Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 6550, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39095365

RESUMEN

The cardiac conduction system (CCS) is a network of specialized cardiomyocytes that coordinates electrical impulse generation and propagation for synchronized heart contractions. Although the components of the CCS, including the sinoatrial node, atrioventricular node, His bundle, bundle branches, and Purkinje fibers, were anatomically discovered more than 100 years ago, their molecular constituents and regulatory mechanisms remain incompletely understood. Here, we demonstrate the transcriptomic landscape of the postnatal mouse CCS at a single-cell resolution with spatial information. Integration of single-cell and spatial transcriptomics uncover region-specific markers and zonation patterns of expression. Network inference shows heterogeneous gene regulatory networks across the CCS. Notably, region-specific gene regulation is recapitulated in vitro using neonatal mouse atrial and ventricular myocytes overexpressing CCS-specific transcription factors, Tbx3 and/or Irx3. This finding is supported by ATAC-seq of different CCS regions, Tbx3 ChIP-seq, and Irx motifs. Overall, this study provides comprehensive molecular profiles of the postnatal CCS and elucidates gene regulatory mechanisms contributing to its heterogeneity.


Asunto(s)
Sistema de Conducción Cardíaco , Proteínas de Homeodominio , Miocitos Cardíacos , Proteínas de Dominio T Box , Animales , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Redes Reguladoras de Genes , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Regulación de la Expresión Génica , Animales Recién Nacidos , Análisis de la Célula Individual , Transcriptoma , Ramos Subendocárdicos/metabolismo , Ramos Subendocárdicos/fisiología , Nodo Atrioventricular/metabolismo , Nodo Sinoatrial/metabolismo , Fascículo Atrioventricular/metabolismo
2.
Brain Imaging Behav ; 13(4): 973-984, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29934818

RESUMEN

This study aims to detect the neural substrate underlying the language impairment in children with developmental language disorder (DLD) using diffusion tensor imaging (DTI) tractography. Deterministic DTI tractography was performed in a group of right-handed children with DLD (N = 17; mean age 10;07 ± 2;01 years) and a typically developing control group matched for age, gender and handedness (N = 22; mean age 11;00 ± 1;11 years) to bilaterally identify the superior longitudinal fascicle, arcuate fascicle, anterior lateral segment and posterior lateral segment (also called dorsal language network) and the middle and inferior longitudinal fascicle, extreme capsule fiber system and uncinate fascicle (also called ventral language network). Language skills were assessed using an extensive, standardized test battery. Differences in language performance, white matter organization and structural lateralization of the language network were statistically analyzed. Children with DLD showed a higher overall volume and higher ADC values for the left-hemispheric language related WM tracts. In addition, in children with DLD, the majority (88%; 7/8) of the studied language related WM tracts did not show a significant left or right lateralization pattern. These structural alterations might underlie the language impairment in children with DLD.


Asunto(s)
Imagen de Difusión Tensora/métodos , Trastornos del Desarrollo del Lenguaje/diagnóstico por imagen , Trastornos del Desarrollo del Lenguaje/fisiopatología , Encéfalo/diagnóstico por imagen , Encéfalo/fisiología , Mapeo Encefálico/métodos , Fascículo Atrioventricular/metabolismo , Fascículo Atrioventricular/fisiología , Corteza Cerebral/fisiología , Niño , Conectoma/métodos , Femenino , Humanos , Lenguaje , Trastornos del Desarrollo del Lenguaje/metabolismo , Imagen por Resonancia Magnética/métodos , Masculino , Red Nerviosa/fisiología , Vías Nerviosas/fisiología , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/fisiología
3.
Development ; 145(17)2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-30042181

RESUMEN

A small network of spontaneously active Tbx3+ cardiomyocytes forms the cardiac conduction system (CCS) in adults. Understanding the origin and mechanism of development of the CCS network are important steps towards disease modeling and the development of biological pacemakers to treat arrhythmias. We found that Tbx3 expression in the embryonic mouse heart is associated with automaticity. Genetic inducible fate mapping revealed that Tbx3+ cells in the early heart tube are fated to form the definitive CCS components, except the Purkinje fiber network. At mid-fetal stages, contribution of Tbx3+ cells was restricted to the definitive CCS. We identified a Tbx3+ population in the outflow tract of the early heart tube that formed the atrioventricular bundle. Whereas Tbx3+ cardiomyocytes also contributed to the adjacent Gja5+ atrial and ventricular chamber myocardium, embryonic Gja5+ chamber cardiomyocytes did not contribute to the Tbx3+ sinus node or to atrioventricular ring bundles. In conclusion, the CCS is established by progressive fate restriction of a Tbx3+ cell population in the early developing heart, which implicates Tbx3 as a useful tool for developing strategies to study and treat CCS diseases.


