Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Circ Res ; 134(7): 892-912, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38415360

RESUMEN

BACKGROUND: Viral cardiac infection represents a significant clinical challenge encompassing several etiological agents, disease stages, complex presentation, and a resulting lack of mechanistic understanding. Myocarditis is a major cause of sudden cardiac death in young adults, where current knowledge in the field is dominated by later disease phases and pathological immune responses. However, little is known regarding how infection can acutely induce an arrhythmogenic substrate before significant immune responses. Adenovirus is a leading cause of myocarditis, but due to species specificity, models of infection are lacking, and it is not understood how adenoviral infection may underlie sudden cardiac arrest. Mouse adenovirus type-3 was previously reported as cardiotropic, yet it has not been utilized to understand the mechanisms of cardiac infection and pathology. METHODS: We have developed mouse adenovirus type-3 infection as a model to investigate acute cardiac infection and molecular alterations to the infected heart before an appreciable immune response or gross cardiomyopathy. RESULTS: Optical mapping of infected hearts exposes decreases in conduction velocity concomitant with increased Cx43Ser368 phosphorylation, a residue known to regulate gap junction function. Hearts from animals harboring a phospho-null mutation at Cx43Ser368 are protected against mouse adenovirus type-3-induced conduction velocity slowing. Additional to gap junction alterations, patch clamping of mouse adenovirus type-3-infected adult mouse ventricular cardiomyocytes reveals prolonged action potential duration as a result of decreased IK1 and IKs current density. Turning to human systems, we find human adenovirus type-5 increases phosphorylation of Cx43Ser368 and disrupts synchrony in human induced pluripotent stem cell-derived cardiomyocytes, indicating common mechanisms with our mouse whole heart and adult cardiomyocyte data. CONCLUSIONS: Together, these findings demonstrate that adenoviral infection creates an arrhythmogenic substrate through direct targeting of gap junction and ion channel function in the heart. Such alterations are known to precipitate arrhythmias and likely contribute to sudden cardiac death in acutely infected patients.


Asunto(s)
Células Madre Pluripotentes Inducidas , Miocarditis , Humanos , Ratones , Animales , Conexina 43/genética , Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Miocitos Cardíacos/fisiología , Uniones Comunicantes , Adenoviridae/genética , Muerte Súbita Cardíaca
2.
Circ Res ; 133(8): 658-673, 2023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37681314

RESUMEN

BACKGROUND: Cardiac conduction is understood to occur through gap junctions. Recent evidence supports ephaptic coupling as another mechanism of electrical communication in the heart. Conduction via gap junctions predicts a direct relationship between conduction velocity (CV) and bulk extracellular resistance. By contrast, ephaptic theory is premised on the existence of a biphasic relationship between CV and the volume of specialized extracellular clefts within intercalated discs such as the perinexus. Our objective was to determine the relationship between ventricular CV and structural changes to micro- and nanoscale extracellular spaces. METHODS: Conduction and Cx43 (connexin43) protein expression were quantified from optically mapped guinea pig whole-heart preparations perfused with the osmotic agents albumin, mannitol, dextran 70 kDa, or dextran 2 MDa. Peak sodium current was quantified in isolated guinea pig ventricular myocytes. Extracellular resistance was quantified by impedance spectroscopy. Intercellular communication was assessed in a heterologous expression system with fluorescence recovery after photobleaching. Perinexal width was quantified from transmission electron micrographs. RESULTS: CV primarily in the transverse direction of propagation was significantly reduced by mannitol and increased by albumin and both dextrans. The combination of albumin and dextran 70 kDa decreased CV relative to albumin alone. Extracellular resistance was reduced by mannitol, unchanged by albumin, and increased by both dextrans. Cx43 expression and conductance and peak sodium currents were not significantly altered by the osmotic agents. In response to osmotic agents, perinexal width, in order of narrowest to widest, was albumin with dextran 70 kDa; albumin or dextran 2 MDa; dextran 70 kDa or no osmotic agent, and mannitol. When compared in the same order, CV was biphasically related to perinexal width. CONCLUSIONS: Cardiac conduction does not correlate with extracellular resistance but is biphasically related to perinexal separation, providing evidence that the relationship between CV and extracellular volume is determined by ephaptic mechanisms under conditions of normal gap junctional coupling.


