2,705 research outputs found

    Altered excitation-contraction coupling in human chronic atrial fibrillation

    Get PDF
    This review focuses on the (mal)adaptive processes in atrial excitation-contraction coupling occurring in patients with chronic atrial fibrillation. Cellular remodeling includes shortening of the atrial action potential duration and effective refractory period, depressed intracellular Ca<sup>2+</sup> transient, and reduced myocyte contractility. Here we summarize the current knowledge of the ionic bases underlying these changes. Understanding the molecular mechanisms of excitation-contraction-coupling remodeling in the fibrillating human atria is important to identify new potential targets for AF therapy

    Constitutively active acetylcholine-dependent potassium current increases atrial defibrillation threshold by favoring post-shock re-initiation

    Get PDF
    Electrical cardioversion (ECV), a mainstay in atrial fibrillation (AF) treatment, is unsuccessful in up to 10-20% of patients. An important aspect of the remodeling process caused by AF is the constitutive activition of the atrium-specific acetylcholine-dependent potassium current (I-K,I-ACh -> I-K,I-ACh-c), which is associated with ECV failure. This study investigated the role of I-K,I-ACh-c in ECV failure and setting the atrial defibrillation threshold (aDFT) in optically mapped neonatal rat cardiomyocyte monolayers. AF was induced by burst pacing followed by application of biphasic shocks of 25-100 V to determine aDFT. Blocking I-K,I-ACh-c by tertiapin significantly decreased DFT, which correlated with a significant increase in wavelength during reentry. Genetic knockdown experiments, using lentiviral vectors encoding a Kcnj5-specific shRNA to modulate I-K,I-ACh-c, yielded similar results. Mechanistically, failed ECV was attributed to incomplete phase singularity (PS) removal or reemergence of PSs (i.e. re-initiation) through unidirectional propagation of shock-induced action potentials. Re-initiation occurred at significantly higher voltages than incomplete PS-removal and was inhibited by I-K,I-ACh-c blockade. Whole-heart mapping confirmed our findings showing a 60% increase in ECV success rate after I-K,I-ACh-c blockade. This study provides new mechanistic insight into failing ECV of AF and identifies I-K,I-ACh-c as possible atrium-specific target to increase ECV effectiveness, while decreasing its harmfulness

    The role of inhibitory G proteins and regulators of G protein signaling in the in vivo control of heart rate and predisposition to cardiac arrhythmias

    Get PDF
    Inhibitory heterotrimeric G proteins and the control of heart rate. The activation of cell signaling pathways involving inhibitory heterotrimeric G proteins acts to slow the heart rate via modulation of ion channels. A large number of Regulators of G protein signalings (RGSs) can act as GTPase accelerating proteins to inhibitory G proteins and thus it is important to understand the network of RGS\G-protein interaction. We will review our recent findings on in vivo heart rate control in mice with global genetic deletion of various inhibitory G protein alpha subunits. We will discuss potential central and peripheral contributions to the phenotype and the controversies in the literature

    Perspective: a dynamics-based classification of ventricular arrhythmias

    Get PDF
    Despite key advances in the clinical management of life-threatening ventricular arrhythmias, culminating with the development of implantable cardioverter-defibrillators and catheter ablation techniques, pharmacologic/biologic therapeutics have lagged behind. The fundamental issue is that biological targets are molecular factors. Diseases, however, represent emergent properties at the scale of the organism that result from dynamic interactions between multiple constantly changing molecular factors. For a pharmacologic/biologic therapy to be effective, it must target the dynamic processes that underlie the disease. Here we propose a classification of ventricular arrhythmias that is based on our current understanding of the dynamics occurring at the subcellular, cellular, tissue and organism scales, which cause arrhythmias by simultaneously generating arrhythmia triggers and exacerbating tissue vulnerability. The goal is to create a framework that systematically links these key dynamic factors together with fixed factors (structural and electrophysiological heterogeneity) synergistically promoting electrical dispersion and increased arrhythmia risk to molecular factors that can serve as biological targets. We classify ventricular arrhythmias into three primary dynamic categories related generally to unstable Ca cycling, reduced repolarization, and excess repolarization, respectively. The clinical syndromes, arrhythmia mechanisms, dynamic factors and what is known about their molecular counterparts are discussed. Based on this framework, we propose a computational-experimental strategy for exploring the links between molecular factors, fixed factors and dynamic factors that underlie life-threatening ventricular arrhythmias. The ultimate objective is to facilitate drug development by creating an in silico platform to evaluate and predict comprehensively how molecular interventions affect not only a single targeted arrhythmia, but all primary arrhythmia dynamics categories as well as normal cardiac excitation-contraction coupling

    Applying computational approaches to the understanding of the consequences and opportunities of ion channel properties in atrial fibrillation