Asunto(s)
Fascículo Atrioventricular/embriología , Miocitos Cardíacos/metabolismo , Proteínas de Dominio T Box/metabolismo , Animales , Fascículo Atrioventricular/metabolismo , Conexinas/metabolismo , Técnicas de Cultivo de Embriones , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/citología , Organogénesis/fisiología , Proteínas de Dominio T Box/genética , Proteína alfa-5 de Unión Comunicante
4.
Elife ; 72018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29565246

RESUMEN

Mammals and birds have a specialized cardiac atrioventricular conduction system enabling rapid activation of both ventricles. This system may have evolved together with high heart rates to support their endothermic state (warm-bloodedness) and is seemingly lacking in ectothermic vertebrates from which first mammals then birds independently evolved. Here, we studied the conduction system in crocodiles (Alligator mississippiensis), the only ectothermic vertebrates with a full ventricular septum. We identified homologues of mammalian conduction system markers (Tbx3-Tbx5, Scn5a, Gja5, Nppa-Nppb) and show the presence of a functional atrioventricular bundle. The ventricular Purkinje network, however, was absent and slow ventricular conduction relied on trabecular myocardium, as it does in other ectothermic vertebrates. We propose the evolution of the atrioventricular bundle followed full ventricular septum formation prior to the development of high heart rates and endothermy. In contrast, the evolution of the ventricular Purkinje network is strongly associated with high heart rates and endothermy.


Asunto(s)
Caimanes y Cocodrilos/fisiología , Sistema de Conducción Cardíaco/fisiología , Frecuencia Cardíaca/fisiología , Corazón/fisiología , Caimanes y Cocodrilos/embriología , Caimanes y Cocodrilos/genética , Animales , Fascículo Atrioventricular/embriología , Fascículo Atrioventricular/metabolismo , Fascículo Atrioventricular/fisiología , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Sistema de Conducción Cardíaco/embriología , Frecuencia Cardíaca/genética , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/metabolismo , Hibridación in Situ , Modelos Cardiovasculares , Ramos Subendocárdicos/embriología , Ramos Subendocárdicos/metabolismo , Ramos Subendocárdicos/fisiología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Tabique Interventricular/embriología , Tabique Interventricular/metabolismo , Tabique Interventricular/fisiología
5.
J Am Heart Assoc ; 2(6): e000246, 2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24356527

RESUMEN

BACKGROUND: The cardiac conduction system consists of the sinus node, nodal extensions, atrioventricular (AV) node, penetrating bundle, bundle branches, and Purkinje fibers. Node-like AV ring tissue also exists at the AV junctions, and the right and left rings unite at the retroaortic node. The study aims were to (1) construct a 3-dimensional anatomical model of the AV rings and retroaortic node, (2) map electrical activation in the right ring and study its action potential characteristics, and (3) examine gene expression in the right ring and retroaortic node. METHODS AND RESULTS: Three-dimensional reconstruction (based on magnetic resonance imaging, histology, and immunohistochemistry) showed the extent and organization of the specialized tissues (eg, how the AV rings form the right and left nodal extensions into the AV node). Multiextracellular electrode array and microelectrode mapping of isolated right ring preparations revealed robust spontaneous activity with characteristic diastolic depolarization. Using laser microdissection gene expression measured at the mRNA level (using quantitative PCR) and protein level (using immunohistochemistry and Western blotting) showed that the right ring and retroaortic node, like the sinus node and AV node but, unlike ventricular muscle, had statistically significant higher expression of key transcription factors (including Tbx3, Msx2, and Id2) and ion channels (including HCN4, Cav3.1, Cav3.2, Kv1.5, SK1, Kir3.1, and Kir3.4) and lower expression of other key ion channels (Nav1.5 and Kir2.1). CONCLUSIONS: The AV rings and retroaortic node possess gene expression profiles similar to that of the AV node. Ion channel expression and electrophysiological recordings show the AV rings could act as ectopic pacemakers and a source of atrial tachycardia.


Asunto(s)
Sistema de Conducción Cardíaco/metabolismo , Potenciales de Acción/fisiología , Animales , Nodo Atrioventricular/anatomía & histología , Nodo Atrioventricular/metabolismo , Nodo Atrioventricular/fisiología , Fascículo Atrioventricular/anatomía & histología , Fascículo Atrioventricular/metabolismo , Fascículo Atrioventricular/fisiología , Sistema de Conducción Cardíaco/anatomía & histología , Sistema de Conducción Cardíaco/fisiología , Modelos Anatómicos , Proteoma , Ramos Subendocárdicos/anatomía & histología , Ramos Subendocárdicos/metabolismo , Ramos Subendocárdicos/fisiología , Ratas , Nodo Sinoatrial/anatomía & histología , Nodo Sinoatrial/metabolismo , Nodo Sinoatrial/fisiología , Transcriptoma
6.
Dev Dyn ; 242(6): 665-77, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23526457

