Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Mol Cell Cardiol ; 167: 1-14, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35304170

RESUMEN

Altered levels of intracellular calcium (Ca2+) are a highly prevalent feature in different forms of cardiac injury, producing changes in contractility, arrhythmias, and mitochondrial dysfunction. In cardiac ischemia-reperfusion injury, mitochondrial Ca2+ overload leads to pathological production of reactive oxygen species (ROS), activates the permeability transition, and cardiomyocyte death. Here we investigated the cardiac phenotype caused by deletion of EF-hand domain-containing protein D1 (Efhd1-/-), a Ca2+-binding mitochondrial protein whose function is poorly understood. Efhd1-/- mice are viable and have no adverse cardiac phenotypes. They feature reductions in basal ROS levels and mitoflash events, both important precursors for mitochondrial injury, though cardiac mitochondria have normal susceptibility to Ca2+ overload. Notably, we also find that Efhd1-/- mice and their cardiomyocytes are resistant to hypoxic injury.


Asunto(s)
Daño por Reperfusión Miocárdica , Miocitos Cardíacos , Animales , Calcio/metabolismo , Isquemia/metabolismo , Ratones , Mitocondrias Cardíacas/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
2.
Sensors (Basel) ; 21(18)2021 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-34577240

RESUMEN

Light-scattering spectroscopy (LSS) is an established optical approach for characterization of biological tissues. Here, we investigated the capabilities of LSS and convolutional neural networks (CNNs) to quantitatively characterize the composition and arrangement of cardiac tissues. We assembled tissue constructs from fixed myocardium and the aortic wall with a thickness similar to that of the atrial free wall. The aortic sections represented fibrotic tissue. Depth, volume fraction, and arrangement of these fibrotic insets were varied. We gathered spectra with wavelengths from 500-1100 nm from the constructs at multiple locations relative to a light source. We used single and combinations of two spectra for training of CNNs. With independently measured spectra, we assessed the accuracy of the CNNs for the classification of tissue constructs from single spectra and combined spectra. Combined spectra, including the spectra from fibers distal from the illumination fiber, typically yielded the highest accuracy. The maximal classification accuracy of the depth detection, volume fraction, and permutated arrangements was (mean ± standard deviation (stddev)) 88.97 ± 2.49%, 76.33 ± 1.51%, and 84.25 ± 1.88%, respectively. Our studies demonstrate the reliability of quantitative characterization of tissue composition and arrangements using a combination of LSS and CNNs. The potential clinical applications of the developed approach include intraoperative quantification and mapping of atrial fibrosis, as well as the assessment of ablation lesions.


Asunto(s)
Miocardio , Redes Neurales de la Computación , Fibrosis , Humanos , Reproducibilidad de los Resultados , Análisis Espectral
3.
J Mol Cell Cardiol ; 139: 113-123, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31982426

RESUMEN

Transient receptor potential canonical 1 (TRPC1) protein is abundantly expressed in cardiomyocytes. While TRPC1 is supposed to be critically involved in cardiac hypertrophy, its physiological role in cardiomyocytes is poorly understood. We investigated the subcellular location of TRPC1 and its contribution to Ca2+ signaling in mammalian ventricular myocytes. Immunolabeling, three-dimensional scanning confocal microscopy and quantitative colocalization analysis revealed an abundant intracellular location of TRPC1 in neonatal rat ventricular myocytes (NRVMs) and adult rabbit ventricular myocytes. TRPC1 was colocalized with intracellular proteins including sarco/endoplasmic reticulum Ca2+ ATPase 2 in the sarcoplasmic reticulum (SR). Colocalization with wheat germ agglutinin, which labels the glycocalyx and thus marks the sarcolemma including the transverse tubular system, was low. Super-resolution and immunoelectron microscopy supported the intracellular location of TRPC1. We investigated Ca2+ signaling in NRVMs after adenoviral TRPC1 overexpression or silencing. In NRVMs bathed in Na+ and Ca2+ free solution, TRPC1 overexpression and silencing was associated with a decreased and increased SR Ca2+ content, respectively. In isolated rabbit cardiomyocytes bathed in Na+ and Ca2+ free solution, we found an increased decay of the cytosolic Ca2+ concentration [Ca2+]i and increased SR Ca2+ content in the presence of the TRPC channel blocker SKF-96365. In a computational model of rabbit ventricular myocytes at physiological pacing rates, Ca2+ leak through SR TRPC channels increased the systolic and diastolic [Ca2+]i with only minor effects on the action potential and SR Ca2+ content. Our studies suggest that TRPC1 channels are localized in the SR, and not present in the sarcolemma of ventricular myocytes. The studies provide evidence for a role of TRPC1 as a contributor to SR Ca2+ leak in cardiomyocytes, which was previously explained by ryanodine receptors only. We propose that the findings will guide us to an understanding of TRPC1 channels as modulators of [Ca2+]i and contractility in cardiomyocytes.