    Full text link
    Cardiac arrhythmias are disorders of the electrical system of the heart and an often clinically-challenging group of disorders. Atrial fibrillation (AF) is the most common cardiac arrhythmia in the general population; it is associated with significant morbidity and mortality. Available antiarrhythmic drugs (AADs) for the treatment of AF are older molecules with sub- optimal efficacy and safety profiles. Recent advances in basic electrophysiology and the development of sophisticated mathematical modeling approaches could help in expanding our understanding of the basic mechanisms of AF and assist in the development of novel AF- selective AADs. The purpose of this thesis was to utilize computational approaches to the understanding of the consequences and opportunities of ion channel properties, with a special emphasis on AF. The cardiac action potential is the basic functional unit of the electrical system of the heart and is the manifestation of coordinated current fluxes through specialized proteins known as ion channels. Antiarrhythmic drugs act through modulation of ion channel properties. We hypothesized that mathematical modeling could be used to study and optimize the pharmacodynamic properties of AADs for the treatment of AF. We demonstrated that the pharmacodynamic properties (binding/unbinding characteristics) of a state-dependent Na+- channel blocker modulate the drug’s anti-/proarrhythmic actions with inactivated-state blockers being optimally AF-selective. The optimized drug’s selectivity for AF was the result of its rate- selectivity (stronger effects at fast vs slow cardiomyocyte activation rates) with relatively mild atrial-selective (stronger effects in atrial vs ventricular cardiomyocytes) actions. We found that the optimally AF-selective Na+-channel blocker had sub-optimal anti-AF efficacy, but that slightly less selective drugs had favorable AF-termination rates. We then sought to explore potential current-block combinations with synergistic AF- selective properties. Using mathematical modeling and laboratory experiments, we demonstrated that the combination of optimized state-dependent Na+-channel block and K+- channel block had synergistic effects, significantly augmenting AF termination rates for any level of AF-selectivity vs pure Na+-channel block. The mechanisms of these synergistic effects were found to be mediated by the functional interaction between the action potential prolonging- v effects of K+-channel block, the Na+-channel blocker’s voltage-dependent binding/unbinding properties and the Na+ channel’s inactivation characteristics, highlighting the non-linear nature of the cardiac action potential’s dynamics. Traditional K+ currents targeted by AADs have significant ventricular proarrhythmic liabilities. Using recent experimental observations, we updated the mathematic formulation for the inactivation dynamics of the ultra-rapid delayed-rectifier K+ current (IKur), an atrial-specific current. Using this model, we showed that, contrary to what had been proposed in the published literature, IKur rate-dependent properties are mediated by its activation properties with minimal contribution from inactivation, under physiological conditions. We also demonstrated that the contribution of IKur to action potential repolarization is preserved, or even increased, in the setting of electrical remodeling-induced IKur downregulation. Finally, we described the mechanisms of the forward rate-dependent of IKur block, mediated by functional non-linear interactions with the rapid delayed inward-rectifier K+ current (IKr), the only K+ current with such properties. Until recently, fibroblasts were considered to be electrically inactive. More recently, experimental work demonstrated the presence of functional ionic current on the fibroblast and possible cardiomyocyte-fibroblast coupling. Here, we described a novel kind of heart failure- induced electrical remodeling involving the fibroblasts ion channels. This was characterized by downregulation of the fibroblast voltage-dependent K+ current (IKv,fb) and upregulation of the fibroblast inward-rectifier K+ current (IKir,fb). We then implemented our experimental findings into a mathematical model of cardiomyocyte-fibroblast coupling and found fibroblast electrical remodeling to have significant effects on the cardiomyocyte’s electrophysiological properties. In a 2-dimension model of simulated AF, downregulation of IKv,fb had an antiarrhythmic effect whereas IKir,fb upregulation was found to be proarrhythmic. The studies presented here utilized mathematical modeling to study non-linear systems in cardiac electrophysiology to tackle questions that would have been difficult to approach with traditional laboratory-based experimentation. They also showcased how theoretical results can help orient and receive confirmation with subsequent experimental work or, conversely, novel experimental findings results be implemented into a mathematical model to investigate potential consequences. Mathematical modeling is a promising tool to help in studying the complex and vi non-linear effects of pharmacological modulation of ion channel properties and assist in the development of optimized antiarrhythmics for the treatment of AF, a major unmet need in clinical medicine. As models increase in sophistication to better represent the cardiomyocyte’s electrophysiology, they will almost certainly play an ever-growing role in expanding our understanding of the mechanisms of complex arrhythmias.Les arythmies cardiaques représentent une famille de pathologies du système électrique cardiaque. La fibrillation auriculaire (FA), est l’arythmie cardiaque la plus fréquente dans la population générale et est associée à un fardeau de morbidité et mortalité cardiovasculaire important. Les médicaments antiarythmiques utilisées dans le traitement de la FA sont de vieilles molécules avec une efficacité sous-optimale et des effets secondaires importants. Les avancées récentes en électrophysiologie cardiaque fondamentale et le développement d’outils de modélisation mathématique ont le potentiel d’élargir notre compréhension des mécanismes pathophysiologiques en FA et contribuer au développement de nouveaux médicaments antiarythmiques optimisés pour le traitement de la FA. L’objectif global de cette thèse est d’utiliser les méthodes de modélisation mathématique pour étudier les conséquences et opportunités thérapeutiques de la modulation des canaux ioniques cardiaques, avec une emphase sur la FA. Le potentiel d’action cardiaque est l’unité fonctionnelle de base du système électrique cardiaque ; il est le résultat du flux coordonné de courants électriques à travers de protéines spécialisées, les canaux ioniques. Les molécules antiarythmiques agissent à travers la modulation des canaux ioniques cardiaques. Nous avons posé l’hypothèse que des modèles mathématiques pourraient être utilisés pour étudier et optimiser les propriétés pharmacodynamiques d’un médicament antiarythmique pour le traitement de la FA. Nous avons démontré que les propriétés pharmacodynamiques (propriétés de liage et déliage) d’un bloqueur des canaux Na+ état-dépendant modulent les effets anti- et pro-arythmiques de la molécule ; un bloqueur Na+ sélectif pour l’état inactivé du canal serait maximalement FA-sélectif. Cette sélectivité pour la FA est la conséquence de la sélectivité pour la fréquence (effet thérapeutique plus important à des fréquences d’activation du cardiomyocyte élevées vs basses) avec une contribution relativement faible de la sélectivité auriculaire (effet thérapeutique plus important sur les cardiomyocytes auriculaires vs ventriculaires). Par la suite, nous avons exploré des combinaisons de bloqueurs ioniques ayant des propriétés anti-FA synergiques. En utilisant des modèles mathématiques et des expériences en laboratoire, nous avons démontré que la combinaison d’un bloqueur des canaux Na+ et d’un iii bloqueur des canaux K+ a des effets synergiques, augmentant de façon importante l’efficacité anti-FA pour un même degré de sélectivité vs un bloqueur des canaux Na+ seul. Le mécanisme de synergie a été élucidé et consiste d’effets fonctionnels médiés par l’interaction du prolongement de la durée du potentiel d’action causé par le bloque des canaux K+, les propriétés voltage-dépendantes du liage et déliage du bloqueur des canaux Na+ ainsi que des propriétés d’inactivation des canaux Na+, démontrant la nature hautement non-linéaire des dynamiques du potentiel d’action cardiaque. Les courants K+ ciblés par les médicaments antiarythmiques ont des effets proarythmiques ventriculaires importants. En utilisant des données expérimentales récentes, nous avons proposé une formulation mise à jour des dynamiques d’inactivation du courant K+ IKur, un courant auriculo-sélectif. En utilisant ce modèle, nous avons démontré que, contrairement à ce qui avait été précédemment proposé, les propriétés fréquence-dépendantes du courant IKur dépendent de ses caractéristiques d’activation avec une contribution négligeable de ses propriétés d’inactivation, sous conditions physiologiques normales. Nous avons également démontré que la contribution de IKur à la repolarisation du potentiel d’action est maintenue, voir augmentée, dans le contexte de la diminution de IKur en situation de remodelage électrique induit par la FA. Finalement, nous avons décrit le mécanisme qui sous-tend les propriétés fréquence-dépendantes du bloque de IKur, l’unique courant K+ avec de telles caractéristiques. Jusqu’à très récemment, les fibroblastes cardiaques étaient considérés comme électriquement inactifs. Des travaux expérimentaux ont démontré la présence de canaux ioniques sur la surface de ces fibroblastes ainsi que la possibilité de couplage électrique entre cardiomyocytes et fibroblastes. Nous avons décrit un nouveau type de remodelage électrique en situation d’insuffisance cardiaque, le remodelage des courants ioniques des fibroblastes cardiaques. Ce remodelage est caractérisé par une diminution du courant K+ voltage-dépendant IKv,fb et une augmentation du courant K+ IKir,fb. Nous avons par la suite incorporé ces trouvailles expérimentales dans un modèle mathématique simulant l’interaction électrique entre cardiomyocytes et fibroblastes et montré que le remodelage électrique des fibroblastes peut avoir un impact important sur les propriétés électrophysiologiques des cardiomyocytes. Dans iv un modèle 2-dimensionel de FA, nous avons trouvé que la diminution de IKv,fb a un effet antiarythmique alors que l’augmentation de IKir,fb a des effets proarythmiques. Les études ici présentées utilisent les méthodes de modélisation mathématique pour l’étude de systèmes non-linéaires en électrophysiologie cardiaque et aborder des avenues de recherche difficilement accessibles aux méthodes de laboratoire traditionnelles. Elles démontrent également comment des résultats théoriques peuvent orienter et trouver confirmation dans des travaux expérimentaux subséquents ou, à l’inverse, des trouvailles expérimentales peuvent être implémentées dans les modèles mathématiques pour investiguer les conséquences de celles-ci. La modélisation mathématique est un outil prometteur pour l’étude des effets complexes et non-linéaires de la modulation pharmacologique des canaux ioniques et ainsi contribuer au développement de médicaments antiarythmiques optimisés pour le traitement de la FA, un besoin clinique majeur