RESUMEN

BACKGROUND: The ventricular conduction system (VCS) coordinates the heartbeat and is composed of central components (the atrioventricular node, bundle, and right and left bundle branches) and a peripheral Purkinje fiber network. Conductive myocytes develop from common progenitor cells with working myocytes in a bimodal process of lineage restriction followed by limited outgrowth. The lineage relationship between progenitor cells giving rise to different components of the VCS is unclear. RESULTS: Cell lineage contributions to different components of the VCS were analysed by a combination of retrospective clonal analysis, regionalized transgene expression studies, and genetic tracing experiments using Connexin40-GFP mice that precisely delineate the VCS. Analysis of a library of hearts containing rare large clusters of clonally related myocytes identifies two VCS lineages encompassing either the right Purkinje fiber network or left bundle branch. Both lineages contribute to the atrioventricular bundle and right bundle branch that segregate early from working myocytes. Right and left VCS lineages share the transcriptional program of the respective ventricular working myocytes and genetic tracing experiments discount alternate progenitor cell contributions to the VCS. CONCLUSIONS: The mammalian VCS is comprised of cells derived from two lineages, supporting a dual contribution of first and second heart field progenitor cells.


Asunto(s)
Linaje de la Célula , Conexinas/genética , Sistema de Conducción Cardíaco/embriología , Ventrículos Cardíacos/embriología , Células Madre/citología , Alelos , Animales , Fascículo Atrioventricular/metabolismo , Conexinas/fisiología , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Microscopía Fluorescente , Miocardio/citología , Miocitos Cardíacos/citología , Factores de Tiempo , Transcripción Genética , Transgenes , Proteína alfa-5 de Unión Comunicante
7.
Dev Biol ; 373(1): 149-62, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23085412

RESUMEN

Regional differences in cardiomyocyte automaticity permit the sinoatrial node (SAN) to function as the leading cardiac pacemaker and the atrioventricular (AV) junction as a subsidiary pacemaker. The regulatory mechanisms controlling the distribution of automaticity within the heart are not understood. To understand regional variation in cardiac automaticity, we carried out an in vivo analysis of cis-regulatory elements that control expression of the hyperpolarization-activated cyclic-nucleotide gated ion channel 4 (Hcn4). Using transgenic mice, we found that spatial and temporal patterning of Hcn4 expression in the AV conduction system required cis-regulatory elements with multiple conserved fragments. One highly conserved region, which contained a myocyte enhancer factor 2C (Mef2C) binding site previously described in vitro, induced reporter expression specifically in the embryonic non-chamber myocardium and the postnatal AV bundle in a Mef2c-dependent manner in vivo. Inhibition of histone deacetylase (HDAC) activity in cultured transgenic embryos showed expansion of reporter activity to working myocardium. In adult animals, hypertrophy induced by transverse aortic constriction, which causes translocation of HDACs out of the nucleus, resulted in ectopic activation of the Hcn4 enhancer in working myocardium, recapitulating pathological electrical remodeling. These findings reveal mechanisms that control the distribution of automaticity among cardiomyocytes during development and in response to stress.


Asunto(s)
Fascículo Atrioventricular/embriología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Histona Desacetilasas/metabolismo , Factores Reguladores Miogénicos/metabolismo , Nodo Sinoatrial/embriología , Animales , Fascículo Atrioventricular/metabolismo , Cardiomegalia/genética , Ensayo de Cambio de Movilidad Electroforética , Elementos de Facilitación Genéticos/genética , Galactósidos , Regulación del Desarrollo de la Expresión Génica/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Inmunohistoquímica , Hibridación in Situ , Indoles , Luciferasas , Factores de Transcripción MEF2 , Ratones , Ratones Transgénicos , Factores Reguladores Miogénicos/genética , Reacción en Cadena de la Polimerasa , Nodo Sinoatrial/metabolismo
8.
Cardiovasc Res ; 95(4): 469-79, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22739121

RESUMEN

AIMS: The aim of this study was to characterize ventricular activation patterns in normal and connexin40-deficient mice in order to dissect the role of connexin40 in developing the conduction system. METHODS AND RESULTS: We performed optical mapping of epicardial activation between ED9.5-18.5 and analysed ventricular activation patterns and times of left ventricular activation. Mouse embryos deficient for connexin40 were compared with normal and heterozygous littermates. Morphology of the primary interventricular ring (PIR) was delineated with the help of T3-LacZ transgene. Four major types of ventricular activation patterns characterized by primary breakthrough in different parts of the heart were detected during development: PIR, left ventricular apex, right ventricular apex, and dual right and left ventricular apices. Activation through PIR was frequently present at the early stages until ED12.5. From ED14.5, the majority of hearts showed dual left and right apical breakthrough, suggesting functionality of both bundle branches. Connexin40-deficient embryos showed initially a delay in left bundle branch function, but the right bundle branch block, previously described in the adults, was not detected in ED14.5 embryos and appeared only gradually with 80% penetrance at ED18.5. CONCLUSION: The switch of function from the early PIR conduction pathway to the mature apex to base activation is dependent upon upregulation of connexin40 expression in the ventricular trabeculae. The early function of right bundle branch does not depend on connexin40. Quantitative analysis of normal mouse embryonic ventricular conduction patterns will be useful for interpretation of effects of mutations affecting the function of the cardiac conduction system.


