44 results on '"Ingrid M. Bonilla"'
Search Results
2. STIM1 ablation impairs exercise-induced physiological cardiac hypertrophy and dysregulates autophagy in mouse hearts
- Author
-
Ingrid M. Bonilla, Stephen Baine, Anastasia Pokrass, Juan Ignacio Elio Mariángelo, Anuradha Kalyanasundaram, Vladimir Bogdanov, Louisa Mezache, Galina Sakuta, Casey M. Beard, Andriy Belevych, Svetlana Tikunova, Radmila Terentyeva, Dmitry Terentyev, Jonathan Davis, Rengasayee Veeraraghavan, Cynthia A. Carnes, and Sandor Györke
- Subjects
Physiology ,Physiology (medical) - Abstract
Store-operated Ca2+ entry (SOCE) has been implicated in pathological cardiac hypertrophy; however, its role in physiological hypertrophy is unknown. Here we report that SOCE is also essential for physiological cardiac hypertrophy and functional adaptations in response to endurance exercise. These adaptations were associated with activation of AKT/mTOR pathway and curtailed cardiac autophagy and degeneration. Thus, SOCE is a common mechanism and an important bifurcation point for signaling paths involved in physiological and pathological hypertrophy.
- Published
- 2023
- Full Text
- View/download PDF
3. MATE1 Deficiency Exacerbates Dofetilide-Induced Proarrhythmia
- Author
-
Muhammad Erfan Uddin, Eric D. Eisenmann, Yang Li, Kevin M. Huang, Dominique A. Garrison, Zahra Talebi, Alice A. Gibson, Yan Jin, Mahesh Nepal, Ingrid M. Bonilla, Qiang Fu, Xinxin Sun, Alec Millar, Mikhail Tarasov, Christopher E. Jay, Xiaoming Cui, Heidi J. Einolf, Ryan M. Pelis, Sakima A. Smith, Przemysław B. Radwański, Douglas H. Sweet, Jörg König, Martin F. Fromm, Cynthia A. Carnes, Shuiying Hu, and Alex Sparreboom
- Subjects
Sulfonamides ,dofetilide ,organic cation transporters ,arrhythmia ,PBPK modeling ,Organic Chemistry ,General Medicine ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,Mice ,Atrial Fibrillation ,Phenethylamines ,Animals ,Humans ,ddc:610 ,Physical and Theoretical Chemistry ,Anti-Arrhythmia Agents ,Molecular Biology ,Spectroscopy - Abstract
Dofetilide is a rapid delayed rectifier potassium current inhibitor widely used to prevent the recurrence of atrial fibrillation and flutter. The clinical use of this drug is associated with increases in QTc interval, which predispose patients to ventricular cardiac arrhythmias. The mechanisms involved in the disposition of dofetilide, including its movement in and out of cardiomyocytes, remain unknown. Using a xenobiotic transporter screen, we identified MATE1 (SLC47A1) as a transporter of dofetilide and found that genetic knockout or pharmacological inhibition of MATE1 in mice was associated with enhanced retention of dofetilide in cardiomyocytes and increased QTc prolongation. The urinary excretion of dofetilide was also dependent on the MATE1 genotype, and we found that this transport mechanism provides a mechanistic basis for previously recorded drug-drug interactions of dofetilide with various contraindicated drugs, including bictegravir, cimetidine, ketoconazole, and verapamil. The translational significance of these observations was examined with a physiologically-based pharmacokinetic model that adequately predicted the drug-drug interaction liabilities in humans. These findings support the thesis that MATE1 serves a conserved cardioprotective role by restricting excessive cellular accumulation and warrant caution against the concurrent administration of potent MATE1 inhibitors and cardiotoxic substrates with a narrow therapeutic window.
- Published
- 2022
- Full Text
- View/download PDF
4. Pyridostigmine improves cardiac function and rhythmicity through RyR2 stabilization and inhibition of STIM1‐mediated calcium entry in heart failure
- Author
-
Andriy E. Belevych, Stephen Baine, Andrei V. Stepanov, Sandor Gyorke, Lisa E. Dorn, Ingrid M. Bonilla, Cynthia A. Carnes, Radmila Terentyeva, Dmitry Terentyev, and Federica Accornero
- Subjects
0301 basic medicine ,Cardiac function curve ,Male ,medicine.medical_specialty ,STIM1 ,Ryanodine receptor 2 ,Muscle hypertrophy ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,medicine ,Myocyte ,echocardiography ,Animals ,Stromal Interaction Molecule 1 ,Pyridostigmine ,Pressure overload ,Heart Failure ,Ejection fraction ,calcium ,Ryanodine receptor ,Chemistry ,phosphorylation ,RyR2 ,Arrhythmias, Cardiac ,Ryanodine Receptor Calcium Release Channel ,Cell Biology ,Original Articles ,medicine.disease ,excitation contraction coupling ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,autonomics ,030220 oncology & carcinogenesis ,Heart failure ,Molecular Medicine ,Original Article ,Cholinesterase Inhibitors ,hypertrophy ,Pyridostigmine Bromide - Abstract
Heart failure (HF) is characterized by asymmetrical autonomic balance. Treatments to restore parasympathetic activity in human heart failure trials have shown beneficial effects. However, mechanisms of parasympathetic‐mediated improvement in cardiac function remain unclear. The present study examined the effects and underpinning mechanisms of chronic treatment with the cholinesterase inhibitor, pyridostigmine (PYR), in pressure overload HF induced by transverse aortic constriction (TAC) in mice. TAC mice exhibited characteristic adverse structural (left ventricular hypertrophy) and functional remodelling (reduced ejection fraction, altered myocyte calcium (Ca) handling, increased arrhythmogenesis) with enhanced predisposition to arrhythmogenic aberrant sarcoplasmic reticulum (SR) Ca release, cardiac ryanodine receptor (RyR2) hyper‐phosphorylation and up‐regulated store‐operated Ca entry (SOCE). PYR treatment resulted in improved cardiac contractile performance and rhythmic activity relative to untreated TAC mice. Chronic PYR treatment inhibited altered intracellular Ca handling by alleviating aberrant Ca release and diminishing pathologically enhanced SOCE in TAC myocytes. At the molecular level, these PYR‐induced changes in Ca handling were associated with reductions of pathologically enhanced phosphorylation of RyR2 serine‐2814 and STIM1 expression in HF myocytes. These results suggest that chronic cholinergic augmentation alleviates HF via normalization of both canonical RyR2‐mediated SR Ca release and non‐canonical hypertrophic Ca signaling via STIM1‐dependent SOCE.
- Published
- 2021
5. Molecular Mechanism and Current Therapies for Catecholaminergic Polymorphic Ventricular Tachycardia
- Author
-
Brian D. Tow, Bin Liu, and Ingrid M. Bonilla
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Chemistry ,030204 cardiovascular system & hematology ,Catecholaminergic polymorphic ventricular tachycardia ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Internal medicine ,cardiovascular system ,Molecular mechanism ,medicine ,Cardiology ,Current (fluid) - Abstract
The rhythmic contraction of the heart relies on tightly regulated calcium (Ca) release from the sarcoplasmic reticulum (SR) Ca release channel, Ryanodine receptor (RyR2). Genetic mutations in components of the calcium release unit such as RyR2, cardiac calsequestrin and other proteins have been shown to cause a genetic arrhythmic syndrome known as catecholaminergic polymorphic ventricular tachycardia (CPVT). This book chapter will focus on the following: (1) to describing CPVT as a stress-induced cardiac arrhythmia syndrome and its genetic causes. (2) Discussing the regulation of SR Ca release, and how dysregulation of Ca release contributes to arrhythmogenesis. (3) Discussing molecular mechanisms of CPVT with a focus on impaired Ca signaling refractoriness as a unifying mechanism underlying different genetic forms of CPVT. (4) Discussing pharmacological approaches as CPVT treatments as well as other potential future therapies. Since dysregulated SR Ca release has been implicated in multiple cardiac disorders including heart failure and metabolic heart diseases, knowledge obtained from CPVT studies will also shed light on the development of therapeutic approaches for these devastating cardiac dysfunctions as a whole.
- Published
- 2022
- Full Text
- View/download PDF
6. Muscarinic-dependent phosphorylation of the cardiac ryanodine receptor by protein kinase G is mediated by PI3K–AKT–nNOS signaling
- Author
-
Sandor Gyorke, Stephen Baine, Rengasayee Veeraraghavan, Ingrid M. Bonilla, Przemysław B. Radwański, Marina S. Ivanova, Justin Thomas, Cynthia A. Carnes, Jiaoni Li, and Andriy E. Belevych
- Subjects
Nitric Oxide Synthase Type I ,Biochemistry ,Ryanodine receptor 2 ,Phosphatidylinositol 3-Kinases ,Cyclic GMP-Dependent Protein Kinases ,Animals ,Myocytes, Cardiac ,Phosphorylation ,Molecular Biology ,Protein kinase B ,Cells, Cultured ,biology ,Chemistry ,Kinase ,Ryanodine receptor ,Ryanodine Receptor Calcium Release Channel ,Cell Biology ,Cell biology ,Mice, Inbred C57BL ,Nitric oxide synthase ,cardiovascular system ,biology.protein ,Signal transduction ,Proto-Oncogene Proteins c-akt ,cGMP-dependent protein kinase ,Signal Transduction - Abstract
Post-translational modifications of proteins involved in calcium handling in myocytes, such as the cardiac ryanodine receptor (RyR2), critically regulate cardiac contractility. Recent studies have suggested that phosphorylation of RyR2 by protein kinase G (PKG) might contribute to the cardioprotective effects of cholinergic stimulation. However, the specific mechanisms underlying these effects remain unclear. Here, using murine ventricular myocytes, immunoblotting, proximity ligation as-says, and nitric oxide imaging, we report that phosphorylation of Ser-2808 in RyR2 induced by the muscarinic receptor agonist carbachol is mediated by a signaling axis comprising phosphoinositide 3-phosphate kinase, Akt Ser/Thr kinase, nitric oxide synthase 1, nitric oxide, soluble guanylate cyclase, cyclic GMP (cGMP), and PKG. We found that this signaling pathway is compartmentalized in myocytes, as it was distinct from atrial natriuretic peptide receptor–cGMP–PKG–RyR2 Ser-2808 signaling and independent of muscarinic-induced phosphorylation of Ser-239 in vasodilator-stimulated phosphoprotein. These results provide detailed insights into muscarinic-induced PKG signaling and the mediators that regulate cardiac RyR2 phosphorylation critical for cardiovascular function.
- Published
- 2020
- Full Text
- View/download PDF
7. Selective ablation of the long splice variant of STIM1 inhibits SOCE, prevents cardiac hypertrophy, and causes severe cardiac impairment and premature death in exercised mice
- Author
-
Benjamin Hernandez Orengo, Ingrid M. Bonilla, Galina Sakuta, Dennison Min, Stephen Baine, Andriy E. Belevych, Cynthia A. Carnes, Przemyslaw Radwanski, and Sandor Gyorke
- Subjects
Biophysics - Published
- 2022
- Full Text
- View/download PDF
8. Calcium-activated potassium current modulates ventricular repolarization in chronic heart failure.
- Author
-
Ingrid M Bonilla, Victor P Long, Pedro Vargas-Pinto, Patrick Wright, Andriy Belevych, Qing Lou, Kent Mowrey, Jae Yoo, Philip F Binkley, Vadim V Fedorov, Sandor Györke, Paulus M L Janssen, Ahmet Kilic, Peter J Mohler, and Cynthia A Carnes
- Subjects
Medicine ,Science - Abstract
The role of I(KCa) in cardiac repolarization remains controversial and varies across species. The relevance of the current as a therapeutic target is therefore undefined. We examined the cellular electrophysiologic effects of I(KCa) blockade in controls, chronic heart failure (HF) and HF with sustained atrial fibrillation. We used perforated patch action potential recordings to maintain intrinsic calcium cycling. The I(KCa) blocker (apamin 100 nM) was used to examine the role of the current in atrial and ventricular myocytes. A canine tachypacing induced model of HF (1 and 4 months, n = 5 per group) was used, and compared to a group of 4 month HF with 6 weeks of superimposed atrial fibrillation (n = 7). A group of age-matched canine controls were used (n = 8). Human atrial and ventricular myocytes were isolated from explanted end-stage failing hearts which were obtained from transplant recipients, and studied in parallel. Atrial myocyte action potentials were unchanged by I(KCa) blockade in all of the groups studied. I(KCa) blockade did not affect ventricular myocyte repolarization in controls. HF caused prolongation of ventricular myocyte action potential repolarization. I(KCa) blockade caused further prolongation of ventricular repolarization in HF and also caused repolarization instability and early afterdepolarizations. SK2 and SK3 expression in the atria and SK3 in the ventricle were increased in canine heart failure. We conclude that during HF, I(KCa) blockade in ventricular myocytes results in cellular arrhythmias. Furthermore, our data suggest an important role for I(KCa) in the maintenance of ventricular repolarization stability during chronic heart failure. Our findings suggest that novel antiarrhythmic therapies should have safety and efficacy evaluated in both atria and ventricles.