    Na/K pump regulation of cardiac repolarization: Insights from a systems biology approach

    Get PDF
    The sodium-potassium pump is widely recognized as the principal mechanism for active ion transport across the cellular membrane of cardiac tissue, being responsible for the creation and maintenance of the transarcolemmal sodium and potassium gradients, crucial for cardiac cell electrophysiology. Importantly, sodium-potassium pump activity is impaired in a number of major diseased conditions, including ischemia and heart failure. However, its subtle ways of action on cardiac electrophysiology, both directly through its electrogenic nature and indirectly via the regulation of cell homeostasis, make it hard to predict the electrophysiological consequences of reduced sodium-potassium pump activity in cardiac repolarization. In this review, we discuss how recent studies adopting the Systems Biology approach, through the integration of experimental and modeling methodologies, have identified the sodium-potassium pump as one of the most\ud important ionic mechanisms in regulating key properties of cardiac repolarization and its rate-dependence, from subcellular to whole organ levels. These include the role of the pump in the biphasic modulation of cellular repolarization and refractoriness, the rate control of intracellular sodium and calcium dynamics and therefore of the adaptation of repolarization to changes in heart rate, as well as its importance in regulating pro-arrhythmic substrates through modulation of dispersion of repolarization and restitution. Theoretical findings are consistent across a variety of cell types and species including human, and widely in agreement with experimental findings. The novel insights and hypotheses on the role of the pump in cardiac electrophysiology obtained through this integrative approach could eventually lead to novel therapeutic and diagnostic strategies

    Differential Modulation of I-K and I-Ca,I-L Channels in High-Fat Diet-Induced Obese Guinea Pig Atria