Asunto(s)
Conexinas/deficiencia , Sistema de Conducción Cardíaco/metabolismo , Ventrículos Cardíacos/metabolismo , Potenciales de Acción , Animales , Fascículo Atrioventricular/embriología , Fascículo Atrioventricular/metabolismo , Bloqueo de Rama/genética , Bloqueo de Rama/metabolismo , Conexinas/genética , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Sistema de Conducción Cardíaco/embriología , Ventrículos Cardíacos/embriología , Operón Lac , Ratones , Ratones Noqueados , Ratones Transgénicos , Morfogénesis , Penetrancia , Imagen de Colorante Sensible al Voltaje , Proteína alfa-5 de Unión Comunicante
9.
Circ Arrhythm Electrophysiol ; 5(1): 163-72, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22247482

RESUMEN

BACKGROUND: Progressive familial heart block type I (PFHBI) is a hereditary arrhythmia characterized by progressive conduction disturbances in the His-Purkinje system. PFHBI has been linked to genes such as SCN5A that influence cardiac excitability but not to genes that influence cell-to-cell communication. Our goal was to explore whether nucleotide substitutions in genes coding for connexin proteins would associate with clinical cases of PFHBI and if so, to establish a genotype-cell phenotype correlation for that mutation. METHODS AND RESULTS: We screened 156 probands with PFHBI. In addition to 12 sodium channel mutations, we found a germ line GJA5 (connexin40 [Cx40]) mutation (Q58L) in 1 family. Heterologous expression of Cx40-Q58L in connexin-deficient neuroblastoma cells resulted in marked reduction of junctional conductance (Cx40-wild type [WT], 22.2±1.7 nS, n=14; Cx40-Q58L, 0.56±0.34 nS, n=14; P<0.001) and diffuse localization of immunoreactive proteins in the vicinity of the plasma membrane without formation of gap junctions. Heteromeric cotransfection of Cx40-WT and Cx40-Q58L resulted in homogenous distribution of proteins in the plasma membrane rather than in membrane plaques in ≈50% of cells; well-defined gap junctions were observed in other cells. Junctional conductance values correlated with the distribution of gap junction plaques. CONCLUSIONS: Mutation Cx40-Q58L impairs gap junction formation at cell-cell interfaces. This is the first demonstration of a germ line mutation in a connexin gene that associates with inherited ventricular arrhythmias and emphasizes the importance of Cx40 in normal propagation in the specialized conduction system.


Asunto(s)
Fascículo Atrioventricular/metabolismo , Conexinas/genética , ADN/genética , Bloqueo Cardíaco/genética , Mutación , Biomarcadores/metabolismo , Western Blotting , Fascículo Atrioventricular/fisiopatología , Trastorno del Sistema de Conducción Cardíaco , Niño , Conexinas/metabolismo , Electrocardiografía , Técnicas Electrofisiológicas Cardíacas , Femenino , Predisposición Genética a la Enfermedad , Bloqueo Cardíaco/metabolismo , Bloqueo Cardíaco/fisiopatología , Frecuencia Cardíaca , Humanos , Inmunohistoquímica , Masculino , Linaje , Reacción en Cadena de la Polimerasa , Pronóstico , Proteína alfa-5 de Unión Comunicante
11.
J Mol Cell Cardiol ; 51(5): 689-701, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21741388

RESUMEN

Functioning of the cardiac conduction system depends critically on its structure and its complement of ion channels. Therefore, the aim of this study was to document both the structure and ion channel expression of the left and right ventricular His-Purkinje networks, as we have previously done for the sinoatrial and atrioventricular nodes. A three-dimensional (3D) anatomical computer model of the His-Purkinje network of the rabbit heart was constructed after staining the network by immunoenzyme labelling of a marker protein, middle neurofilament. The bundle of His is a ribbon-like structure and the architecture of the His-Purkinje network differs between the left and right ventricles. The 3D model is able to explain the breakthrough points of the action potential on the ventricular epicardium during sinus rhythm. Using quantitative PCR, the expression levels of the major ion channels were measured in the free running left and right Purkinje fibres of the rabbit heart. Expression of ion channels differs from that of the working myocardium and can explain the specialised electrical activity of the Purkinje fibres as suggested by computer simulations; the expression profile of the left Purkinje fibres is more specialised than that of the right Purkinje fibres. The structure and ion channel expression of the Purkinje fibres are highly specialised and tailored to the functioning of the system. The His-Purkinje network in the left ventricle is more developed than that in the right ventricle and this may explain its greater clinical importance.


Asunto(s)
Potenciales de Acción/fisiología , Ventrículos Cardíacos , Imagenología Tridimensional/métodos , Canales Iónicos/metabolismo , Imagen Molecular/métodos , Miocardio/metabolismo , Ramos Subendocárdicos , Animales , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Fascículo Atrioventricular/anatomía & histología , Fascículo Atrioventricular/metabolismo , Conexinas/genética , Conexinas/metabolismo , Expresión Génica/fisiología , Perfilación de la Expresión Génica , Ventrículos Cardíacos/anatomía & histología , Ventrículos Cardíacos/metabolismo , Inmunohistoquímica , Canales Iónicos/genética , Masculino , Ramos Subendocárdicos/anatomía & histología , Ramos Subendocárdicos/metabolismo , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
Heart Rhythm ; 6(5): 672-80, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19328044

RESUMEN

BACKGROUND: The so-called specialized tissues within the heart are the sinus node, the atrioventricular conduction system, and the Purkinje network. Further structures with the characteristics of specialized tissue are also found within the atrioventricular junction, although they are less well described. OBJECTIVE: The purpose of this study was to demonstrate the location and extent of these atrioventricular ring specialized tissues, showing their relationship with the normal atrioventricular conduction system. METHODS: We identified the tissues using histology combined with immunohistochemical labeling with connexin43 (Cx43), the major gap junction in heart, and HCN4, the major isoform of the funny channel. RESULTS: We observed rings of specialized tissue mainly in hearts from rats, mice, and guinea pigs, negative for Cx43 but positive for HCN4. Each ring takes its origin from an inferior extension of the atrioventricular node. The rightward ring runs around the vestibule of the tricuspid valve, whereas the leftward ring encircles the mitral valve. On returning toward the atrial septum, the tricuspid ring crosses over the penetrating part of the atrioventricular conduction system, reuniting with the mitral ring to form a superiorly located retroaortic node. The atrioventricular conduction system itself continues beyond the origin of the right and left bundle branches, forming an aortic ring that ascends toward the retroaortic node but fails to make contact because of the intervening area of aortic-to-mitral valvar fibrous continuity. CONCLUSION: Rings of conduction tissue take their origin from inferior extensions of the atrioventricular node, passing rightward and leftward to encircle the orifices of the tricuspid and mitral valves and reuniting to form an extensive retroaortic node. Thus, a ring with morphologic features justifying a definition of specialized conduction tissue surrounds the atrioventricular junctions, although its function has yet to be established.


Asunto(s)
Aorta Torácica/inervación , Nodo Atrioventricular/citología , Fascículo Atrioventricular/citología , Animales , Aorta Torácica/citología , Aorta Torácica/metabolismo , Nodo Atrioventricular/metabolismo , Fascículo Atrioventricular/metabolismo , Caveolina 3/metabolismo , Tamaño de la Célula , Conexina 43/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Cobayas , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Inmunohistoquímica , Masculino , Ratones , Canales de Potasio/metabolismo , Ratas , Ratas Wistar
14.
Basic Res Cardiol ; 104(5): 511-22, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19255801

RESUMEN

Cardiac sodium channels are responsible for conduction in the normal and diseased heart. We aimed to investigate regional and transmural distribution of sodium channel expression and function in the myocardium. Sodium channel Scn5a mRNA and Na(v)1.5 protein distribution was investigated in adult and embryonic mouse heart through immunohistochemistry and in situ hybridization. Functional sodium channel availability in subepicardial and subendocardial myocytes was assessed using patch-clamp technique. Adult and embryonic (ED14.5) mouse heart sections showed low expression of Na(v)1.5 in the HCN4-positive sinoatrial and atrioventricular nodes. In contrast, high expression levels of Na(v)1.5 were observed in the HCN4-positive and Cx43-negative AV or His bundle, bundle branches and Purkinje fibers. In both ventricles, a transmural gradient was observed, with a low Na(v)1.5 labeling intensity in the subepicardium as compared to the subendocardium. Similar Scn5a mRNA expression patterns were observed on in situ hybridization of embryonic and adult tissue. Maximal action potential upstroke velocity was significantly lower in subepicardial myocytes (mean +/- SEM 309 +/- 32 V/s; n = 14) compared to subendocardial myocytes (394 +/- 32 V/s; n = 11; P < 0.05), indicating decreased sodium channel availability in subepicardium compared to subendocardium. Scn5a and Na(v)1.5 show heterogeneous distribution patterns within the cardiac conduction system and across the ventricular wall. This differential distribution of the cardiac sodium channel may have profound consequences for conduction disease phenotypes and arrhythmogenesis in the setting of sodium channel disease.


Asunto(s)
Sistema de Conducción Cardíaco/metabolismo , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Canales de Sodio/metabolismo , Potenciales de Acción , Animales , Nodo Atrioventricular/metabolismo , Fascículo Atrioventricular/metabolismo , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Sistema de Conducción Cardíaco/embriología , Ventrículos Cardíacos/metabolismo , Humanos , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Proteínas Musculares/genética , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp , Ramos Subendocárdicos/metabolismo , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Canales de Sodio/genética , Transfección
15.
Heart Rhythm ; 4(4): 499-507, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17399641

RESUMEN

BACKGROUND: Pulmonary veins (PVs) and the coronary sinus (CS) play pivotal roles in triggering some episodes of atrial fibrillation. In isolated rabbit right or left atrial preparations, a 3-hour intermittent burst pacing protocol shortens action potential duration (APD) in CS and PV, but not in sinus node (SN) and left Bachmann bundle (BB) regions. OBJECTIVE: The purpose of this study was to use patch clamp techniques to study the rapidly inactivating (I(to)) and sustained (I(sus)) K(+) currents as well as Ca(2+) currents (I(Ca)) in cells dispersed from intermittent burst pacing and sham PV, BB, CS, and SN regions to determine whether changes in these currents contributed to APD shortening. METHODS: Real-time polymerase chain reaction was performed for transient outward K(+) and Ca(2+) channel subunit mRNAs to determine if intermittent burst pacing affected expression levels. RESULTS: I(to) densities were unaffected by intermittent burst pacing in PV and Bachmann bundle cells. mRNA levels of K(V)4.3, K(V)4.2, K(V)1.4, and KChIP2 subunits of I(to) in both regions were stable. In CS cells, I(to) densities in intermittent burst pacing were greater than in sham (P <.05), but there were no parallel mRNA changes. I(Ca) density of PV cells was reduced from 14.27 +/- 2.08 pA/pF (at -5 mV) in sham to 7.52 +/- 1.65 pA/pF in intermittent burst pacing PV cells (P <.05) due to a significant shift in voltage dependence of activation. These results were seen in the absence of mRNA changes in alpha(1C) and alpha(1D) Ca(2+) channel subunits. In contrast, intermittent burst pacing had no effect on Ca(2+) current densities and kinetics of CS cells, but decreased alpha(1)C and alpha(1)D mRNA levels. CONCLUSION: There is region-specific remodeling of I(to) and I(Ca) by intermittent burst pacing protocols in rabbit atrium. Increased I(to) in CS cells could account for the APD shortening observed with intermittent burst pacing, whereas an intermittent burst pacing-induced shift in voltage dependence of activation may contribute to APD shortening in PV cells.


Asunto(s)
Potenciales de Acción , Función Atrial , Canales de Calcio/metabolismo , Estimulación Cardíaca Artificial , Canales de Potasio con Entrada de Voltaje/metabolismo , Potenciales de Acción/efectos de los fármacos , Análisis de Varianza , Animales , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Fibrilación Atrial/terapia , Función Atrial/efectos de los fármacos , Fascículo Atrioventricular/citología , Fascículo Atrioventricular/metabolismo , Fascículo Atrioventricular/fisiopatología , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Estimulación Eléctrica , Electrodos Implantados , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Atrios Cardíacos/fisiopatología , Proteínas de Interacción con los Canales Kv/metabolismo , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa , Bloqueadores de los Canales de Potasio/farmacología , Venas Pulmonares/citología , Venas Pulmonares/metabolismo , Venas Pulmonares/fisiopatología , ARN Mensajero/metabolismo , Conejos , Tiempo de Reacción/efectos de los fármacos , Proyectos de Investigación , Nodo Sinoatrial/citología , Nodo Sinoatrial/metabolismo , Nodo Sinoatrial/fisiopatología
16.
Circulation ; 112(13): 1927-35, 2005 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-16172272

RESUMEN

BACKGROUND: The SCN5A sodium channel is a major determinant for cardiac impulse propagation. We used epicardial mapping of the atria, ventricles, and septae to investigate conduction velocity (CV) in Scn5a heterozygous young and old mice. METHODS AND RESULTS: Mice were divided into 4 groups: (1) young (3 to 4 months) wild-type littermates (WT); (2) young heterozygous Scn5a-knockout mice (HZ); (3) old (12 to 17 months) WT; and (4) old HZ. In young HZ hearts, CV in the right but not the left ventricle was reduced in agreement with a rightward rotation in the QRS axes; fibrosis was virtually absent in both ventricles, and the pattern of connexin43 (Cx43) expression was similar to that of WT mice. In old WT animals, the right ventricle transversal CV was slightly reduced and was associated with interstitial fibrosis. In old HZ hearts, right and left ventricle CVs were severely reduced both in the transversal and longitudinal direction; multiple areas of severe reactive fibrosis invaded the myocardium, accompanied by markedly altered Cx43 expression. The right and left bundle-branch CVs were comparable to those of WT animals. The atria showed only mild fibrosis, with heterogeneously disturbed Cx40 and Cx43 expression. CONCLUSIONS: A 50% reduction in Scn5a expression alone or age-related interstitial fibrosis only slightly affects conduction. In aged HZ mice, reduced Scn5a expression is accompanied by the presence of reactive fibrosis and disarrangement of gap junctions, which results in profound conduction impairment.


Asunto(s)
Envejecimiento , Conexinas/metabolismo , Sistema de Conducción Cardíaco/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Canales de Sodio/deficiencia , Animales , Fascículo Atrioventricular/metabolismo , Conexina 43/metabolismo , Electrocardiografía , Fibrosis , Atrios Cardíacos , Ventrículos Cardíacos , Heterocigoto , Técnicas In Vitro , Ratones , Ratones Noqueados , Factores de Tiempo , Función Ventricular , Canales de Sodio Activados por Voltaje , Proteína alfa-5 de Unión Comunicante
17.
Cardiovasc Res ; 63(1): 77-86, 2004 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15194464

RESUMEN

OBJECTIVE: The aim of this work was to target a vital reporter gene in the mouse cardiac conduction system (CS) to distinguish this tissue from the surrounding myocardium in the adult heart. METHODS: A transgenic mouse line has been created in which EGFP is expressed under the control of the Cx40 gene. Correlative investigations associating EGFP imaging and electrophysiological techniques were carried out on the adult heart and isolated cardiomyocytes. RESULTS: In the heart of the Cx40(EGFP/+) mice, EGFP signal was seen in the coronary arteries, the atria, the atrioventricular (AV) node and the His-Purkinje system. The latter was found to be structurally and functionally asymmetrical. The anatomical asymmetry was apparent in both the number of strands or fasciculi making up the His bundle branches (BBs) (1 strand on the right, 20 or so on the left), and the density (low on the right, high on the left) of the network of Purkinje fibers (PFs) that extends over the ventricular wall surfaces. The profiles of the electrical activation patterns recorded on the right and left flanks of the septum were also asymmetrical, mirroring the architecture of the branches. EGFP made it easy to identify the Purkinje cells in populations of dissociated cardiomyocytes and they were investigated using the patch-clamp technique. The hyperpolarization-activated current (If) was recorded in all spontaneously active Purkinje cells. CONCLUSIONS: This investigation provides positive evidence of the asymmetry of the His-Purkinje system of the adult mouse, and the first patch-clamp recording data on murine cardiac Purkinje cells. This mouse model opens up new perspectives for investigating the contribution of specific genes to the morphology and function of the His-Purkinje system.


Asunto(s)
Conexinas/genética , Conexinas/metabolismo , Sistema de Conducción Cardíaco/anatomía & histología , Potenciales de Acción , Animales , Fascículo Atrioventricular/anatomía & histología , Fascículo Atrioventricular/metabolismo , Expresión Génica , Proteínas Fluorescentes Verdes , Sistema de Conducción Cardíaco/metabolismo , Proteínas Luminiscentes/genética , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Ramos Subendocárdicos/anatomía & histología , Ramos Subendocárdicos/metabolismo , Proteína alfa-5 de Unión Comunicante
18.
Circulation ; 109(9): 1172-9, 2004 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-14769705

RESUMEN

BACKGROUND: The 3D structure of the atrioventricular conduction axis incorporating detailed cellular and molecular composition, especially that relating to gap-junctional proteins, is still unclear, impeding mechanistic understanding of cardiac rhythmic disorders. METHODS AND RESULTS: A 3D model of the rabbit atrioventricular conduction axis was reconstructed by combining histological and immunofluorescence staining on serial sections. The exact cellular boundaries, especially those between transitional cells and atrial myocardium, were demarcated by a dense and irregular desmin-labeling pattern in conductive myocardium. The model demonstrates that the atrioventricular conduction axis is segregated into 2 connecting compartments, 1 predominantly expressing connexin45 (compact node and transitional cells) and the other predominantly coexpressing connexin43 and connexin45 (His bundle, lower nodal cells, and posterior nodal extension). The transitional zone shows unique features of spatial complexity, including a bridging bilayer structure (a deep transitional zone connecting with a superficial atrial-transitional overlay) and asymmetrical continuity (wider atrial-transitional interfaces and shorter atrial-axial distances in the hisian portion than in the ostial portion). In the latter compartment, the His bundle, lower nodal cells, and posterior nodal extension form a continual axis and longitudinal transitional-axial interface. CONCLUSIONS: Key findings of the present study are the demonstration of a distinct anatomical border between transitional and atrial cells, connection between transitional cells and both lower nodal cells and posterior nodal extension, and distinctive connexin expression patterns in different compartments of the rabbit atrioventricular conduction axis. These features, synthesized in a novel 3D model, provide a structural framework for the interpretation of nodal function.