- Published
- 2014
- Full Text
- View/download PDF
9. Targeting OCT3 attenuates doxorubicin-induced cardiac injury
- Author
-
Megan E. Zavorka Thomas, Eric D. Eisenmann, Muhammad Erfan Uddin, Sherry H. Xia, Vincenzo Coppola, Markus Keiser, Yan Jin, Pearlly S. Yan, Paul W. Burridge, Sharyn D. Baker, Duncan DiGiacomo, Kevin M. Huang, Ralf Bundschuh, Tarek Magdy, Sakima A. Smith, Anne T. Nies, Taosheng Chen, Alexander Pan, Kara N. Corps, Daniel Addison, Moray J. Campbell, Qiang Fu, Alice A. Gibson, Alex Sparreboom, Stefan Oswald, Joanne Wang, Cynthia A. Carnes, Shuiying Hu, Maryam B. Lustberg, Yang Li, Marcus Otter, and Ingrid M. Bonilla
- Subjects
Side effect ,Organic Anion Transporters, Sodium-Independent ,SLC22A3 ,Mice ,Neoplasms ,Medicine ,Animals ,Humans ,Doxorubicin ,Myocytes, Cardiac ,Molecular Targeted Therapy ,Child ,Cardiotoxicity ,Multidisciplinary ,Organic cation transport proteins ,biology ,business.industry ,Sequence Analysis, RNA ,Transporter ,Biological Sciences ,medicine.disease ,Leukemia ,Pyrimidines ,Nilotinib ,Gene Expression Regulation ,Heart Injuries ,Cancer research ,biology.protein ,business ,medicine.drug - Abstract
Doxorubicin is a commonly used anticancer agent that can cause debilitating and irreversible cardiac injury. The initiating mechanisms contributing to this side effect remain unknown, and current preventative strategies offer only modest protection. Using stem-cell–derived cardiomyocytes from patients receiving doxorubicin, we probed the transcriptomic landscape of solute carriers and identified organic cation transporter 3 (OCT3) (SLC22A3) as a critical transporter regulating the cardiac accumulation of doxorubicin. Functional validation studies in heterologous overexpression models confirmed that doxorubicin is transported into cardiomyocytes by OCT3 and that deficiency of OCT3 protected mice from acute and chronic doxorubicin-related changes in cardiovascular function and genetic pathways associated with cardiac damage. To provide proof-of-principle and demonstrate translational relevance of this transport mechanism, we identified several pharmacological inhibitors of OCT3, including nilotinib, and found that pharmacological targeting of OCT3 can also preserve cardiovascular function following treatment with doxorubicin without affecting its plasma levels or antitumor effects in multiple models of leukemia and breast cancer. Finally, we identified a previously unrecognized, OCT3-dependent pathway of doxorubicin-induced cardiotoxicity that results in a downstream signaling cascade involving the calcium-binding proteins S100A8 and S100A9. These collective findings not only shed light on the etiology of doxorubicin-induced cardiotoxicity, but also are of potential translational relevance and provide a rationale for the implementation of a targeted intervention strategy to prevent this debilitating side effect.
- Published
- 2021
10. Dietary omega-3 fatty acids promote arrhythmogenic remodeling of cellular Ca2+ handling in a postinfarction model of sudden cardiac death.
- Author
-
Andriy E Belevych, Hsiang-Ting Ho, Radmila Terentyeva, Ingrid M Bonilla, Dmitry Terentyev, Cynthia A Carnes, Sandor Gyorke, and George E Billman
- Subjects
Medicine ,Science - Abstract
It has been proposed that dietary omega-3 polyunsaturated fatty acids (n-3 PUFAs) can reduce the risk of ventricular arrhythmias in post-MI patients. Abnormal Ca(2+) handling has been implicated in the genesis of post-MI ventricular arrhythmias. Therefore, we tested the hypothesis that dietary n-3 PUFAs alter the vulnerability of ventricular myocytes to cellular arrhythmia by stabilizing intracellular Ca(2+) cycling. To test this hypothesis, we used a canine model of post-MI ventricular fibrillation (VF) and assigned the animals to either placebo (1 g/day corn oil) or n-3 PUFAs (1-4 g/day) groups. Using Ca(2+) imaging techniques, we examined the intracellular Ca(2+) handling in myocytes isolated from post-MI hearts resistant (VF-) and susceptible (VF+) to VF. Frequency of occurrence of diastolic Ca(2+) waves (DCWs) in VF+ myocytes from placebo group was significantly higher than in placebo-treated VF- myocytes. n-3 PUFA treatment did not decrease frequency of DCWs in VF+ myocytes. In contrast, VF- myocytes from the n-3 PUFA group had a significantly higher frequency of DCWs than myocytes from the placebo group. In addition, n-3 PUFA treatment increased beat-to-beat alterations in the amplitude of Ca(2+) transients (Ca(2+) alternans) in VF- myocytes. These n-3 PUFAs effects in VF- myocytes were associated with an increased Ca(2+) spark frequency and reduced sarcoplasmic reticulum Ca(2+) content, indicative of increased activity of ryanodine receptors. Thus, dietary n-3 PUFAs do not alleviate intracellular Ca(2+) cycling remodeling in myocytes isolated from post-MI VF+ hearts. Furthermore, dietary n-3 PUFAs increase vulnerability of ventricular myocytes to cellular arrhythmia in post-MI VF- hearts by destabilizing intracellular Ca(2+) handling.
- Published
- 2013
- Full Text
- View/download PDF
11. Abstract 14035: Renal Tubular Secretion and Cardiac Distribution of Dofetilide is Dependent on MATE1 Function
- Author
-
Ingrid M. Bonilla, Shuiying Hu, Cynthia A. Carnes, Alice A. Gibson, Yan Jin, Muhammad Erfan Uddin, and Alex Sparreboom
- Subjects
Cardioprotection ,medicine.medical_specialty ,business.industry ,Long QT syndrome ,Dofetilide ,Atrial fibrillation ,medicine.disease ,QT interval ,Potassium channel ,Renal tubular secretion ,Physiology (medical) ,Internal medicine ,medicine ,Cardiology ,Distribution (pharmacology) ,Cardiology and Cardiovascular Medicine ,business ,medicine.drug - Abstract
Introduction: Dofetilide is a delayed rectifier potassium channel inhibitor used to treat patients with atrial fibrillation and flutter, and its use is associated with a risk of QT prolongation and Torsades de Pointes . The mechanisms involved in dofetilide’s renal tubular secretion and its uptake into cardiomyocytes remain unknown. Previously reported drug-drug interaction (DDI) studies suggest the involvement of organic cation transporters. Here, we investigated the contribution of organic cation transporters (OCT2 and MATE1) to the pharmacokinetics of dofetilide to gain insight into its DDI potential. Hypothesis: Based on known DDIs with dofetilide, we hypothesize that OCT2 and/or MATE1 play a key role in the inter-individual variability in pharmacokinetics and pharmacodynamics of dofetilide. Methods: In vitro and ex vivo transport kinetics of dofetilide were determined in HEK293 cells stably transfected with OCT2 or MATE1, and in isolated cardiomyocytes, respectively. In vivo studies were performed in wild-type, OCT2-, and MATE1-deficient mice (n=5) receiving dofetilide (5 mg/kg, p.o., 2.5 mg/kg, i.v.), with or without several contraindicated drugs. Dofetilide concentrations in plasma and urine were determined by UPLC-MS/MS. Results: In vitro studies demonstrated that dofetilide is a good substrate of MATE1 but not OCT2. Deficiency of MATE1 was associated with increased plasma concentrations of dofetilide and with a significantly reduced urinary excretion (3-fold in females and 5-fold in males, respectively). Dofetilide accumulation in cardiomyocytes was increased by 2-fold in MATE1-deficient females, and pre-incubation with the MATE1 inhibitor cimetidine significantly reduced dofetilide uptake in wild-type cardiomyocytes. Several contraindicated drugs listed in the dofetilide prescribing information, including cimetidine, ketoconazole, increased dofetilide plasma exposure in wild-type mice by >2.8-fold. Conclusion: Renal secretion of dofetilide is mediated by MATE1 and is highly sensitive to inhibition by many widely used prescription drugs that can cause clinically relevant DDIs. Deficiency of MATE1 also increases accumulation in the heart which may contribute to individual variation in response to dofetilide.
- Published
- 2020
- Full Text
- View/download PDF
12. Nitric Oxide synthases and atrial fibrillation
- Author
-
Ingrid M Bonilla, Arun eSridhar, Sandor eGyorke, Arturo J Cardounel, and Cynthia Ann Carnes
- Subjects
Atrial Fibrillation ,Electrophysiology ,Oxidative Stress ,Therapeutics ,nitric oxide synthases ,Physiology ,QP1-981 - Abstract
Oxidative stress has been implicated in the pathogenesis of atrial fibrillation. There are multiple systems in the myocardium which contribute to redox homeostasis, and loss of homeostasis can result in oxidative stress. Potential sources of oxidants include nitric oxide synthases, which normally produce nitric oxide in the heart. Two nitric oxide synthase isoforms (1 and 3) are normally expressed in the heart. During pathologies such as heart failure, there is induction of nitric oxide synthase 2 in multiple cell types in the myocardium. In certain conditions, the NOS enzymes may become uncoupled, shifting from production of nitric oxide to superoxide anion, a potent free radical and oxidant. Multiple lines of evidence suggest a role for nitric oxide synthases in the pathogenesis of atrial fibrillation. Therapeutic approaches to reduce atrial fibrillation by modulation of nitric oxide synthase activity may be beneficial, although further investigation of this strategy is needed.
- Published
- 2012
- Full Text
- View/download PDF
13. Tetrodotoxin-sensitive Navs contribute to early and delayed afterdepolarizations in long QT arrhythmia models
- Author
-
Sandor Gyorke, Bjorn C. Knollmann, Stephen Baine, Justin Thomas, Naveed Zaman, Przemysław B. Radwański, Rengasayee Veeraraghavan, Megan Koleske, and Ingrid M. Bonilla
- Subjects
0301 basic medicine ,Physiology ,Chemistry ,Sodium channel ,Torsades de pointes ,030204 cardiovascular system & hematology ,Pharmacology ,Catecholaminergic polymorphic ventricular tachycardia ,medicine.disease ,Calsequestrin ,QT interval ,3. Good health ,Afterdepolarization ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine ,Repolarization ,Anti-Arrhythmia Agents - Abstract
Recent evidence suggests that neuronal Na+ channels (nNavs) contribute to catecholamine-promoted delayed afterdepolarizations (DADs) and catecholaminergic polymorphic ventricular tachycardia (CPVT). The newly identified overlap between CPVT and long QT (LQT) phenotypes has stoked interest in the cross-talk between aberrant Na+ and Ca2+ handling and its contribution to early afterdepolarizations (EADs) and DADs. Here, we used Ca2+ imaging and electrophysiology to investigate the role of Na+ and Ca2+ handling in DADs and EADs in wild-type and cardiac calsequestrin (CASQ2)-null mice. In experiments, repolarization was impaired using 4-aminopyridine (4AP), whereas the L-type Ca2+ and late Na+ currents were augmented using Bay K 8644 (BayK) and anemone toxin II (ATX-II), respectively. The combination of 4AP and isoproterenol prolonged action potential duration (APD) and promoted aberrant Ca2+ release, EADs, and DADs in wild-type cardiomyocytes. Similarly, BayK in the absence of isoproterenol induced the same effects in CASQ2-null cardiomyocytes. In vivo, it prolonged the QT interval and, upon catecholamine challenge, precipitated wide QRS polymorphic ventricular tachycardia that resembled human torsades de pointes. Treatment with ATX-II produced similar effects at both the cellular level and in vivo. Importantly, nNav inhibition with riluzole or 4,9-anhydro-tetrodotoxin reduced the incidence of ATX-II–, BayK-, or 4AP-induced EADs, DADs, aberrant Ca2+ release, and VT despite only modestly mitigating APD prolongation. These data reveal the contribution of nNaVs to triggered arrhythmias in murine models of LQT and CPVT-LQT overlap phenotypes. We also demonstrate the antiarrhythmic impact of nNaV inhibition, independent of action potential and QT interval duration, and provide a basis for a mechanistically driven antiarrhythmic strategy.