    Full text link
    [EN] Obesity mechanisms that make atrial tissue vulnerable to arrhythmia are poorly understood. Voltage-dependent potassium (I-K, I-Kur, and I-K1) and L-type calcium currents (I-Ca,I- L) are electrically relevant and represent key substrates for modulation in obesity. We investigated whether electrical remodeling produced by high-fat diet (HFD) alone or in concert with acute atrial stimulation were different. Electrophysiology was used to assess atrial electrical function after short-term HFD-feeding in guinea pigs. HFD atria displayed spontaneous beats, increased I-K (I-Kr + I-Ks) and decreased I-Ca,I- L densities. Only with pacing did a reduction in I-Kur and increased I-K1 phenotype emerge, leading to a further shortening of action potential duration. Computer modeling studies further indicate that the measured changes in potassium and calcium current densities contribute prominently to shortened atrial action potential duration in human heart. Our data are the first to show that multiple mechanisms (shortened action potential duration, early after depolarizations and increased incidence of spontaneous beats) may underlie initiation of supraventricular arrhythmias in obese guinea pig hearts. These results offer different mechanistic insights with implications for obese patients harboring supraventricular arrhythmias.This study was supported by an AHA (13SDG16850065 to AA), NIH (R01 HL147044 to AA), and Programa Prometeu de la Conselleria d Educació, Formació I Ocupació de la Generalitat Valenciana, award number PROMETEU/2016/088.Martínez-Mateu, L.; Saiz Rodríguez, FJ.; Aromolaran, A. (2019). Differential Modulation of I-K and I-Ca,I-L Channels in High-Fat Diet-Induced Obese Guinea Pig Atria. Frontiers in Physiology. 10:1-18. https://doi.org/10.3389/fphys.2019.01212S11810Abed, H. S., & Wittert, G. A. (2013). Obesity and atrial fibrillation. Obesity Reviews, 14(11), 929-938. doi:10.1111/obr.12056Angelin, B., Olivecrona, H., Reihnér, E., Rudling, M., Ståhlberg, D., Eriksson, M., … Einarsson, K. (1992). Hepatic cholesterol metabolism in estrogen-treated men. Gastroenterology, 103(5), 1657-1663. doi:10.1016/0016-5085(92)91192-7Aoki, Y., Hatakeyama, N., Yamamoto, S., Kinoshita, H., Matsuda, N., Hattori, Y., & Yamazaki, M. (2012). Role of ion channels in sepsis-induced atrial tachyarrhythmias in guinea pigs. British Journal of Pharmacology, 166(1), 390-400. doi:10.1111/j.1476-5381.2011.01769.xAromolaran, A. S., & Boutjdir, M. (2017). Cardiac Ion Channel Regulation in Obesity and the Metabolic Syndrome: Relevance to Long QT Syndrome and Atrial Fibrillation. Frontiers in Physiology, 8. doi:10.3389/fphys.2017.00431Aromolaran, A. S., Colecraft, H. M., & Boutjdir, M. (2016). High-fat diet-dependent modulation of the delayed rectifier K + current in adult guinea pig atrial myocytes. Biochemical and Biophysical Research Communications, 474(3), 554-559. doi:10.1016/j.bbrc.2016.04.113Aromolaran, A. S., Subramanyam, P., Chang, D. D., Kobertz, W. R., & Colecraft, H. M. (2014). LQT1 mutations in KCNQ1 C-terminus assembly domain suppress IKs using different mechanisms. Cardiovascular Research, 104(3), 501-511. doi:10.1093/cvr/cvu231Ashrafi, R., Yon, M., Pickavance, L., Yanni Gerges, J., Davis, G., Wilding, J., … Boyett, M. (2016). Altered Left Ventricular Ion Channel Transcriptome in a High-Fat-Fed Rat Model of Obesity: Insight into Obesity-Induced Arrhythmogenesis. Journal of Obesity, 2016, 1-12. doi:10.1155/2016/7127898Bai, J., Gladding, P. A., Stiles, M. K., Fedorov, V. V., & Zhao, J. (2018). Ionic and cellular mechanisms underlying TBX5/PITX2 insufficiency-induced atrial fibrillation: Insights from mathematical models of human atrial cells. Scientific Reports, 8(1). doi:10.1038/s41598-018-33958-yBarana, A., Matamoros, M., Dolz-Gaitón, P., Pérez-Hernández, M., Amorós, I., Núñez, M., … Caballero, R. (2014). Chronic Atrial Fibrillation Increases MicroRNA-21 in Human Atrial Myocytes Decreasing L-Type Calcium Current. Circulation: Arrhythmia and Electrophysiology, 7(5), 861-868. doi:10.1161/circep.114.001709Bar�, I., & Escande, D. (1989). A Ca2+-activated K+ current in guinea-pig atrial myocytes. Pfl�gers Archiv European Journal of Physiology, 414(S1), S168-S168. doi:10.1007/bf00582286Bar�, I., & Escande, D. (1989). A long lasting Ca2+ -activated outward current in guinea-pig atrial myocytes. Pfl�gers Archiv European Journal of Physiology, 415(1), 63-71. doi:10.1007/bf00373142Bhuyan, R., & Seal, A. (2016). Dynamics and modulation studies of human voltage gated Kv1.5 channel. Journal of Biomolecular Structure and Dynamics, 35(2), 380-398. doi:10.1080/07391102.2016.1144528Boden, G., She, P., Mozzoli, M., Cheung, P., Gumireddy, K., Reddy, P., … Ruderman, N. (2005). Free Fatty Acids Produce Insulin Resistance and Activate the Proinflammatory Nuclear Factor- B Pathway in Rat Liver. Diabetes, 54(12), 3458-3465. doi:10.2337/diabetes.54.12.3458Bosch, R. F., Schneck, A. C., Csillag, S., Eigenberger, B., Gerlach, U., Brendel, J., … Kühlkamp, V. (2003). Effects of the chromanol HMR 1556 on potassium currents in atrial myocytes. Naunyn-Schmiedeberg’s Archives of Pharmacology, 367(3), 281-288. doi:10.1007/s00210-002-0672-5BOUTJDIR, M., HEUZEY, J. Y., LAVERGNE, T., CHAUVAUD, S., GUIZE, L., CARPENTIER, A., & PERONNEAU, P. (1986). Inhomogeneity of Cellular Refractoriness in Human Atrium: Factor of Arrhythmia?. Pacing and Clinical Electrophysiology, 