe the transitions in the gene expression profiles of 4 KCNJ genes from healthy or dilated cardiomyopathic human hearts. In the healthy adult ventricles, KCNJ2, KCNJ12, and KCNJ4 (Kir2.1-2.3, respectively) genes were expressed at high levels, while expression of the KCNJ14 (Kir2.4) gene was low. In DCM ventricles, the levels of Kir2.1 and Kir2.3 were upregulated, but those of Kir2.2 channels were downregulated. Additionally, the expression of the DLG1 gene coding for the synapse-associated protein 97 (SAP97) anchoring molecule exhibited a 2-fold decline with increasing age in normal hearts, and it was robustly downregulated in young DCM patients. These adaptations could offer a new aspect for the explanation of the generally observed physiological and molecular alterations found in DCM.
Collapse
Affiliation(s)
- Viktoria Szuts
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Walmsley J, Rodriguez JF, Mirams GR, Burrage K, Efimov IR, Rodriguez B. mRNA expression levels in failing human hearts predict cellular electrophysiological remodeling: a population-based simulation study. PLoS One 2013; 8:e56359. [PMID: 23437117 PMCID: PMC3577832 DOI: 10.1371/journal.pone.0056359] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 01/08/2013] [Indexed: 01/06/2023] Open
Abstract
Differences in mRNA expression levels have been observed in failing versus non-failing human hearts for several membrane channel proteins and accessory subunits. These differences may play a causal role in electrophysiological changes observed in human heart failure and atrial fibrillation, such as action potential (AP) prolongation, increased AP triangulation, decreased intracellular calcium transient (CaT) magnitude and decreased CaT triangulation. Our goal is to investigate whether the information contained in mRNA measurements can be used to predict cardiac electrophysiological remodeling in heart failure using computational modeling. Using mRNA data recently obtained from failing and non-failing human hearts, we construct failing and non-failing cell populations incorporating natural variability and up/down regulation of channel conductivities. Six biomarkers are calculated for each cell in each population, at cycle lengths between 1500 ms and 300 ms. Regression analysis is performed to determine which ion channels drive biomarker variability in failing versus non-failing cardiomyocytes. Our models suggest that reported mRNA expression changes are consistent with AP prolongation, increased AP triangulation, increased CaT duration, decreased CaT triangulation and amplitude, and increased delay between AP and CaT upstrokes in the failing population. Regression analysis reveals that changes in AP biomarkers are driven primarily by reduction in I, and changes in CaT biomarkers are driven predominantly by reduction in I and SERCA. In particular, the role of I is pacing rate dependent. Additionally, alternans developed at fast pacing rates for both failing and non-failing cardiomyocytes, but the underlying mechanisms are different in control and heart failure.
Collapse
Affiliation(s)
- John Walmsley
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Jose F. Rodriguez
- Aragón Institute of Engineering Research, University of Zaragoza, Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Zaragoza, Spain
| | - Gary R. Mirams
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Kevin Burrage
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Igor R. Efimov
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Blanca Rodriguez
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
49
|
Ambrosi CM, Yamada KA, Nerbonne JM, Efimov IR. Gender differences in electrophysiological gene expression in failing and non-failing human hearts. PLoS One 2013; 8:e54635. [PMID: 23355885 PMCID: PMC3552854 DOI: 10.1371/journal.pone.0054635] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 12/13/2012] [Indexed: 12/19/2022] Open
Abstract
The increasing availability of human cardiac tissues for study are critically important in increasing our understanding of the impact of gender, age, and other parameters, such as medications and cardiac disease, on arrhythmia susceptibility. In this study, we aimed to compare the mRNA expression of 89 ion channel subunits, calcium handling proteins, and transcription factors important in cardiac conduction and arrhythmogenesis in the left atria (LA) and ventricles (LV) of failing and nonfailing human hearts of both genders. Total RNA samples, prepared from failing male (n = 9) and female (n = 7), and from nonfailing male (n = 9) and female (n = 9) hearts, were probed using custom-designed Taqman gene arrays. Analyses were performed to explore the relationships between gender, failure state, and chamber expression. Hierarchical cluster analysis revealed chamber specific expression patterns, but failed to identify disease- or gender-dependent clustering. Gender-specific analysis showed lower expression levels in transcripts encoding for Kv4.3, KChIP2, Kv1.5, and Kir3.1 in the failing female as compared with the male LA. Analysis of LV transcripts, however, did not reveal significant differences based on gender. Overall, our data highlight the differential expression and transcriptional remodeling of ion channel subunits in the human heart as a function of gender and cardiac disease. Furthermore, the availability of such data sets will allow for the development of disease-, gender-, and, most importantly, patient-specific cardiac models, with the ability to utilize such information as mRNA expression to predict cardiac phenotype.
Collapse
Affiliation(s)
- Christina M. Ambrosi
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Kathryn A. Yamada
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jeanne M. Nerbonne
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Igor R. Efimov
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
50
|
Goldoni D, Yarham J, McGahon M, O’Connor A, Guduric-Fuchs J, Edgar K, McDonald D, Simpson D, Collins A. A novel dual-fluorescence strategy for functionally validating microRNA targets in 3' untranslated regions: regulation of the inward rectifier potassium channel K(ir)2.1 by miR-212. Biochem J 2012; 448:103-13. [PMID: 22880819 PMCID: PMC3475433 DOI: 10.1042/bj20120578] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 08/03/2012] [Accepted: 08/13/2012] [Indexed: 01/16/2023]
Abstract
Gene targeting by microRNAs is important in health and disease. We developed a functional assay for identifying microRNA targets and applied it to the K(+) channel K(ir)2.1 [KCNJ2 (potassium inwardly-rectifying channel, subfamily J, member 2)] which is dysregulated in cardiac and vascular disorders. The 3'UTR (untranslated region) was inserted downstream of the mCherry red fluorescent protein coding sequence in a mammalian expression plasmid. MicroRNA sequences were inserted into the pSM30 expression vector which provides enhanced green fluorescent protein as an indicator of microRNA expression. HEK (human embryonic kidney)-293 cells were co-transfected with the mCherry-3'UTR plasmid and a pSM30-based plasmid with a microRNA insert. The principle of the assay is that functional targeting of the 3'UTR by the microRNA results in a decrease in the red/green fluorescence intensity ratio as determined by automated image analysis. The method was validated with miR-1, a known down-regulator of K(ir)2.1 expression, and was used to investigate the targeting of the K(ir)2.1 3'UTR by miR-212. The red/green ratio was lower in miR-212-expressing cells compared with the non-targeting controls, an effect that was attenuated by mutating the predicted target site. miR-212 also reduced inward rectifier current and K(ir)2.1 protein in HeLa cells. This novel assay has several advantages over traditional luciferase-based assays including larger sample size, amenability to time course studies and adaptability to high-throughput screening.
Collapse
Key Words
- hela cell
- hek-293 cell
- image analysis
- microrna
- patch clamp
- cmv, cytomegalovirus
- dmem, dulbecco’s modified eagle’s medium
- egfp, enhanced green fluorescent protein
- gapdh, glyceraldehyde-3-phosphate dehydrogenase
- hek, human embryonic kidney
- hprt1, hypoxanthine–phosphoribosyltransferase 1
- ik1, inward-rectifier k+ current
- kcnj2, potassium inwardly-rectifying channel, subfamily j, member 2
- mirna, microrna
- qrt–pcr, quantitative reverse transcription pcr
- race, rapid amplification of cdna ends
- sirna, short interfering rna
- utr, untranslated region
Collapse
Affiliation(s)
- Dana Goldoni
- Centre for Vision and Vascular Science, Queen's University of Belfast, Institute of Clinical Science, Block A, Royal Victoria Hospital, Grosvenor Road, Belfast, BT12 6BA, U.K
| | - Janet M. Yarham
- Centre for Vision and Vascular Science, Queen's University of Belfast, Institute of Clinical Science, Block A, Royal Victoria Hospital, Grosvenor Road, Belfast, BT12 6BA, U.K
| | - Mary K. McGahon
- Centre for Vision and Vascular Science, Queen's University of Belfast, Institute of Clinical Science, Block A, Royal Victoria Hospital, Grosvenor Road, Belfast, BT12 6BA, U.K
| | - Anna O’Connor
- Centre for Vision and Vascular Science, Queen's University of Belfast, Institute of Clinical Science, Block A, Royal Victoria Hospital, Grosvenor Road, Belfast, BT12 6BA, U.K
| | - Jasenka Guduric-Fuchs
- Centre for Vision and Vascular Science, Queen's University of Belfast, Institute of Clinical Science, Block A, Royal Victoria Hospital, Grosvenor Road, Belfast, BT12 6BA, U.K
| | - Kevin Edgar
- Centre for Vision and Vascular Science, Queen's University of Belfast, Institute of Clinical Science, Block A, Royal Victoria Hospital, Grosvenor Road, Belfast, BT12 6BA, U.K
| | - Denise M. McDonald
- Centre for Vision and Vascular Science, Queen's University of Belfast, Institute of Clinical Science, Block A, Royal Victoria Hospital, Grosvenor Road, Belfast, BT12 6BA, U.K
| | - David A. Simpson
- Centre for Vision and Vascular Science, Queen's University of Belfast, Institute of Clinical Science, Block A, Royal Victoria Hospital, Grosvenor Road, Belfast, BT12 6BA, U.K
| | - Anthony Collins
- Centre for Vision and Vascular Science, Queen's University of Belfast, Institute of Clinical Science, Block A, Royal Victoria Hospital, Grosvenor Road, Belfast, BT12 6BA, U.K
| |
Collapse
|