- Published
- 2018
- Full Text
- View/download PDF
14. Hyperactivity of RyR2 in Cardiac Disease is Exacerbated by Calcium Leak-Induced Mitochondrial ROS
- Author
-
Dmitry Terentyev, Bjorn C. Knollmann, Shanna Hamilton, Sandor Gyorke, Andriy E. Belevych, Andrei V. Stepanov, Jiaoni Li, Przemysław B. Radwański, Ingrid M. Bonilla, and Radmila Terentyeva
- Subjects
Mitochondrial ROS ,medicine.medical_specialty ,Leak ,business.industry ,Biophysics ,chemistry.chemical_element ,Disease ,Calcium ,Ryanodine receptor 2 ,Endocrinology ,chemistry ,Internal medicine ,medicine ,business - Published
- 2020
- Full Text
- View/download PDF
15. Increased RyR2 activity is exacerbated by calcium leak-induced mitochondrial ROS
- Author
-
Dmitry Terentyev, Bjorn C. Knollmann, Jiaoni Li, Przemysław B. Radwański, Andrei V. Stepanov, Andriy E. Belevych, Benjamin Martin, Ingrid M. Bonilla, Shanna Hamilton, Radmila Terentyeva, Fruzsina Perger, and Sandor Gyorke
- Subjects
Mitochondrial ROS ,Male ,Heart Diseases ,Physiology ,Mitochondrion ,Catecholaminergic polymorphic ventricular tachycardia ,Ryanodine receptor 2 ,Rats, Sprague-Dawley ,Mice ,Physiology (medical) ,medicine ,Animals ,Myocytes, Cardiac ,Uniporter ,Chemistry ,Ryanodine receptor ,Depolarization ,Ryanodine Receptor Calcium Release Channel ,Original Contribution ,medicine.disease ,Cell biology ,Mitochondria ,Rats ,Mice, Inbred C57BL ,Mitochondrial matrix ,cardiovascular system ,Ventricular arrhythmia ,Calcium ,Female ,Cardiology and Cardiovascular Medicine ,Reactive oxygen species ,Ca2+ leak - Abstract
Cardiac disease is associated with deleterious emission of mitochondrial reactive oxygen species (mito-ROS), as well as enhanced oxidation and activity of the sarcoplasmic reticulum (SR) Ca2+ release channel, the ryanodine receptor (RyR2). The transfer of Ca2+ from the SR via RyR2 to mitochondria is thought to play a key role in matching increased metabolic demand during stress. In this study, we investigated whether augmented RyR2 activity results in self-imposed exacerbation of SR Ca2+ leak, via altered SR-mitochondrial Ca2+ transfer and elevated mito-ROS emission. Fluorescent indicators and spatially restricted genetic ROS probes revealed that both pharmacologically and genetically enhanced RyR2 activity, in ventricular myocytes from rats and catecholaminergic polymorphic ventricular tachycardia (CPVT) mice, respectively, resulted in increased ROS emission under β-adrenergic stimulation. Expression of mitochondrial Ca2+ probe mtRCamp1h revealed diminished net mitochondrial [Ca2+] with enhanced SR Ca2+ leak, accompanied by depolarization of the mitochondrial matrix. While this may serve as a protective mechanism to prevent mitochondrial Ca2+ overload, protection is not complete and enhanced mito-ROS emission resulted in oxidation of RyR2, further amplifying proarrhythmic SR Ca2+ release. Importantly, the effects of augmented RyR2 activity could be attenuated by mitochondrial ROS scavenging, and experiments with dominant-negative paralogs of the mitochondrial Ca2+ uniporter (MCU) supported the hypothesis that SR-mitochondria Ca2+ transfer is essential for the increase in mito-ROS. We conclude that in a process whereby leak begets leak, augmented RyR2 activity modulates mitochondrial Ca2+ handling, promoting mito-ROS emission and driving further channel activity in a proarrhythmic feedback cycle in the diseased heart.
- Published
- 2019
16. Chronic heart failure increases negative chronotropic effects of adenosine in canine sinoatrial cells via A1R stimulation and GIRK-mediated I
- Author
-
Ingrid M. Bonilla, Sanjay Kumar, Stephen Baine, Vadim V. Fedorov, Patric Glynn, Kent Mowrey, Raul Weiss, Nam Y. Lee, Victor P. Long, Sandor Gyorke, Karsten E. Schober, Cynthia A. Carnes, Thomas J. Hund, and Peter J. Mohler
- Subjects
0301 basic medicine ,Chronotropic ,Male ,Adenosine ,Patch-Clamp Techniques ,Action Potentials ,Pharmacology ,Adenosine A1 Receptor Antagonists ,In Vitro Techniques ,030226 pharmacology & pharmacy ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Adenosine A1 receptor ,0302 clinical medicine ,Dogs ,Biological Clocks ,Heart Rate ,medicine ,Potassium Channel Blockers ,Animals ,G protein-coupled inwardly-rectifying potassium channel ,General Pharmacology, Toxicology and Pharmaceutics ,Sinoatrial Node ,Heart Failure ,Chemistry ,Sinoatrial node ,Receptor, Adenosine A1 ,General Medicine ,Membrane hyperpolarization ,Diastolic depolarization ,Hyperpolarization (biology) ,Adenosine A1 Receptor Agonists ,Bee Venoms ,030104 developmental biology ,medicine.anatomical_structure ,G Protein-Coupled Inwardly-Rectifying Potassium Channels ,Xanthines ,Chronic Disease ,Female ,medicine.drug - Abstract
AIMS: Bradycardia contributes to tachy-brady arrhythmias or sinus arrest during heart failure (HF). Sinoatrial node (SAN) adenosine A1 receptors (ADO A1Rs) are upregulated in HF, and adenosine is known to exert negative chronotropic effects on the SAN. Here, we investigated the role of A1R signaling at physiologically relevant ADO concentrations on HF SAN pacemaker cells. MAIN METHODS: Dogs with tachypacing-induced chronic HF and normal controls (CTL) were studied. SAN tissue was collected for A1R and GIRK mRNA quantification. SAN cells were isolated for perforated patch clamp recordings and firing rate (bpm), slope of slow diastolic depolarization (SDD), and maximum diastolic potential (MDP) were measured. Action potentials (APs) and currents were recorded before and after addition of 1 and 10 μM ADO. To assess contributions of A1R and G protein-coupled Inward Rectifier Potassium Current (GIRK) to ADO effects, APs were measured after the addition of DPCPX (selective A1R antagonist) or TPQ (selective GIRK blocker). KEY FINDINGS: A1R and GIRK mRNA expression were significantly increased in HF. In addition, ADO induced greater rate slowing and membrane hyperpolarization in HF vs CTL (p
- Published
- 2019
17. Enhancement of Cardiac Store Operated Calcium Entry (SOCE) within Novel Intercalated Disk Microdomains in Arrhythmic Disease
- Author
-
G. A. Sakuta, Rengasayee Veeraraghavan, Stephen Baine, Sandor Gyorke, Cynthia A. Carnes, Tom Bodnar, Andriy E. Belevych, Silvia G. Priori, Ingrid M. Bonilla, Noah Weisleder, Bin Liu, Louisa Mezache, Andrei V. Stepanov, Przemysław B. Radwański, and Pompeo Volpe
- Subjects
0301 basic medicine ,Male ,ORAI1 Protein ,Calcium and vitamin D ,lcsh:Medicine ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,Myocyte ,Animals ,Myocytes, Cardiac ,Calcium Signaling ,Gene Knock-In Techniques ,Stromal Interaction Molecule 1 ,lcsh:Science ,Excitation Contraction Coupling ,Catecholaminergic ,Multidisciplinary ,Chemistry ,Myocardium ,lcsh:R ,Calcium signalling ,Wild type ,Arrhythmias, Cardiac ,Store-operated calcium entry ,Cell biology ,Coupling (electronics) ,Mice, Inbred C57BL ,Sarcoplasmic Reticulum ,030104 developmental biology ,cardiovascular system ,Cholinergic ,lcsh:Q ,Calcium ,Female ,Calcium Channels ,030217 neurology & neurosurgery ,Intracellular - Abstract
Store-operated Ca2+ entry (SOCE), a major Ca2+ signaling mechanism in non-myocyte cells, has recently emerged as a component of Ca2+ signaling in cardiac myocytes. Though it has been reported to play a role in cardiac arrhythmias and to be upregulated in cardiac disease, little is known about the fundamental properties of cardiac SOCE, its structural underpinnings or effector targets. An even greater question is how SOCE interacts with canonical excitation-contraction coupling (ECC). We undertook a multiscale structural and functional investigation of SOCE in cardiac myocytes from healthy mice (wild type; WT) and from a genetic murine model of arrhythmic disease (catecholaminergic ventricular tachycardia; CPVT). Here we provide the first demonstration of local, transient Ca2+entry (LoCE) events, which comprise cardiac SOCE. Although infrequent in WT myocytes, LoCEs occurred with greater frequency and amplitude in CPVT myocytes. CPVT myocytes also evidenced characteristic arrhythmogenic spontaneous Ca2+ waves under cholinergic stress, which were effectively prevented by SOCE inhibition. In a surprising finding, we report that both LoCEs and their underlying protein machinery are concentrated at the intercalated disk (ID). Therefore, localization of cardiac SOCE in the ID compartment has important implications for SOCE-mediated signaling, arrhythmogenesis and intercellular mechanical and electrical coupling in health and disease.
- Published
- 2019
18. Renal Tubular Secretion of Dofetilide is Dependent on MATE1 Function
- Author
-
Qiang Fu, Alice A. Gibson, Alex Sparreboom, Shuiying Hu, Muhammad Erfan Uddin, Ingrid M. Bonilla, and Cynthia A. Carnes
- Subjects
medicine.medical_specialty ,Renal tubular secretion ,Endocrinology ,Chemistry ,Internal medicine ,Genetics ,medicine ,Dofetilide ,Molecular Biology ,Biochemistry ,Function (biology) ,Biotechnology ,medicine.drug - Published
- 2020
- Full Text
- View/download PDF
19. SOCE Contributes to Normal Calcium Homeostasis and Rythmic Activity of Atrial Myocardium
- Author
-
Silvia G. Priori, Ingrid M. Bonilla, Stephen Baine, Sandor Gyorke, Andriy E. Belevych, Jiaoni Li, Andrei V. Stepanov, Przemysław B. Radwański, P. Volpe, and Dmitry Terentyev
- Subjects
medicine.medical_specialty ,Endocrinology ,Chemistry ,Normal calcium ,Internal medicine ,Biophysics ,medicine ,Atrial myocardium ,Homeostasis - Published
- 2020
- Full Text
- View/download PDF
20. Pyridostigmine Reduces Arrhythmogenic Store Operated Calcium Entry in a Transverse Aortic Constriction HF Model in Mice
- Author
-
Stephen Baine, Andriy E. Belevych, Sandor Gyorke, and Ingrid M. Bonilla
- Subjects
Transverse plane ,medicine.medical_specialty ,Pyridostigmine ,business.industry ,Aortic constriction ,Internal medicine ,Biophysics ,Cardiology ,Medicine ,business ,Store-operated calcium entry ,medicine.drug - Published
- 2020
- Full Text
- View/download PDF
21. Muscarinic Receptor Stimulation Differentially Regulates Nucleoplasmic Calcium in Atrial and Ventricular Myocytes
- Author
-
Ingrid M. Bonilla, Jiaoni Li, Dmitry Terentyev, Sandor Gyorke, Andriy E. Belevych, and Andrei V. Stepanov
- Subjects
medicine.medical_specialty ,Endocrinology ,chemistry ,Internal medicine ,Muscarinic acetylcholine receptor ,Biophysics ,medicine ,chemistry.chemical_element ,Stimulation ,Ventricular myocytes ,Calcium - Published
- 2020
- Full Text
- View/download PDF
22. Gene Transfer of Engineered Calmodulin Alleviates Ventricular Arrhythmias in a Calsequestrin‐Associated Mouse Model of Catecholaminergic Polymorphic Ventricular Tachycardia
- Author
-
Vikram Shettigar, Przemysław B. Radwański, Andriy E. Belevych, Svetlana B. Tikunova, Sandor Gyorke, Bjorn C. Knollmann, Shane D. Walton, Ingrid M. Bonilla, Silvia G. Priori, Hsiang-Ting Ho, Jonathan P. Davis, Pompeo Volpe, and Bin Liu
- Subjects
0301 basic medicine ,Male ,calmodulin ,030204 cardiovascular system & hematology ,Pharmacology ,Arrhythmias ,Ventricular tachycardia ,Calsequestrin ,Ryanodine receptor 2 ,Sudden Cardiac Death ,Calcium Cycling/Excitation-Contraction Coupling ,0302 clinical medicine ,Heart Rate ,Myocyte ,Medicine ,Myocytes, Cardiac ,Arrhythmia and Electrophysiology ,Original Research ,Mice, Knockout ,Ryanodine receptor ,arrhythmia (mechanisms) ,calcium channel ,calcium signaling ,gene therapy ,Gene Transfer Techniques ,3. Good health ,Phenotype ,cardiovascular system ,Cardiology and Cardiovascular Medicine ,Diastole ,Catecholaminergic polymorphic ventricular tachycardia ,Sudden death ,03 medical and health sciences ,Animals ,Diseases of the circulatory (Cardiovascular) system ,Genetic Predisposition to Disease ,business.industry ,Ryanodine Receptor Calcium Release Channel ,Genetic Therapy ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Animal Models of Human Disease ,RC666-701 ,Tachycardia, Ventricular ,business ,Basic Science Research - Abstract
Background Catecholaminergic polymorphic ventricular tachycardia ( CPVT ) is a familial arrhythmogenic syndrome characterized by sudden death. There are several genetic forms of CPVT associated with mutations in genes encoding the cardiac ryanodine receptor (RyR2) and its auxiliary proteins including calsequestrin ( CASQ 2) and calmodulin (CaM). It has been suggested that impairment of the ability of RyR2 to stay closed (ie, refractory) during diastole may be a common mechanism for these diseases. Here, we explore the possibility of engineering CaM variants that normalize abbreviated RyR2 refractoriness for subsequent viral‐mediated delivery to alleviate arrhythmias in non–CaM‐related CPVT . Methods and Results To that end, we have designed a CaM protein ( GSH ‐M37Q; dubbed as therapeutic CaM or T‐CaM) that exhibited a slowed N‐terminal Ca dissociation rate and prolonged RyR2 refractoriness in permeabilized myocytes derived from CPVT mice carrying the CASQ 2 mutation R33Q. This T‐CaM was introduced to the heart of R33Q mice through recombinant adeno‐associated viral vector serotype 9. Eight weeks postinfection, we performed confocal microscopy to assess Ca handling and recorded surface ECGs to assess susceptibility to arrhythmias in vivo. During catecholamine stimulation with isoproterenol, T‐CaM reduced isoproterenol‐promoted diastolic Ca waves in isolated CPVT cardiomyocytes. Importantly, T‐CaM exposure abolished ventricular tachycardia in CPVT mice challenged with catecholamines. Conclusions Our results suggest that gene transfer of T‐CaM by adeno‐associated viral vector serotype 9 improves myocyte Ca handling and alleviates arrhythmias in a calsequestrin‐associated CPVT model, thus supporting the potential of a CaM‐based antiarrhythmic approach as a therapeutic avenue for genetically distinct forms of CPVT .
- Published
- 2018
- Full Text
- View/download PDF
23. Cardiac Store Operated Calcium Entry (SOCE) is Compartmentalized at Intercalated Disks and Linked to Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT)
- Author
-
Mercado, Ingrid M. Bonilla, primary, Belevych, Andriy, additional, Baine, Stephen, additional, Bodnar, Tom, additional, Liu, Bin, additional, Radwanski, Przemyslaw, additional, Veeraraghavan, Rengansayee, additional, Volpe, Pompeo, additional, Priori, Silvia, additional, Weisleder, Noah, additional, and Gyorke, Sandor, additional
- Published
- 2019
- Full Text
- View/download PDF
24. Heart failure duration progressively modulates the arrhythmia substrate through structural and electrical remodeling
- Author
-
Murugesan Velayutham, Kent Mowrey, Jay L. Zweier, Yoshinori Nishijima, Ingrid M. Bonilla, Sanjay Kumar, Chun Li, Nam Y. Lee, Sandor Gyorke, Peter J. Mohler, Pedro Vargas-Pinto, Patrick Wright, Arun Sridhar, Victor P. Long, and Cynthia A. Carnes
- Subjects
medicine.medical_specialty ,Patch-Clamp Techniques ,Time Factors ,Immunoblotting ,Action Potentials ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,General Biochemistry, Genetics and Molecular Biology ,Muscle hypertrophy ,Electrocardiography ,Dogs ,Ventricular hypertrophy ,Fibrosis ,Amphotericin B ,Internal medicine ,medicine ,Animals ,Repolarization ,Potassium Channels, Inwardly Rectifying ,General Pharmacology, Toxicology and Pharmaceutics ,Ventricular remodeling ,DNA Primers ,Heart Failure ,Analysis of Variance ,Ventricular Remodeling ,medicine.diagnostic_test ,Electron Spin Resonance Spectroscopy ,Arrhythmias, Cardiac ,General Medicine ,medicine.disease ,Myocardial Contraction ,Electrophysiology ,Endocrinology ,Heart failure ,Cardiology ,Reactive Oxygen Species ,Delayed Rectifier Potassium Channels - Abstract
Ventricular arrhythmias are a common cause of death in patients with heart failure (HF). Structural and electrical abnormalities in the heart provide a substrate for such arrhythmias. Canine tachypacing-induced HF models of 4-6 weeks duration are often used to study pathophysiology and therapies for HF. We hypothesized that a chronic canine model of HF would result in greater electrical and structural remodeling than a short term model, leading to a more arrhythmogenic substrate.HF was induced by ventricular tachypacing for one (short-term) or four (chronic) months to study remodeling.Left ventricular contractility was progressively reduced, while ventricular hypertrophy and interstitial fibrosis were evident at 4 month but not 1 month of HF. Left ventricular myocyte action potentials were prolonged after 4 (p0.05) but not 1 month of HF. Repolarization instability and early afterdepolarizations were evident only after 4 months of HF (p0.05), coinciding with a prolonged QTc interval (p0.05). The transient outward potassium current was reduced in both HF groups (p0.05). The outward component of the inward rectifier potassium current was reduced only in the 4 month HF group (p0.05). The delayed rectifier potassium currents were reduced in 4 (p0.05) but not 1 month of HF. Reactive oxygen species were increased at both 1 and 4 months of HF (p0.05).Reduced Ito, outward IK1, IKs, and IKr in HF contribute to EAD formation. Chronic, but not short term canine HF, results in the altered electrophysiology and repolarization instability characteristic of end-stage human HF.
- Published
- 2015
- Full Text
- View/download PDF
25. The role of spatial organization of Ca2+ release sites in the generation of arrhythmogenic diastolic Ca2+ release in myocytes from failing hearts
- Author
-
Ingrid M. Bonilla, Karsten E. Schober, Dmitry Terentyev, Hsiang-Ting Ho, Cynthia A. Carnes, Sandor Gyorke, Andriy E. Belevych, and Radmila Terentyeva
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Physiology ,Refractory period ,Ryanodine receptor ,Chemistry ,Phosphatase ,Stimulation ,030204 cardiovascular system & hematology ,Ryanodine receptor 2 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,Physiology (medical) ,Ca2+/calmodulin-dependent protein kinase ,Internal medicine ,cardiovascular system ,Biophysics ,medicine ,Myocyte ,Cardiology and Cardiovascular Medicine ,Receptor - Abstract
In heart failure (HF), dysregulated cardiac ryanodine receptors (RyR2) contribute to the generation of diastolic Ca2+ waves (DCWs), thereby predisposing adrenergically stressed failing hearts to life-threatening arrhythmias. However, the specific cellular, subcellular, and molecular defects that account for cardiac arrhythmia in HF remain to be elucidated. Patch-clamp techniques and confocal Ca2+ imaging were applied to study spatially defined Ca2+ handling in ventricular myocytes isolated from normal (control) and failing canine hearts. Based on their activation time upon electrical stimulation, Ca2+ release sites were categorized as coupled, located in close proximity to the sarcolemmal Ca2+ channels, and uncoupled, the Ca2+ channel-free non-junctional Ca2+ release units. In control myocytes, stimulation of β-adrenergic receptors with isoproterenol (Iso) resulted in a preferential increase in Ca2+ spark rate at uncoupled sites. This site-specific effect of Iso was eliminated by the phosphatase inhibitor okadaic acid, which caused similar facilitation of Ca2+ sparks at coupled and uncoupled sites. Iso-challenged HF myocytes exhibited increased predisposition to DCWs compared to control myocytes. In addition, the overall frequency of Ca2+ sparks was increased in HF cells due to preferential stimulation of coupled sites. Furthermore, coupled sites exhibited accelerated recovery from functional refractoriness in HF myocytes compared to control myocytes. Spatially resolved subcellular Ca2+ mapping revealed that DCWs predominantly originated from coupled sites. Inhibition of CaMKII suppressed DCWs and prevented preferential stimulation of coupled sites in Iso-challenged HF myocytes. These results suggest that CaMKII- (and phosphatase)-dependent dysregulation of junctional Ca2+ release sites contributes to Ca2+-dependent arrhythmogenesis in HF.
- Published
- 2017
- Full Text
- View/download PDF
26. In Utero Particulate Matter Exposure Produces Heart Failure, Electrical Remodeling, and Epigenetic Changes at Adulthood
- Author
-
Markus Velten, Cynthia A. Carnes, Mark T. Ziolo, Stephen Baine, Clayton M. Eichenseer, Matthew W. Gorr, Victor P. Long, Loren E. Wold, Ingrid M. Bonilla, Vikram Shettigar, Vineeta Tanwar, and Jonathan P. Davis
- Subjects
0301 basic medicine ,air pollution ,Action Potentials ,heart failure ,030204 cardiovascular system & hematology ,Calcium Cycling/Excitation-Contraction Coupling ,Ventricular Function, Left ,Molecular Cardiology ,DNA Methyltransferase 3A ,Epigenesis, Genetic ,Mice ,Sirtuin 2 ,0302 clinical medicine ,Sirtuin 1 ,Heart Rate ,Pregnancy ,Myocytes, Cardiac ,DNA (Cytosine-5-)-Methyltransferases ,Phosphorylation ,Original Research ,myocyte ,Inhalation Exposure ,Ventricular Remodeling ,Age Factors ,myocardial ,Particulates ,Maternal Exposure ,In utero ,Prenatal Exposure Delayed Effects ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,Cardiovascular outcomes ,DNA (Cytosine-5-)-Methyltransferase 1 ,medicine.medical_specialty ,cardiac ,Gestational Age ,Calcium-Transporting ATPases ,calcium signaling ,in utero ,Sodium-Calcium Exchanger ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Electrical Remodeling ,Epigenetics ,Particle Size ,cardiovascular function ,particulate matter ,business.industry ,Calcium-Binding Proteins ,Arrhythmias, Cardiac ,Atrial Remodeling ,medicine.disease ,030104 developmental biology ,Endocrinology ,Animals, Newborn ,Animal Models of Human Disease ,Heart failure ,Contractile function ,business - Abstract
Background Particulate matter (PM; PM 2.5 [PM with diameters of PM 2.5 exposure alone could alter cardiac structure and function at adulthood. Methods and Results Female FVB mice were exposed either to filtered air or PM 2.5 at an average concentration of 73.61 μg/m 3 for 6 h/day, 7 days/week throughout pregnancy. After birth, animals were analyzed at 12 weeks of age. Echocardiographic (n=9–10 mice/group) and pressure‐volume loop analyses (n=5 mice/group) revealed reduced fractional shortening, increased left ventricular end‐systolic and ‐diastolic diameters, reduced left ventricular posterior wall thickness, end‐systolic elastance, contractile reserve ( dP /dt max /end‐systolic volume), frequency‐dependent acceleration of relaxation), and blunted contractile response to β‐adrenergic stimulation in PM 2.5 ‐exposed mice. Isolated cardiomyocyte (n=4–5 mice/group) function illustrated reduced peak shortening, ± dL / dT , and prolonged action potential duration at 90% repolarization. Histological left ventricular analyses (n=3 mice/group) showed increased collagen deposition in in utero PM 2.5 ‐exposed mice at adulthood. Cardiac interleukin ( IL) ‐6, IL ‐1ß, collagen‐1, matrix metalloproteinase ( MMP ) 9, and MMP 13 gene expressions were increased at birth in in utero PM 2.5 ‐exposed mice (n=4 mice/group). In adult hearts (n=5 mice/group), gene expressions of sirtuin (Sirt) 1 and Sirt2 were decreased, DNA methyltransferase (Dnmt) 1, Dnmt3a, and Dnmt3b were increased, and protein expression (n=6 mice/group) of Ca 2+ ‐ATPase, phosphorylated phospholamban, and Na + /Ca 2+ exchanger were decreased. Conclusions In utero PM 2.5 exposure triggers an acute inflammatory response, chronic matrix remodeling, and alterations in Ca 2+ handling proteins, resulting in global adult cardiac dysfunction. These results also highlight the potential involvement of epigenetics in priming of adult cardiac disease.
- Published
- 2017
- Full Text
- View/download PDF
27. The role of spatial organization of Ca
- Author
-
Andriy E, Belevych, Hsiang-Ting, Ho, Ingrid M, Bonilla, Radmila, Terentyeva, Karsten E, Schober, Dmitry, Terentyev, Cynthia A, Carnes, and Sándor, Györke
- Subjects
Heart Failure ,Male ,Time Factors ,Calcium Channels, L-Type ,Refractory Period, Electrophysiological ,Sus scrofa ,Cardiac Pacing, Artificial ,Arrhythmias, Cardiac ,Ryanodine Receptor Calcium Release Channel ,Adrenergic beta-Agonists ,Ventricular Function, Left ,Article ,Membrane Potentials ,Disease Models, Animal ,Dogs ,Membrane Microdomains ,Sarcolemma ,Diastole ,Heart Rate ,Animals ,Calcium ,Female ,Myocytes, Cardiac ,Calcium Signaling ,Calcium-Calmodulin-Dependent Protein Kinase Type 2 - Abstract
In heart failure (HF), dysregulated cardiac ryanodine receptors (RyR2) contribute to the generation of diastolic Ca2+ waves (DCWs), thereby predisposing adrenergically stressed failing hearts to life-threatening arrhythmias. However, the specific cellular, subcellular, and molecular defects that account for cardiac arrhythmia in HF remain to be elucidated. Patch-clamp techniques and confocal Ca2+ imaging were applied to study spatially defined Ca2+ handling in ventricular myocytes isolated from normal (control) and failing canine hearts. Based on their activation time upon electrical stimulation, Ca2+ release sites were categorized as coupled, located in close proximity to the sarcolemmal Ca2+ channels, and uncoupled, the Ca2+ channel-free non-junctional Ca2+ release units. In control myocytes, stimulation of β-adrenergic receptors with isoproterenol (Iso) resulted in a preferential increase in Ca2+ spark rate at uncoupled sites. This site-specific effect of Iso was eliminated by the phosphatase inhibitor okadaic acid, which caused similar facilitation of Ca2+ sparks at coupled and uncoupled sites. Iso-challenged HF myocytes exhibited increased predisposition to DCWs compared to control myocytes. In addition, the overall frequency of Ca2+ sparks was increased in HF cells due to preferential stimulation of coupled sites. Furthermore, coupled sites exhibited accelerated recovery from functional refractoriness in HF myocytes compared to control myocytes. Spatially resolved subcellular Ca2+ mapping revealed that DCWs predominantly originated from coupled sites. Inhibition of CaMK∏ suppressed DCWs and prevented preferential stimulation of coupled sites in Iso-challenged HF myocytes. These results suggest that CaMK∏-(and phosphatase)-dependent dysregulation of junctional Ca2+ release sites contributes to Ca2+-dependent arrhythmogenesis in HF.
- Published
- 2017
28. Cardiac Store Operated Calcium Entry (SOCE) is Compartmentalized at Intercalated Disks and Linked to Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT)
- Author
-
Tom Bodnar, Noah Weisleder, Stephen Baine, Andriy E. Belevych, Ingrid M. Bonilla Mercado, Przemysław B. Radwański, Rengansayee Veeraraghavan, Sandor Gyorke, Pompeo Volpe, Silvia G. Priori, and Bin Liu
- Subjects
medicine.medical_specialty ,Chemistry ,Internal medicine ,Biophysics ,medicine ,Cardiology ,Catecholaminergic polymorphic ventricular tachycardia ,medicine.disease ,Store-operated calcium entry - Published
- 2019
- Full Text
- View/download PDF
29. Ibandronate and Ventricular Arrhythmia Risk
- Author
-
Pedro Vargas-Pinto, Ingrid M. Bonilla, Adriana Pedraza-Toscano, Yoshinori Nishijima, Patric Glynn, Hsiang-Ting Ho, Robert L. Hamlin, Raul Weiss, Victor P. Long, Andriy E. Belevych, Thomas J. Hund, Sandor Gyorke, Cynthia A. Carnes, Troy E. Rhodes, and Mahmoud Houmsse
- Subjects
Proarrhythmia ,medicine.medical_specialty ,Ryanodine receptor ,business.industry ,chemistry.chemical_element ,Calcium ,medicine.disease ,QT interval ,Calcium in biology ,Afterdepolarization ,Endocrinology ,chemistry ,Physiology (medical) ,Internal medicine ,Ventricular fibrillation ,cardiovascular system ,medicine ,Repolarization ,Cardiology and Cardiovascular Medicine ,business - Abstract
Ibandronate-Induced Proarrhythmia Introduction Bisphosphonates, including ibandronate, are used in the prevention and treatment of osteoporosis. Methods and Results We report a case of suspected ibandronate-associated arrhythmia, following a single dose of ibandronate in a 55-year-old female. ECG at presentation revealed frequent ectopy and QT/QTc interval prolongation; at follow-up 9 months later the QT/QTc intervals were normalized. Proarrhythmic potential of ibandronate was assessed with a combination of in vivo and in vitro approaches in canines and canine ventricular myocytes. We observed late onset in vivo repolarization instability after ibandronate treatment. Myocytes superfused with ibandronate exhibited action potential duration (APD) prolongation and variability, increased early afterdepolarizations (EADs) and reduced Ito (P
- Published
- 2013
- Full Text
- View/download PDF
30. Differential Effects of the Peroxynitrite Donor, SIN-1, on Atrial and Ventricular Myocyte Electrophysiology
- Author
-
Arturo J. Cardounel, Sandor Gyorke, Arun Sridhar, Yoshinori Nishijima, Cynthia A. Carnes, and Ingrid M. Bonilla
- Subjects
Male ,medicine.medical_specialty ,SERCA ,Action Potentials ,Article ,chemistry.chemical_compound ,Dogs ,Internal medicine ,Animals ,Ventricular Function ,Medicine ,Myocyte ,Repolarization ,Myocytes, Cardiac ,Nitric Oxide Donors ,cardiovascular diseases ,Patch clamp ,Enzyme Inhibitors ,Pharmacology ,business.industry ,Atrial fibrillation ,Atrial Function ,medicine.disease ,medicine.anatomical_structure ,chemistry ,Ventricle ,Molsidomine ,Heart failure ,cardiovascular system ,Cardiology ,Thapsigargin ,Female ,Cardiology and Cardiovascular Medicine ,business ,Peroxynitrite - Abstract
Oxidative stress has been implicated in the pathogenesis of heart failure and atrial fibrillation and can result in increased peroxynitrite production in the myocardium. Atrial and ventricular canine cardiac myocytes were superfused with 3-morpholinosydnonimine-N-ethylcarbamide (SIN-1), a peroxynitrite donor, to evaluate the acute electrophysiologic effects of peroxynitrite. Perforated whole-cell patch clamp techniques were used to record action potentials. SIN-1 (200 µM) increased the action potential duration (APD) in atrial and ventricular myocytes; however, in the atria, APD prolongation was rate independent, whereas in the ventricle APD, prolongation was rate dependent. In addition to prolongation of the action potential, beat-to-beat variability of repolarization was significantly increased in ventricular but not in atrial myocytes. We examined the contribution of intracellular calcium cycling to the effects of SIN-1 by treating myocytes with the SERCA blocker, thapsigargin (5-10 µM). Inhibition of calcium cycling prevented APD prolongation in the atrial and ventricular myocytes, and prevented the SIN-1-induced increase in ventricular beat-to-beat APD variability. Collectively, these data demonstrate that peroxynitrite affects atrial and ventricular electrophysiology differentially. A detailed understanding of oxidative modulation of electrophysiology in specific chambers is critical to optimize therapeutic approaches for cardiac diseases.
- Published
- 2013
- Full Text
- View/download PDF
31. Chronic Omega-3 Polyunsaturated Fatty Acid Treatment Variably Affects Cellular Repolarization in a Healed Post-MI Arrhythmia Model
- Author
-
George E. Billman, Yoshinori Nishijima, Arun Sridhar, Pedro Vargas-Pinto, Ingrid M. Bonilla, Chun Li, Cynthia A. Carnes, and Stephen Baine
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Physiology ,030204 cardiovascular system & hematology ,digestive system ,Sudden cardiac death ,03 medical and health sciences ,0302 clinical medicine ,potassium current ,Physiology (medical) ,Internal medicine ,medicine ,Repolarization ,Myocyte ,Myocardial infarction ,Original Research ,omega-3 polyunsaturated fatty acids ,chemistry.chemical_classification ,business.industry ,ventricular fibrillation ,electrophysiology ,medicine.disease ,3. Good health ,Potassium current ,Electrophysiology ,myocardial infarction ,030104 developmental biology ,chemistry ,Ventricular fibrillation ,Cardiology ,business ,Polyunsaturated fatty acid - Abstract
Introduction: Over the last 40 years omega-3 polyunsaturated fatty acids (PUFAs) have been shown to be anti-arrhythmic or pro-arrhythmic depending on the method and duration of administration and model studied. We previously reported that omega-3 PUFAs do not confer anti-arrhythmic properties and are pro-arrhythmic in canine model of sudden cardiac death. Here we evaluated the effects of chronic omega-3 PUFA treatment in post- MI animals susceptible (VF+) or resistant (VF-) to ventricular tachyarrhythmias. Methods: Perforated patch clamp techniques were used to measure cardiomyocyte action potential durations at 50 and 90% repolarization and short term variability of repolarization. The early repolarizing transient outward potassium current Ito was also studied. Results Omega-3 PUFAs prolonged the action potential in VF- myocytes at both 50% and 90% repolarization. Short term variability of repolarization was increased in both untreated and treated VF- myocytes vs. controls. Ito was unaffected by omega-3 PUFA treatment. Omega-3 PUFA treatment attenuated the action potential prolongation in VF+ myocytes, but did not return repolarization to control values. Conclusions: Omega-3 PUFAs do not confer anti-arrhythmic properties in the setting of healed myocardial infarction in a canine model of sudden cardiac death. In canines previously resistant to ventricular fibrillation (VF-), omega-3 PUFA treatment prolonged the action potential in VF- myocytes and may contribute to pro-arrhythmic responses.
- Published
- 2016
- Full Text
- View/download PDF
32. Muscarinic Stimulation Facilitates Sarcoplasmic Reticulum Ca Release by Modulating Ryanodine Receptor 2 Phosphorylation Through Protein Kinase G and Ca/Calmodulin-Dependent Protein Kinase II
- Author
-
Cynthia A. Carnes, Karsten E. Schober, Bin Liu, Andriy E. Belevych, Sandor Gyorke, Przemysław B. Radwański, I. V. Kubasov, Ingrid M. Bonilla, Héctor H. Valdivia, and Hsiang-Ting Ho
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Cholinergic Agents ,030204 cardiovascular system & hematology ,Ryanodine receptor 2 ,Sensitivity and Specificity ,Article ,Dephosphorylation ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Dogs ,Internal medicine ,Ca2+/calmodulin-dependent protein kinase ,Muscarinic acetylcholine receptor ,Internal Medicine ,medicine ,Animals ,Myocytes, Cardiac ,Phosphorylation ,Cells, Cultured ,Excitation Contraction Coupling ,Heart Failure ,Chemistry ,Ryanodine receptor ,Endoplasmic reticulum ,Ryanodine Receptor Calcium Release Channel ,Receptors, Muscarinic ,Cell biology ,Disease Models, Animal ,Sarcoplasmic Reticulum ,030104 developmental biology ,Endocrinology ,cardiovascular system ,Calcium ,Calcium-Calmodulin-Dependent Protein Kinase Type 2 ,cGMP-dependent protein kinase - Abstract
Although the effects and the underlying mechanism of sympathetic stimulation on cardiac Ca handling are relatively well established both in health and disease, the modes of action and mechanisms of parasympathetic modulation are poorly defined. Here, we demonstrate that parasympathetic stimulation initiates a novel mode of excitation–contraction coupling that enhances the efficiency of cardiac sarcoplasmic reticulum Ca store utilization. This efficient mode of excitation–contraction coupling involves reciprocal changes in the phosphorylation of ryanodine receptor 2 at Ser-2808 and Ser-2814. Specifically, Ser-2808 phosphorylation was mediated by muscarinic receptor subtype 2 and activation of PKG (protein kinase G), whereas dephosphorylation of Ser-2814 involved activation of muscarinic receptor subtype 3 and decreased reactive oxygen species–dependent activation of CaMKII (Ca/calmodulin-dependent protein kinase II). The overall effect of these changes in phosphorylation of ryanodine receptor 2 is an increase in systolic Ca release at the low sarcoplasmic reticulum Ca content and a paradoxical reduction in aberrant Ca leak. Accordingly, cholinergic stimulation of cardiomyocytes isolated from failing hearts improved Ca cycling efficiency by restoring altered ryanodine receptor 2 phosphorylation balance.
- Published
- 2016
33. Prolonged Action Potential and After depolarizations Are Not due to Changes in Potassium Currents in NOS3 Knockout Ventricular Myocytes
- Author
-
Ingrid M. Bonilla, Mark T. Ziolo, Mark J. Kohr, Quanhua He, Xin Huang, Cynthia A. Carnes, and Honglan Wang
- Subjects
medicine.medical_specialty ,Article Subject ,Inward-rectifier potassium ion channel ,Potassium ,Prolonged action ,chemistry.chemical_element ,Stimulation ,Cell Biology ,medicine.disease ,Biochemistry ,Nitric oxide ,body regions ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Endocrinology ,chemistry ,TheoryofComputation_ANALYSISOFALGORITHMSANDPROBLEMCOMPLEXITY ,Heart failure ,Internal medicine ,cardiovascular system ,medicine ,Myocyte ,cardiovascular diseases ,Ventricular myocytes ,Research Article - Abstract
Ventricular myocytes deficient in endothelial nitric oxide synthase (NOS3−/−) exhibit prolonged action potential (AP) duration and enhanced spontaneous activity (early and delayed afterdepolarizations) during β-adrenergic (β-AR) stimulation. Studies have shown that nitric oxide is able to regulate various K+ channels. Our objective was to examine if NOS3-/- myocytes had altered K+ currents. APs, transient outward (), sustained (), and inward rectifier () K+ currents were measured in NOS3-/- and wild-type (WT) myocytes. During β-AR stimulation, AP duration (measured as 90% repolarization-APD90) was prolonged in NOS3−/− compared to WT myocytes. Nevertheless, we did not observe differences in , , or between WT and NOS3−/− myocytes. Our previous work showed that NOS3−/− myocytes had a greater Ca2+ influx via L-type Ca2+ channels with β-AR stimulation. Thus, we measured β-AR-stimulated SR Ca2+ load and found a greater increase in NOS3−/− versus WT myocytes. Hence, our data suggest that the prolonged AP in NOS3−/− myocytes is not due to changes in , , or . Furthermore, the increase in spontaneous activity in NOS3−/− myocytes may be due to a greater increase in SR Ca2+ load. This may have important implications for heart failure patients, where arrhythmias are increased and NOS3 expression is decreased.
- Published
- 2012
- Full Text
- View/download PDF
34. Shortened Ca 2+ Signaling Refractoriness Underlies Cellular Arrhythmogenesis in a Postinfarction Model of Sudden Cardiac Death
- Author
-
George E. Billman, Hsiang-Ting Ho, Ingrid M. Bonilla, Andriy E. Belevych, Dmitry Terentyev, Cynthia A. Carnes, Radmila Terentyeva, Sandor Gyorke, and Inna Györke
- Subjects
Male ,Agonist ,Benzylamines ,medicine.medical_specialty ,Time Factors ,Physiology ,medicine.drug_class ,Refractory period ,Myocardial Infarction ,Biology ,Ryanodine receptor 2 ,Article ,Dogs ,Caffeine ,Ca2+/calmodulin-dependent protein kinase ,Internal medicine ,Reaction Time ,medicine ,Animals ,Myocyte ,Myocytes, Cardiac ,Calcium Signaling ,Phosphorylation ,Receptor ,Protein Kinase Inhibitors ,Excitation Contraction Coupling ,Sulfonamides ,Ryanodine receptor ,Cardiac Pacing, Artificial ,Isoproterenol ,Tiopronin ,Ryanodine Receptor Calcium Release Channel ,Adrenergic beta-Agonists ,medicine.disease ,Calcium Channel Agonists ,Disease Models, Animal ,Sarcoplasmic Reticulum ,Death, Sudden, Cardiac ,Endocrinology ,Reducing Agents ,Ventricular Fibrillation ,Ventricular fibrillation ,Cardiology ,Female ,Calcium-Calmodulin-Dependent Protein Kinase Type 2 ,Cardiology and Cardiovascular Medicine ,Oxidation-Reduction - Abstract
Rationale: Diastolic spontaneous Ca 2+ waves (DCWs) are recognized as important contributors to triggered arrhythmias. DCWs are thought to arise when [Ca 2+ ] in sarcoplasmic reticulum ([Ca 2+ ] SR ) reaches a certain threshold level, which might be reduced in cardiac disease as a consequence of sensitization of ryanodine receptors (RyR2s) to luminal Ca 2+ . Objective: We investigated the mechanisms of DCW generation in myocytes from normal and diseased hearts, using a canine model of post–myocardial infarction ventricular fibrillation (VF). Methods and Results: The frequency of DCWs, recorded during periodic pacing in the presence of a β-adrenergic receptor agonist isoproterenol, was significantly higher in VF myocytes than in normal controls. Rather than occurring immediately on reaching a final [Ca 2+ ] SR , DCWs arose with a distinct time delay after attaining steady [Ca 2+ ] SR in both experimental groups. Although the rate of [Ca 2+ ] SR recovery after the SR Ca 2+ release was similar between the groups, in VF myocytes the latency to DCWs was shorter, and the [Ca 2+ ] SR at DCW initiation was lower. The restitution of depolarization-induced Ca 2+ transients, assessed by a 2-pulse protocol, was significantly faster in VF myocytes than in controls. The VF-related alterations in myocyte Ca 2+ cycling were mimicked by the RyR2 agonist, caffeine. The reducing agent, mercaptopropionylglycine, or the CaMKII inhibitor, KN93, decreased DCW frequency and normalized restitution of Ca 2+ release in VF myocytes. Conclusions: The attainment of a certain threshold [Ca 2+ ] SR is not sufficient for the generation of DCWs. Postrelease Ca 2+ signaling refractoriness critically influences the occurrence of DCWs. Shortened Ca 2+ signaling refractoriness due to RyR2 phosphorylation and oxidation is responsible for the increased rate of DCWs observed in VF myocytes and could provide a substrate for synchronization of arrhythmogenic events at the tissue level in hearts prone to VF.
- Published
- 2012
- Full Text
- View/download PDF
35. Tetrahydrobiopterin depletion and NOS2 uncoupling contribute to heart failure-induced alterations in atrial electrophysiology
- Author
-
Terry S. Elton, Arturo J. Cardounel, Jay L. Zweier, Cynthia A. Carnes, Murugesan Velayutham, Arun Sridhar, Chun Li, Periannan Kuppusamy, Yoshinori Nishijima, Radmila Terentyeva, Dmitry Terentyev, Sandor Gyorke, Mahmood Khan, and Ingrid M. Bonilla
- Subjects
Male ,medicine.medical_specialty ,Patch-Clamp Techniques ,Atrial action potential ,Physiology ,Action Potentials ,Nitric Oxide Synthase Type II ,Biopterin ,Arginine ,Nitric Oxide ,medicine.disease_cause ,Ventricular Function, Left ,Nitric oxide ,chemistry.chemical_compound ,Dogs ,Superoxides ,Physiology (medical) ,Internal medicine ,Atrial Fibrillation ,medicine ,Animals ,Heart Atria ,cardiovascular diseases ,Atrium (heart) ,Heart Failure ,business.industry ,Myocardium ,P wave ,Cardiac Pacing, Artificial ,Atrial fibrillation ,medicine.disease ,Up-Regulation ,Disease Models, Animal ,Kinetics ,Oxidative Stress ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Heart failure ,cardiovascular system ,Cardiology ,Atrial Function, Left ,Female ,Cardiology and Cardiovascular Medicine ,business ,Oxidative stress - Abstract
Aims Heart failure is a common antecedent to atrial fibrillation; both heart failure and atrial fibrillation are associated with increased myocardial oxidative stress. Chronic canine heart failure reduces atrial action potential duration and atrial refractoriness. We hypothesized that inducible nitric oxide synthase 2 (NOS2) contributes to atrial oxidative stress and electrophysiologic alterations. Methods and results A 16-week canine tachypacing model of heart failure was used ( n = 21). At 10 weeks, dogs were randomized to either placebo ( n = 12) or active treatment ( n = 9) with NOS cofactor, tetrahydrobiopterin (BH4, 50 mg), and NOS substrate (l-arginine, 3 g) twice daily for 6 weeks. A group of matched controls ( n = 7) was used for comparison. Heart failure increased atrial NOS2 and reduced atrial BH4, while l-arginine was unchanged. Treatment reduced inducible atrial fibrillation and normalized the heart failure-induced shortening of the left atrial myocyte action potential duration. Treatment increased atrial [BH4] while [l-arginine] was unchanged. Treatment did not improve left ventricular function or dimensions. Heart failure-induced reductions in atrial [BH4] resulted in NOS uncoupling, as measured by NO and superoxide anion (O2·−) production, while BH4 and l-arginine treatment normalized NO and O2·−. Heart failure resulted in left atrial oxidative stress, which was attenuated by BH4 and l-arginine treatment. Conclusion Chronic non-ischaemic heart failure results in atrial oxidative stress and electrophysiologic abnormalities by depletion of BH4 and uncoupling of NOS2. Modulation of NOS2 activity by repletion of BH4 may be a safe and effective approach to reduce the frequency of atrial arrhythmias during heart failure.
- Published
- 2011
- Full Text
- View/download PDF
36. Abstract 17344: Increasing Calcium-activated Potassium Current Shortens and Stabilizes Repolarization in Chronic Heart Failure
- Author
-
Ingrid M Bonilla, Victor Long, Kent Mowrey, Karsten Schober, Raul Weiss, Sandor Gyorke, and Cynthia Carnes
- Subjects
Physiology (medical) ,Cardiology and Cardiovascular Medicine - Abstract
Heart failure (HF) is a chronic disease resulting in abnormal prolongation and instability of ventricular repolarization. IKCa has been suggested to stabilize repolarization in HF as IKca blockade has shown pro-arrhythmic effects in the failing ventricle. We tested the hypothesis that an SK channel (IKCa) agonist CyPPA, would shorten and stabilize ventricular repolarization in HF ventricular myocytes. Methods: A tachypacing - induced 4 month HF canine model was used (LVFS: 15.9 ± 2.5%), and LV midwall myocytes were isolated and compared to normal controls. Action potential duration at 50 (APD50), and 90% (APD90) repolarization and APD instability (beat to beat variability of repolarization, a measure of arrhythmia risk) were measured before and after the application of CyPPA (0.001nM-20uM) in control ventricular, and in 4 months HF ventricular myocytes (n=10-15 per group). Results: In control myocytes, CyPPA shortened action potential APD50 and APD90 in a concentration dependent manner (0.001nM-20uM, p
- Published
- 2015
- Full Text
- View/download PDF
37. Dysfunction in the βII Spectrin-Dependent Cytoskeleton Underlies Human Arrhythmia
- Author
-
Ingrid M. Bonilla, Ahmet Kilic, Thomas J. Hund, Iuliia Polina, Amy C. Sturm, Raul Weiss, Langston D. Hughes, Robert S.D. Higgins, Crystal F. Kline, Victor P. Long, Mingzhai Sun, Philip F. Binkley, Tyler R. Webb, Patrick Wright, Peter J. Mohler, Matthew N. Rasband, Dobromir Dobrev, Jerry Curran, Matteo Vatta, Zhiyi Wei, Niels Voigt, Cynthia A. Carnes, Michael A. Makara, Deepak Bhakta, Mingjie Zhang, Chuansheng Zhang, Paul M. Janssen, Katherine G. Spoonamore, Sakima A. Smith, Sean C. Little, and Jianjie Ma
- Subjects
medicine.medical_specialty ,Medizin ,Biology ,Article ,Physiology (medical) ,Internal medicine ,medicine ,Myocyte ,Ankyrin ,Animals ,Humans ,Spectrin ,Myocytes, Cardiac ,Cytoskeleton ,chemistry.chemical_classification ,Cardiac arrhythmia ,Arrhythmias, Cardiac ,Microfilament Protein ,Transport protein ,Cell biology ,Endocrinology ,Membrane protein ,chemistry ,Cardiology and Cardiovascular Medicine - Abstract
Background— The cardiac cytoskeleton plays key roles in maintaining myocyte structural integrity in health and disease. In fact, human mutations in cardiac cytoskeletal elements are tightly linked to cardiac pathologies, including myopathies, aortopathies, and dystrophies. Conversely, the link between cytoskeletal protein dysfunction and cardiac electric activity is not well understood and often overlooked in the cardiac arrhythmia field. Methods and Results— Here, we uncover a new mechanism for the regulation of cardiac membrane excitability. We report that βII spectrin, an actin-associated molecule, is essential for the posttranslational targeting and localization of critical membrane proteins in heart. βII spectrin recruits ankyrin-B to the cardiac dyad, and a novel human mutation in the ankyrin-B gene disrupts the ankyrin-B/βII spectrin interaction, leading to severe human arrhythmia phenotypes. Mice lacking cardiac βII spectrin display lethal arrhythmias, aberrant electric and calcium handling phenotypes, and abnormal expression/localization of cardiac membrane proteins. Mechanistically, βII spectrin regulates the localization of cytoskeletal and plasma membrane/sarcoplasmic reticulum protein complexes, including the Na/Ca exchanger, ryanodine receptor 2, ankyrin-B, actin, and αII spectrin. Finally, we observe accelerated heart failure phenotypes in βII spectrin–deficient mice. Conclusions— Our findings identify βII spectrin as critical for normal myocyte electric activity, link this molecule to human disease, and provide new insight into the mechanisms underlying cardiac myocyte biology.
- Published
- 2015
38. Calcium-activated potassium current modulates ventricular repolarization in chronic heart failure
- Author
-
Ahmet Kilic, Victor P. Long, Qing Lou, Sándor Györke, Jae Yoo, Peter J. Mohler, Pedro Vargas-Pinto, Patrick J. Wright, Philip F. Binkley, Ingrid M. Bonilla, Paulus M. L. Janssen, Andriy E. Belevych, Vladim V. Fedorov, Cynthia A. Carnes, and Kent Mowrey
- Subjects
medicine.medical_specialty ,Atrial action potential ,Science ,Heart Ventricles ,Cardiology ,Action Potentials ,Pharmacology ,Cardiovascular Pharmacology ,Afterdepolarization ,Dogs ,Internal medicine ,Atrial Fibrillation ,Medicine and Health Sciences ,Medicine ,Repolarization ,Myocyte ,Animals ,Humans ,Myocytes, Cardiac ,cardiovascular diseases ,Heart Failure ,Multidisciplinary ,business.industry ,Myocardium ,Atrial fibrillation ,Heart ,medicine.disease ,3. Good health ,Blockade ,Disease Models, Animal ,medicine.anatomical_structure ,Ventricle ,Cardiovascular Diseases ,Heart failure ,cardiovascular system ,Potassium ,Calcium ,business ,Arrhythmia ,Research Article - Abstract
The role of I(KCa) in cardiac repolarization remains controversial and varies across species. The relevance of the current as a therapeutic target is therefore undefined. We examined the cellular electrophysiologic effects of I(KCa) blockade in controls, chronic heart failure (HF) and HF with sustained atrial fibrillation. We used perforated patch action potential recordings to maintain intrinsic calcium cycling. The I(KCa) blocker (apamin 100 nM) was used to examine the role of the current in atrial and ventricular myocytes. A canine tachypacing induced model of HF (1 and 4 months, n = 5 per group) was used, and compared to a group of 4 month HF with 6 weeks of superimposed atrial fibrillation (n = 7). A group of age-matched canine controls were used (n = 8). Human atrial and ventricular myocytes were isolated from explanted end-stage failing hearts which were obtained from transplant recipients, and studied in parallel. Atrial myocyte action potentials were unchanged by I(KCa) blockade in all of the groups studied. I(KCa) blockade did not affect ventricular myocyte repolarization in controls. HF caused prolongation of ventricular myocyte action potential repolarization. I(KCa) blockade caused further prolongation of ventricular repolarization in HF and also caused repolarization instability and early afterdepolarizations. SK2 and SK3 expression in the atria and SK3 in the ventricle were increased in canine heart failure. We conclude that during HF, I(KCa) blockade in ventricular myocytes results in cellular arrhythmias. Furthermore, our data suggest an important role for I(KCa) in the maintenance of ventricular repolarization stability during chronic heart failure. Our findings suggest that novel antiarrhythmic therapies should have safety and efficacy evaluated in both atria and ventricles.
- Published
- 2014
39. Dietary omega-3 fatty acids promote arrhythmogenic remodeling of cellular Ca2+ handling in a postinfarction model of sudden cardiac death
- Author
-
Radmila Terentyeva, Hsiang-Ting Ho, Sandor Gyorke, Cynthia A. Carnes, George E. Billman, Dmitry Terentyev, Ingrid M. Bonilla, and Andriy E. Belevych
- Subjects
medicine.medical_specialty ,Science ,Diastole ,Myocardial Infarction ,Sudden cardiac death ,Dogs ,Internal medicine ,Fatty Acids, Omega-3 ,Medicine ,Myocyte ,Animals ,Myocytes, Cardiac ,Calcium metabolism ,chemistry.chemical_classification ,Multidisciplinary ,business.industry ,Ryanodine receptor ,food and beverages ,Arrhythmias, Cardiac ,Heart ,Ryanodine Receptor Calcium Release Channel ,medicine.disease ,Diet ,Sarcoplasmic Reticulum ,Death, Sudden, Cardiac ,chemistry ,Ventricular fibrillation ,Ventricular Fibrillation ,Cardiology ,Calcium ,business ,Intracellular ,Polyunsaturated fatty acid ,Research Article - Abstract
It has been proposed that dietary omega-3 polyunsaturated fatty acids (n-3 PUFAs) can reduce the risk of ventricular arrhythmias in post-MI patients. Abnormal Ca(2+) handling has been implicated in the genesis of post-MI ventricular arrhythmias. Therefore, we tested the hypothesis that dietary n-3 PUFAs alter the vulnerability of ventricular myocytes to cellular arrhythmia by stabilizing intracellular Ca(2+) cycling. To test this hypothesis, we used a canine model of post-MI ventricular fibrillation (VF) and assigned the animals to either placebo (1 g/day corn oil) or n-3 PUFAs (1-4 g/day) groups. Using Ca(2+) imaging techniques, we examined the intracellular Ca(2+) handling in myocytes isolated from post-MI hearts resistant (VF-) and susceptible (VF+) to VF. Frequency of occurrence of diastolic Ca(2+) waves (DCWs) in VF+ myocytes from placebo group was significantly higher than in placebo-treated VF- myocytes. n-3 PUFA treatment did not decrease frequency of DCWs in VF+ myocytes. In contrast, VF- myocytes from the n-3 PUFA group had a significantly higher frequency of DCWs than myocytes from the placebo group. In addition, n-3 PUFA treatment increased beat-to-beat alterations in the amplitude of Ca(2+) transients (Ca(2+) alternans) in VF- myocytes. These n-3 PUFAs effects in VF- myocytes were associated with an increased Ca(2+) spark frequency and reduced sarcoplasmic reticulum Ca(2+) content, indicative of increased activity of ryanodine receptors. Thus, dietary n-3 PUFAs do not alleviate intracellular Ca(2+) cycling remodeling in myocytes isolated from post-MI VF+ hearts. Furthermore, dietary n-3 PUFAs increase vulnerability of ventricular myocytes to cellular arrhythmia in post-MI VF- hearts by destabilizing intracellular Ca(2+) handling.