9(6), 1095-1100. doi:10.1111/j.1540-8159.1986.tb06676.xBrundel, B. J. J. M., Van Gelder, I. C., Henning, R. H., Tieleman, R. G., Tuinenburg, A. E., Wietses, M., … Crijns, H. J. G. M. (2001). Ion Channel Remodeling Is Related to Intraoperative Atrial Effective Refractory Periods in Patients With Paroxysmal and Persistent Atrial Fibrillation. Circulation, 103(5), 684-690. doi:10.1161/01.cir.103.5.684Bünemann, M., Liliom, K., Brandts, B. K., Pott, L., Tseng, J. L., Desiderio, D. M., … Tigyi, G. (1996). A novel membrane receptor with high affinity for lysosphingomyelin and sphingosine 1-phosphate in atrial myocytes. The EMBO Journal, 15(20), 5527-5534. doi:10.1002/j.1460-2075.1996.tb00937.xCaballero, R., de la Fuente, M. G., Gómez, R., Barana, A., Amorós, I., Dolz-Gaitón, P., … Delpón, E. (2010). In Humans, Chronic Atrial Fibrillation Decreases the Transient Outward Current and Ultrarapid Component of the Delayed Rectifier Current Differentially on Each Atria and Increases the Slow Component of the Delayed Rectifier Current in Both. Journal of the American College of Cardiology, 55(21), 2346-2354. doi:10.1016/j.jacc.2010.02.028Caillier, B., Pilote, S., Patoine, D., Levac, X., Couture, C., Daleau, P., … Drolet, B. (2012). Metabolic syndrome potentiates the cardiac action potential-prolonging action of drugs: A possible ‘anti-proarrhythmic’ role for amlodipine. Pharmacological Research, 65(3), 320-327. doi:10.1016/j.phrs.2011.11.015Chiu, H.-C., Kovacs, A., Ford, D. A., Hsu, F.-F., Garcia, R., Herrero, P., … Schaffer, J. E. (2001). A novel mouse model of lipotoxic cardiomyopathy. Journal of Clinical Investigation, 107(7), 813-822. doi:10.1172/jci10947Christ, T., Boknik, P., Wöhrl, S., Wettwer, E., Graf, E. M., Bosch, R. F., … Dobrev, D. (2004). L-Type Ca2+Current Downregulation in Chronic Human Atrial Fibrillation Is Associated With Increased Activity of Protein Phosphatases. Circulation, 110(17), 2651-2657. doi:10.1161/01.cir.0000145659.80212.6aCourtemanche, M., Ramirez, R. J., & Nattel, S. (1998). Ionic mechanisms underlying human atrial action potential properties: insights from a mathematical model. American Journal of Physiology-Heart and Circulatory Physiology, 275(1), H301-H321. doi:10.1152/ajpheart.1998.275.1.h301Czick, M. E., Shapter, C. L., & Silverman, D. I. (2016). Atrial Fibrillation: The Science behind Its Defiance. Aging and Disease, 7(5), 635. doi:10.14336/ad.2016.0211Dan, G.-A., & Dobrev, D. (2018). Antiarrhythmic drugs for atrial fibrillation: Imminent impulses are emerging. IJC Heart & Vasculature, 21, 11-15. doi:10.1016/j.ijcha.2018.08.005Daoud, E. G., Knight, B. P., Weiss, R., Bahu, M., Paladino, W., Goyal, R., … Morady, F. (1997). Effect of Verapamil and Procainamide on Atrial Fibrillation–Induced Electrical Remodeling in Humans. Circulation, 96(5), 1542-1550. doi:10.1161/01.cir.96.5.1542De Sensi, F., Costantino, S., Limbruno, U., & Paneni, F. (2019). Atrial fibrillation in the cardiometabolic patient. Minerva Medica, 110(2). doi:10.23736/s0026-4806.18.05882-2Dey, S., DeMazumder, D., Sidor, A., Foster, D. B., & O’Rourke, B. (2018). Mitochondrial ROS Drive Sudden Cardiac Death and Chronic Proteome Remodeling in Heart Failure. Circulation Research, 123(3), 356-371. doi:10.1161/circresaha.118.312708Diness, J. G., Sørensen, U. S., Nissen, J. D., Al-Shahib, B., Jespersen, T., Grunnet, M., & Hansen, R. S. (2010). Inhibition of Small-Conductance Ca 2+ -Activated K + Channels Terminates and Protects Against Atrial Fibrillation. Circulation: Arrhythmia and Electrophysiology, 3(4), 380-390. doi:10.1161/circep.110.957407Djoussé, L., Benkeser, D., Arnold, A., Kizer, J. R., Zieman, S. J., Lemaitre, R. N., … Ix, J. H. (2013). Plasma Free Fatty Acids and Risk of Heart Failure. Circulation: Heart Failure, 6(5), 964-969. doi:10.1161/circheartfailure.113.000521Ehrlich, J. R., Ocholla, H., Ziemek, D., Rütten, H., Hohnloser, S. H., & Gögelein, H. (2008). Characterization of Human Cardiac Kv1.5 Inhibition by the Novel Atrial-selective Antiarrhythmic Compound AVE1231. Journal of Cardiovascular Pharmacology, 51(4), 380-387. doi:10.1097/fjc.0b013e3181669030Feng, J., Yue, L., Wang, Z., & Nattel, S. (1998). Ionic Mechanisms of Regional Action Potential Heterogeneity in the Canine Right Atrium. Circulation Research, 83(5), 541-551. doi:10.1161/01.res.83.5.541Fernandez, M. L., Conde, A. K., Ruiz, L. R., Montano, C., Ebner, J., & McNamara, D. J. (1995). Carbohydrate type and amount alter intravascular processing and catabolism of plasma lipoproteins in guinea pigs. Lipids, 30(7), 619-626. doi:10.1007/bf02536998Fretts, A. M., Mozaffarian, D., Siscovick, D. S., Djousse, L., Heckbert, S. R., King, I. B., … Lemaitre, R. N. (2014). Plasma Phospholipid Saturated Fatty Acids and Incident Atrial Fibrillation: The Cardiovascular Health Study. Journal of the American Heart Association, 3(3). doi:10.1161/jaha.114.000889Gaborit, N., Steenman, M., Lamirault, G., Le Meur, N., Le Bouter, S., Lande, G., … Demolombe, S. (2005). Human Atrial Ion Channel and Transporter Subunit Gene-Expression Remodeling Associated With Valvular Heart Disease and Atrial Fibrillation. Circulation, 112(4), 471-481. doi:10.1161/circulationaha.104.506857Garnvik, L. E., Malmo, V., Janszky, I., Wisløff, U., Loennechen, J. P., & Nes, B. M. (2018). Physical activity modifies the risk of atrial fibrillation in obese individuals: The HUNT3 study. European Journal of Preventive Cardiology, 25(15), 1646-1652. doi:10.1177/2047487318784365Gaspo, R., Bosch, R. F., Talajic, M., & Nattel, S. (1997). Functional Mechanisms Underlying Tachycardia-Induced Sustained Atrial Fibrillation in a Chronic Dog Model. Circulation, 96(11), 4027-4035. doi:10.1161/01.cir.96.11.4027Goette, A., Honeycutt, C., & Langberg, J. J. (1996). Electrical Remodeling in Atrial Fibrillation. Circulation, 94(11), 2968-2974. doi:10.1161/01.cir.94.11.2968González de la Fuente, M., Barana, A., Gómez, R., Amorós, I., Dolz-Gaitón, P., Sacristán, S., … Delpón, E. (2012). Chronic atrial fibrillation up-regulates β1-Adrenoceptors affecting repolarizing currents and action potential duration. Cardiovascular Research, 97(2), 379-388. doi:10.1093/cvr/cvs313Grandi, E., Dobrev, D., & Heijman, J. (2019). Computational modeling: What does it tell us about atrial fibrillation therapy? International Journal of Cardiology, 287, 155-161. doi:10.1016/j.ijcard.2019.01.077Grandi, E., Pandit, S. V., Voigt, N., Workman, A. J., Dobrev, D., Jalife, J., & Bers, D. M. (2011). Human Atrial Action Potential and Ca2+Model. Circulation Research, 109(9), 1055-1066. doi:10.1161/circresaha.111.253955HADIAN, D., ZIPES, D. P., OLGIN, J. E., & MILLER, J. M. (2002). Short-Term Rapid Atrial Pacing Produces Electrical Remodeling of Sinus Node Function in Humans. Journal of Cardiovascular Electrophysiology, 13(6), 584-586. doi:10.1046/j.1540-8167.2002.00584.xHeijman, J., Guichard, J.-B., Dobrev, D., & Nattel, S. (2018). Translational Challenges in Atrial Fibrillation. Circulation Research, 122(5), 752-773. doi:10.1161/circresaha.117.311081Huang, H., Amin, V., Gurin, M., Wan, E., Thorp, E., Homma, S., & Morrow, J. P. (2013). Diet-induced obesity causes long QT and reduces transcription of voltage-gated potassium channels. Journal of Molecular and Cellular Cardiology, 59, 151-158. doi:10.1016/j.yjmcc.2013.03.007Hume, J. R., & Uehara, A. (1985). Ionic basis of the different action potential configurations of single guinea-pig atrial and ventricular myocytes. The Journal of Physiology, 368(1), 525-544. doi:10.1113/jphysiol.1985.sp015874INOUE, M., INOUE, D., ISHIBASHI, K., SAKAI, R., OMORI, I., YAMAHARA, Y., … NAKAGAWA, M. (1993). Effects of Pilsicainide on the Atrial Fibrillation Threshold in Guinea Kg Atria. A Comparative Study with Disopyramide, Lidocaine and Flecainide. Japanese Heart Journal, 34(3), 301-312. doi:10.1536/ihj.34.301Inoue, D., Shirayama, T., Omori, I., Inoue, M., Sakai, R., Ishibashi, K., … Nakagawa, M. (1993). Electrophysiological effects of flecainide acetate on stretched guinea pig left atrial muscle fibers. Cardiovascular Drugs and Therapy, 7(3), 373-378. doi:10.1007/bf00880161Iwasaki, Y., Nishida, K., Kato, T., & Nattel, S. (2011). Atrial Fibrillation Pathophysiology. Circulation, 124(20), 2264-2274. doi:10.1161/circulationaha.111.019893Jensen, M. D., Ryan, D. H., Apovian, C. M., Ard, J. D., Comuzzie, A. G., Donato, K. A., … Yanovski, S. Z. (2013). 2013 AHA/ACC/TOS Guideline for the Management of Overweight and Obesity in Adults. Circulation, 129(25 suppl 2), S102-S138. doi:10.1161/01.cir.0000437739.71477.eeJi, Y., Varkevisser, R., Opacic, D., Bossu, A., Kuiper, M., Beekman, J. D. M., … van der Heyden, M. A. G. (2017). The inward rectifier current inhibitor PA-6 terminates atrial fibrillation and does not cause ventricular arrhythmias in goat and dog models. British Journal of Pharmacology, 174(15), 2576-2590. doi:10.1111/bph.13869Kanner, S. A., Jain, A., & Colecraft, H. M. (2018). Development of a High-Throughput Flow Cytometry Assay to Monitor Defective Trafficking and Rescue of Long QT2 Mutant hERG Channels. Frontiers in Physiology, 9. doi:10.3389/fphys.2018.00397Killeen, M. J., Sabir, I. N., Grace, A. A., & Huang, C. L.-H. (2008). Dispersions of repolarization and ventricular arrhythmogenesis: Lessons from animal models. Progress in Biophysics and Molecular Biology, 98(2-3), 219-229. doi:10.1016/j.pbiomolbio.2008.10.008Killeen, M. J., Thomas, G., Sabir, I. N., Grace, A. A., & Huang, C. L.-H. (2008). Mouse models of human arrhythmia syndromes. Acta Physiologica, 192(4), 455-469. doi:10.1111/j.1748-1716.2007.01822.xKoivumäki, J. T., Korhonen, T., & Tavi, P. (2011). Impact of Sarcoplasmic Reticulum Calcium Release on Calcium Dynamics and Action Potential Morphology in Human Atrial Myocytes: A Computational Study. PLoS Computational Biology, 7(1), e1001067. doi:10.1371/journal.pcbi.1001067LAU, C.-P., TSE, H.-F., SIU, C.-W., & GBADEBO, D. (2012). Atrial Electrical and Structural Remodeling: Implications for Racial Differences in Atrial Fibrillation. Journal of Cardiovascular Electrophysiology, 23, s36-s40. doi:10.1111/jce.12022Leopoldo, A. S., Lima-Leopoldo, A. P., Sugizaki, M. M., Nascimento, A. F. do, de Campos, D. H. S., Luvizotto, R. de A. M., … Cicogna, A. C. (2011). Involvement of L-type calcium channel and serca2a in myocardial dysfunction induced by obesity. Journal of Cellular Physiology, 226(11), 2934-2942. doi:10.1002/jcp.22643Lima-Leopoldo, A. P., Leopoldo, A. S., Silva, D. C. T., Nascimento, A. F. do, Campos, D. H. S. de, Luvizotto, R. de A. M., … Cicogna, A. C. (2013). Influence of Long-Term Obesity on Myocardial Gene Expression. Arquivos Brasileiros de Cardiologia, 100(3). doi:10.5935/abc.20130045Lima-Leopoldo, A. P., Sugizaki, M. M., Leopoldo, A. S., Carvalho, R. F., Nogueira, C. R., Nascimento, A. F., … Cicogna, A. C. (2008). Obesity induces upregulation of genes