Asunto(s)
Ventrículos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Animales Recién Nacidos , Calcio/metabolismo , Proteínas del Citoesqueleto/metabolismo , Modelos Biológicos , Miocitos Cardíacos/ultraestructura , Conejos , Ratas , Sarcolema/metabolismo , Retículo Sarcoplasmático/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Canales Catiónicos TRPC/ultraestructura
4.
Biophys J ; 116(8): 1386-1393, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30979553

RESUMEN

In mammalian ventricular cardiomyocytes, invaginations of the surface membrane form the transverse tubular system (T-system), which consists of transverse tubules (TTs) that align with sarcomeres and Z-lines as well as longitudinal tubules (LTs) that are present between Z-lines in some species. In many cardiac disease etiologies, the T-system is perturbed, which is believed to promote spatially heterogeneous, dyssynchronous Ca2+ release and inefficient contraction. In general, T-system characterization approaches have been directed primarily at isolated cells and do not detect subcellular T-system heterogeneity. Here, we present MatchedMyo, a matched-filter-based algorithm for subcellular T-system characterization in isolated cardiomyocytes and millimeter-scale myocardial sections. The algorithm utilizes "filters" representative of TTs, LTs, and T-system absence. Application of the algorithm to cardiomyocytes isolated from rat disease models of myocardial infarction (MI), dilated cardiomyopathy induced via aortic banding, and sham surgery confirmed and quantified heterogeneous T-system structure and remodeling. Cardiomyocytes from post-MI hearts exhibited increasing T-system disarray as proximity to the infarct increased. We found significant (p < 0.05, Welch's t-test) increases in LT density within cardiomyocytes proximal to the infarct (12 ± 3%, data reported as mean ± SD, n = 3) versus sham (4 ± 2%, n = 5), but not distal to the infarct (7 ± 1%, n = 3). The algorithm also detected decreases in TTs within 5° of the myocyte minor axis for isolated aortic banding (36 ± 9%, n = 3) and MI cardiomyocytes located intermediate (37 ± 4%, n = 3) and proximal (34 ± 4%, n = 3) to the infarct versus sham (57 ± 12%, n = 5). Application of bootstrapping to rabbit MI tissue revealed distal sections comprised 18.9 ± 1.0% TTs, whereas proximal sections comprised 10.1 ± 0.8% TTs (p < 0.05), a 46.6% decrease. The matched-filter approach therefore provides a robust and scalable technique for T-system characterization from isolated cells through millimeter-scale myocardial sections.


Asunto(s)
Algoritmos , Procesamiento de Imagen Asistido por Computador/métodos , Espacio Intracelular/metabolismo , Miocitos Cardíacos/citología , Animales , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/diagnóstico por imagen , Ratas
5.
PLoS Comput Biol ; 14(10): e1006438, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30303952

RESUMEN

The cardiac muscarinic receptor (M2R) regulates heart rate, in part, by modulating the acetylcholine (ACh) activated K+ current IK,ACh through dissociation of G-proteins, that in turn activate KACh channels. Recently, M2Rs were noted to exhibit intrinsic voltage sensitivity, i.e. their affinity for ligands varies in a voltage dependent manner. The voltage sensitivity of M2R implies that the affinity for ACh (and thus the ACh effect) varies throughout the time course of a cardiac electrical cycle. The aim of this study was to investigate the contribution of M2R voltage sensitivity to the rate and shape of the human sinus node action potentials in physiological and pathophysiological conditions. We developed a Markovian model of the IK,ACh modulation by voltage and integrated it into a computational model of human sinus node. We performed simulations with the integrated model varying ACh concentration and voltage sensitivity. Low ACh exerted a larger effect on IK,ACh at hyperpolarized versus depolarized membrane voltages. This led to a slowing of the pacemaker rate due to an attenuated slope of phase 4 depolarization with only marginal effect on action potential duration and amplitude. We also simulated the theoretical effects of genetic variants that alter the voltage sensitivity of M2R. Modest negative shifts in voltage sensitivity, predicted to increase the affinity of the receptor for ACh, slowed the rate of phase 4 depolarization and slowed heart rate, while modest positive shifts increased heart rate. These simulations support our hypothesis that altered M2R voltage sensitivity contributes to disease and provide a novel mechanistic foundation to study clinical disorders such as atrial fibrillation and inappropriate sinus tachycardia.


Asunto(s)
Modelos Cardiovasculares , Receptor Muscarínico M2/fisiología , Nodo Sinoatrial/fisiología , Acetilcolina/metabolismo , Biología Computacional , Humanos , Cadenas de Markov
6.
Circulation ; 135(17): 1632-1645, 2017 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-28073805

RESUMEN

BACKGROUND: Cardiac recovery in response to mechanical unloading by left ventricular assist devices (LVADs) has been demonstrated in subgroups of patients with chronic heart failure (HF). Hallmarks of HF are depletion and disorganization of the transverse tubular system (t-system) in cardiomyocytes. Here, we investigated remodeling of the t-system in human end-stage HF and its role in cardiac recovery. METHODS: Left ventricular biopsies were obtained from 5 donors and 26 patients with chronic HF undergoing implantation of LVADs. Three-dimensional confocal microscopy and computational image analysis were applied to assess t-system structure, density, and distance of ryanodine receptor clusters to the sarcolemma, including the t-system. Recovery of cardiac function in response to mechanical unloading was assessed by echocardiography during turndown of the LVAD. RESULTS: The majority of HF myocytes showed remarkable t-system remodeling, particularly sheet-like invaginations of the sarcolemma. Circularity of t-system components was decreased in HF versus controls (0.37±0.01 versus 0.46±0.02; P<0.01), and the volume/length ratio was increased in HF (0.36±0.01 versus 0.25±0.02 µm2; P<0.0001). T-system density was reduced in HF, leading to increased ryanodine receptor-sarcolemma distances (0.96±0.05 versus 0.64±0.1 µm; P<0.01). Low ryanodine receptor-sarcolemma distances at the time of LVAD implantation predicted high post-LVAD left ventricular ejection fractions (P<0.01) and ejection fraction increases during unloading (P<0.01). Ejection fraction in patients with pre-LVAD ryanodine receptor-sarcolemma distances >1 µm did not improve after mechanical unloading. In addition, calcium transients were recorded in field-stimulated isolated human cardiomyocytes and analyzed with respect to local t-system density. Calcium release in HF myocytes was restricted to regions proximal to the sarcolemma. Local calcium upstroke was delayed (23.9±4.9 versus 10.3±1.7 milliseconds; P<0.05) and more asynchronous (18.1±1.5 versus 8.9±2.2 milliseconds; P<0.01) in HF cells with low t-system density versus cells with high t-system density. CONCLUSIONS: The t-system in end-stage human HF presents a characteristic novel phenotype consisting of sheet-like invaginations of the sarcolemma. Our results suggest that the remodeled t-system impairs excitation-contraction coupling and functional recovery during chronic LVAD unloading. An intact t-system at the time of LVAD implantation may constitute a precondition and predictor for functional cardiac recovery after mechanical unloading.


Asunto(s)
Acoplamiento Excitación-Contracción , Insuficiencia Cardíaca/terapia , Corazón Auxiliar , Contracción Miocárdica , Miocitos Cardíacos/patología , Función Ventricular Izquierda , Remodelación Ventricular , Adulto , Anciano , Biopsia , Estudios de Casos y Controles , Ecocardiografía , Femenino , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Microscopía Confocal , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Estudios Prospectivos , Diseño de Prótesis , Recuperación de la Función , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Factores de Tiempo , Resultado del Tratamiento
7.
J Theor Biol ; 412: 61-73, 2017 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-27590324

RESUMEN

Gap junction channels are formed out of connexin isoforms, which enable molecule and ion selective diffusion amongst neighboring cells. HeLa cells expressing distinct connexins (Cx) allow the formation of heterotypic channels, where we observed a molecular charge-independent preferential flux of large fluorescent molecules in the Cx45 to Cx43 direction. We hypothesize that the pore's shape is a significant factor along-side charge and transjunctional voltages for this asymmetric flux. To test this hypothesis, we developed a 3D computational model simulating Brownian diffusion of large molecules in a gap junction channel pore. The basic pore contour was derived from x-ray crystallographic structures of Cx43 and Cx26 and approximated using basic geometric shapes. Lucifer yellow dye molecules and cesium counter-ions were modeled as spheres using their respective Stokes radii. Our simulation results from simple diffusion and constant concentration gradient experiments showed that only charged particles yield asymmetric fluxes in heterotypic pores. While increasing the inner mouth size resulted in a near-quadratic rise in flux, the rise was asymptotic for outer mouth radii increase. Probability maps and average force per particle per pore section explain the asymmetric flux with variation in pore shape. Furthermore, the simulation results are in agreement with our in vitro experimental results with HeLa cells in Cx43-Cx45 heterotypic configurations. The presence of asymmetric fluxes can help us to understand effects of the molecular structure of the pore and predict potential differences in vivo.


Asunto(s)
Conexina 43/química , Conexinas/química , Uniones Comunicantes/química , Modelos Moleculares , Conexina 26 , Conexina 43/metabolismo , Conexinas/metabolismo , Uniones Comunicantes/metabolismo , Células HeLa , Humanos
8.
Mol Pharmacol ; 90(4): 469-82, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27502018

RESUMEN

Ginsenoside 20(S)-Rg3 (Rg3) is a steroid glycoside that induces human ether-à-go-go-related gene type 1 (hERG1, Kv11.1) channels to activate at more negative potentials and to deactivate more slowly than normal. However, it is unknown whether this action is unique to hERG1 channels. Here we compare and contrast the mechanisms of actions of Rg3 on hERG1 with three other members of the ether-à-go-go (EAG) K(+) channel gene family, including EAG1 (Kv10.1), ERG3 (Kv11.3), and ELK1 (Kv12.1). All four channel types were heterologously expressed in Xenopus laevis oocytes, and K(+) currents were measured using the two-microelectrode voltage-clamp technique. At a maximally effective concentration, Rg3 shifted the half-point of voltage-dependent activation of currents by -14 mV for ERG1 (EC50 = 414 nM), -20 mV for ERG3 (EC50 = 374 nM), -28 mV for EAG1 (EC50 = 1.18 µM), and more than -100 mV for ELK1 (EC50 = 197 nM) channels. Rg3 also induced slowing of ERG1, ERG3, and ELK1 channel deactivation and accelerated the rate of EAG1 channel activation. A Markov model was developed to simulate gating and the effects of Rg3 on the voltage dependence of activation of hELK1 channels. Understanding the mechanism underlying the action of Rg3 may facilitate the development of more potent and selective EAG family channel activators as therapies for cardiovascular and neural disorders.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/metabolismo , Ginsenósidos/farmacología , Activación del Canal Iónico/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Canales de Potasio Éter-A-Go-Go/química , Humanos , Modelos Biológicos , Ratas , Eliminación de Secuencia , Xenopus laevis
9.
J Cardiovasc Electrophysiol ; 27(10): 1220-1229, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27447370

RESUMEN

INTRODUCTION: Large animal models of progressive atrial fibrosis would provide an attractive platform to study relationship between structural and electrical remodeling in atrial fibrillation (AF). Here we established a new transgenic goat model of AF with cardiac specific overexpression of TGF-ß1 and investigated the changes in the cardiac structure and function leading to AF. METHODS AND RESULTS: Transgenic goats with cardiac specific overexpression of constitutively active TGF-ß1 were generated by somatic cell nuclear transfer. We examined myocardial tissue, ECGs, echocardiographic data, and AF susceptibility in transgenic and wild-type control goats. Transgenic goats exhibited significant increase in fibrosis and myocyte diameters in the atria compared to controls, but not in the ventricles. P-wave duration was significantly greater in transgenic animals starting at 12 months of age, but no significant chamber enlargement was detected, suggesting conduction slowing in the atria. Furthermore, this transgenic goat model exhibited a significant increase in AF vulnerability. Six of 8 transgenic goats (75%) were susceptible to AF induction and exhibited sustained AF (>2 minutes), whereas none of 6 controls displayed sustained AF (P < 0.01). Length of induced AF episodes was also significantly greater in the transgenic group compared to controls (687 ± 212.02 seconds vs. 2.50 ± 0.88 seconds, P < 0.0001), but no persistent or permanent AF was observed. CONCLUSION: A novel transgenic goat model with a substrate for AF was generated. In this model, cardiac overexpression of TGF-ß1 led to an increase in fibrosis and myocyte size in the atria, and to progressive P-wave prolongation. We suggest that these factors underlie increased AF susceptibility.


Asunto(s)
Fibrilación Atrial/metabolismo , Remodelación Atrial , Cabras/genética , Atrios Cardíacos/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Potenciales de Acción , Animales , Animales Modificados Genéticamente , Fibrilación Atrial/genética , Fibrilación Atrial/patología , Fibrilación Atrial/fisiopatología , Biopsia , Ecocardiografía , Electrocardiografía , Fibrosis , Predisposición Genética a la Enfermedad , Atrios Cardíacos/patología , Atrios Cardíacos/fisiopatología , Frecuencia Cardíaca , Humanos , Microscopía Confocal , Fenotipo , Factor de Crecimiento Transformador beta1/genética
10.
NMR Biomed ; 29(10): 1338-49, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27485033

RESUMEN

Diffusion tensor imaging (DTI) has emerged as a promising method for noninvasive quantification of myocardial microstructure. However, the origin and behavior of DTI measurements during myocardial normal development and remodeling remain poorly understood. In this work, conventional and bicompartmental DTI in addition to three-dimensional histological correlation were performed in a sheep model of myocardial development from third trimester to postnatal 5 months of age. Comparing the earliest time points in the third trimester with the postnatal 5 month group, the scalar transverse diffusivities preferentially increased in both left ventricle (LV) and right ventricle (RV): secondary eigenvalues D2 increased by 54% (LV) and 36% (RV), whereas tertiary eigenvalues D3 increased by 85% (LV) and 67% (RV). The longitudinal diffusivity D1 changes were small, which led to a decrease in fractional anisotropy by 41% (LV) and 33% (RV) in 5 month versus fetal hearts. Histological analysis suggested that myocardial development is associated with hyperplasia in the early stages of the third trimester followed by myocyte growth in the later stages up to 5 months of age (increased average myocyte width by 198%, myocyte length by 128%, and decreased nucleus density by 70% between preterm and postnatal 5 month hearts.) In a few histological samples (N = 6), correlations were observed between DTI longitudinal diffusivity and myocyte length (r = 0.86, P < 0.05), and transverse diffusivity and myocyte width (r = 0.96, P < 0.01). Linear regression analysis showed that transverse diffusivities are more affected by changes in myocyte size and nucleus density changes than longitudinal diffusivities, which is consistent with predictions of classical models of diffusion in porous media. Furthermore, primary and secondary DTI eigenvectors during development changed significantly. Collectively, the findings demonstrate a role for DTI to monitor and quantify myocardial development, and potentially cardiac disease. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Envejecimiento/patología , Envejecimiento/fisiología , Imagen de Difusión Tensora/métodos , Corazón Fetal/anatomía & histología , Corazón Fetal/embriología , Animales , Corazón Fetal/diagnóstico por imagen , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Ovinos
11.
PLoS Biol ; 11(12): e1001727, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24311987

RESUMEN

The 3-O-sulfotransferase (3-OST) family catalyzes rare modifications of glycosaminoglycan chains on heparan sulfate proteoglycans, yet their biological functions are largely unknown. Knockdown of 3-OST-7 in zebrafish uncouples cardiac ventricular contraction from normal calcium cycling and electrophysiology by reducing tropomyosin4 (tpm4) expression. Normal 3-OST-7 activity prevents the expansion of BMP signaling into ventricular myocytes, and ectopic activation of BMP mimics the ventricular noncontraction phenotype seen in 3-OST-7 depleted embryos. In 3-OST-7 morphants, ventricular contraction can be rescued by overexpression of tropomyosin tpm4 but not by troponin tnnt2, indicating that tpm4 serves as a lynchpin for ventricular sarcomere organization downstream of 3-OST-7. Contraction can be rescued by expression of 3-OST-7 in endocardium, or by genetic loss of bmp4. Strikingly, BMP misregulation seen in 3-OST-7 morphants also occurs in multiple cardiac noncontraction models, including potassium voltage-gated channel gene, kcnh2, affected in Romano-Ward syndrome and long-QT syndrome, and cardiac troponin T gene, tnnt2, affected in human cardiomyopathies. Together these results reveal 3-OST-7 as a key component of a novel pathway that constrains BMP signaling from ventricular myocytes, coordinates sarcomere assembly, and promotes cardiac contractile function.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Contracción Miocárdica/fisiología , Sulfotransferasas/fisiología , Proteínas de Pez Cebra/fisiología , Potenciales de Acción/fisiología , Animales , Técnicas de Silenciamiento del Gen , Desarrollo de Músculos/fisiología , Miocitos Cardíacos/fisiología , Sarcómeros/fisiología , Transducción de Señal/fisiología , Tropomiosina/fisiología , Pez Cebra
12.
J Mol Cell Cardiol ; 80: 186-95, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25633834

RESUMEN

Gain-of-function mutations in CACNA1C, encoding the L-type Ca(2+) channel Cav1.2, cause Timothy syndrome (TS), a multi-systemic disorder with dysmorphic features, long-QT syndrome (LQTS) and autism spectrum disorders. TS patients have heterozygous mutations (G402S and G406R) located in the alternatively spliced exon 8, causing a gain-of-function by reduced voltage-dependence of inactivation. Screening 540 unrelated patients with non-syndromic forms of LQTS, we identified six functional relevant CACNA1C mutations in different regions of the channel. All these mutations caused a gain-of-function combining different mechanisms, including changes in current amplitude, rate of inactivation and voltage-dependence of activation or inactivation, similar as in TS. Computer simulations support the theory that the novel CACNA1C mutations prolong action potential duration. We conclude that genotype-negative LQTS patients should be investigated for mutations in CACNA1C, as a gain-of-function in Cav1.2 is likely to cause LQTS and only specific and rare mutations, i.e. in exon 8, cause the multi-systemic TS.


Asunto(s)
Canales de Calcio Tipo L/genética , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/fisiopatología , Mutación , Potenciales de Acción , Adolescente , Adulto , Sustitución de Aminoácidos , Trastorno Autístico/genética , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Línea Celular , Niño , Preescolar , Análisis Mutacional de ADN , Electrocardiografía , Femenino , Expresión Génica , Variación Genética , Humanos , Lactante , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/metabolismo , Masculino , Linaje , Polimorfismo de Nucleótido Simple , Dominios y Motivos de Interacción de Proteínas , Sindactilia/genética , Adulto Joven
13.
J Mol Cell Cardiol ; 68: 1-11, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24389341

RESUMEN

Excitation-contraction coupling in cardiomyocytes requires Ca(2+) influx through dihydropyridine receptors in the sarcolemma, which gates Ca(2+) release through sarcoplasmic ryanodine receptors (RyRs). Ca(2+) influx, release and diffusion produce a cytosolic Ca(2+) transient. Here, we investigated the relationship between Ca(2+) transients and the spatial arrangement of the sarcolemma including the transverse tubular system (t-system). To accomplish this, we studied isolated ventricular myocytes of rabbit, which exhibit a heterogeneously distributed t-system. We developed protocols for fluorescent labeling and triggered two-dimensional confocal microscopic imaging with high spatiotemporal resolution. From sequences of microscopic images, we measured maximal upstroke velocities and onset times of local Ca(2+) transients together with their distance from the sarcolemma. Analyses indicate that not only sarcolemmal release sites, but also those that are within 1 µm of the sarcolemma actively release Ca(2+). Our data also suggest that release does not occur at sites further than 2.5 µm from the sarcolemma. The experimental data are in agreement with results from a mathematical model of Ca(2+) release and diffusion. Our findings can be explained by a modified local control model, which constrains the region of regenerative activation of non-junctional RyR clusters. We believe that this model will be useful for describing excitation-contraction coupling in cardiac myocytes with a sparse t-system, which includes those from diseased heart tissue as well as atrial myocytes of some species.


Asunto(s)
Acoplamiento Excitación-Contracción , Miocitos Cardíacos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Sarcolema/metabolismo , Animales , Células Cultivadas , Simulación por Computador , Ventrículos Cardíacos/citología , Modelos Biológicos , Contracción Miocárdica , Miocitos Cardíacos/ultraestructura , Conejos , Sarcolema/ultraestructura
14.
J Mol Cell Cardiol ; 72: 186-95, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24657727

RESUMEN

Sarcomeres are the basic contractile units of cardiac myocytes. Recent studies demonstrated remodeling of sarcomeric proteins in several diseases, including genetic defects and heart failure. Here we investigated remodeling of sarcomeric α-actinin in two models of heart failure, synchronous (SHF) and dyssynchronous heart failure (DHF), as well as a model of cardiac resynchronization therapy (CRT). We applied three-dimensional confocal microscopy and quantitative methods of image analysis to study isolated cells from our animal models. 3D Fourier analysis revealed a decrease of the spatial regularity of the α-actinin distribution in both SHF and DHF versus control cells. The spatial regularity of α-actinin in DHF cells was reduced when compared with SHF cells. The spatial regularity of α-actinin was partially restored after CRT. We found longitudinal depositions of α-actinin in SHF, DHF and CRT cells. These depositions spanned adjacent Z-disks and exhibited a lower density of α-actinin than in the Z-disk. Differences in the occurrence of depositions between the SHF, CRT and DHF models versus control were significant. Also, CRT cells exhibited a higher occurrence of depositions versus SHF, but not DHF cells. Other sarcomeric proteins did not accumulate in the depositions to the same extent as α-actinin. We did not find differences in the expression of α-actinin protein and its encoding gene in our animal models. In summary, our studies indicate that HF is associated with two different types of remodeling of α-actinin and only one of those was reversed after CRT. We suggest that these results can guide us to an understanding of remodeling of structures and function associated with sarcomeres.


Asunto(s)
Actinina/química , Terapia de Resincronización Cardíaca , Citoesqueleto/ultraestructura , Insuficiencia Cardíaca/terapia , Ventrículos Cardíacos/ultraestructura , Miocitos Cardíacos/ultraestructura , Actinina/genética , Actinina/metabolismo , Animales , Citoesqueleto/metabolismo , Citoesqueleto/patología , Perros , Expresión Génica , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Masculino , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Sarcómeros/metabolismo , Sarcómeros/patología , Sarcómeros/ultraestructura , Remodelación Ventricular
15.
NMR Biomed ; 27(11): 1378-86, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25200106

RESUMEN

Non-invasive imaging techniques are highly desirable as an alternative to conventional biopsy for the characterization of the remodeling of tissues associated with disease progression, including end-stage heart failure. Cardiac diffusion tensor imaging (DTI) has become an established method for the characterization of myocardial microstructure. However, the relationships between diffuse myocardial fibrosis, which is a key biomarker for staging and treatment planning of the failing heart, and measured DTI parameters have yet to be investigated systematically. In this study, DTI was performed on left ventricular specimens collected from patients with chronic end-stage heart failure as a result of idiopathic dilated cardiomyopathy (n = 14) and from normal donors (n = 5). Scalar DTI parameters, including fractional anisotropy (FA) and mean (MD), primary (D1 ), secondary (D2 ) and tertiary (D3 ) diffusivities, were correlated with collagen content measured by digital microscopy. Compared with hearts from normal subjects, the FA in failing hearts decreased by 22%, whereas the MD, D2 and D3 increased by 12%, 14% and 24%, respectively (P < 0.01). No significant change was detected for D1 between the two groups. Furthermore, significant correlation was observed between the DTI scalar indices and quantitative histological measurements of collagen (i.e. fibrosis). Pearson's correlation coefficients (r) between collagen content and FA, MD, D2 and D3 were -0.51, 0.59, 0.56 and 0.62 (P < 0.05), respectively. The correlation between D1 and collagen content was not significant (r = 0.46, P = 0.05). Computational modeling analysis indicated that the behaviors of the DTI parameters as a function of the degree of fibrosis were well explained by compartmental exchange between myocardial and collagenous tissues. Combined, these findings suggest that scalar DTI parameters can be used as metrics for the non-invasive assessment of diffuse fibrosis in failing hearts.


Asunto(s)
Imagen de Difusión Tensora/métodos , Insuficiencia Cardíaca/patología , Miocardio/patología , Adulto , Anciano , Anisotropía , Biopsia , Cardiomiopatía Dilatada/complicaciones , Cardiomiopatía Dilatada/patología , Colágeno/análisis , Simulación por Computador , Femenino , Fibrosis , Ventrículos Cardíacos/química , Ventrículos Cardíacos/patología , Humanos , Masculino , Persona de Mediana Edad , Modelos Cardiovasculares , Método de Montecarlo , Miocardio/química , Adulto Joven
16.
Circ Res ; 110(4): 588-97, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22253411

RESUMEN

RATIONALE: Cardiac resynchronization therapy (CRT) is an established treatment for patients with chronic heart failure. However, CRT-associated structural and functional remodeling at cellular and subcellular levels is only partly understood. OBJECTIVE: To investigate the effects of CRT on subcellular structures and protein distributions associated with excitation-contraction coupling of ventricular cardiomyocytes. METHODS AND RESULTS: Our studies revealed remodeling of the transverse tubular system (t-system) and the spatial association of ryanodine receptor (RyR) clusters in a canine model of dyssynchronous heart failure (DHF). We did not find this remodeling in a synchronous heart failure model based on atrial tachypacing. Remodeling in DHF ranged from minor alterations in anterior left ventricular myocytes to nearly complete loss of the t-system and dissociation of RyRs from sarcolemmal structures in lateral cells. After CRT, we found a remarkable and almost complete reverse remodeling of these structures despite persistent left ventricular dysfunction. Studies of whole-cell Ca(2+) transients showed that the structural remodeling and restoration were accompanied with remodeling and restoration of Ca(2+) signaling. CONCLUSIONS: DHF is associated with regional remodeling of the t-system. Myocytes undergo substantial structural and functional restoration after only 3 weeks of CRT. The finding suggests that t-system status can provide an early marker of the success of this therapy. The results could also guide us to an understanding of the loss and remodeling of proteins associated with the t-system. The steep relationship between free Ca(2+) and contraction suggests that some restoration of Ca(2+) release units will have a disproportionately large effect on contractility.


Asunto(s)
Calcio/metabolismo , Terapia de Resincronización Cardíaca , Acoplamiento Excitación-Contracción , Insuficiencia Cardíaca/terapia , Miocitos Cardíacos/metabolismo , Disfunción Ventricular Izquierda/terapia , Función Ventricular Izquierda , Remodelación Ventricular , Animales , Modelos Animales de Enfermedad , Perros , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Masculino , Potenciales de la Membrana , Contracción Miocárdica , Miocitos Cardíacos/patología , Recuperación de la Función , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Sarcolema/metabolismo , Factores de Tiempo , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
17.
J Biomed Opt ; 29(6): 066005, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38841076

RESUMEN

Significance: Damage to the cardiac conduction system remains one of the most significant risks associated with surgical interventions to correct congenital heart disease. This work demonstrates how light-scattering spectroscopy (LSS) can be used to non-destructively characterize cardiac tissue regions. Aim: To present an approach for associating tissue composition information with location-specific LSS data and further evaluate an LSS and machine learning system as a method for non-destructive tissue characterization. Approach: A custom LSS probe was used to gather spectral data from locations across 14 excised human pediatric nodal tissue samples (8 sinus nodes, 6 atrioventricular nodes). The LSS spectra were used to train linear and neural-network-based regressor models to predict tissue composition characteristics derived from the 3D models. Results: Nodal tissue region nuclear densities were reported. A linear model trained to regress nuclear density from spectra achieved a prediction r-squared of 0.64 and a concordance correlation coefficient of 0.78. Conclusions: These methods build on previous studies suggesting that LSS measurements combined with machine learning signal processing can provide clinically relevant cardiac tissue composition.


