37 results on '"Ponce-Balbuena D"'
Search Results
2. Extracellular Matrix-Mediated Maturation of Human Pluripotent Stem Cell-Derived Cardiac Monolayer Structure and Electrophysiological Function
- Author
-
Herron T, Da Rocha A, Campbell K, Ponce-Balbuena D, Willis B, Guerrero-Serna G, Liu Q, Klos M, Musa H, Zarzoso M, Bizy A, Furness J, Anumonwo J, Mironov S, and Jalife J
- Published
- 2016
3. Structural bases for the different anti-fibrillatory effects of chloroquine and quinidine
- Author
-
Noujaim, S. F., primary, Stuckey, J. A., additional, Ponce-Balbuena, D., additional, Ferrer-Villada, T., additional, Lopez-Izquierdo, A., additional, Pandit, S. V., additional, Sanchez-Chapula, J. A., additional, and Jalife, J., additional
- Published
- 2011
- Full Text
- View/download PDF
4. Constitutive Intracellular Na+ Excess in Purkinje Cells Promotes Arrhythmogenesis at Lower Levels of Stress than Ventricular Myocytes from Mice with Catecholaminergic Polymorphic Ventricular Tachycardia
- Author
-
Cicero Willis, Pandit S, Ponce-Balbuena D, Zarzoso M, Guerrero-Serna G, Limbu B, Deo M, Cammors E, Rj, Ramirez, Mironov S, Tj, Herron, Hh, Valdivia, and Jalife J
5. Paradoxical SERCA dysregulation contributes to atrial fibrillation in a model of diet-induced obesity.
- Author
-
Ponce-Balbuena D, Tyrrell DJ, Cruz-Cortés C, Guerrero-Serna G, Da Rocha AM, Herron TJ, Song J, Raza DS, Anumonwo J, Goldstein DR, and Espinoza-Fonseca LM
- Abstract
Obesity is a major risk factor for atrial fibrillation (AF) the most common serious cardiac arrhythmia, but the molecular mechanisms underlying diet-induced AF remain unclear. In this study, we subjected mice to a chronic high-fat diet and acute sympathetic activation ('two-hit' model) to study the mechanisms by which diet-induced obesity promotes AF. Surface electrocardiography revealed that diet-induced obesity and sympathetic activation synergize during intracardiac tachypacing to induce AF. At the cellular level, diet-induced obesity and acute adrenergic stimulation facilitate the formation of delayed afterdepolarizations in atrial myocytes, implicating altered Ca
2+ dynamics as the underlying cause of AF. We found that diet-induced obesity does not alter the expression of major Ca2+ -handling proteins in atria, including the sarcoplasmic reticulum Ca2+ -ATPase (SERCA), a major component of beat-to-beat Ca2+ cycling in the heart. Paradoxically, obesity reduces phospholamban phosphorylation, suggesting decreased SERCA activity, yet atrial myocytes from obese mice showed a significantly increased Ca2+ transient amplitude and SERCA-mediated Ca2+ uptake. Adrenergic stimulation further increases the Ca2+ transient amplitude but does not affect Ca2+ reuptake in atrial myocytes from obese mice. Transcriptomics analysis showed that a high-fat diet prompts upregulation of neuronatin, a protein that has been implicated in obesity and is known to stimulate SERCA activity. We propose a mechanism in which obesity primes SERCA for paradoxical activation, and adrenergic stimulation facilitates AF conversion through a Ca2+ -induced Ca2+ release gain in atrial myocytes. Overall, this study links obesity, altered Ca2+ signaling, and AF, and targeting this mechanism may prove effective for treating obesity-induced AF., Competing Interests: Competing Interest Statement: D.P.-B., D.J.T., C.C.-C., G.G.S., A.M.D.R., J.S., D.S.R., J.A., D.R.G., and L.M.E.-F. declare no competing interests. T.J.H is co-founder of Cartox, Inc., and scientific advisor to StemBioSys, Inc.- Published
- 2024
- Full Text
- View/download PDF
6. p38γ/δ activation alters cardiac electrical activity and predisposes to ventricular arrhythmia.
- Author
-
Romero-Becerra R, Cruz FM, Mora A, Lopez JA, Ponce-Balbuena D, Allan A, Ramos-Mondragón R, González-Terán B, León M, Rodríguez ME, Leiva-Vega L, Guerrero-Serna G, Jimenez-Vazquez EN, Filgueiras-Rama D, Vázquez J, Jalife J, and Sabio G
- Subjects
- Animals, Action Potentials drug effects, Action Potentials physiology, Disease Models, Animal, Mice, Inbred C57BL, Mitogen-Activated Protein Kinase 13 metabolism, Mitogen-Activated Protein Kinase 13 genetics, Phosphorylation, Myocytes, Cardiac metabolism, Male, Enzyme Activation, Mice, Ventricular Premature Complexes physiopathology, Ventricular Premature Complexes genetics, Ventricular Premature Complexes metabolism, Sarcoplasmic Reticulum metabolism, Mice, Knockout, Humans, Calcium Signaling, Age Factors, Mitogen-Activated Protein Kinase 12 metabolism, Mitogen-Activated Protein Kinase 12 genetics, Ryanodine Receptor Calcium Release Channel metabolism, Ryanodine Receptor Calcium Release Channel genetics, Ventricular Fibrillation physiopathology, Ventricular Fibrillation metabolism, Ventricular Fibrillation genetics
- Abstract
Ventricular fibrillation (VF) is a leading immediate cause of sudden cardiac death. There is a strong association between aging and VF, although the mechanisms are unclear, limiting the availability of targeted therapeutic interventions. Here we found that the stress kinases p38γ and p38δ are activated in the ventricles of old mice and mice with genetic or drug-induced arrhythmogenic conditions. We discovered that, upon activation, p38γ and p38δ cooperatively increase the susceptibility to stress-induced VF. Mechanistically, our data indicate that activated p38γ and p38δ phosphorylate ryanodine receptor 2 (RyR2) disrupt Kv4.3 channel localization, promoting sarcoplasmic reticulum calcium leak, I
to current reduction and action potential duration prolongation. In turn, this led to aberrant intracellular calcium handling, premature ventricular complexes and enhanced susceptibility to VF. Blocking this pathway protected genetically modified animals from VF development and reduced the VF duration in aged animals. These results indicate that p38γ and p38δ are a potential therapeutic target for sustained VF prevention., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
7. Decreased FAM13B Expression Increases Atrial Fibrillation Susceptibility by Regulating Sodium Current and Calcium Handling.
- Author
-
Tchou G, Ponce-Balbuena D, Liu N, Gore-Panter S, Hsu J, Liu F, Opoku E, Brubaker G, Schumacher SM, Moravec CS, Barnard J, Van Wagoner DR, Chung MK, and Smith JD
- Abstract
A specific genetic variant associated with atrial fibrillation risk, rs17171731, was identified as a regulatory variant responsible for controlling FAM13B expression. The atrial fibrillation risk allele decreases FAM13B expression, whose knockdown alters the expression of many genes in stem cell-derived cardiomyocytes, including SCN2B , and led to pro-arrhythmogenic changes in the late sodium current and Ca
2+ cycling. Fam13b knockout mice had increased P-wave and QT interval duration and were more susceptible to pacing-induced arrhythmias vs control mice. FAM13B expression, its regulation, and downstream effects are potential targets for investigation of patient-specific therapeutics., Competing Interests: This work was supported by the National Institutes of Health (NIH) grants RO1 HL 111314 and P01HL158502 to Drs Chung, Van Wagoner, Barnard, and Smith; an American Heart Association Strategically Focused Research Network grant 18SFRN34110067 to Drs Chung, Van Wagoner, Barnard, and Smith; the NIH National Center for Research Resources for Case Western Reserve University and Cleveland Clinic Clinical and Translational Science Award UL1-RR024989; the Cleveland Clinic Department of Cardiovascular Medicine philanthropy research funds; and the Tomsich Atrial Fibrillation Research Fund. Dr Ponce-Balbuena was supported by an American Heart Association Career Development Award 20CDA35320040. Dr Hsu was supported by NIH training grant T32 GM 088088. Dr Smith was supported by the Geoffrey Gund Endowed Chair for Cardiovascular Research. The Fusion Lumos LC-MS instrument was purchased from the NIH shared instrument grant 1S10OD023436. The authors have reported that they have no relationships relevant to the contents of this paper to disclose., (© 2023 The Authors.)- Published
- 2023
- Full Text
- View/download PDF
8. SNTA1 gene rescues ion channel function and is antiarrhythmic in cardiomyocytes derived from induced pluripotent stem cells from muscular dystrophy patients.
- Author
-
Jimenez-Vazquez EN, Arad M, Macías Á, Vera-Pedrosa ML, Cruz FM, Gutierrez LK, Cuttitta AJ, Monteiro da Rocha A, Herron TJ, Ponce-Balbuena D, Guerrero-Serna G, Binah O, Michele DE, and Jalife J
- Subjects
- Action Potentials, Arrhythmias, Cardiac metabolism, Dystrophin genetics, Female, Humans, Male, Myocytes, Cardiac metabolism, Calcium-Binding Proteins genetics, Cardiomyopathies metabolism, Induced Pluripotent Stem Cells metabolism, Membrane Proteins genetics, Muscle Proteins genetics, Muscular Dystrophy, Duchenne genetics, Muscular Dystrophy, Duchenne metabolism, Potassium Channels, Inwardly Rectifying genetics, Potassium Channels, Inwardly Rectifying metabolism
- Abstract
Background: Patients with cardiomyopathy of Duchenne Muscular Dystrophy (DMD) are at risk of developing life-threatening arrhythmias, but the mechanisms are unknown. We aimed to determine the role of ion channels controlling cardiac excitability in the mechanisms of arrhythmias in DMD patients., Methods: To test whether dystrophin mutations lead to defective cardiac Na
V 1.5-Kir2.1 channelosomes and arrhythmias, we generated iPSC-CMs from two hemizygous DMD males, a heterozygous female, and two unrelated control males. We conducted studies including confocal microscopy, protein expression analysis, patch-clamping, non-viral piggy-bac gene expression, optical mapping and contractility assays., Results: Two patients had abnormal ECGs with frequent runs of ventricular tachycardia. iPSC-CMs from all DMD patients showed abnormal action potential profiles, slowed conduction velocities, and reduced sodium (INa ) and inward rectifier potassium (IK1 ) currents. Membrane NaV 1.5 and Kir2.1 protein levels were reduced in hemizygous DMD iPSC-CMs but not in heterozygous iPSC-CMs. Remarkably, transfecting just one component of the dystrophin protein complex (α1-syntrophin) in hemizygous iPSC-CMs from one patient restored channelosome function, INa and IK1 densities, and action potential profile in single cells. In addition, α1-syntrophin expression restored impulse conduction and contractility and prevented reentrant arrhythmias in hiPSC-CM monolayers., Conclusions: We provide the first demonstration that iPSC-CMs reprogrammed from skin fibroblasts of DMD patients with cardiomyopathy have a dysfunction of the NaV 1.5-Kir2.1 channelosome, with consequent reduction of cardiac excitability and conduction. Altogether, iPSC-CMs from patients with DMD cardiomyopathy have a NaV 1.5-Kir2.1 channelosome dysfunction, which can be rescued by the scaffolding protein α1-syntrophin to restore excitability and prevent arrhythmias., Funding: Supported by National Institutes of Health R01 HL122352 grant; 'la Caixa' Banking Foundation (HR18-00304); Fundación La Marató TV3: Ayudas a la investigación en enfermedades raras 2020 (LA MARATO-2020); Instituto de Salud Carlos III/FEDER/FSE; Horizon 2020 - Research and Innovation Framework Programme GA-965286 to JJ; the CNIC is supported by the Instituto de Salud Carlos III (ISCIII), the Ministerio de Ciencia e Innovación (MCIN) and the Pro CNIC Foundation), and is a Severo Ochoa Center of Excellence (grant CEX2020-001041-S funded by MICIN/AEI/10.13039/501100011033). American Heart Association postdoctoral fellowship 19POST34380706s to JVEN. Israel Science Foundation to OB and MA [824/19]. Rappaport grant [01012020RI]; and Niedersachsen Foundation [ZN3452] to OB; US-Israel Binational Science Foundation (BSF) to OB and TH [2019039]; Dr. Bernard Lublin Donation to OB; and The Duchenne Parent Project Netherlands (DPPNL 2029771) to OB. National Institutes of Health R01 AR068428 to DM and US-Israel Binational Science Foundation Grant [2013032] to DM and OB., Competing Interests: EJ, MA, ÁM, MV, FC, LG, AC, DP, GG, OB, DM No competing interests declared, AM Stembiosys consulting fees and stock options, TH, JJ Stembiosys stock options, (© 2022, Jimenez-Vazquez et al.)- Published
- 2022
- Full Text
- View/download PDF
9. Paclitaxel mitigates structural alterations and cardiac conduction system defects in a mouse model of Hutchinson-Gilford progeria syndrome.
- Author
-
Macías Á, Díaz-Larrosa JJ, Blanco Y, Fanjul V, González-Gómez C, Gonzalo P, Andrés-Manzano MJ, da Rocha AM, Ponce-Balbuena D, Allan A, Filgueiras-Rama D, Jalife J, and Andrés V
- Subjects
- Action Potentials drug effects, Animals, Arrhythmias, Cardiac genetics, Arrhythmias, Cardiac metabolism, Arrhythmias, Cardiac physiopathology, Cytoskeleton metabolism, Cytoskeleton pathology, Disease Models, Animal, Female, Genetic Predisposition to Disease, Heart Conduction System metabolism, Heart Conduction System physiopathology, Lamin Type A genetics, Lamin Type A metabolism, Male, Mice, Mutant Strains, Mutation, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Progeria genetics, Progeria metabolism, Progeria physiopathology, Refractory Period, Electrophysiological drug effects, Swine, Swine, Miniature, Tubulin metabolism, Mice, Anti-Arrhythmia Agents pharmacology, Arrhythmias, Cardiac drug therapy, Cytoskeleton drug effects, Excitation Contraction Coupling drug effects, Heart Conduction System drug effects, Heart Rate drug effects, Myocytes, Cardiac drug effects, Paclitaxel pharmacology, Progeria drug therapy
- Abstract
Aims: Hutchinson-Gilford progeria syndrome (HGPS) is an ultrarare laminopathy caused by expression of progerin, a lamin A variant, also present at low levels in non-HGPS individuals. HGPS patients age and die prematurely, predominantly from cardiovascular complications. Progerin-induced cardiac repolarization defects have been described previously, although the underlying mechanisms are unknown., Methods and Results: We conducted studies in heart tissue from progerin-expressing LmnaG609G/G609G (G609G) mice, including microscopy, intracellular calcium dynamics, patch-clamping, in vivo magnetic resonance imaging, and electrocardiography. G609G mouse cardiomyocytes showed tubulin-cytoskeleton disorganization, t-tubular system disruption, sarcomere shortening, altered excitation-contraction coupling, and reductions in ventricular thickening and cardiac index. G609G mice exhibited severe bradycardia, and significant alterations of atrio-ventricular conduction and repolarization. Most importantly, 50% of G609G mice had altered heart rate variability, and sinoatrial block, both significant signs of premature cardiac aging. G609G cardiomyocytes had electrophysiological alterations, which resulted in an elevated action potential plateau and early afterdepolarization bursting, reflecting slower sodium current inactivation and long Ca+2 transient duration, which may also help explain the mild QT prolongation in some HGPS patients. Chronic treatment with low-dose paclitaxel ameliorated structural and functional alterations in G609G hearts., Conclusions: Our results demonstrate that tubulin-cytoskeleton disorganization in progerin-expressing cardiomyocytes causes structural, cardiac conduction, and excitation-contraction coupling defects, all of which can be partially corrected by chronic treatment with low dose paclitaxel., (© The Author(s) 2021. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2022
- Full Text
- View/download PDF
10. In vitro model of ischemic heart failure using human induced pluripotent stem cell-derived cardiomyocytes.
- Author
-
Davis J, Chouman A, Creech J, Monteiro da Rocha A, Ponce-Balbuena D, Jimenez Vazquez EN, Nichols R, Lozhkin A, Madamanchi NR, Campbell KF, and Herron TJ
- Subjects
- Action Potentials physiology, Cell Differentiation physiology, Cells, Cultured, Humans, Heart Failure physiopathology, Induced Pluripotent Stem Cells physiology, Models, Cardiovascular, Myocardial Ischemia physiopathology, Myocytes, Cardiac physiology
- Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been used extensively to model inherited heart diseases, but hiPSC-CM models of ischemic heart disease are lacking. Here, our objective was to generate an hiPSC-CM model of ischemic heart disease. To this end, hiPSCs were differentiated into functional hiPSC-CMs and then purified using either a simulated ischemia media or by using magnetic antibody-based purification targeting the nonmyocyte population for depletion from the cell population. Flow cytometry analysis confirmed that each purification approach generated hiPSC-CM cultures that had more than 94% cTnT+ cells. After purification, hiPSC-CMs were replated as confluent syncytial monolayers for electrophysiological phenotype analysis and protein expression by Western blotting. The phenotype of metabolic stress-selected hiPSC-CM monolayers recapitulated many of the functional and structural hallmarks of ischemic CMs, including elevated diastolic calcium, diminished calcium transient amplitude, prolonged action potential duration, depolarized resting membrane potential, hypersensitivity to chemotherapy-induced cardiotoxicity, depolarized mitochondrial membrane potential, depressed SERCA2a expression, reduced maximal oxygen consumption rate, and abnormal response to β1-adrenergic receptor stimulation. These findings indicate that metabolic selection of hiPSC-CMs generated cell populations with phenotype similar to what is well known to occur in the setting of ischemic heart failure and thus provide a opportunity for study of human ischemic heart disease.
- Published
- 2021
- Full Text
- View/download PDF
11. Human influenza A virus causes myocardial and cardiac-specific conduction system infections associated with early inflammation and premature death.
- Author
-
Filgueiras-Rama D, Vasilijevic J, Jalife J, Noujaim SF, Alfonso JM, Nicolas-Avila JA, Gutierrez C, Zamarreño N, Hidalgo A, Bernabé A, Cop CP, Ponce-Balbuena D, Guerrero-Serna G, Calle D, Desco M, Ruiz-Cabello J, Nieto A, and Falcon A
- Subjects
- Animals, Connexins genetics, Cytokines metabolism, Disease Models, Animal, Dogs, Extracellular Matrix metabolism, Extracellular Matrix virology, Female, Fibrosis, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Heart Conduction System metabolism, Heart Conduction System pathology, Host-Pathogen Interactions, Humans, Inflammation Mediators metabolism, Alphainfluenzavirus genetics, Alphainfluenzavirus growth & development, Kinetics, Lung virology, Madin Darby Canine Kidney Cells, Mice, Inbred BALB C, Mice, Transgenic, Mutation, Myocarditis metabolism, Myocarditis pathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac virology, Orthomyxoviridae Infections metabolism, Orthomyxoviridae Infections pathology, Purkinje Fibers metabolism, Purkinje Fibers virology, Viral Load, Virulence, Virus Replication, Gap Junction alpha-5 Protein, Mice, Heart Conduction System virology, Alphainfluenzavirus pathogenicity, Myocarditis virology, Orthomyxoviridae Infections virology
- Abstract
Aims: Human influenza A virus (hIAV) infection is associated with important cardiovascular complications, although cardiac infection pathophysiology is poorly understood. We aimed to study the ability of hIAV of different pathogenicity to infect the mouse heart, and establish the relationship between the infective capacity and the associated in vivo, cellular and molecular alterations., Methods and Results: We evaluated lung and heart viral titres in mice infected with either one of several hIAV strains inoculated intranasally. 3D reconstructions of infected cardiac tissue were used to identify viral proteins inside mouse cardiomyocytes, Purkinje cells, and cardiac vessels. Viral replication was measured in mouse cultured cardiomyocytes. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were used to confirm infection and study underlying molecular alterations associated with the in vivo electrophysiological phenotype. Pathogenic and attenuated hIAV strains infected and replicated in cardiomyocytes, Purkinje cells, and hiPSC-CMs. The infection was also present in cardiac endothelial cells. Remarkably, lung viral titres did not statistically correlate with viral titres in the mouse heart. The highly pathogenic human recombinant virus PAmut showed faster replication, higher level of inflammatory cytokines in cardiac tissue and higher viral titres in cardiac HL-1 mouse cells and hiPSC-CMs compared with PB2mut-attenuated virus. Correspondingly, cardiac conduction alterations were especially pronounced in PAmut-infected mice, associated with high mortality rates, compared with PB2mut-infected animals. Consistently, connexin43 and NaV1.5 expression decreased acutely in hiPSC-CMs infected with PAmut virus. YEM1L protease also decreased more rapidly and to lower levels in PAmut-infected hiPSC-CMs compared with PB2mut-infected cells, consistent with mitochondrial dysfunction. Human IAV infection did not increase myocardial fibrosis at 4-day post-infection, although PAmut-infected mice showed an early increase in mRNAs expression of lysyl oxidase., Conclusion: Human IAV can infect the heart and cardiac-specific conduction system, which may contribute to cardiac complications and premature death., (© The Author(s) 2020. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2021
- Full Text
- View/download PDF
12. Long QT syndrome - Bench to bedside.
- Author
-
Ponce-Balbuena D and Deschênes I
- Abstract
Long QT syndrome (LQTS) is a cardiovascular disorder characterized by an abnormality in cardiac repolarization leading to a prolonged QT interval and T-wave irregularities on the surface electrocardiogram. It is commonly associated with syncope, seizures, susceptibility to torsades de pointes, and risk for sudden death. LQTS is a rare genetic disorder and a major preventable cause of sudden cardiac death in the young. The availability of therapy for this lethal disease emphasizes the importance of early and accurate diagnosis. Additionally, understanding of the molecular mechanisms underlying LQTS could help to optimize genotype-specific treatments to prevent deaths in LQTS patients. In this review, we briefly summarize current knowledge regarding molecular underpinning of LQTS, in particular focusing on LQT1, LQT2, and LQT3, and discuss novel strategies to study ion channel dysfunction and drug-specific therapies in LQT1, LQT2, and LQT3 syndromes., (© 2021 Heart Rhythm Society. Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
13. Human perinatal stem cell derived extracellular matrix enables rapid maturation of hiPSC-CM structural and functional phenotypes.