involved in myocardial Ca2+ handling. Brazilian Journal of Medical and Biological Research, 41(7), 615-620. doi:10.1590/s0100-879x2008000700011Liu, T., Takimoto, E., Dimaano, V. L., DeMazumder, D., Kettlewell, S., Smith, G., … O’Rourke, B. (2014). Inhibiting Mitochondrial Na + /Ca 2+ Exchange Prevents Sudden Death in a Guinea Pig Model of Heart Failure. Circulation Research, 115(1), 44-54. doi:10.1161/circresaha.115.303062Mancarella, S., Yue, Y., Karnabi, E., Qu, Y., El-Sherif, N., & Boutjdir, M. (2008). Impaired Ca2+ homeostasis is associated with atrial fibrillation in the α1D L-type Ca2+ channel KO mouse. American Journal of Physiology-Heart and Circulatory Physiology, 295(5), H2017-H2024. doi:10.1152/ajpheart.00537.2008Mangoni, M. E., Couette, B., Bourinet, E., Platzer, J., Reimer, D., Striessnig, J., & Nargeot, J. (2003). Functional role of L-type Cav1.3 Ca2+ channels in cardiac pacemaker activity. Proceedings of the National Academy of Sciences, 100(9), 5543-5548. doi:10.1073/pnas.0935295100Martinez-Mateu, L., Romero, L., Ferrer-Albero, A., Sebastian, R., Rodríguez Matas, J. F., Jalife, J., … Saiz, J. (2018). Factors affecting basket catheter detection of real and phantom rotors in the atria: A computational study. PLOS Computational Biology, 14(3), e1006017. doi:10.1371/journal.pcbi.1006017Matafome, P., & Seiça, R. (2017). Function and Dysfunction of Adipose Tissue. Obesity and Brain Function, 3-31. doi:10.1007/978-3-319-63260-5_1Matsimra, H., & Ehara, T. (1997). Selective Enhancement of the Slow Component of Delayed Rectifier K+Current in Guinea-Pig Atrial Cells by External ATP. The Journal of Physiology, 503(1), 45-54. doi:10.1111/j.1469-7793.1997.045bi.xMichael, G., Xiao, L., Qi, X.-Y., Dobrev, D., & Nattel, S. (2008). Remodelling of cardiac repolarization: how homeostatic responses can lead to arrhythmogenesis. Cardiovascular Research, 81(3), 491-499. doi:10.1093/cvr/cvn266Mickelson, A. V., & Chandra, M. (2017). Hypertrophic cardiomyopathy mutation in cardiac troponin T (R95H) attenuates length-dependent activation in guinea pig cardiac muscle fibers. American Journal of Physiology-Heart and Circulatory Physiology, 313(6), H1180-H1189. doi:10.1152/ajpheart.00369.2017Nakaya, H., Furusawa, Y., Ogura, T., Tamagawa, M., & Uemura, H. (2000). Inhibitory effects of JTV-519, a novel cardioprotective drug, on potassium currents and experimental atrial fibrillation in guinea-pig hearts. British Journal of Pharmacology, 131(7), 1363-1372. doi:10.1038/sj.bjp.0703713Nattel, S. (2002). New ideas about atrial fibrillation 50 years on. Nature, 415(6868), 219-226. doi:10.1038/415219aNattel, S., & Dobrev, D. (2017). Controversies About Atrial Fibrillation Mechanisms. Circulation Research, 120(9), 1396-1398. doi:10.1161/circresaha.116.310489Nattel, S., & Harada, M. (2014). Atrial Remodeling and Atrial Fibrillation. Journal of the American College of Cardiology, 63(22), 2335-2345. doi:10.1016/j.jacc.2014.02.555Nerbonne, J. M., & Kass, R. S. (2005). Molecular Physiology of Cardiac Repolarization. Physiological Reviews, 85(4), 1205-1253. doi:10.1152/physrev.00002.2005Ni, H., Whittaker, D. G., Wang, W., Giles, W. R., Narayan, S. M., & Zhang, H. (2017). Synergistic Anti-arrhythmic Effects in Human Atria with Combined Use of Sodium Blockers and Acacetin. Frontiers in Physiology, 8. doi:10.3389/fphys.2017.00946O’Connell, R. P., Musa, H., Gomez, M. S. M., Avula, U. M., Herron, T. J., Kalifa, J., & Anumonwo, J. M. B. (2015). Free Fatty Acid Effects on the Atrial Myocardium: Membrane Ionic Currents Are Remodeled by the Disruption of T-Tubular Architecture. PLOS ONE, 10(8), e0133052. doi:10.1371/journal.pone.0133052OCHI, R., MOMOSE, Y., OYAMA, K., & GILES, W. (2006). Sphingosine-1-phosphate effects on guinea pig atrial myocytes: Alterations in action potentials and K+ currents. Cardiovascular Research, 70(1), 88-96. doi:10.1016/j.cardiores.2006.01.010O’Hara, T., & Rudy, Y. (2012). Quantitative comparison of cardiac ventricular myocyte electrophysiology and response to drugs in human and nonhuman species. American Journal of Physiology-Heart and Circulatory Physiology, 302(5), H1023-H1030. doi:10.1152/ajpheart.00785.2011Osadchii, O. E. (2012). Electrophysiological determinants of arrhythmic susceptibility upon endocardial and epicardial pacing in guinea-pig heart. Acta Physiologica, 205(4), 494-506. doi:10.1111/j.1748-1716.2012.02428.xPatoine, D., Levac, X., Pilote, S., Drolet, B., & Simard, C. (2013). Decreased CYP3A Expression and Activity in Guinea Pig Models of Diet-Induced Metabolic Syndrome: Is Fatty Liver Infiltration Involved? Drug Metabolism and Disposition, 41(5), 952-957. doi:10.1124/dmd.112.050641Paulino, E. C., Ferreira, J. C. B., Bechara, L. R., Tsutsui, J. M., Mathias, W., Lima, F. B., … Negrão, C. E. (2010). Exercise Training and Caloric Restriction Prevent Reduction in Cardiac Ca 2+ -Handling Protein Profile in Obese Rats. Hypertension, 56(4), 629-635. doi:10.1161/hypertensionaha.110.156141Pérez-Hernández, M., Matamoros, M., Barana, A., Amorós, I., Gómez, R., Núñez, M., … Caballero, R. (2015). Pitx2c increases in atrial myocytes from chronic atrial fibrillation patients enhancingIKsand decreasingICa,L. Cardiovascular Research, 109(3), 431-441. doi:10.1093/cvr/cvv280A, P., H, C., MC, F., L, B., JN, W., & HS, K. (2018). Atrial Fibrillation Initiated by Early Afterdepolarization-Mediated Triggered Activity during Acute Oxidative Stress: Eff

    New Paradigm of Defibrillation: Towards Painless Therapy

    Get PDF
    Sudden cardiac death: SCD) causes approximately 300,000 - 400,000 deaths a year in the United States. It usually starts as ventricular tachycardia: VT) and then degenerates into ventricular fibrillation: VF). Implantable cardioverter defibrillator: ICD) therapy is the only reliable treatment of VT/VF and has been shown to effectively reduce mortality by many clinical trials. However, high-voltage ICD shocks could result in myocardial dysfunction and damage. The majority of patients receiving ICD therapy have a history of coronary disease; their hearts develop myocardium infarction, which could provide a substrate for reentrant tachy-arrhythmias. Other than lethal ventricular tachycardia, atrial fibrillation: AF) became the most common arrhythmia by affecting 2.2 to 5.6 millions of Americans. The complications of AF include an increased rate of mortality, heart failure, stroke, etc. In this dissertation, we explore mechanisms of sustained ventricular and atrial tachyarrhythmias and the mechanisms of defibrillation using the conventional high-voltage single shock. Through the use of novel fluorescent optical mapping techniques and several animal models of ventricular and atrial arrhythmias, we develop and validate several novel low-voltage defibrillation therapies for atrial and ventricular arrhythmias. Several important previous studies on mechanisms of arrhythmia maintenance and termination using mathematical and experimental models are overviewed in Chapter 2. A study on multiple monophasic shocks improving electrotherapy of ventricular tachycardia in rabbit model of chronic infarction is presented in Chapter 3. Ventricular arrhythmias and low-voltage defibrillation therapy are studied in a more clinically-relevent in vivo canine model of healing myocardial infarction in Chapter 4. Finally, Chapter 5 presents a novel multi-stage low-energy defibrillation therapy for atrial fibrillation in in vivo canine hearts

    Chronic myocardial infarction promotes atrial action potential alternans, afterdepolarisations and fibrillation

    Get PDF
    Aims: Atrial fibrillation (AF) is increased in patients with heart failure resulting from myocardial infarction (MI). We aimed to determine the effects of chronic ventricular MI in rabbits on the susceptibility to AF, and underlying atrial electrophysiological and Ca2+-handling mechanisms. Methods and results: In Langendorff-perfused rabbit hearts, under beta-adrenergic-stimulation with isoproterenol (1 µM; ISO), 8 weeks MI decreased AF threshold, indicating increased AF-susceptibility. This was associated with increased atrial action potential duration-alternans at 90% repolarisation, by 147%, and no significant change in mean APD or atrial global conduction velocity (n=6-13 non-MI hearts, 5-12 MI). In atrial isolated myocytes, also under beta-stimulation, L-type Ca2+ current (ICaL) density and intracellular Ca2+-transient amplitude were decreased by MI, by 35% and 41%, respectively, and the frequency of spontaneous depolarisations (SDs) was substantially increased. MI increased atrial myocyte size and capacity, and markedly decreased transverse-tubule density. In non-MI hearts perfused with ISO, the ICaL-blocker nifedipine, at a concentration (0.02 µM) causing an equivalent ICaL-reduction (35%) to that from the MI, did not affect AF-susceptibility, and decreased APD. Conclusion: chronic MI in rabbits remodels atrial structure, electrophysiology and intracellular Ca2+-handling. Increased susceptibility to AF by MI, under beta-adrenergic-stimulation, may result from associated production of atrial APD-alternans and SDs, since steady-state APD and global conduction velocity were unchanged under these conditions, and may be unrelated to the associated reduction in whole-cell ICaL. Future studies may clarify potential contributions of local conduction changes, and cellular and sub-cellular mechanisms of alternans, to the increased AF-susceptibility
    corecore