Asunto(s)
Dispersión de Radiación , Análisis Espectral , Humanos , Análisis Espectral/métodos , Aprendizaje Automático , Luz , Corazón/diagnóstico por imagen , Miocardio/química
18.
Cardiovasc Pathol ; 70: 107626, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38458505

RESUMEN

Iatrogenic damage to the cardiac conduction system (CCS) remains a significant risk during congenital heart surgery. Current surgical best practice involves using superficial anatomical landmarks to locate and avoid damaging the CCS. Prior work indicates inherent variability in the anatomy of the CCS and supporting tissues. This study introduces high-resolution, 3D models of the CCS in normal pediatric human hearts to evaluate variability in the nodes and surrounding structures. Human pediatric hearts were obtained with an average donor age of 2.7 days. A pipeline was developed to excise, section, stain, and image atrioventricular (AVN) and sinus nodal (SN) tissue regions. A convolutional neural network was trained to enable precise multi-class segmentation of whole-slide images, which were subsequently used to generate high- resolution 3D tissue models. Nodal tissue region models were created. All models (10 AVN, 8 SN) contain tissue composition of neural tissue, vasculature, and nodal tissues at micrometer resolution. We describe novel nodal anatomical variations. We found that the depth of the His bundle in females was on average 304 µm shallower than those of male patients. These models provide surgeons with insight into the heterogeneity of the nodal regions and the intricate relationships between the CCS and surrounding structures.


Asunto(s)
Nodo Atrioventricular , Imagenología Tridimensional , Humanos , Femenino , Masculino , Recién Nacido , Nodo Atrioventricular/anatomía & histología , Modelos Cardiovasculares , Nodo Sinoatrial/anatomía & histología , Fascículo Atrioventricular/fisiopatología , Redes Neurales de la Computación , Factores Sexuales , Factores de Edad , Sistema de Conducción Cardíaco/fisiopatología
19.
J Physiol ; 591(17): 4273-86, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23652593

RESUMEN

Choline (Ch) is a precursor and metabolite of the neurotransmitter acetylcholine (ACh). In canine and guinea pig atrial myocytes, Ch was shown to activate an outward K(+) current in a delayed rectifier fashion. This current has been suggested to modulate cardiac electrical activity and to play a role in atrial fibrillation pathophysiology. However, the exact nature and identity of this current has not been convincingly established. We recently described the unique ligand- and voltage-dependent properties of muscarinic activation of ACh-activated K(+) current (IKACh) and showed that, in contrast to ACh, pilocarpine induces a current with delayed rectifier-like properties with membrane depolarization. Here, we tested the hypothesis that Ch activates IKACh in feline atrial myocytes in a voltage-dependent manner similar to pilocarpine. Single-channel recordings, biophysical profiles, specific pharmacological inhibition and computational data indicate that the current activated by Ch is IKACh. Moreover, we show that membrane depolarization increases the potency and efficacy of IKACh activation by Ch and thus gives the appearance of a delayed rectifier activating K(+) current at depolarized potentials. Our findings support the emerging concept that IKACh modulation is both voltage- and ligand-specific and reinforce the importance of these properties in understanding cardiac physiology.


Asunto(s)
Potenciales de Acción , Canales de Potasio de Tipo Rectificador Tardío/metabolismo , Atrios Cardíacos/metabolismo , Miocitos Cardíacos/metabolismo , Receptor Muscarínico M2/metabolismo , Animales , Gatos , Colina/farmacología , Femenino , Atrios Cardíacos/citología , Masculino , Potenciales de la Membrana , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Pilocarpina/farmacología
20.
Exp Physiol ; 98(3): 746-57, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23180812

RESUMEN

Insulin resistance, which characterizes type 2 diabetes, is associated with reduced translocation of glucose transporter 4 (GLUT4) to the plasma membrane following insulin stimulation, and diabetic patients with insulin resistance show a higher incidence of ischaemia, arrhythmias and sudden cardiac death. The aim of this study was to examine whether GLUT4 deficiency leads to more severe alterations in cardiac electrical activity during cardiac stress due to hypoxia. To fulfil this aim, we compared cardiac electrical activity from cardiac-selective GLUT4-ablated (G4H-/-) mouse hearts and corresponding control (CTL) littermates. A custom-made cylindrical 'cage' electrode array measured potentials (Ves) from the epicardium of isolated, perfused mouse hearts. The normalized average of the maximal downstroke of Ves ( (|d Ves/dt(min)|na), which we previously introduced as an index of electrical activity in normal, ischaemic and hypoxic hearts, was used to assess the effects of GLUT4 deficiency on electrical activity. The |d Ves/dt(min)|na of G4H −/− and CTL hearts decreased by 75 and 47%, respectively (P < 0.05), 30 min after the onset of hypoxia. Administration of insulin attenuated decreases in values of |d Ves/dt(min)|na in G4H −/− hearts as well as in CTL hearts, during hypoxia. In general, however, G4H −/− hearts showed a severe alteration of the propagation sequence and a prolonged total activation time. Results of this study demonstrate that reduced glucose availability associated with insulin resistance and a reduction in GLUT4-mediated glucose transport impairs electrical activity during hypoxia, and may contribute to cardiac vulnerability to arrhythmias in diabetic patients.


Asunto(s)
Transportador de Glucosa de Tipo 4/deficiencia , Corazón/fisiología , Hipoxia/fisiopatología , Potenciales de Acción/fisiología , Animales , Femenino , Glucosa/metabolismo , Técnicas In Vitro , Resistencia a la Insulina/fisiología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA