Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Med ; 9(2)2020 Feb 10.
Article in English | MEDLINE | ID: mdl-32050722

ABSTRACT

BACKGROUND: Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a rare, inheritable cardiac disorder characterized by ventricular tachyarrhythmias, progressive loss of cardiomyocytes with fibrofatty replacement and sudden cardiac death. The exact underlying mechanisms are unclear. METHODS: This study investigated the possible roles of nucleoside diphosphate kinase B (NDPK-B) and SK4 channels in the arrhythmogenesis of ARVC by using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). RESULTS: In hiPSC-CMs from a patient with ARVC, the expression levels of NDPK-B and SK4 channels were upregulated, the cell automaticity was increased and the occurrence rate of arrhythmic events was enhanced. Recombinant NDPK-B applied into hiPSC-CMs from either healthy donors or the patient enhanced SK4 channel current (ISK4), cell automaticity and the occurrence of arrhythmic events, whereas protein histidine phosphatase 1 (PHP-1), a counter actor of NDPK-B, prevented the NDPK-B effect. Application of PHP-1 alone or a SK4 channel blocker also reduced cell automaticity and arrhythmic events. CONCLUSION: This study demonstrated that the elevated NDPK-B expression, via activating SK4 channels, contributes to arrhythmogenesis in ARVC, and hence, NDPK-B may be a potential therapeutic target for treating arrhythmias in patients with ARVC.

2.
Stem Cells Int ; 2018: 6067096, 2018.
Article in English | MEDLINE | ID: mdl-29535773

ABSTRACT

BACKGROUND: Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are providing new possibilities for the biological study, cell therapies, and drug discovery. However, the ion channel expression and functions as well as regulations in hiPSC-CMs still need to be fully characterized. METHODS: Cardiomyocytes were derived from hiPS cells that were generated from two healthy donors. qPCR and patch clamp techniques were used for the study. RESULTS: In addition to the reported ion channels, INa, ICa-L, ICa-T, If, INCX, IK1, Ito, IKr, IKs IKATP, IK-pH, ISK1-3, and ISK4, we detected both the expression and currents of ACh-activated (KACh) and Na+-activated (KNa) K+, volume-regulated and calcium-activated (Cl-Ca) Cl-, and TRPV channels. All the detected ion currents except IK1, IKACh, ISK, IKNa, and TRPV1 currents contribute to AP duration. Isoprenaline increased ICa-L, If, and IKs but reduced INa and INCX, without an effect on Ito, IK1, ISK1-3, IKATP, IKr, ISK4, IKNa, ICl-Ca, and ITRPV1. Carbachol alone showed no effect on the tested ion channel currents. CONCLUSION: Our data demonstrate that most ion channels, which are present in healthy or diseased cardiomyocytes, exist in hiPSC-CMs. Some of them contribute to action potential performance and are regulated by adrenergic stimulation.

3.
Europace ; 20(FI1): f46-f56, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29566126

ABSTRACT

Aims: Our aim is to investigate the arrhythmogenic mechanism in arrhythmogenic right ventricular cardiomyopathy (ARVC)-patients by using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Methods and results: Human-induced pluripotent stem cell-derived cardiomyocytes were generated from human skin fibroblasts of two healthy donors and an ARVC-patient with a desmoglein-2 (DSG2) mutation. Patch clamp, quantitative polymerase chain reaction, and calcium imaging techniques were employed for the study. The amplitude and maximal upstroke velocity (Vmax) of action potential (AP) in ARVC-cells were smaller than that in healthy donor cells, whereas the resting potential and AP duration (APD) was not changed. The reduced Vmax resulted from decreased peak sodium current. The reason for undetected changes in APD may be the counter-action of reduced transient outward, small conductance Ca2+-activated, adenosine triphosphate-sensitive, Na/Ca exchanger (INCX) currents, and enhanced rapidly delayed rectifier currents. Isoprenaline (Iso) reduced INCX and shortened APD in both donor and ARVC-hiPSC-CMs. However, the effects of Iso in ARVC-cells are significantly larger than that in donor cells. In addition, ARVC-hiPSC-CMs showed more frequently than donor cells arrhythmogenic events induced by adrenergic stimulation. Conclusion: Cardiomyocytes derived from the ARVC patient with a DSG2 mutation displayed multiple ion channel dysfunctions and abnormal cellular electrophysiology as well as enhanced sensitivity to adrenergic stimulation. These may underlie the arrhythmogenesis in ARVC patients.