Asunto(s)
Conexina 43 , Dextranos , Animales , Cobayas , Dextranos/metabolismo , Conexina 43/metabolismo , Miocitos Cardíacos/metabolismo , Sodio/metabolismo , Uniones Comunicantes/metabolismo , Albúminas/metabolismo , Manitol/farmacología , Manitol/metabolismo , Potenciales de Acción
3.
Am J Physiol Heart Circ Physiol ; 326(1): H25-H31, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37889255

RESUMEN

Since 2010, the number of life science doctoral graduates opting into academic postdoctoral employment has steadily declined. In recent years, this decline has made routine headlines in academic news cycles, and faculty members, universities, and funding bodies alike have begun to take notice. In November 2022, the National Institutes of Health (NIH) convened a special interest group to address the problems in postdoctoral recruitment and retention. In response, the American Physiological Society Science Policy Committee highlighted several key issues in postdoctoral training and working conditions and offered the NIH solutions to consider. There are known issues that affect postdoctoral recruitment and retention efforts: low wages relative to other employment sectors, a heavy workload, and poor job prospects to name a few. Unfortunately, these concerns are frequently dismissed as "the price of doing business in academia," and postdoctoral scholars are promised that if they overcome the trials and tribulations of this training period, the reward at the end, a career with academic freedom to pursue your own interests, justifies the means. However, academic freedom cannot and should not be used as the band-aid in a system where most of us will never actually experience academic freedom. Instead, we should systematically embrace solutions that improve the personal and professional health of early career researchers in all levels of training and independence if the goal is to truly shore up the academic workforce.


Asunto(s)
Investigadores , Condiciones de Trabajo , Humanos , Estados Unidos , Recursos Humanos , Investigadores/educación
4.
Am J Physiol Heart Circ Physiol ; 323(3): H363-H377, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35749719

RESUMEN

In recent decades, the scientific community has seen an increased interest in rigor and reproducibility. In 2017, concerns about methodological thoroughness and reporting practices were implicated as significant barriers to reproducibility within the preclinical cardiovascular literature, particularly in studies using animal research. The Langendorff, whole heart technique has proven to be an invaluable research tool, being modified in a myriad of ways to probe questions across the spectrum of physiological and pathophysiological functions of the heart. As a result, significant variability in the application of the Langendorff technique exists. This literature review quantifies the different methods employed in the implementation of the Langendorff technique and provides brief examples of how individual parametric differences can impact the outcomes and interpretation of studies. From 2017 to 2020, significant variability of animal models, anesthesia, cannulation time, perfusate composition, pH, and temperature demonstrate that the technique has diversified to meet new challenges and answer different scientific questions. The review also reveals which individual methods are most frequently reported, even if there is no explicit agreement upon which parameters should be reported. The analysis of methods related to the Langendorff technique suggests a framework for considering methodological approach when interpreting seemingly contradictory results, rather than concluding that results are irreproducible.


Asunto(s)
Corazón , Proyectos de Investigación , Animales , Corazón/fisiología , Modelos Animales , Reproducibilidad de los Resultados , Vísceras
5.
Pflugers Arch ; 473(3): 557-571, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33660028

RESUMEN

The relationship between cardiac conduction velocity (CV) and extracellular potassium (K+) is biphasic, with modest hyperkalemia increasing CV and severe hyperkalemia slowing CV. Recent studies from our group suggest that elevating extracellular sodium (Na+) and calcium (Ca2+) can enhance CV by an extracellular pathway parallel to gap junctional coupling (GJC) called ephaptic coupling that can occur in the gap junction adjacent perinexus. However, it remains unknown whether these same interventions modulate CV as a function of K+. We hypothesize that Na+, Ca2+, and GJC can attenuate conduction slowing consequent to severe hyperkalemia. Elevating Ca2+ from 1.25 to 2.00 mM significantly narrowed perinexal width measured by transmission electron microscopy. Optically mapped, Langendorff-perfused guinea pig hearts perfused with increasing K+ revealed the expected biphasic CV-K+ relationship during perfusion with different Na+ and Ca2+ concentrations. Neither elevating Na+ nor Ca2+ alone consistently modulated the positive slope of CV-K+ or conduction slowing at 10-mM K+; however, combined Na+ and Ca2+ elevation significantly mitigated conduction slowing at 10-mM K+. Pharmacologic GJC inhibition with 30-µM carbenoxolone slowed CV without changing the shape of CV-K+ curves. A computational model of CV predicted that elevating Na+ and narrowing clefts between myocytes, as occur with perinexal narrowing, reduces the positive and negative slopes of the CV-K+ relationship but do not support a primary role of GJC or sodium channel conductance. These data demonstrate that combinatorial effects of Na+ and Ca2+ differentially modulate conduction during hyperkalemia, and enhancing determinants of ephaptic coupling may attenuate conduction changes in a variety of physiologic conditions.


Asunto(s)
Calcio/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Corazón/fisiología , Potasio/metabolismo , Sodio/metabolismo , Animales , Cobayas , Preparación de Corazón Aislado
6.
Am J Physiol Heart Circ Physiol ; 321(6): H1042-H1055, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34623182

RESUMEN

Cardiac voltage-gated sodium channel gain-of-function prolongs repolarization in the long-QT syndrome type 3 (LQT3). Previous studies suggest that narrowing the perinexus within the intercalated disc, leading to rapid sodium depletion, attenuates LQT3-associated action potential duration (APD) prolongation. However, it remains unknown whether extracellular sodium concentration modulates APD prolongation during sodium channel gain-of-function. We hypothesized that elevated extracellular sodium concentration and widened perinexus synergistically prolong APD in LQT3. LQT3 was induced with sea anemone toxin (ATXII) in Langendorff-perfused guinea pig hearts (n = 34). Sodium concentration was increased from 145 to 160 mM. Perinexal expansion was induced with mannitol or the sodium channel ß1-subunit adhesion domain antagonist (ßadp1). Epicardial ventricular action potentials were optically mapped. Individual and combined effects of varying clefts and sodium concentrations were simulated in a computational model. With ATXII, both mannitol and ßadp1 significantly widened the perinexus and prolonged APD, respectively. The elevated sodium concentration alone significantly prolonged APD as well. Importantly, the combination of elevated sodium concentration and perinexal widening synergistically prolonged APD. Computational modeling results were consistent with animal experiments. Concurrently elevating extracellular sodium and increasing intercalated disc edema prolongs repolarization more than the individual interventions alone in LQT3. This synergistic effect suggests an important clinical implication that hypernatremia in the presence of cardiac edema can markedly increase LQT3-associated APD prolongation. Therefore, to our knowledge, this is the first study to provide evidence of a tractable and effective strategy to mitigate LQT3 phenotype by means of managing sodium levels and preventing cardiac edema in patients.NEW & NOTEWORTHY This is the first study to demonstrate that the long-QT syndrome type 3 (LQT3) phenotype can be exacerbated or concealed by regulating extracellular sodium concentrations and/or the intercalated disc separation. The animal experiments and computational modeling in the current study reveal a critically important clinical implication: sodium dysregulation in the presence of edema within the intercalated disc can markedly increase the risk of arrhythmia in LQT3. These findings strongly suggest that maintaining extracellular sodium within normal physiological limits may be an effective and inexpensive therapeutic option for patients with congenital or acquired sodium channel gain-of-function diseases.


Asunto(s)
Potenciales de Acción , Edema Cardíaco/complicaciones , Edema Cardíaco/metabolismo , Frecuencia Cardíaca , Hipernatremia/sangre , Hipernatremia/complicaciones , Síndrome de QT Prolongado/metabolismo , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Sodio/sangre , Animales , Venenos de Cnidarios , Simulación por Computador , Modelos Animales de Enfermedad , Edema Cardíaco/patología , Edema Cardíaco/fisiopatología , Cobayas , Hipernatremia/fisiopatología , Preparación de Corazón Aislado , Síndrome de QT Prolongado/inducido químicamente , Síndrome de QT Prolongado/fisiopatología , Masculino , Modelos Cardiovasculares , Miocitos Cardíacos/patología
7.
Int J Mol Sci ; 22(19)2021 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-34638526

RESUMEN

Gap junctions (GJ) and connexins play integral roles in cellular physiology and have been found to be involved in multiple pathophysiological states from cancer to cardiovascular disease. Studies over the last 60 years have demonstrated the utility of altering GJ signaling pathways in experimental models, which has led to them being attractive targets for therapeutic intervention. A number of different mechanisms have been proposed to regulate GJ signaling, including channel blocking, enhancing channel open state, and disrupting protein-protein interactions. The primary mechanism for this has been through the design of numerous peptides as therapeutics, that are either currently in early development or are in various stages of clinical trials. Despite over 25 years of research into connexin targeting peptides, the overall mechanisms of action are still poorly understood. In this overview, we discuss published connexin targeting peptides, their reported mechanisms of action, and the potential for these molecules in the treatment of disease.


Asunto(s)
Conexinas/metabolismo , Péptidos/metabolismo , Péptidos/farmacología , Animales , Uniones Comunicantes/metabolismo , Humanos , Proteínas del Tejido Nervioso/metabolismo , Isoformas de Proteínas/metabolismo , Transducción de Señal
8.
Am J Physiol Heart Circ Physiol ; 316(4): H849-H861, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30707595

RESUMEN

We previously demonstrated that altering extracellular sodium (Nao) and calcium (Cao) can modulate a form of electrical communication between cardiomyocytes termed "ephaptic coupling" (EpC), especially during loss of gap junction coupling. We hypothesized that altering Nao and Cao modulates conduction velocity (CV) and arrhythmic burden during ischemia. Electrophysiology was quantified by optically mapping Langendorff-perfused guinea pig ventricles with modified Nao (147 or 155 mM) and Cao (1.25 or 2.0 mM) during 30 min of simulated metabolic ischemia (pH 6.5, anoxia, aglycemia). Gap junction-adjacent perinexal width ( WP), a candidate cardiac ephapse, and connexin (Cx)43 protein expression and Cx43 phosphorylation at S368 were quantified by transmission electron microscopy and Western immunoblot analysis, respectively. Metabolic ischemia slowed CV in hearts perfused with 147 mM Nao and 2.0 mM Cao; however, theoretically increasing EpC with 155 mM Nao was arrhythmogenic, and CV could not be measured. Reducing Cao to 1.25 mM expanded WP, as expected during ischemia, consistent with reduced EpC, but attenuated CV slowing while delaying arrhythmia onset. These results were further supported by osmotically reducing WP with albumin, which exacerbated CV slowing and increased early arrhythmias during ischemia, whereas mannitol expanded WP, permitted conduction, and delayed the onset of arrhythmias. Cx43 expression patterns during the various interventions insufficiently correlated with observed CV changes and arrhythmic burden. In conclusion, decreasing perfusate calcium during metabolic ischemia enhances perinexal expansion, attenuates conduction slowing, and delays arrhythmias. Thus, perinexal expansion may be cardioprotective during metabolic ischemia. NEW & NOTEWORTHY This study demonstrates, for the first time, that modulating perfusate ion composition can alter cardiac electrophysiology during simulated metabolic ischemia.


Asunto(s)
Calcio/farmacología , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/fisiopatología , Isquemia Miocárdica/fisiopatología , Sodio/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/fisiopatología , Conexina 43/metabolismo , Uniones Comunicantes/efectos de los fármacos , Cobayas , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Técnicas In Vitro , Masculino , Concentración Osmolar
10.
Am J Physiol Heart Circ Physiol ; 313(6): H1190-H1198, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28939646

RESUMEN

With the sudden increase in affordable manufacturing technologies, the relationship between experimentalists and the designing process for laboratory equipment is rapidly changing. While experimentalists are still dependent on engineers and manufacturers for precision electrical, mechanical, and optical equipment, it has become a realistic option for in house manufacturing of other laboratory equipment with less precise design requirements. This is possible due to decreasing costs and increasing functionality of desktop three-dimensional (3-D) printers and 3-D design software. With traditional manufacturing methods, iterative design processes are expensive and time consuming, and making more than one copy of a custom piece of equipment is prohibitive. Here, we provide an overview to design a tissue bath and stabilizer for a customizable, suspended, whole heart optical mapping apparatus that can be produced significantly faster and less expensive than conventional manufacturing techniques. This was accomplished through a series of design steps to prevent fluid leakage in the areas where the optical imaging glass was attached to the 3-D printed bath. A combination of an acetone dip along with adhesive was found to create a water tight bath. Optical mapping was used to quantify cardiac conduction velocity and action potential duration to compare 3-D printed baths to a bath that was designed and manufactured in a machine shop. Importantly, the manufacturing method did not significantly affect conduction, action potential duration, or contraction, suggesting that 3-D printed baths are equally effective for optical mapping experiments.NEW & NOTEWORTHY This article details three-dimensional printable equipment for use in suspended whole heart optical mapping experiments. This equipment is less expensive than conventional manufactured equipment as well as easily customizable to the experimentalist. The baths can be waterproofed using only a three-dimensional printer, acetone, a glass microscope slide, c-clamps, and adhesive.


Asunto(s)
Potenciales de Acción , Corazón/fisiología , Preparación de Corazón Aislado/instrumentación , Poliésteres/química , Impresión Tridimensional , Imagen de Colorante Sensible al Voltaje/instrumentación , Animales , Estimulación Cardíaca Artificial , Diseño Asistido por Computadora , Análisis Costo-Beneficio , Electroencefalografía/instrumentación , Diseño de Equipo , Cobayas , Preparación de Corazón Aislado/economía , Masculino , Ensayo de Materiales , Marcapaso Artificial , Impresión Tridimensional/economía , Reproducibilidad de los Resultados , Factores de Tiempo , Imagen de Colorante Sensible al Voltaje/economía
11.
JACC Clin Electrophysiol ; 10(5): 829-842, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38430092

RESUMEN

BACKGROUND: Sudden unexpected death in epilepsy (SUDEP) is a fatal complication experienced by otherwise healthy epilepsy patients. Dravet syndrome (DS) is an inherited epileptic disorder resulting from loss of function of the voltage-gated sodium channel, NaV 1.1, and is associated with particularly high SUDEP risk. Evidence is mounting that NaVs abundant in the brain also occur in the heart, suggesting that the very molecular mechanisms underlying epilepsy could also precipitate cardiac arrhythmias and sudden death. Despite marked reduction of NaV 1.1 functional expression in DS, pathogenic late sodium current (INa,L) is paradoxically increased in DS hearts. However, the mechanisms by which DS directly impacts the heart to promote sudden death remain unclear. OBJECTIVES: In this study, the authors sought to provide evidence implicating remodeling of Na+ - and Ca2+ -handling machinery, including NaV 1.6 and Na+/Ca2+exchanger (NCX) within transverse (T)-tubules in DS-associated arrhythmias. METHODS: The authors undertook scanning ion conductance microscopy (SICM)-guided patch clamp, super-resolution microscopy, confocal Ca2+ imaging, and in vivo electrocardiography studies in Scn1a haploinsufficient murine model of DS. RESULTS: DS promotes INa,L in T-tubular nanodomains, but not in other subcellular regions. Consistent with increased NaV activity in these regions, super-resolution microscopy revealed increased NaV 1.6 density near Ca2+release channels, the ryanodine receptors (RyR2) and NCX in DS relative to WT hearts. The resulting INa,L in these regions promoted aberrant Ca2+ release, leading to ventricular arrhythmias in vivo. Cardiac-specific deletion of NaV 1.6 protects adult DS mice from increased T-tubular late NaV activity and the resulting arrhythmias, as well as sudden death. CONCLUSIONS: These data demonstrate that NaV 1.6 undergoes remodeling within T-tubules of adult DS hearts serving as a substrate for Ca2+ -mediated cardiac arrhythmias and may be a druggable target for the prevention of SUDEP in adult DS subjects.


Asunto(s)
Epilepsias Mioclónicas , Canal de Sodio Activado por Voltaje NAV1.6 , Animales , Femenino , Humanos , Masculino , Ratones , Arritmias Cardíacas/genética , Calcio/metabolismo , Epilepsias Mioclónicas/genética , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.6/genética , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Intercambiador de Sodio-Calcio/genética , Intercambiador de Sodio-Calcio/metabolismo , Muerte Súbita e Inesperada en la Epilepsia
12.
bioRxiv ; 2023 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-36824727

RESUMEN

During each heartbeat, the propagation of action potentials through the heart coordinates the contraction of billions of individual cardiomyocytes and is thus, a critical life process. Unsurprisingly, intercalated discs, which are cell-cell contact sites specialized to provide electrical and mechanical coupling between adjacent cardiomyocytes, have been the focus of much investigation. Slowed or disrupted propagation leads to potentially life-threatening arrhythmias in a wide range of pathologies, where intercalated disc remodeling is a common finding. Hence, the importance and urgency of understanding intercalated disc structure and its influence on action potential propagation. Surprisingly, however, conventional modeling approaches cannot predict changes in propagation elicited by perturbations that alter intercalated disc ultrastructure or molecular organization, owing to lack of quantitative structural data at subcellular through nano scales. In order to address this critical gap in knowledge, we sought to quantify intercalated disc structure at these finer spatial scales in the healthy adult mouse heart and relate them to function in a chamber-specific manner as a precursor to understanding the impacts of pathological intercalated disc remodeling. Using super-resolution light microscopy, electron microscopy, and computational image analysis, we provide here the first ever systematic, multiscale quantification of intercalated disc ultrastructure and molecular organization. By incorporating these data into a rule-based model of cardiac tissue with realistic intercalated disc structure, and comparing model predictions of electrical propagation with experimental measures of conduction velocity, we reveal that atrial intercalated discs can support faster conduction than their ventricular counterparts, which is normally masked by inter-chamber differences in myocyte geometry. Further, we identify key ultrastructural and molecular organization features underpinning the ability of atrial intercalated discs to support faster conduction. These data provide the first stepping stone to elucidating chamber-specific impacts of pathological intercalated disc remodeling, as occurs in many arrhythmic diseases.

13.
JACC Clin Electrophysiol ; 9(12): 2425-2443, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37498248

RESUMEN

BACKGROUND: Propagation of action potentials through the heart coordinates the heartbeat. Thus, intercalated discs, specialized cell-cell contact sites that provide electrical and mechanical coupling between cardiomyocytes, are an important target for study. Impaired propagation leads to arrhythmias in many pathologies, where intercalated disc remodeling is a common finding, hence the importance and urgency of understanding propagation dependence on intercalated disc structure. Conventional modeling approaches cannot predict changes in propagation elicited by perturbations that alter intercalated disc ultrastructure or molecular organization, because of lack of quantitative structural data at subcellular through nano scales. OBJECTIVES: This study sought to quantify intercalated disc structure at these spatial scales in the healthy adult mouse heart and relate them to chamber-specific properties of propagation as a precursor to understanding the effects of pathological intercalated disc remodeling. METHODS: Using super-resolution light microscopy, electron microscopy, and computational image analysis, we provide here the first ever systematic, multiscale quantification of intercalated disc ultrastructure and molecular organization. RESULTS: By incorporating these data into a rule-based model of cardiac tissue with realistic intercalated disc structure, and comparing model predictions of electrical propagation with experimental measures of conduction velocity, we reveal that atrial intercalated discs can support faster conduction than their ventricular counterparts, which is normally masked by interchamber differences in myocyte geometry. Further, we identify key ultrastructural and molecular organization features underpinning the ability of atrial intercalated discs to support faster conduction. CONCLUSIONS: These data provide the first stepping stone to elucidating chamber-specific effects of pathological intercalated disc remodeling, as occurs in many arrhythmic diseases.