- Author
-
Block T, Creech J, da Rocha AM, Marinkovic M, Ponce-Balbuena D, Jiménez-Vázquez EN, Griffey S, and Herron TJ
- Subjects
- Amniotic Fluid metabolism, Cell Differentiation, Cell Shape, Cells, Cultured, Collagen pharmacology, Drug Combinations, Extracellular Matrix metabolism, Humans, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Laminin pharmacology, Mitochondria metabolism, Myocytes, Cardiac metabolism, Phenotype, Proteoglycans pharmacology, Toxicity Tests methods, Troponin I genetics, Troponin I metabolism, Amniotic Fluid cytology, Cellular Reprogramming Techniques methods, Extracellular Matrix Proteins pharmacology, Induced Pluripotent Stem Cells drug effects, Myocytes, Cardiac cytology
- Abstract
The immature phenotype of human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) is a major limitation to the use of these valuable cells for pre-clinical toxicity testing and for disease modeling. Here we tested the hypothesis that human perinatal stem cell derived extracellular matrix (ECM) promotes hiPSC-CM maturation to a greater extent than mouse cell derived ECM. We refer to the human ECM as Matrix Plus (Matrix Plus) and compare effects to commercially available mouse ECM (Matrigel). hiPSC-CMs cultured on Matrix Plus mature functionally and structurally seven days after thaw from cryopreservation. Mature hiPSC-CMs showed rod-shaped morphology, highly organized sarcomeres, elevated cTnI expression and mitochondrial distribution and function like adult cardiomyocytes. Matrix Plus also promoted mature hiPSC-CM electrophysiological function and monolayers' response to hERG ion channel specific blocker was Torsades de Pointes (TdP) reentrant arrhythmia activations in 100% of tested monolayers. Importantly, Matrix Plus enabled high throughput cardiotoxicity screening using mature human cardiomyocytes with validation utilizing reference compounds recommended for the evolving Comprehensive In Vitro Proarrhythmia Assay (CiPA) coordinated by the Health and Environmental Sciences Institute (HESI). Matrix Plus offers a solution to the commonly encountered problem of hiPSC-CM immaturity that has hindered implementation of these human based cell assays for pre-clinical drug discovery.
- Published
- 2020
- Full Text
- View/download PDF
14. Kir2.1 Interactome Mapping Uncovers PKP4 as a Modulator of the Kir2.1-Regulated Inward Rectifier Potassium Currents.
- Author
-
Park SS, Ponce-Balbuena D, Kuick R, Guerrero-Serna G, Yoon J, Mellacheruvu D, Conlon KP, Basrur V, Nesvizhskii AI, Jalife J, and Rual JF
- Subjects
- Action Potentials drug effects, Action Potentials physiology, Andersen Syndrome genetics, Andersen Syndrome physiopathology, Chromatography, Liquid, Desmosomes drug effects, Desmosomes metabolism, HEK293 Cells, Humans, Lysosomes metabolism, Molecular Chaperones metabolism, Mutation, Myocytes, Cardiac drug effects, Patch-Clamp Techniques, Potassium Channels, Inwardly Rectifying genetics, Protein Transport genetics, Protein Transport physiology, Signal Transduction genetics, Signal Transduction physiology, Somatomedins metabolism, Tandem Mass Spectrometry, Utrophin metabolism, Andersen Syndrome metabolism, Myocytes, Cardiac metabolism, Plakophilins metabolism, Potassium metabolism, Potassium Channels, Inwardly Rectifying metabolism, Protein Interaction Maps genetics, Protein Interaction Maps physiology
- Abstract
Kir2.1, a strong inward rectifier potassium channel encoded by the KCNJ2 gene, is a key regulator of the resting membrane potential of the cardiomyocyte and plays an important role in controlling ventricular excitation and action potential duration in the human heart. Mutations in KCNJ2 result in inheritable cardiac diseases in humans, e.g. the type-1 Andersen-Tawil syndrome (ATS1). Understanding the molecular mechanisms that govern the regulation of inward rectifier potassium currents by Kir2.1 in both normal and disease contexts should help uncover novel targets for therapeutic intervention in ATS1 and other Kir2.1-associated channelopathies. The information available to date on protein-protein interactions involving Kir2.1 channels remains limited. Additional efforts are necessary to provide a comprehensive map of the Kir2.1 interactome. Here we describe the generation of a comprehensive map of the Kir2.1 interactome using the proximity-labeling approach BioID. Most of the 218 high-confidence Kir2.1 channel interactions we identified are novel and encompass various molecular mechanisms of Kir2.1 function, ranging from intracellular trafficking to cross-talk with the insulin-like growth factor receptor signaling pathway, as well as lysosomal degradation. Our map also explores the variations in the interactome profiles of Kir2.1
WT versus Kir2.1Δ314-315 , a trafficking deficient ATS1 mutant, thus uncovering molecular mechanisms whose malfunctions may underlie ATS1 disease. Finally, using patch-clamp analysis, we validate the functional relevance of PKP4, one of our top BioID interactors, to the modulation of Kir2.1-controlled inward rectifier potassium currents. Our results validate the power of our BioID approach in identifying functionally relevant Kir2.1 interactors and underline the value of our Kir2.1 interactome as a repository for numerous novel biological hypotheses on Kir2.1 and Kir2.1-associated diseases., Competing Interests: Conflict of interest—Authors declare no competing interests., (© 2020 Park et al.)- Published
- 2020
- Full Text
- View/download PDF
15. Molecular and Electrophysiological Analyses of ATP2B4 Gene Variants in Bilateral Adrenal Hyperaldosteronism.
- Author
-
Hattangady NG, Foster J, Lerario AM, Ponce-Balbuena D, Rege J, Monticone S, Rainey WE, Mulatero P, and Else T
- Subjects
- Cell Line, Tumor, Female, Humans, Male, Electrophysiology methods, Genetic Variation genetics, High-Throughput Nucleotide Sequencing methods, Hyperaldosteronism genetics, Plasma Membrane Calcium-Transporting ATPases genetics
- Abstract
Primary aldosteronism (PA) is the most common cause of secondary hypertension with a high prevalence among patients with resistant hypertension. Despite the recent discovery of somatic variants in aldosterone-producing adenoma (APA)-associated PA, causes for PA due to bilateral aldosterone production (bilateral hyperaldosteronism; BHA) remain unknown. Herein, we identified rare gene variants in ATP2B4, in a cohort of patients with BHA. ATP2B4 belongs to the same family of Ca-ATPases as ATP2B3, which is involved in the pathogenesis of APA. Endogenous ATP2B4 expression was characterized in adrenal tissue, and the gene variants were functionally analyzed for effects on aldosterone synthase (CYP11B2) expression, steroid production in basal and agonist-stimulated conditions, and for changes in biophysical properties of channel properties. Knockdown of ATP2B4 in HAC15 exhibited reduced angiotensin II stimulation in one of four shRNA clones. Stable HAC15 cell lines with doxycycline (dox) - inducible wild-type and variant forms of ATP2B4 - were generated, and dox-induced upregulation of ATP2B4 mRNA and protein was confirmed. However, ATP2B4 variants did not alter basal or agonist-stimulated CYP11B2 expression. Whole-cell recordings in HAC15 cells indicated robust endogenous ATP2B4 conductance in native cells but reduced conductance with overexpressed WT and variant ATP2B4. The previously defined PA-causing ATP2B3 variant served as a positive control and exhibited elevated CYP11B2 mRNA. In conclusion, while this study did not confirm a pathogenic role for ATP2B4 variants in BHA, we describe the sequencing analysis for familial and sporadic BHA and outline a template for the thorough in vitro characterization of gene variants.
- Published
- 2020
- Full Text
- View/download PDF
16. Brugada syndrome trafficking-defective Nav1.5 channels can trap cardiac Kir2.1/2.2 channels.
- Author
-
Pérez-Hernández M, Matamoros M, Alfayate S, Nieto-Marín P, Utrilla RG, Tinaquero D, de Andrés R, Crespo T, Ponce-Balbuena D, Willis BC, Jiménez-Vazquez EN, Guerrero-Serna G, da Rocha AM, Campbell K, Herron TJ, Díez-Guerra FJ, Tamargo J, Jalife J, Caballero R, and Delpón E
- Subjects
- Animals, Arrhythmias, Cardiac metabolism, CHO Cells, Cricetulus, Golgi Matrix Proteins, Humans, Induced Pluripotent Stem Cells, Male, Mice, Mice, Knockout, Mice, Transgenic, Myocytes, Cardiac metabolism, NAV1.5 Voltage-Gated Sodium Channel genetics, Potassium Channels metabolism, Potassium Channels, Inwardly Rectifying genetics, Rats, Rats, Sprague-Dawley, Sodium Channels metabolism, Brugada Syndrome metabolism, NAV1.5 Voltage-Gated Sodium Channel metabolism, Potassium Channels, Inwardly Rectifying metabolism
- Abstract
Cardiac Nav1.5 and Kir2.1-2.3 channels generate Na (INa) and inward rectifier K (IK1) currents, respectively. The functional INa and IK1 interplay is reinforced by the positive and reciprocal modulation between Nav15 and Kir2.1/2.2 channels to strengthen the control of ventricular excitability. Loss-of-function mutations in the SCN5A gene, which encodes Nav1.5 channels, underlie several inherited arrhythmogenic syndromes, including Brugada syndrome (BrS). We investigated whether the presence of BrS-associated mutations alters IK1 density concomitantly with INa density. Results obtained using mouse models of SCN5A haploinsufficiency, and the overexpression of native and mutated Nav1.5 channels in expression systems - rat ventricular cardiomyocytes and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) - demonstrated that endoplasmic reticulum (ER) trafficking-defective Nav1.5 channels significantly decreased IK1, since they did not positively modulate Kir2.1/2.2 channels. Moreover, Golgi trafficking-defective Nav1.5 mutants produced a dominant negative effect on Kir2.1/2.2 and thus an additional IK1 reduction. Moreover, ER trafficking-defective Nav1.5 channels can be partially rescued by Kir2.1/2.2 channels through an unconventional secretory route that involves Golgi reassembly stacking proteins (GRASPs). Therefore, cardiac excitability would be greatly affected in subjects harboring Nav1.5 mutations with Golgi trafficking defects, since these mutants can concomitantly trap Kir2.1/2.2 channels, thus unexpectedly decreasing IK1 in addition to INa.
- Published
- 2018
- Full Text
- View/download PDF
17. Eplerenone Reduces Atrial Fibrillation Burden Without Preventing Atrial Electrical Remodeling.
- Author
-
Takemoto Y, Ramirez RJ, Kaur K, Salvador-Montañés O, Ponce-Balbuena D, Ramos-Mondragón R, Ennis SR, Guerrero-Serna G, Berenfeld O, and Jalife J
- Subjects
- Animals, Atrial Fibrillation pathology, Cardiac Pacing, Artificial, Eplerenone, Fibrosis, Male, Sheep, Spironolactone therapeutic use, Atrial Fibrillation prevention & control, Atrial Remodeling drug effects, Mineralocorticoid Receptor Antagonists therapeutic use, Spironolactone analogs & derivatives
- Abstract
Background: The aldosterone inhibitor eplerenone (EPL) has been shown to reduce the incidence of atrial fibrillation (AF) in patients with systolic heart failure, but the mechanism is unknown., Objectives: This study hypothesized that by reducing atrial dilation and fibrosis in the absence of heart failure, EPL also reduces AF burden and prevents AF perpetuation., Methods: The authors conducted a randomized controlled study in 34 sheep that were atrially tachypaced (13 ± 1 week). They compared daily oral EPL (n = 19) versus sugar pill (SP) treatment (n = 15) from the start of tachypacing. The endpoint was a continuous 7-day stretch of persistent AF (n = 29) or completion of 23 weeks tachypacing (n = 5)., Results: EPL significantly reduced the rate of left atrial dilation increase during AF progression. Atria from EPL-treated sheep had less smooth muscle actin protein, collagen-III expression, interstitial atrial fibrosis, and cell hypertrophy than SP-treated sheep atria did. However, EPL did not modify the AF-induced increase in the rate of dominant frequency and ion channel densities seen under SP treatment, but rather prolonged the time to persistent AF in 26% of animals. It also reduced the degree of fibrillatory conduction, AF inducibility, and AF burden., Conclusions: In the sheep model, EPL mitigates fibrosis and atrial dilation, modifies AF inducibility and AF complexity, and prolongs the transition to persistent AF in 26% of animals, but it does not prevent AF-induced electrical remodeling or AF persistence. The results highlight structural remodeling as a central upstream target to reduce AF burden, and the need to prevent electrical remodeling to avert AF perpetuation., (Copyright © 2017 American College of Cardiology Foundation. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
18. Tbx20 controls the expression of the KCNH2 gene and of hERG channels.
- Author
-
Caballero R, Utrilla RG, Amorós I, Matamoros M, Pérez-Hernández M, Tinaquero D, Alfayate S, Nieto-Marín P, Guerrero-Serna G, Liu QH, Ramos-Mondragón R, Ponce-Balbuena D, Herron T, Campbell KF, Filgueiras-Rama D, Peinado R, López-Sendón JL, Jalife J, Delpón E, and Tamargo J
- Subjects
- Action Potentials genetics, Animals, Arrhythmias, Cardiac genetics, Arrhythmias, Cardiac metabolism, CHO Cells, Cell Line, Cricetulus, Heterozygote, Humans, Induced Pluripotent Stem Cells metabolism, Long QT Syndrome genetics, Long QT Syndrome metabolism, Male, Mice, Mutation genetics, Myocytes, Cardiac metabolism, Rats, Rats, Sprague-Dawley, ERG1 Potassium Channel genetics, ERG1 Potassium Channel metabolism, Ether-A-Go-Go Potassium Channels genetics, Ether-A-Go-Go Potassium Channels metabolism, T-Box Domain Proteins genetics, T-Box Domain Proteins metabolism
- Abstract
Long QT syndrome (LQTS) exhibits great phenotype variability among family members carrying the same mutation, which can be partially attributed to genetic factors. We functionally analyzed the KCNH2 (encoding for Kv11.1 or hERG channels) and TBX20 (encoding for the transcription factor Tbx20) variants found by next-generation sequencing in two siblings with LQTS in a Spanish family of African ancestry. Affected relatives harbor a heterozygous mutation in KCNH2 that encodes for p.T152HfsX180 Kv11.1 (hERG). This peptide, by itself, failed to generate any current when transfected into Chinese hamster ovary (CHO) cells but, surprisingly, exerted "chaperone-like" effects over native hERG channels in both CHO cells and mouse atrial-derived HL-1 cells. Therefore, heterozygous transfection of native (WT) and p.T152HfsX180 hERG channels generated a current that was indistinguishable from that generated by WT channels alone. Some affected relatives also harbor the p.R311C mutation in Tbx20. In human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), Tbx20 enhanced human KCNH2 gene expression and hERG currents (I
hERG ) and shortened action-potential duration (APD). However, Tbx20 did not modify the expression or activity of any other channel involved in ventricular repolarization. Conversely, p.R311C Tbx20 did not increase KCNH2 expression in hiPSC-CMs, which led to decreased IhERG and increased APD. Our results suggest that Tbx20 controls the expression of hERG channels responsible for the rapid component of the delayed rectifier current. On the contrary, p.R311C Tbx20 specifically disables the Tbx20 protranscriptional activity over KCNH2 Therefore, TBX20 can be considered a KCNH2-modifying gene., Competing Interests: The authors declare no conflict of interest.- Published
- 2017
- Full Text
- View/download PDF
19. Mutated KCNJ5 activates the acute and chronic regulatory steps in aldosterone production.
- Author
-
Hattangady NG, Karashima S, Yuan L, Ponce-Balbuena D, Jalife J, Gomez-Sanchez CE, Auchus RJ, Rainey WE, and Else T
- Subjects
- Adrenal Glands metabolism, Calcium Channel Blockers pharmacology, Cell Line, Tumor, Cytochrome P-450 CYP11B2 genetics, Cytochrome P-450 CYP11B2 metabolism, G Protein-Coupled Inwardly-Rectifying Potassium Channels metabolism, Gene Expression, Gene Expression Regulation drug effects, Genetic Vectors genetics, Humans, RNA, Messenger genetics, RNA, Messenger metabolism, Steroids biosynthesis, Transcription, Genetic, Transduction, Genetic, Aldosterone biosynthesis, G Protein-Coupled Inwardly-Rectifying Potassium Channels genetics, Mutation
- Abstract
Somatic and germline mutations in the inward-rectifying K(+) channel (KCNJ5) are a common cause of primary aldosteronism (PA) in aldosterone-producing adenoma and familial hyperaldosteronism type III, respectively. Dysregulation of adrenal cell calcium signaling represents one mechanism for mutated KCNJ5 stimulation of aldosterone synthase (CYP11B2) expression and aldosterone production. However, the mechanisms stimulating acute and chronic production of aldosterone by mutant KCNJ5 have not been fully characterized. Herein, we defined the effects of the T158A KCNJ5 mutation (KCNJ5(T158A)) on acute and chronic regulation of aldosterone production using an adrenal cell line with a doxycycline-inducible KCNJ5(T158A) gene (HAC15-TRE-KCNJ5(T158A)). Doxycycline incubation caused a time-dependent increase in KCNJ5(T158A) and CYP11B2 mRNA and protein levels. Electrophysiological analyses confirm the loss of inward rectification and increased Na(+) permeability in KCNJ5(T158A)-expressing cells. KCNJ5(T158A) expression also led to the activation of CYP11B2 transcriptional regulators, NURR1 and ATF2. Acutely, KCNJ5(T158A) stimulated the expression of total and phosphorylated steroidogenic acute regulatory protein (StAR). KCNJ5(T158A) expression increased the synthesis of aldosterone and the hybrid steroids 18-hydroxycortisol and 18-oxocortisol, measured with liquid chromatography-tandem mass spectrometry (LC-MS/MS). All of these stimulatory effects of KCNJ5(T158A) were inhibited by the L-type Ca(2+) channel blocker, verapamil. Overall, KCNJ5(T158A)increases CYP11B2 expression and production of aldosterone, corticosterone and hybrid steroids by upregulating both acute and chronic regulatory events in aldosterone production, and verapamil blocks KCNJ5(T158A)-mediated pathways leading to aldosterone production., Competing Interests: DECLARATION OF INTEREST: We hereby declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported., (© 2016 Society for Endocrinology.)
- Published
- 2016
- Full Text
- View/download PDF
20. Constitutive Intracellular Na+ Excess in Purkinje Cells Promotes Arrhythmogenesis at Lower Levels of Stress Than Ventricular Myocytes From Mice With Catecholaminergic Polymorphic Ventricular Tachycardia.
- Author
-
Willis BC, Pandit SV, Ponce-Balbuena D, Zarzoso M, Guerrero-Serna G, Limbu B, Deo M, Camors E, Ramirez RJ, Mironov S, Herron TJ, Valdivia HH, and Jalife J
- Subjects
- Animals, Calcium Signaling, Humans, Mice, Purkinje Cells, Calcium metabolism, Myocytes, Cardiac physiology, Sodium metabolism, Tachycardia, Ventricular metabolism
- Abstract
Background: In catecholaminergic polymorphic ventricular tachycardia (CPVT), cardiac Purkinje cells (PCs) appear more susceptible to Ca(2+) dysfunction than ventricular myocytes (VMs). The underlying mechanisms remain unknown. Using a CPVT mouse (RyR2(R4496C+/Cx40eGFP)), we tested whether PC intracellular Ca(2+) ([Ca(2+)]i) dysregulation results from a constitutive [Na(+)]i surplus relative to VMs., Methods and Results: Simultaneous optical mapping of voltage and [Ca(2+)]i in CPVT hearts showed that spontaneous Ca(2+) release preceded pacing-induced triggered activity at subendocardial PCs. On simultaneous current-clamp and Ca(2+) imaging, early and delayed afterdepolarizations trailed spontaneous Ca(2+) release and were more frequent in CPVT PCs than CPVT VMs. As a result of increased activity of mutant ryanodine receptor type 2 channels, sarcoplasmic reticulum Ca(2+) load, measured by caffeine-induced Ca(2+) transients, was lower in CPVT VMs and PCs than respective controls, and sarcoplasmic reticulum fractional release was greater in both CPVT PCs and VMs than respective controls. [Na(+)]i was higher in both control and CPVT PCs than VMs, whereas the density of the Na(+)/Ca(2+) exchanger current was not different between PCs and VMs. Computer simulations using a PC model predicted that the elevated [Na(+)]i of PCs promoted delayed afterdepolarizations, which were always preceded by spontaneous Ca(2+) release events from hyperactive ryanodine receptor type 2 channels. Increasing [Na(+)]i monotonically increased delayed afterdepolarization frequency. Confocal imaging experiments showed that postpacing Ca(2+) spark frequency was highest in intact CPVT PCs, but such differences were reversed on saponin-induced membrane permeabilization, indicating that differences in [Na(+)]i played a central role., Conclusions: In CPVT mice, the constitutive [Na(+)]i excess of PCs promotes triggered activity and arrhythmogenesis at lower levels of stress than VMs., (© 2016 The Authors.)
- Published
- 2016
- Full Text
- View/download PDF
21. Nav1.5 N-terminal domain binding to α1-syntrophin increases membrane density of human Kir2.1, Kir2.2 and Nav1.5 channels.
- Author
-
Matamoros M, Pérez-Hernández M, Guerrero-Serna G, Amorós I, Barana A, Núñez M, Ponce-Balbuena D, Sacristán S, Gómez R, Tamargo J, Caballero R, Jalife J, and Delpón E
- Subjects
- Animals, Cricetinae, Heart Ventricles metabolism, Humans, Mutagenesis, Site-Directed, Myocytes, Cardiac metabolism, Rats, Calcium-Binding Proteins metabolism, Membrane Proteins metabolism, Muscle Proteins metabolism, NAV1.5 Voltage-Gated Sodium Channel metabolism, Potassium Channels, Inwardly Rectifying metabolism
- Abstract
Aims: Cardiac excitability and refractoriness are largely determined by the function and number of inward rectifier K⁺ channels (Kir2.1-2.3), which are differentially expressed in the atria and ventricles, and Nav1.5 channels. We have focused on how Nav1.5 and Kir2.x function within a macromolecular complex by elucidating the molecular determinants that govern Nav1.5/Kir2.x reciprocal modulation., Methods and Results: The results demonstrate that there is an unexpected 'internal' PDZ-like binding domain located at the N-terminus of the Nav1.5 channel that mediates its binding to α1-syntrophin. Nav1.5 N-terminal domain, by itself (the 132 aa peptide) (Nter), exerts a 'chaperone-like' effect that increases sodium (I(Na)) and inward rectifier potassium (I(K1)) currents by enhancing the expression of Nav1.5, Kir2.1, and Kir2.2 channels as demonstrated in Chinese hamster ovary (CHO) cells and in rat cardiomyocytes. Site-directed mutagenesis analysis demonstrates that the Nter chaperone-like effect is determined by Serine 20. Nav1.5-Kir2.x reciprocal positive interactions depend on a specific C-terminal PDZ-binding domain sequence (SEI), which is present in Kir2.1 and Kir2.2 channels but not in Kir2.3. Therefore, in human atrial myocytes, the presence of Kir2.3 isoforms precludes reciprocal I(K1)-INa density modulation. Moreover, results in rat and human atrial myocytes demonstrate that binding to α1-syntrophin is necessary for the Nav1.5-Kir2.x-positive reciprocal modulation., Conclusions: The results demonstrate the critical role of the N-terminal domain of Nav1.5 channels in Nav1.5-Kir2.x-reciprocal interactions and suggest that the molecular mechanisms controlling atrial and ventricular cellular excitability may be different., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author 2016. For permissions please email: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
22. Extracellular Matrix-Mediated Maturation of Human Pluripotent Stem Cell-Derived Cardiac Monolayer Structure and Electrophysiological Function.
- Author
-
Herron TJ, Rocha AM, Campbell KF, Ponce-Balbuena D, Willis BC, Guerrero-Serna G, Liu Q, Klos M, Musa H, Zarzoso M, Bizy A, Furness J, Anumonwo J, Mironov S, and Jalife J
- Subjects
- Action Potentials physiology, Cell Differentiation, Cell Line, Humans, Induced Pluripotent Stem Cells cytology, Myocytes, Cardiac cytology, Signal Transduction, Electrophysiological Phenomena physiology, Extracellular Matrix physiology, Induced Pluripotent Stem Cells metabolism, Myocytes, Cardiac metabolism
- Abstract
Background: Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) monolayers generated to date display an immature embryonic-like functional and structural phenotype that limits their utility for research and cardiac regeneration. In particular, the electrophysiological function of hPSC-CM monolayers and bioengineered constructs used to date are characterized by slow electric impulse propagation velocity and immature action potential profiles., Methods and Results: Here, we have identified an optimal extracellular matrix for significant electrophysiological and structural maturation of hPSC-CM monolayers. hPSC-CM plated in the optimal extracellular matrix combination have impulse propagation velocities ≈2× faster than previously reported (43.6±7.0 cm/s; n=9) and have mature cardiomyocyte action potential profiles, including hyperpolarized diastolic potential and rapid action potential upstroke velocity (146.5±17.7 V/s; n=5 monolayers). In addition, the optimal extracellular matrix promoted hypertrophic growth of cardiomyocytes and the expression of key mature sarcolemmal (SCN5A, Kir2.1, and connexin43) and myofilament markers (cardiac troponin I). The maturation process reported here relies on activation of integrin signaling pathways: neutralization of β1 integrin receptors via blocking antibodies and pharmacological blockade of focal adhesion kinase activation prevented structural maturation., Conclusions: Maturation of human stem cell-derived cardiomyocyte monolayers is achieved in a 1-week period by plating cardiomyocytes on PDMS (polydimethylsiloxane) coverslips rather than on conventional 2-dimensional cell culture formats, such as glass coverslips or plastic dishes. Activation of integrin signaling and focal adhesion kinase is essential for significant maturation of human cardiac monolayers., (© 2016 American Heart Association, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
23. Galectin-3 Regulates Atrial Fibrillation Remodeling and Predicts Catheter Ablation Outcomes.
- Author
-
Takemoto Y, Ramirez RJ, Yokokawa M, Kaur K, Ponce-Balbuena D, Sinno MC, Willis BC, Ghanbari H, Ennis SR, Guerrero-Serna G, Henzi BC, Latchamsetty R, Ramos-Mondragon R, Musa H, Martins RP, Pandit SV, Noujaim SF, Crawford T, Jongnarangsin K, Pelosi F, Bogun F, Chugh A, Berenfeld O, Morady F, Oral H, and Jalife J
- Abstract
Objectives: To determine whether Gal-3 mediates sustained atrial fibrillation (AF)-induced atrial structural and electrical remodeling and contributes to AF perpetuation., Background: Galectin-3 (Gal-3) mediates extracellular matrix remodeling in heart failure, but its role in AF progression remains unexplored., Methods: We examined intracardiac blood samples from patients with AF ( N=55 ) to identify potential biomarkers of AF recurrence. In a sheep model of tachypacing-induced AF ( N=20 ), we tested the effects of Gal-3 inhibition during AF progression., Results: In patients, intracardiac serum Gal-3 levels were greater in persistent than paroxysmal AF and independently predicted atrial tachyarrhythmia recurrences after a single ablation procedure. In the sheep model, both Gal-3 and TGF-β1 were elevated in the atria of persistent AF animals. The Gal-3 inhibitor GM-CT-01 (GMCT) reduced both Gal-3 and TGF-β1-induced sheep atrial fibroblast migration and proliferation in vitro . GMCT (12 mg/kg twice/week) prevented the increase in serum procollagen type III N-terminal peptide seen during progression to persistent AF, and also mitigated atrial dilatation, myocyte hypertrophy, fibrosis, and the expected increase in dominant frequency of excitation. Atria of GMCT-treated animals had significantly less TGF-β1-Smad2/3 signaling pathway activation and expression of α-smooth muscle actin and collagen than saline-treated animals. Ex-vivo hearts from GMCT-treated animals had significantly longer action potential durations and fewer rotors and wavebreaks during AF, and myocytes had lower functional expression of inward rectifier K
+ channel (Kir2.3) than saline-treated animals. Importantly, GMCT increased the probability of spontaneous AF termination, decreased AF inducibility and reduced overall AF burden., Conclusions: Inhibiting Gal-3 during AF progression might be useful as an adjuvant treatment to improve outcomes of catheter ablation for persistent AF. Gal-3 inhibition may be a potential new upstream therapy for prevention of AF progression.- Published
- 2016
- Full Text
- View/download PDF
24. Protein assemblies of sodium and inward rectifier potassium channels control cardiac excitability and arrhythmogenesis.
- Author
-
Willis BC, Ponce-Balbuena D, and Jalife J
- Subjects
- Action Potentials, Animals, Arrhythmias, Cardiac genetics, Arrhythmias, Cardiac physiopathology, Humans, Membrane Microdomains metabolism, Multiprotein Complexes, Signal Transduction, Arrhythmias, Cardiac metabolism, Myocytes, Cardiac metabolism, NAV1.5 Voltage-Gated Sodium Channel metabolism, Potassium Channels, Inwardly Rectifying metabolism
- Abstract
The understanding of how cardiac ion channels function in the normal and the diseased heart has greatly increased over the last four decades thanks to the advent of patch-clamp technology and, more recently, the emergence of genetics, as well as cellular and molecular cardiology. However, our knowledge of how these membrane-embedded proteins physically interact with each other within macromolecular complexes remains incomplete. This review focuses on how the main cardiac inward sodium channel (NaV1.5) and the strong inward rectifier potassium channel (Kir2.1) function within macromolecular complexes to control cardiac excitability. It has become increasingly clear that these two important ion channel proteins physically interact with multiple other protein partners and with each other from early stages of protein trafficking and targeting through membrane anchoring, recycling, and degradation. Recent findings include compartmentalized regulation of NaV1.5 channel expression and function through a PDZ (postsynaptic density protein, Drosophila disc large tumor suppressor, and zonula occludens-1 protein) domain-binding motif, and interaction of caveolin-3 with Kir2.1 and ankyrin-G as a molecular platform for NaV1.5 signaling. At the cardiomyocyte membrane, NaV1.5 and Kir2.1 interact through at least two distinct PDZ domain-scaffolding proteins (synapse-associated protein-97 and α1-syntrophin), thus modulating reciprocally their cell-surface expression at two different microdomains. Emerging evidence also shows that inheritable mutations in plakophilin-2, ankyrin-G, dystrophin, syntrophin, synapse-associated protein-97, and caveolin-3, among others, modify functional expression and/or localization in the cardiac cell of NaV1.5, Kir2.1 or both to give rise to arrhythmogenic diseases. Unveiling the mechanistic underpinnings of macromolecular interactions should increase our understanding of inherited and acquired arrhythmogenic cardiac diseases and may lead to advances in therapy., (Copyright © 2015 the American Physiological Society.)
- Published
- 2015
- Full Text
- View/download PDF
25. Dominant frequency increase rate predicts transition from paroxysmal to long-term persistent atrial fibrillation.
- Author
-
Martins RP, Kaur K, Hwang E, Ramirez RJ, Willis BC, Filgueiras-Rama D, Ennis SR, Takemoto Y, Ponce-Balbuena D, Zarzoso M, O'Connell RP, Musa H, Guerrero-Serna G, Avula UM, Swartz MF, Bhushal S, Deo M, Pandit SV, Berenfeld O, and Jalife J
- Subjects
- Animals, Cardiac Pacing, Artificial, Disease Models, Animal, Electrophysiologic Techniques, Cardiac, Hypertrophy, Myocytes, Cardiac pathology, Patch-Clamp Techniques, Sheep, Time Factors, Action Potentials physiology, Atrial Fibrillation physiopathology, Calcium Channels, L-Type physiology, Disease Progression, Heart Rate physiology, Potassium Channels, Inwardly Rectifying physiology, Sinoatrial Node physiopathology, Sodium Channels physiology
- Abstract
Background: Little is known about the mechanisms underlying the transition from paroxysmal to persistent atrial fibrillation (AF). In an ovine model of long-standing persistent AF we tested the hypothesis that the rate of electric and structural remodeling, assessed by dominant frequency (DF) changes, determines the time at which AF becomes persistent., Methods and Results: Self-sustained AF was induced by atrial tachypacing. Seven sheep were euthanized 11.5±2.3 days after the transition to persistent AF and without reversal to sinus rhythm; 7 sheep were euthanized after 341.3±16.7 days of long-standing persistent AF. Seven sham-operated animals were in sinus rhythm for 1 year. DF was monitored continuously in each group. Real-time polymerase chain reaction, Western blotting, patch clamping, and histological analyses were used to determine the changes in functional ion channel expression and structural remodeling. Atrial dilatation, mitral valve regurgitation, myocyte hypertrophy, and atrial fibrosis occurred progressively and became statistically significant after the transition to persistent AF, with no evidence for left ventricular dysfunction. DF increased progressively during the paroxysmal-to-persistent AF transition and stabilized when AF became persistent. Importantly, the rate of DF increase correlated strongly with the time to persistent AF. Significant action potential duration abbreviation, secondary to functional ion channel protein expression changes (CaV1.2, NaV1.5, and KV4.2 decrease; Kir2.3 increase), was already present at the transition and persisted for 1 year of follow up., Conclusions: In the sheep model of long-standing persistent AF, the rate of DF increase predicts the time at which AF stabilizes and becomes persistent, reflecting changes in action potential duration and densities of sodium, L-type calcium, and inward rectifier currents.
- Published
- 2014
- Full Text
- View/download PDF
26. ABCC9 is a novel Brugada and early repolarization syndrome susceptibility gene.
- Author
-
Hu D, Barajas-Martínez H, Terzic A, Park S, Pfeiffer R, Burashnikov E, Wu Y, Borggrefe M, Veltmann C, Schimpf R, Cai JJ, Nam GB, Deshmukh P, Scheinman M, Preminger M, Steinberg J, López-Izquierdo A, Ponce-Balbuena D, Wolpert C, Haïssaguerre M, Sánchez-Chapula JA, and Antzelevitch C
- Subjects
- Adolescent, Adult, Aged, Amino Acid Sequence, Animals, Brugada Syndrome diagnosis, Female, HEK293 Cells, Humans, Male, Mice, Middle Aged, Molecular Sequence Data, Protein Structure, Secondary, Rabbits, Rats, Sulfonylurea Receptors chemistry, Young Adult, Brugada Syndrome epidemiology, Brugada Syndrome genetics, Genetic Predisposition to Disease epidemiology, Genetic Predisposition to Disease genetics, Mutation genetics, Sulfonylurea Receptors genetics
- Abstract
Background: Genetic defects in KCNJ8, encoding the Kir6.1 subunit of the ATP-sensitive K(+) channel (I(K-ATP)), have previously been associated with early repolarization (ERS) and Brugada (BrS) syndromes. Here we test the hypothesis that genetic variants in ABCC9, encoding the ATP-binding cassette transporter of IK-ATP (SUR2A), are also associated with both BrS and ERS., Methods and Results: Direct sequencing of all ERS/BrS susceptibility genes was performed on 150 probands and family members. Whole-cell and inside-out patch-clamp methods were used to characterize mutant channels expressed in TSA201-cells. Eight ABCC9 mutations were uncovered in 11 male BrS probands. Four probands, diagnosed with ERS, carried a highly-conserved mutation, V734I-ABCC9. Functional expression of the V734I variant yielded a Mg-ATP IC₅₀ that was 5-fold that of wild-type (WT). An 18-y/o male with global ERS inherited an SCN5A-E1784K mutation from his mother, who displayed long QT intervals, and S1402C-ABCC9 mutation from his father, who displayed an ER pattern. ABCC9-S1402C likewise caused a gain of function of IK-ATP with a shift of ATP IC₅₀ from 8.5 ± 2 mM to 13.4 ± 5 μM (p<0.05). The SCN5A mutation reduced peak INa to 39% of WT (p<0.01), shifted steady-state inactivation by -18.0 mV (p<0.01) and increased late I(Na) from 0.14% to 2.01% of peak I(Na) (p<0.01)., Conclusion: Our study is the first to identify ABCC9 as a susceptibility gene for ERS and BrS. Our findings also suggest that a gain-of-function in I(K-ATP) when coupled with a loss-of-function in SCN5A may underlie type 3 ERS, which is associated with a severe arrhythmic phenotype., (Copyright © 2013 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
27. Myosin light chain 2-based selection of human iPSC-derived early ventricular cardiac myocytes.
- Author
-
Bizy A, Guerrero-Serna G, Hu B, Ponce-Balbuena D, Willis BC, Zarzoso M, Ramirez RJ, Sener MF, Mundada LV, Klos M, Devaney EJ, Vikstrom KL, Herron TJ, and Jalife J
- Subjects
- Adenoviridae genetics, Cardiac Myosins genetics, Cell Differentiation, Cell Lineage, Flow Cytometry, Genes, Reporter, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Humans, Induced Pluripotent Stem Cells metabolism, Myocytes, Cardiac metabolism, Myosin Light Chains genetics, Phenotype, Promoter Regions, Genetic, Protein Isoforms genetics, Protein Isoforms metabolism, Cardiac Myosins metabolism, Cell Separation methods, Heart Ventricles cytology, Induced Pluripotent Stem Cells cytology, Myocytes, Cardiac cytology, Myosin Light Chains metabolism
- Abstract
Applications of human induced pluripotent stem cell derived-cardiac myocytes (hiPSC-CMs) would be strengthened by the ability to generate specific cardiac myocyte (CM) lineages. However, purification of lineage-specific hiPSC-CMs is limited by the lack of cell marking techniques. Here, we have developed an iPSC-CM marking system using recombinant adenoviral reporter constructs with atrial- or ventricular-specific myosin light chain-2 (MLC-2) promoters. MLC-2a and MLC-2v selected hiPSC-CMs were purified by fluorescence-activated cell sorting and their biochemical and electrophysiological phenotypes analyzed. We demonstrate that the phenotype of both populations remained stable in culture and they expressed the expected sarcomeric proteins, gap junction proteins and chamber-specific transcription factors. Compared to MLC-2a cells, MLC-2v selected CMs had larger action potential amplitudes and durations. In addition, by immunofluorescence, we showed that MLC-2 isoform expression can be used to enrich hiPSC-CM consistent with early atrial and ventricular myocyte lineages. However, only the ventricular myosin light chain-2 promoter was able to purify a highly homogeneous population of iPSC-CMs. Using this approach, it is now possible to develop ventricular-specific disease models using iPSC-CMs while atrial-specific iPSC-CM cultures may require additional chamber-specific markers., (© 2013.)
- Published
- 2013
- Full Text
- View/download PDF
28. TGF-β1, released by myofibroblasts, differentially regulates transcription and function of sodium and potassium channels in adult rat ventricular myocytes.
- Author
-
Kaur K, Zarzoso M, Ponce-Balbuena D, Guerrero-Serna G, Hou L, Musa H, and Jalife J
- Subjects
- Animals, Cells, Cultured, Culture Media, Conditioned pharmacology, Enzyme-Linked Immunosorbent Assay, Male, Potassium Channels genetics, Rats, Sodium Channels genetics, Myocytes, Cardiac metabolism, Myofibroblasts metabolism, Potassium Channels metabolism, Sodium Channels metabolism, Transforming Growth Factor beta1 metabolism, Transforming Growth Factor beta1 pharmacology
- Abstract
Cardiac injury promotes fibroblasts activation and differentiation into myofibroblasts, which are hypersecretory of multiple cytokines. It is unknown whether any of such cytokines are involved in the electrophysiological remodeling of adult cardiomyocytes. We cultured adult cardiomyocytes for 3 days in cardiac fibroblast conditioned medium (FCM) from adult rats. In whole-cell voltage-clamp experiments, FCM-treated myocytes had 41% more peak inward sodium current (I(Na)) density at -40 mV than myocytes in control medium (p<0.01). In contrast, peak transient outward current (I(to)) was decreased by ∼55% at 60 mV (p<0.001). Protein analysis of FCM demonstrated that the concentration of TGF-β1 was >3 fold greater in FCM than control, which suggested that FCM effects could be mediated by TGF-β1. This was confirmed by pre-treatment with TGF-β1 neutralizing antibody, which abolished the FCM-induced changes in both I(Na) and I(to). In current-clamp experiments TGF-β1 (10 ng/ml) prolonged the action potential duration at 30, 50, and 90 repolarization (p<0.05); at 50 ng/ml it gave rise to early afterdepolarizations. In voltage-clamp experiments, TGF-β1 increased I(Na) density in a dose-dependent manner without affecting voltage dependence of activation or inactivation. I(Na) density was -36.25±2.8 pA/pF in control, -59.17±6.2 pA/pF at 0.1 ng/ml (p<0.01), and -58.22±6.6 pA/pF at 1 ng/ml (p<0.01). In sharp contrast, I(to) density decreased from 22.2±1.2 pA/pF to 12.7±0.98 pA/pF (p<0.001) at 10 ng/ml. At 1 ng/ml TGF-β1 significantly increased SCN5A (Na(V)1.5) (+73%; p<0.01), while reducing KCNIP2 (Kchip2; -77%; p<0.01) and KCND2 (K(V)4.2; -50% p<0.05) mRNA levels. Further, the TGF-β1-induced increase in I(Na) was mediated through activation of the PI3K-AKT pathway via phosphorylation of FOXO1 (a negative regulator of SCN5A). TGF-β1 released by myofibroblasts differentially regulates transcription and function of the main cardiac sodium channel and of the channel responsible for the transient outward current. The results provide new mechanistic insight into the electrical remodeling associated with myocardial injury.
- Published
- 2013
- Full Text
- View/download PDF
29. Molecular mechanisms of chloroquine inhibition of heterologously expressed Kir6.2/SUR2A channels.
- Author
-
Ponce-Balbuena D, Rodríguez-Menchaca AA, López-Izquierdo A, Ferrer T, Kurata HT, Nichols CG, and Sánchez-Chapula JA
- Subjects
- Animals, Binding Sites, HEK293 Cells, Humans, Mice, Mutation, Patch-Clamp Techniques, Phosphatidylinositol 4,5-Diphosphate pharmacology, Potassium Channels, Inwardly Rectifying genetics, Spermine pharmacology, Sulfonylurea Receptors, Transfection, ATP-Binding Cassette Transporters antagonists & inhibitors, Antimalarials pharmacology, Chloroquine pharmacology, Potassium Channels, Inwardly Rectifying antagonists & inhibitors, Receptors, Drug antagonists & inhibitors
- Abstract
Chloroquine and related compounds can inhibit inwardly rectifying potassium channels by multiple potential mechanisms, including pore block and allosteric effects on channel gating. Motivated by reports that chloroquine inhibition of cardiac ATP-sensitive inward rectifier K(+) current (I(KATP)) is antifibrillatory in rabbit ventricle, we investigated the mechanism of chloroquine inhibition of ATP-sensitive potassium (K(ATP)) channels (Kir6.2/SUR2A) expressed in human embryonic kidney 293 cells, using inside-out patch-clamp recordings. We found that chloroquine inhibits the Kir6.2/SUR2A channel by interacting with at least two different sites and by two mechanisms of action. A fast-onset effect is observed at depolarized membrane voltages and enhanced by the N160D mutation in the central cavity, probably reflecting direct channel block resulting from the drug entering the channel pore from the cytoplasmic side. Conversely, a slow-onset, voltage-independent inhibition of I(KATP) is regulated by chloroquine interaction with a different site and probably involves disruption of interactions between Kir6.2/SUR2A and phosphatidylinositol 4,5-bisphosphate. Our findings reveal multiple mechanisms of K(ATP) channel inhibition by chloroquine, highlighting the numerous convergent regulatory mechanisms of these ligand-dependent ion channels.
- Published
- 2012
- Full Text
- View/download PDF
30. Mechanisms for Kir channel inhibition by quinacrine: acute pore block of Kir2.x channels and interference in PIP2 interaction with Kir2.x and Kir6.2 channels.
- Author
-
López-Izquierdo A, Aréchiga-Figueroa IA, Moreno-Galindo EG, Ponce-Balbuena D, Rodríguez-Martínez M, Ferrer-Villada T, Rodríguez-Menchaca AA, van der Heyden MA, and Sánchez-Chapula JA
- Subjects
- HEK293 Cells, Humans, Quinacrine pharmacology, Myocytes, Cardiac physiology, Phosphatidylinositol 4,5-Diphosphate physiology, Potassium Channels, Inwardly Rectifying drug effects, Potassium Channels, Inwardly Rectifying physiology
- Abstract
Cardiac inward rectifier potassium currents determine the resting membrane potential and contribute repolarization capacity during phase 3 repolarization. Quinacrine is a cationic amphiphilic drug. In this work, the effects of quinacrine were studied on cardiac Kir channels expressed in HEK 293 cells and on the inward rectifier potassium currents, I(K1) and I(KATP), in cardiac myocytes. We found that quinacrine differentially inhibited Kir channels, Kir6.2 ∼ Kir2.3 > Kir2.1. In addition, we found in cardiac myocytes that quinacrine inhibited I(KATP) > I(K1). We presented evidence that quinacrine displays a double action towards strong inward rectifier Kir2.x channels, i.e., direct pore block and interference in phosphatidylinositol 4,5-bisphosphate, PIP(2)-Kir channel interaction. Pore block is evident in Kir2.1 and 2.3 channels as rapid block; channel block involves residues E224 and E299 facing the cytoplasmic pore of Kir2.1. The interference of the drug with the interaction of Kir2.x and Kir6.2/SUR2A channels and PIP(2) is suggested from four sources of evidence: (1) Slow onset of current block when quinacrine is applied from either the inside or the outside of the channel. (2) Mutation of Kir2.3(I213L) and mutation of Kir6.2(C166S) increase their affinity for PIP(2) and lowers its sensitivity for quinacrine. (3) Mutations of Kir2.1(L222I and K182Q) which decreased its affinity for PIP(2) increased its sensitivity for quinacrine. (4) Co-application of quinacrine with PIP(2) lowers quinacrine-mediated current inhibition. In conclusion, our data demonstrate how an old drug provides insight into a dual a blocking mechanism of Kir carried inward rectifier channels.
- Published
- 2011
- Full Text
- View/download PDF
31. Carvedilol inhibits Kir2.3 channels by interference with PIP₂-channel interaction.
- Author
-
Ferrer T, Ponce-Balbuena D, López-Izquierdo A, Aréchiga-Figueroa IA, de Boer TP, van der Heyden MA, and Sánchez-Chapula JA
- Subjects
- Carvedilol, HEK293 Cells, Humans, Point Mutation, Potassium Channels, Inwardly Rectifying genetics, Protein Binding drug effects, Carbazoles pharmacology, Phosphatidylinositol 4,5-Diphosphate metabolism, Potassium Channel Blockers pharmacology, Potassium Channels, Inwardly Rectifying antagonists & inhibitors, Potassium Channels, Inwardly Rectifying metabolism, Propanolamines pharmacology
- Abstract
Carvedilol, a β- and α-adrenoceptor blocker, is used to treat congestive heart failure, mild to moderate hypertension, and myocardial infarction. It has been proposed to block K(ATP) channels by binding to the bundle crossing region at a domain including cysteine at position 166, and thereby plugging the pore region. However, carvedilol was reported not to affect Kir2.1 channels, which lack 166 Cys. Here, we demonstrate that carvedilol inhibits Kir2.3 carried current by an alternative mechanism. Carvedilol inhibited Kir2.3 channels with at least 100 fold higher potency (IC(50)=0.49 μM) compared to that for Kir2.1 (IC(50)>50 μM). Kir2.3 channel inhibition was concentration-dependent and voltage-independent. Increasing Kir2.3 channel affinity for PIP(2), by a I213L point mutation, decreased the inhibitory effect of carvedilol more than twentyfold (IC(50)=11.1 μM). In the presence of exogenous PIP(2), Kir2.3 channel inhibition by carvedilol was strongly reduced (80 vs. 2% current inhibition). These results suggest that carvedilol, as other cationic amphiphilic drugs, inhibits Kir2.3 channels by interfering with the PIP(2)-channel interaction., (Copyright © 2011 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
32. The antimalarial drug mefloquine inhibits cardiac inward rectifier K+ channels: evidence for interference in PIP2-channel interaction.
- Author
-
López-Izquierdo A, Ponce-Balbuena D, Moreno-Galindo EG, Aréchiga-Figueroa IA, Rodríguez-Martínez M, Ferrer T, Rodríguez-Menchaca AA, and Sánchez-Chapula JA
- Subjects
- Animals, Antimalarials administration & dosage, Cats, HEK293 Cells, Humans, Inhibitory Concentration 50, KATP Channels antagonists & inhibitors, Mefloquine administration & dosage, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Patch-Clamp Techniques, Potassium Channel Blockers administration & dosage, Potassium Channel Blockers pharmacology, Antimalarials pharmacology, Mefloquine pharmacology, Phosphatidylinositol 4,5-Diphosphate metabolism, Potassium Channels, Inwardly Rectifying antagonists & inhibitors
- Abstract
The antimalarial drug mefloquine was found to inhibit the KATP channel by an unknown mechanism. Because mefloquine is a Cationic amphiphilic drug and is known to insert into lipid bilayers, we postulate that mefloquine interferes with the interaction between PIP2 and Kir channels resulting in channel inhibition. We studied the inhibitory effects of mefloquine on Kir2.1, Kir2.3, Kir2.3(I213L), and Kir6.2/SUR2A channels expressed in HEK-293 cells, and on IK1 and IKATP from feline cardiac myocytes. The order of mefloquine inhibition was Kir6.2/SUR2A ≈ Kir2.3 (IC50 ≈ 2 μM) > Kir2.1 (IC50 > 30 μM). Similar results were obtained in cardiac myocytes. The Kir2.3(I213L) mutant, which enhances the strength of interaction with PIP2 (compared to WT), was significantly less sensitive (IC50 = 9 μM). In inside-out patches, continuous application of PIP2 strikingly prevented the mefloquine inhibition. Our results support the idea that mefloquine interferes with PIP2-Kir channels interactions.
- Published
- 2011
- Full Text
- View/download PDF
33. Specific residues of the cytoplasmic domains of cardiac inward rectifier potassium channels are effective antifibrillatory targets.
- Author
-
Noujaim SF, Stuckey JA, Ponce-Balbuena D, Ferrer-Villada T, López-Izquierdo A, Pandit S, Calvo CJ, Grzeda KR, Berenfeld O, Chapula JA, and Jalife J
- Subjects
- Animals, Chloroquine chemistry, Cytoplasm drug effects, Mice, Models, Molecular, Potassium Channels, Inwardly Rectifying chemistry, Potassium Channels, Inwardly Rectifying drug effects, Rabbits, Receptors, KIR antagonists & inhibitors, Receptors, KIR metabolism, Sheep, Tachycardia, Ventricular drug therapy, Tachycardia, Ventricular pathology, Ventricular Fibrillation drug therapy, Ventricular Fibrillation pathology, Anti-Arrhythmia Agents pharmacology, Chloroquine pharmacology, Heart drug effects, Potassium Channels, Inwardly Rectifying antagonists & inhibitors
- Abstract
Atrial and ventricular tachyarrhythmias can be perpetuated by up-regulation of inward rectifier potassium channels. Thus, it may be beneficial to block inward rectifier channels under conditions in which their function becomes arrhythmogenic (e.g., inherited gain-of-function mutation channelopathies, ischemia, and chronic and vagally mediated atrial fibrillation). We hypothesize that the antimalarial quinoline chloroquine exerts potent antiarrhythmic effects by interacting with the cytoplasmic domains of Kir2.1 (I(K1)), Kir3.1 (I(KACh)), or Kir6.2 (I(KATP)) and reducing inward rectifier potassium currents. In isolated hearts of three different mammalian species, intracoronary chloroquine perfusion reduced fibrillatory frequency (atrial or ventricular), and effectively terminated the arrhythmia with resumption of sinus rhythm. In patch-clamp experiments chloroquine blocked I(K1), I(KACh), and I(KATP). Comparative molecular modeling and ligand docking of chloroquine in the intracellular domains of Kir2.1, Kir3.1, and Kir6.2 suggested that chloroquine blocks or reduces potassium flow by interacting with negatively charged amino acids facing the ion permeation vestibule of the channel in question. These results open a novel path toward discovering antiarrhythmic pharmacophores that target specific residues of the cytoplasmic domain of inward rectifier potassium channels.
- Published
- 2010
- Full Text
- View/download PDF
34. Thiopental inhibits function of different inward rectifying potassium channel isoforms by a similar mechanism.
- Author
-
López-Izquierdo A, Ponce-Balbuena D, Ferrer T, Rodríguez-Menchaca AA, and Sánchez-Chapula JA
- Subjects
- Cell Line, Transformed, Dose-Response Relationship, Drug, Drug Interactions, Humans, Patch-Clamp Techniques, Phosphatidylinositol 4,5-Diphosphate pharmacology, Point Mutation, Potassium Channels, Inwardly Rectifying genetics, Protein Isoforms, Thiopental antagonists & inhibitors, Membrane Potentials drug effects, Potassium Channels, Inwardly Rectifying antagonists & inhibitors, Thiopental pharmacology
- Abstract
Thiopental is a well-known intravenous barbiturate anesthetic with important cardiac side effects. The actions of thiopental on the transmembrane ionic currents that determine the resting potential and action potential duration in cardiomyocytes have been studied widely. We aimed at elucidating the characteristics and mechanism of inhibition by thiopental on members of the subfamily of inward rectifying Kir2.x (Kir2.1, 2.2 and 2.3), Kir1.1 and Kir6.2/SUR2A channels. These inward rectifier potassium channels were transfected in HEK-293 cells and macroscopic currents were recorded in the whole-cell and inside-out configurations of the patch-clamp technique. Thiopental inhibited Kir2.1, Kir2.2, Kir2.3, Kir1.1 and Kir6.2/SUR2A currents with similar potency; in whole-cell experiments 30 microM thiopental decreased Kir2.1, Kir2.2, Kir2.3 and Kir1.1 currents to 55+/-6, 39+/-8, 42+/-5 and 49+/-5% at -120 mV, respectively. Point mutations on Kir2.3 (I213L) or Kir2.1 (L222I) did not modify the potency of block. Thiopental inhibited all Kir channels in a concentration-dependent and voltage-independent manner. Also, the time course of thiopental inhibition was slow (T(1/2) approximately 4 min) and independent of external or internal drug application. However, in the presence of PIP(2), inhibition by thiopental on Kir2.1 was significantly decreased. Thiopental at clinically relevant concentrations significantly inhibited all Kir channels evaluated in this work. The reduction of thiopental effects during PIP(2) treatment suggests that thiopental inhibition on Kir2.1 channels is related to channel-PIP(2) interaction., ((c) 2010 Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
35. Tamoxifen inhibits cardiac ATP-sensitive and acetylcholine-activated K+ currents in part by interfering with phosphatidylinositol 4,5-bisphosphate-channel interaction.
- Author
-
Ponce-Balbuena D, Moreno-Galindo EG, López-Izquierdo A, Ferrer T, and Sánchez-Chapula JA
- Subjects
- Animals, Cats, Cell Line, Dose-Response Relationship, Drug, Drug Interactions, G Protein-Coupled Inwardly-Rectifying Potassium Channels genetics, Humans, KATP Channels genetics, Membrane Potentials genetics, Mutagenesis, Site-Directed methods, Myocytes, Cardiac drug effects, Patch-Clamp Techniques, Phosphatidylinositol 4,5-Diphosphate, Tamoxifen antagonists & inhibitors, G Protein-Coupled Inwardly-Rectifying Potassium Channels drug effects, KATP Channels drug effects, Membrane Potentials drug effects, Phosphatidylinositol Phosphates pharmacology, Tamoxifen pharmacology
- Abstract
Tamoxifen inhibits transmembrane currents of the Kir2.x inward rectifier potassium channels by interfering with the interaction of the channels with membrane phosphatidylinositol 4,5-bisphosphate (PIP(2)). We tested the hypothesis that Kir channels with low affinity for PIP(2), like the adenosine triphosphate (ATP)-sensitive K(+) channel (K(ATP)) and acetylcholine (ACh)-activated K(+) channel (K(ACh)), have at least the same sensitivity to tamoxifen as Kir2.3. We investigated the effects of tamoxifen (0.1 - 10 microM) on Kir6.2/SUR2A (K(ATP)) and Kir3.1/3.4 (K(ACh)) channels expressed in HEK-293 cells and ATP-sensitive K(+) current (I(KATP)) and ACh-activated K(+) current (I(KACh)) in feline atrial myocytes. The onset of tamoxifen inhibition of both I(KATP) and I(KACh) was slow (T(1/2) approximately 3.5 min) and concentration-dependent but voltage-independent. The time course and degree of inhibition was independent of external or internal drug application. Tamoxifen interacts with the pore forming subunit, Kir6.2, rather than with the SUR subunit. The inhibitory potency of tamoxifen on the Kir6.2/SUR2A channel was decreased by the mutation (C166S) on Kir6.2 and in the continuous presence of PIP(2). In atrial myocytes, the mechanism and potency of the effects of tamoxifen on K(ATP) and K(ACh) channels were comparable to those in HEK-293 cells. These data suggest that, similar to its effects on Kir2.x currents, tamoxifen inhibits K(ATP) and K(ACh) currents by interfering with the interaction between the channel and PIP(2).
- Published
- 2010
- Full Text
- View/download PDF
36. Tamoxifen inhibits inward rectifier K+ 2.x family of inward rectifier channels by interfering with phosphatidylinositol 4,5-bisphosphate-channel interactions.
- Author
-
Ponce-Balbuena D, López-Izquierdo A, Ferrer T, Rodríguez-Menchaca AA, Aréchiga-Figueroa IA, and Sánchez-Chapula JA
- Subjects
- Animals, Cats, Cell Line, Chloroquine pharmacology, Electrophysiology, Heart Atria metabolism, Heart Ventricles metabolism, Humans, Ion Channel Gating drug effects, Ion Channels drug effects, Kinetics, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Patch-Clamp Techniques, Potassium Channels, Inwardly Rectifying genetics, Raloxifene Hydrochloride pharmacology, Transfection, Estrogen Antagonists pharmacology, Ion Channels metabolism, Phosphatidylinositol 4,5-Diphosphate metabolism, Potassium Channel Blockers pharmacology, Potassium Channels, Inwardly Rectifying antagonists & inhibitors, Tamoxifen pharmacology
- Abstract
Tamoxifen, an estrogen receptor antagonist used in the treatment of breast cancer, inhibits the inward rectifier potassium current (I(K1)) in cardiac myocytes by an unknown mechanism. We characterized the inhibitory effects of tamoxifen on Kir2.1, Kir2.2, and Kir2.3 potassium channels that underlie cardiac I(K1). We also studied the effects of 4-hydroxytamoxifen and raloxifene. All three drugs inhibited inward rectifier K(+) 2.x (Kir2.x) family members. The order of inhibition for all three drugs was Kir2.3 > Kir2.1 approximately Kir2.2. The onset of inhibition of Kir2.x current by these compounds was slow (T(1/2) approximately 6 min) and only partially recovered after washout ( approximately 30%). Kir2.x inhibition was concentration-dependent but voltage-independent. The time course and degree of inhibition was independent of external or internal drug application. We tested the hypothesis that tamoxifen interferes with the interaction between the channel and the membrane-delimited channel activator, phosphatidylinositol 4,5-bisphosphate (PIP(2)). Inhibition of Kir2.3 currents was significantly reduced by a single point mutation of I213L, which enhances Kir2.3 interaction with membrane PIP(2). Pretreatment with PIP(2) significantly decreased the inhibition induced by tamoxifen, 4-hydroxytamoxifen, and raloxifene on Kir2.3 channels. Pretreatment with spermine (100 microM) decreased the inhibitory effect of tamoxifen on Kir2.1, probably by strengthening the channel's interaction with PIP(2). In cat atrial and ventricular myocytes, 3 microM tamoxifen inhibited I(K1), but the effect was greater in the former than the latter. The data strongly suggest that tamoxifen, its metabolite, and the estrogen receptor inhibitor raloxifene inhibit Kir2.x channels indirectly by interfering with the interaction between the channel and PIP(2).
- Published
- 2009
- Full Text
- View/download PDF
37. Chloroquine blocks a mutant Kir2.1 channel responsible for short QT syndrome and normalizes repolarization properties in silico.
- Author
-
Lopez-Izquierdo A, Ponce-Balbuena D, Ferrer T, Sachse FB, Tristani-Firouzi M, and Sanchez-Chapula JA
- Subjects
- Action Potentials drug effects, Action Potentials genetics, Cell Line, Computer Simulation, Dose-Response Relationship, Drug, Electrophysiology, Heart Ventricles cytology, Humans, Kidney cytology, Membrane Potentials drug effects, Membrane Potentials genetics, Membrane Potentials physiology, Models, Statistical, Mutation drug effects, Myocytes, Cardiac drug effects, Patch-Clamp Techniques, Potassium Channels, Inwardly Rectifying physiology, Refractory Period, Electrophysiological drug effects, Antimalarials pharmacology, Chloroquine pharmacology, Potassium Channel Blockers pharmacology, Potassium Channels, Inwardly Rectifying genetics
- Abstract
Short QT Syndrome (SQTS) is a novel clinical entity characterized by markedly rapid cardiac repolarization and lethal arrhythmias. A mutation in the Kir2.1 inward rectifier K+ channel (D172N) causes one form of SQTS (SQT3). Pharmacologic block of Kir2.1 channels may hold promise as potential therapy for SQT3. We recently reported that the anti-malarial drug chloroquine blocks Kir2.1 channels by plugging the cytoplasmic pore domain. In this study, we tested whether chloroquine blocks D172N Kir2.1 channels in a heterologous expression system and if chloroquine normalizes repolarization properties using a mathematical model of a human ventricular myocyte. Chloroquine caused a dose- and voltage-dependent reduction in wild-type (WT), D172N and WT-D172N heteromeric Kir2.1 current. The potency and kinetics of chloroquine block of D172N and WT-D172N Kir2.1 current were similar to WT. In silico modeling of the heterozygous WT-D172N Kir2.1 condition predicted that 3 microM chloroquine normalized inward rectifier K+ current magnitude, action potential duration and effective refractory period. Our results suggest that therapeutic concentrations of chloroquine might lengthen cardiac repolarization in SQT3., (Copyright (c) 2009 S. Karger AG, Basel.)
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.