Asunto(s)
Nodo Atrioventricular/anatomía & histología , Fascículo Atrioventricular/anatomía & histología , Conexinas/análisis , Desmina/análisis , Modelos Anatómicos , Animales , Nodo Atrioventricular/química , Nodo Atrioventricular/metabolismo , Fascículo Atrioventricular/química , Fascículo Atrioventricular/metabolismo , Conexinas/inmunología , Conexinas/metabolismo , Desmina/inmunología , Histocitoquímica , Imagenología Tridimensional , Inmunohistoquímica , Microscopía Confocal , Conejos
19.
Basic Res Cardiol ; 97(4): 286-94, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12111038

RESUMEN

OBJECTIVE: Despite the pathophysiological and therapeutic significance of the negative dromotropic effect of adenosine, its underlying ionic mechanism, and specifically the role of the adenosine-activated K(+) current (I(K,ADO)) is not experimentally defined. Therefore, we studied the contribution of I(K,ADO) to the negative dromotropic effect of adenosine. METHODS: Effects of adenosine on single atrioventricular nodal and left atrial myocytes from rabbits were studied using the whole cell configuration of the patch clamp technique. Complementary experiments were done in rabbit and guinea pig isolated hearts instrumented to measure the atrium-to-His bundle interval. RESULTS: In contrast to its effect in atrial myocytes, Ba(2+) selectively and completely blocked I(K,ADO) at membrane potentials from -70 to 0 mV in atrioventricular nodal myocytes and abolished the adenosine-induced leftward shift of the reversal membrane potential. Ba(2+) alone did not significantly prolong the A-H interval, but markedly attenuated the A-H interval prolongation caused by adenosine. In guinea pig heart, EC(50) values ( pD(2) +/- SEM) for adenosine-induced atrium-to-His bundle interval prolongation were 3.3 micromol/L (5.48 +/- 0.04) and 13.2 micromol/L (4.88 +/- 0.05, P < 0.001) in the absence and presence of Ba(2+), respectively. Despite species-dependent differences in sensitivities to adenosine (guinea pig > rabbit), the relative contribution of adenosine-activated K(+) current to the atrium-to-His bundle interval prolongation was nearly identical. In guinea pig hearts it ranged from 37.8 % (P = 0.013) to 72.5 % (P < 0.001) at 2 to 6 micromol/L adenosine, respectively. CONCLUSION: I(K,ADO) contributes significantly to the negative dromotropic effect of adenosine, but predominantly at relatively high concentrations of the nucleoside.


Asunto(s)
Adenosina/farmacología , Nodo Atrioventricular/metabolismo , Fascículo Atrioventricular/metabolismo , Canales de Potasio/metabolismo , Animales , Nodo Atrioventricular/citología , Bario/farmacología , Fascículo Atrioventricular/citología , Bloqueadores de los Canales de Calcio/farmacología , Diltiazem/farmacología , Interacciones Farmacológicas , Cobayas , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Perfusión , Potasio/farmacocinética , Conejos
20.
Adv Clin Path ; 5(3): 79-86, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11753879

RESUMEN

Aim of this study is to determine the expression of apoptosis and Proliferating Cell Nuclear Antigen (PCNA) in the cardiac conduction system in crib death and explained death (ED) cases. Postnatal morphogenesis of the conducting tissue is an important part of its normal development. In the atrio-ventricular node (AVN) and His bundle (HB) it consists of degeneration, cell death and replacing in an orderly programmed way. However, its nature and its relation to crib death is not yet fully explained. Apoptosis and PCNA were investigated in 8 heart conduction systems of infants dying of crib death and in 3 conduction systems of infants dying of ED as controls. The cardiac conduction system was removed in two blocks: the first included the sino-atrial node (SAN) and the crista terminalis, the second contained the atrio-ventricular node (AVN), His bundle (HB), bifurcation, and bundle branches. In the conduction systems as well as in the common myocardium the PCNA Labeling Index (PCNA-LI) was found to be negative in all cases. The apoptotic indices (AI) in SIDS and in ED were found to have no statistically significant differences (p>0.05). The SAN, in both groups, showed an AI similar to the one detected in common myocardium. In almost all cases, TUNEL labeling was detected in peripheral region of the AVN, close to the atrial myocardium. The AI was higher in the AVN, HB and the initial tract of bundle branches than in the common myocardium (p<0.05; Student's t test).


Asunto(s)
Apoptosis , Nodo Atrioventricular/metabolismo , Fascículo Atrioventricular/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Muerte Súbita del Lactante/patología , Nodo Atrioventricular/patología , Fascículo Atrioventricular/patología , División Celular , ADN/análisis , Femenino , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Lactante , Masculino , Miocardio/metabolismo , Miocardio/patología , Muerte Súbita del Lactante/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...