Asunto(s)
Miocardio , Miocitos Cardíacos , Ratones , Animales , Frecuencia Cardíaca , Miocitos Cardíacos/fisiología , Arritmias Cardíacas
14.
Compr Physiol ; 12(4): 3833-3867, 2022 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-35959755

RESUMEN

The arterial vasculature can be divided into large conduit arteries, intermediate contractile arteries, resistance arteries, arterioles, and capillaries. Resistance arteries and arterioles primarily function to control systemic blood pressure. The resistance arteries are composed of a layer of endothelial cells oriented parallel to the direction of blood flow, which are separated by a matrix layer termed the internal elastic lamina from several layers of smooth muscle cells oriented perpendicular to the direction of blood flow. Cells within the vessel walls communicate in a homocellular and heterocellular fashion to govern luminal diameter, arterial resistance, and blood pressure. At rest, potassium currents govern the basal state of endothelial and smooth muscle cells. Multiple stimuli can elicit rises in intracellular calcium levels in either endothelial cells or smooth muscle cells, sourced from intracellular stores such as the endoplasmic reticulum or the extracellular space. In general, activation of endothelial cells results in the production of a vasodilatory signal, usually in the form of nitric oxide or endothelial-derived hyperpolarization. Conversely, activation of smooth muscle cells results in a vasoconstriction response through smooth muscle cell contraction. © 2022 American Physiological Society. Compr Physiol 12: 1-35, 2022.


Asunto(s)
Células Endoteliales , Músculo Liso Vascular , Comunicación Celular , Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Humanos , Músculo Liso Vascular/fisiología , Vasoconstricción/fisiología , Vasodilatación/fisiología
15.
Sci Rep ; 10(1): 17289, 2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-33057157

RESUMEN

Recent studies revealed that relatively small changes in perfusate sodium ([Na+]o) composition significantly affect cardiac electrical conduction and stability in contraction arrested ex vivo Langendorff heart preparations before and during simulated ischemia. Additionally, [Na+]o modulates cardiomyocyte contractility via a sodium-calcium exchanger (NCX) mediated pathway. It remains unknown, however, whether modest changes to [Na+]o that promote electrophysiologic stability similarly improve mechanical function during baseline and ischemia-reperfusion conditions. The purpose of this study was to quantify cardiac mechanical function during ischemia-reperfusion with perfusates containing 145 or 155 mM Na+ in Langendorff perfused isolated rat heart preparations. Relative to 145 mM Na+, perfusion with 155 mM [Na+]o decreased the amplitude of left-ventricular developed pressure (LVDP) at baseline and accelerated the onset of ischemic contracture. Inhibiting NCX with SEA0400 abolished LVDP depression caused by increasing [Na+]o at baseline and reduced the time to peak ischemic contracture. Ischemia-reperfusion decreased LVDP in all hearts with return of intrinsic activity, and reperfusion with 155 mM [Na+]o further depressed mechanical function. In summary, elevating [Na+]o by as little as 10 mM can significantly modulate mechanical function under baseline conditions, as well as during ischemia and reperfusion. Importantly, clinical use of Normal Saline, which contains 155 mM [Na+]o, with cardiac ischemia may require further investigation.


Asunto(s)
Corazón/fisiopatología , Isquemia Miocárdica/metabolismo , Sodio/metabolismo , Animales , Humanos , Masculino , Contracción Miocárdica , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/cirugía , Miocardio/metabolismo , Ratas , Ratas Sprague-Dawley , Reperfusión , Intercambiador de Sodio-Calcio/metabolismo
16.
Annu Int Conf IEEE Eng Med Biol Soc ; 2018: 5446-5459, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30441569

RESUMEN

Cardiac resynchronization therapy (CRT) can substantially improve dyssynchronous heart failure and reduce mortality. However, one-third of the CRT patients derive no measurable benefit from CRT, due to suboptimal placement of the left ventricular (LV) lead. We introduce a pipeline for improved CRT-therapy by creating an electromechanical model using patient-specific geometric parameters allowing individualization of therapy. The model successfully mimics expected changes when variables for tension, stiffness, and conduction are entered. Changing LV pacing site had a notable effect on maximum pressure gradient (dP/dtmax) in the presence of cardiac scarring, causing non-uniform excitation propagation through the LV. Tailoring CRT to the individual requires simulations with patient-specific biventricular meshes including cardiac geometry and conductivity properties.