Subject(s)
Action Potentials , Arrhythmogenic Right Ventricular Dysplasia/metabolism , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Action Potentials/drug effects , Adrenergic beta-Agonists/pharmacology , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/pathology , Arrhythmogenic Right Ventricular Dysplasia/physiopathology , Calcium Signaling , Case-Control Studies , Cells, Cultured , Delayed Rectifier Potassium Channels/metabolism , Desmoglein 2/genetics , Desmoglein 2/metabolism , Genetic Predisposition to Disease , Heart Rate , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Isoproterenol/pharmacology , Kinetics , Male , Middle Aged , Mutation , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phenotype , Sodium-Calcium Exchanger/metabolism
4.
J Am Heart Assoc ; 7(7)2018 03 24.
Article in English | MEDLINE | ID: mdl-29574456

ABSTRACT

BACKGROUND: Short QT syndrome (SQTS), a disorder associated with characteristic ECG QT-segment abbreviation, predisposes affected patients to sudden cardiac death. Despite some progress in assessing the organ-level pathophysiology and genetic changes of the disorder, the understanding of the human cellular phenotype and discovering of an optimal therapy has lagged because of a lack of appropriate human cellular models of the disorder. The objective of this study was to establish a cellular model of SQTS using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). METHODS AND RESULTS: This study recruited 1 patient with short QT syndrome type 1 carrying a mutation (N588K) in KCNH2 as well as 2 healthy control subjects. We generated hiPSCs from their skin fibroblasts, and differentiated hiPSCs into cardiomyocytes (hiPSC-CMs) for physiological and pharmacological studies. The hiPSC-CMs from the patient showed increased rapidly activating delayed rectifier potassium channel current (IKr) density and shortened action potential duration compared with healthy control hiPSC-CMs. Furthermore, they demonstrated abnormal calcium transients and rhythmic activities. Carbachol increased the arrhythmic events in SQTS but not in control cells. Gene and protein expression profiling showed increased KCNH2 expression in SQTS cells. Quinidine but not sotalol or metoprolol prolonged the action potential duration and abolished arrhythmic activity induced by carbachol. CONCLUSIONS: Patient-specific hiPSC-CMs are able to recapitulate single-cell phenotype features of SQTS and provide novel opportunities to further elucidate the cellular disease mechanism and test drug effects.


Subject(s)
Action Potentials , Arrhythmias, Cardiac/metabolism , ERG1 Potassium Channel/metabolism , Heart Conduction System/abnormalities , Heart Defects, Congenital/metabolism , Heart Rate , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Action Potentials/drug effects , Adult , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Calcium Signaling , Case-Control Studies , Cell Differentiation , Cell Lineage , Cells, Cultured , ERG1 Potassium Channel/genetics , Genetic Predisposition to Disease , Heart Conduction System/metabolism , Heart Conduction System/physiopathology , Heart Defects, Congenital/drug therapy , Heart Defects, Congenital/genetics , Heart Defects, Congenital/physiopathology , Humans , Induced Pluripotent Stem Cells/drug effects , Kinetics , Male , Mutation, Missense , Myocytes, Cardiac/drug effects , Phenotype
5.
Int J Cardiol ; 254: 195-202, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29407091

ABSTRACT

BACKGROUND AND PURPOSE: Previous studies revealed that Takotsubo cardiomyopathy (TTC), a transient disorder of ventricular dysfunction affecting predominantly postmenopausal women, is associated with acquired long QT syndrome and arrhythmias, but the exact pathophysiologic mechanism is unknown. Our aim is to investigate the electrophysiological mechanism for QT-prolongation in TTC-patients by using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). METHODS: hiPSC-CMs, which were generated from human skin fibroblasts of three healthy donors, were treated by estradiol (10µM for one week) and a toxic concentration of isoprenaline (Iso, 1mM for 2h). Patch clamp techniques, qPCR and fluorescence-activated cell sorting (FACS) were employed for the study. KEY RESULTS: Iso enhanced late INa and suppressed Ito and thus prolonged the action potential duration (APD), suggesting possible reasons for arrhythmias in TTC. Iso elevated the production of reactive oxygen species (ROS). N-acetylcystein (1mM), a ROS-blocker, abolished the effects of Iso on late INa and Ito. H2O2 (100µM) mimicked Iso effects on late INa and Ito. These data indicate that the effects of Iso were mediated by ROS. Metoprolol (1mM), a beta-blocker, prevented the effects of Iso on late INa and APD, confirming the adrenoceptor-dependent effects of Iso. Estradiol treatment prevented the APD-prolongation, attenuated the enhancement of INa, diminished the reduction of Ito, suppressed ROS-production induced by Iso and reduced the expression levels of adrenoceptors, suggesting protective effects of estragon against toxic effects of catecholamine. CONCLUSIONS: Estradiol has protective effects against catecholamine excess and hence reduction in estrogen level may increase the risk of acquired long QT syndrome in TTC.


Subject(s)
Action Potentials/drug effects , Catecholamines/toxicity , Cytoprotection/drug effects , Estradiol/pharmacology , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Action Potentials/physiology , Cells, Cultured , Cytoprotection/physiology , Estradiol/therapeutic use , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , Induced Pluripotent Stem Cells/physiology , Long QT Syndrome/drug therapy , Long QT Syndrome/physiopathology , Myocytes, Cardiac/physiology , Reactive Oxygen Species/metabolism , Takotsubo Cardiomyopathy/drug therapy , Takotsubo Cardiomyopathy/physiopathology
6.
Sci Rep ; 7(1): 2935, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28592841

ABSTRACT

Severe infections like sepsis lead frequently to cardiomyopathy. The mechanisms are unclear and an optimal therapy for septic cardiomyopathy still lacks. The aim of this study is to establish an endotoxin-induced inflammatory model using human induced pluripotent stem cell (hiPSC) derived cardiomyocytes (hiPSC-CMs) for mechanistic and therapeutic studies. hiPSC-CMs were treated by lipopolysaccharide (LPS) in different concentrations for different times. ELISA, FACS, qPCR, and patch-clamp techniques were used for the study. TLR4 (Toll-like receptor 4) and its associated proteins, CD14, LBP (lipopolysaccharide binding protein), TIRAP (toll-interleukin 1 receptor domain containing adaptor protein), Ly96 (lymphocyte antigen 96) and nuclear factor kappa B as well as some pro-and anti-inflammatory factors are expressed in hiPSC-CMs. LPS-treatment for 6 hours increased the expression levels of pro-inflammatory and chemotactic cytokines (TNF-a, IL-1ß, IL-6, CCL2, CCL5, IL-8), whereas 48 hour-treatment elevated the expression of anti-inflammatory factors (IL-10 and IL-6). LPS led to cell injury resulting from exaggerated cell apoptosis and necrosis. Finally, LPS inhibited small conductance Ca2+-activated K+ channel currents, enhanced Na+/Ca2+-exchanger currents, prolonged action potential duration, suggesting cellular electrical dysfunctions. Our data demonstrate that hiPSC-CMs possess the functional reaction system involved in endotoxin-induced inflammation and can model some bacterium-induced inflammatory responses in cardiac myocytes.


Subject(s)
Electrophysiological Phenomena , Induced Pluripotent Stem Cells/cytology , Lipopolysaccharides/immunology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Action Potentials/drug effects , Action Potentials/immunology , Biomarkers , Cell Differentiation , Cell Line , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/metabolism , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Ion Channels/genetics , Ion Channels/metabolism , Lipopolysaccharides/adverse effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...