- Published
- 2013
40. Endurance exercise training normalizes repolarization and calcium-handling abnormalities, preventing ventricular fibrillation in a model of sudden cardiac death
- Author
-
Quanhua He, Andriy E. Belevych, George E. Billman, Ingrid M. Bonilla, Bin Liu, Arun Sridhar, Cynthia A. Carnes, Radmila Terentyeva, Sandor Gyorke, Yoshinori Nishijima, Dmitry Terentyev, Victor P. Long, Hsiang-Ting Ho, and Monica Kukielka
- Subjects
medicine.medical_specialty ,Patch-Clamp Techniques ,Potassium Channels ,Time Factors ,Physiology ,Myocardial Infarction ,Action Potentials ,Sudden cardiac death ,Electrocardiography ,Dogs ,Endurance training ,Heart Rate ,Physiology (medical) ,Internal medicine ,Heart rate ,medicine ,Repolarization ,Animals ,Myocytes, Cardiac ,Myocardial infarction ,Calcium Signaling ,Phosphorylation ,medicine.diagnostic_test ,business.industry ,Ryanodine Receptor Calcium Release Channel ,Articles ,medicine.disease ,Exercise Therapy ,Disease Models, Animal ,Death, Sudden, Cardiac ,Anesthesia ,Ventricular fibrillation ,Ventricular Fibrillation ,Cardiology ,cardiovascular system ,Physical Endurance ,Potassium ,Myocardial infarction complications ,business - Abstract
The risk of sudden cardiac death is increased following myocardial infarction. Exercise training reduces arrhythmia susceptibility, but the mechanism is unknown. We used a canine model of sudden cardiac death (healed infarction, with ventricular tachyarrhythmias induced by an exercise plus ischemia test, VF+); we previously reported that endurance exercise training was antiarrhythmic in this model (Billman GE. Am J Physiol Heart Circ Physiol 297: H1171–H1193, 2009). A total of 41 VF+ animals were studied, after random assignment to 10 wk of endurance exercise training (EET; n = 21) or a matched sedentary period ( n = 20). Following (>1 wk) the final attempted arrhythmia induction, isolated myocytes were used to test the hypotheses that the endurance exercise-induced antiarrhythmic effects resulted from normalization of cellular electrophysiology and/or normalization of calcium handling. EET prevented VF and shortened in vivo repolarization ( P < 0.05). EET normalized action potential duration and variability compared with the sedentary group. EET resulted in a further decrement in transient outward current compared with the sedentary VF+ group ( P < 0.05). Sedentary VF+ dogs had a significant reduction in repolarizing K+ current, which was restored by exercise training ( P < 0.05). Compared with controls, myocytes from the sedentary VF+ group displayed calcium alternans, increased calcium spark frequency, and increased phosphorylation of S2814 on ryanodine receptor 2. These abnormalities in intracellular calcium handling were attenuated by exercise training ( P < 0.05). Exercise training prevented ischemically induced VF, in association with a combination of beneficial effects on cellular electrophysiology and calcium handling.
- Published
- 2012
41. Repolarization abnormalities and afterdepolarizations in a canine model of sudden cardiac death
- Author
-
Gail A. Robertson, Yoshinori Nishijima, George E. Billman, Rebecca Uelmen, Monica Kukielka, Dmitry Terentyev, Sandor Gyorke, Ingrid M. Bonilla, Radmila Terentyeva, Arun Sridhar, and Cynthia A. Carnes
- Subjects
Male ,medicine.medical_specialty ,Potassium Channels ,Physiology ,Ischemia ,Myocardial Infarction ,Action Potentials ,Down-Regulation ,Cell Separation ,In Vitro Techniques ,Sudden death ,Sudden cardiac death ,Afterdepolarization ,Electrocardiography ,Dogs ,Physiology (medical) ,Internal medicine ,Potassium Channel Blockers ,Medicine ,Repolarization ,Animals ,Myocytes, Cardiac ,Myocardial infarction ,cardiovascular diseases ,Risk factor ,4-Aminopyridine ,medicine.diagnostic_test ,business.industry ,Translational Physiology ,medicine.disease ,Electrophysiology ,Death, Sudden, Cardiac ,Anesthesia ,Cardiology ,Tachycardia, Ventricular ,Female ,business - Abstract
Ventricular tachyarrhythmias are the most common cause of sudden cardiac death (SCD); a healed myocardial infarction increases the risk of SCD. We determined the contribution of specific repolarization abnormalities to ventricular tachyarrhythmias in a postinfarction model of SCD. For our methods, we used a postinfarction canine model of SCD, where an exercise and ischemia test was used to stratify animals as either susceptible (VF+) or resistant (VF−) to sustained ventricular tachyarrhythmias. Our results show no changes in global left ventricular contractility or volumes occurred after infarction. At 8–10 wk postmyocardial infarction, myocytes were isolated from the left ventricular midmyocardial wall and studied. In the VF+ animals, myocyte action potential (AP) prolongation occurred at 50 and 90% repolarization ( P < 0.05) and was associated with increased variability of AP duration and afterdepolarizations. Multiple repolarizing K+ currents ( IKr, Ito) and inward IK1 were also reduced ( P < 0.05) in myocytes from VF+ animals compared with control, noninfarcted dogs. In contrast, only Ito was reduced in VF− myocytes compared with controls ( P < 0.05). While afterdepolarizations were not elicited at baseline in myocytes from VF− animals, afterdepolarizations were consistently elicited after the addition of an IKr blocker. In conclusion, the loss of repolarization reserve via reductions in multiple repolarizing currents in the VF+ myocytes leads to AP prolongation, repolarization instability, and afterdepolarizations in myocytes from animals susceptible to SCD. These abnormalities may provide a substrate for initiation of postmyocardial infarction ventricular tachyarrhythmias.
- Published
- 2008
42. Properties of Atrial Myocyte Calcium Handling in Canine Model of Chronic Heart Failure
- Author
-
Karsten E. Schober, Kent Mowrey, Ingrid M. Bonilla, Sandor Gyorke, Raul Weiss, Lucia Brunello, Andriy E. Belevych, Cynthia A. Carnes, Qing Lou, and Hsiang-Ting Ho
- Subjects
0303 health sciences ,medicine.medical_specialty ,Sarcolemma ,SERCA ,Endoplasmic reticulum ,fungi ,Biophysics ,010402 general chemistry ,medicine.disease ,01 natural sciences ,Calcium in biology ,0104 chemical sciences ,3. Good health ,Calcium ATPase ,03 medical and health sciences ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Heart failure ,Internal medicine ,medicine ,Myocyte ,Cyclopiazonic acid ,030304 developmental biology - Abstract
Weakened atrial contractility is a common comorbidity in heart failure (HF) that increases severity of HF symptoms. The role of intracellular calcium (Ca) handling remodeling in pathogenesis of ventricular contractile failure has been well established. However, the relationship between Ca signaling and atrial dysfunction in HF is not well recognized. We studied intracellular Ca handling in atrial myocytes isolated from control dogs and dogs with tachypacing-induced chronic HF. Ca transients and cellular shortening were reduced, while sarcoplasmic reticulum (SR) Ca content was increased in field-stimulated myocytes from HF compared to controls. Both control and HF myocytes have scarce t-tubule density, and Ca transients originated predominantly at the periphery of myocytes in both groups. Diminished Ca transients in HF myocytes were associated with decreased peripheral Ca release and impaired propagation of Ca signal from sarcolemma to the central areas of the myocyte. Peak density of the L-type Ca currents and gain of excitation-contraction coupling were reduced in voltage-clamped HF myocytes compared to control myocytes. Analysis of decay of caffeine- and electrically-induced Ca transients suggests reduced Na/Ca exchanger activity and increased SR Ca ATPase (SERCA) activity in HF myocytes. SERCA inhibition with cyclopiazonic acid did not improve centripetal propagation of Ca signal in HF myocytes. In contrast, L-type Ca channel agonist BayK 8644 increased peripheral Ca release, improved centripetal Ca wave propagation and restored shortening of HF myocytes to control levels.We conclude that defective activation and impaired propagation of the SR Ca release in atrial myocytes contribute to compromised atrial contractility in chronic HF. Our results suggest L-type Ca channels as a therapeutic target for treatment of atrial contractile dysfunction in HF.
- Published
- 2015
- Full Text
- View/download PDF
43. Atrial Electrophysiologic Remodeling in Canine Heart Failure: Modulation by Atrial Fibrillation
- Author
-
Ingrid M. Bonilla, Raul Weiss, Victor P. Long, P. Vargas Pinto, J. Green, Yoshinori Nishijima, K. Mowrey, Vadim V. Fedorov, and Cynthia A. Carnes
- Subjects
medicine.medical_specialty ,business.industry ,Atrial fibrillation ,medicine.disease ,Canine heart ,Left atrial ,Physiology (medical) ,Internal medicine ,Anesthesia ,Heart failure ,cardiovascular system ,medicine ,Cardiology ,Repolarization ,Myocyte ,Cardiology and Cardiovascular Medicine ,business ,Atrial flutter - Abstract
Background Heart failure (HF) is a common antecedent to atrial fibrillation (AF). Concomitant HF and AF increases morbidity and mortality over that of either disease alone. AF- and HF-induced atrial electrophysiologic remodeling is known to favor the progression of AF. AF results in accelerated repolarization and reductions in multiple ion currents, including transient outward K + current (I to ), changes associated with AF maintenance. Methods Dogs had tachypacing-induced HF for 10 weeks and then were assigned to 6 weeks of atrial tachypacing (60 seconds of pacing at 10 Hz, repeated every 70 seconds) until sustained AF/atrial flutter was induced (HF+AF, n=3), or continued right ventricular tachypacing (n = 6). Baseline LVFS was reduced ( P to were made at 35° ± 0.5°C. Results HF significantly increased I to vs control, whereas HF+AF significantly reduced I to vs control and HF (Figure A). At 2 Hz, HF shortened action potential durations at 50% repolarization (APD 50 ); HF+AF lengthened APD 50 vs HF alone, and did not differ from control (Figure B). HF shortened APD at 90% repolarization (APD 90 ) vs controls, whereas HF+AF had similar shortening of APD 90 . Conclusions Chronic HF results in increased I to and faster repolarization of left atrial appendage myocytes. The addition of AF to chronic HF reduced I to and lengthened APD 50 without changing APD 90 . The effects of chronic HF and chronic HF+AF on I to and APD morphology are variable, suggesting complex disease-induced effects on the substrate for AF.
- Published
- 2012
- Full Text
- View/download PDF
44. Ibandronate and Ventricular Arrhythmia Risk
- Author
-
Mahmoud Houmsse, Pedro Vargas-Pinto, Hsiang-Ting Ho, Andriy E. Belevych, Robert L. Hamlin, J. Smith, Ingrid M. Bonilla, Sandor Gyorke, Raul Weiss, Yoshinori Nishijima, Victor P. Long, Cynthia A. Carnes, Troy E. Rhodes, and Adriana Pedraza-Toscano
- Subjects
Proarrhythmia ,medicine.medical_specialty ,business.industry ,Ryanodine receptor ,medicine.disease ,QT interval ,Hypokalemia ,Afterdepolarization ,Endocrinology ,Physiology (medical) ,Internal medicine ,cardiovascular system ,Myocyte ,Medicine ,Repolarization ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Perfusion - Abstract
Background Ibandronate is given monthly or quarterly to postmenopausal women to treat or reduce osteoporosis. We report a case of cardiac arrest in an otherwise healthy 55-year-old female, 2 weeks after her first dose of ibandronate, with associated hypokalemia and QTc prolongation (575 ms). The QT/QTc remained prolonged after correction of hypokalemia but returned toward normal after drug discontinuation. We examined potential proarrhythmic mechanisms of ibandronate. Methods A 3-mg injection was given to 4 dogs; ECG was monitored for 6 weeks by telemetry. Adult canine ventricular myocytes were used to study action potentials (0.5, 1, and 2 Hz), K + currents, and Ca 2+ handling during perfusion with ibandronate (0.001–1000 μg/L), and washout. Results Three of 4 dogs exhibited increased short-term variability in repolarization starting 4+ weeks post dose. In myocytes, ibandronate caused reverse use–dependent prolongation of the action potential (APD 50 and APD 90 ) as well as increased beat-to-beat variability in APD 90 ( P to conductance ∼30% ( P Kr or I Ks was observed. Buffering Ca 2+ with BAPTA prevented ibandronate-induced APD 90 instability and afterdepolarizations. In separate experiments, ibandronate increased sarcoplasmic reticulum Ca 2+ load ( P 2+ sparks and waves were increased ( P Conclusions Ibandronate alters myocyte Ca 2+ regulation (reversible inhibition of ryanodine receptor that is lost upon washout) and ventricular myocyte electrophysiology to induce cellular arrhythmias; in vivo data are consistent with late onset of torsadogenic repolarization instability. We conclude that current paradigms to detect proarrhythmia risk may not be sufficiently sensitive.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.