Asunto(s)
Terapia de Resincronización Cardíaca , Insuficiencia Cardíaca , Frecuencia Cardíaca , Ventrículos Cardíacos , Humanos , Resultado del Tratamiento
17.
Elife ; 72018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30106376

RESUMEN

Computational modeling indicates that cardiac conduction may involve ephaptic coupling - intercellular communication involving electrochemical signaling across narrow extracellular clefts between cardiomyocytes. We hypothesized that ß1(SCN1B) -mediated adhesion scaffolds trans-activating NaV1.5 (SCN5A) channels within narrow (<30 nm) perinexal clefts adjacent to gap junctions (GJs), facilitating ephaptic coupling. Super-resolution imaging indicated preferential ß1 localization at the perinexus, where it co-locates with NaV1.5. Smart patch clamp (SPC) indicated greater sodium current density (INa) at perinexi, relative to non-junctional sites. A novel, rationally designed peptide, ßadp1, potently and selectively inhibited ß1-mediated adhesion, in electric cell-substrate impedance sensing studies. ßadp1 significantly widened perinexi in guinea pig ventricles, and selectively reduced perinexal INa, but not whole cell INa, in myocyte monolayers. In optical mapping studies, ßadp1 precipitated arrhythmogenic conduction slowing. In summary, ß1-mediated adhesion at the perinexus facilitates action potential propagation between cardiomyocytes, and may represent a novel target for anti-arrhythmic therapies.


Asunto(s)
Arritmias Cardíacas/tratamiento farmacológico , Comunicación Celular/genética , Uniones Comunicantes/ultraestructura , Miocitos Cardíacos/fisiología , Potenciales de Acción , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Adhesión Celular/genética , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Biología Computacional , Impedancia Eléctrica , Uniones Comunicantes/fisiología , Cobayas , Humanos , Ratones , Modelos Cardiovasculares , Miocitos Cardíacos/ultraestructura , Canal de Sodio Activado por Voltaje NAV1.5/genética , Técnicas de Placa-Clamp , Péptidos/química , Sodio/metabolismo , Subunidad beta-1 de Canal de Sodio Activado por Voltaje/genética
18.
Front Physiol ; 9: 398, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29780324

RESUMEN

Aims: Atrial fibrillation (AF) is the most common sustained arrhythmia. Previous evidence in animal models suggests that the gap junction (GJ) adjacent nanodomain - perinexus - is a site capable of independent intercellular communication via ephaptic transmission. Perinexal expansion is associated with slowed conduction and increased ventricular arrhythmias in animal models, but has not been studied in human tissue. The purpose of this study was to characterize the perinexus in humans and determine if perinexal expansion associates with AF. Methods: Atrial appendages from 39 patients (pts) undergoing cardiac surgery were fixed for immunofluorescence and transmission electron microscopy (TEM). Intercalated disk distribution of the cardiac sodium channel Nav1.5, its ß1 subunit, and connexin43 (C×43) was determined by confocal immunofluorescence. Perinexal width (Wp) from TEM was manually segmented by two blinded observers using ImageJ software. Results: Nav1.5, ß1, and C×43 are co-adjacent within intercalated disks of human atria, consistent with perinexal protein distributions in ventricular tissue of other species. TEM revealed that the GJ adjacent intermembrane separation in an individual perinexus does not change at distances greater than 30 nm from the GJ edge. Importantly, Wp is significantly wider in patients with a history of AF than in patients with no history of AF by approximately 3 nm, and Wp correlates with age (R = 0.7, p < 0.05). Conclusion: Human atrial myocytes have voltage-gated sodium channels in a dynamic intercellular cleft adjacent to GJs that is consistent with previous descriptions of the perinexus. Further, perinexal width is greater in patients with AF undergoing cardiac surgery than in those without.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA