Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 394
Filtrar
1.
Nature ; 619(7971): 801-810, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37438528

RESUMEN

The function of a cell is defined by its intrinsic characteristics and its niche: the tissue microenvironment in which it dwells. Here we combine single-cell and spatial transcriptomics data to discover cellular niches within eight regions of the human heart. We map cells to microanatomical locations and integrate knowledge-based and unsupervised structural annotations. We also profile the cells of the human cardiac conduction system1. The results revealed their distinctive repertoire of ion channels, G-protein-coupled receptors (GPCRs) and regulatory networks, and implicated FOXP2 in the pacemaker phenotype. We show that the sinoatrial node is compartmentalized, with a core of pacemaker cells, fibroblasts and glial cells supporting glutamatergic signalling. Using a custom CellPhoneDB.org module, we identify trans-synaptic pacemaker cell interactions with glia. We introduce a druggable target prediction tool, drug2cell, which leverages single-cell profiles and drug-target interactions to provide mechanistic insights into the chronotropic effects of drugs, including GLP-1 analogues. In the epicardium, we show enrichment of both IgG+ and IgA+ plasma cells forming immune niches that may contribute to infection defence. Overall, we provide new clarity to cardiac electro-anatomy and immunology, and our suite of computational approaches can be applied to other tissues and organs.


Asunto(s)
Microambiente Celular , Corazón , Multiómica , Miocardio , Humanos , Comunicación Celular , Fibroblastos/citología , Ácido Glutámico/metabolismo , Corazón/anatomía & histología , Corazón/inervación , Canales Iónicos/metabolismo , Miocardio/citología , Miocardio/inmunología , Miocardio/metabolismo , Miocitos Cardíacos/citología , Neuroglía/citología , Pericardio/citología , Pericardio/inmunología , Células Plasmáticas/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Nodo Sinoatrial/anatomía & histología , Nodo Sinoatrial/citología , Nodo Sinoatrial/fisiología , Sistema de Conducción Cardíaco/anatomía & histología , Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/metabolismo
2.
Res Vet Sci ; 138: 137-147, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34144281

RESUMEN

Histologically, the cardiac conduction network is formed of electrically isolated subendocardial fibers that comprise specialized cells with fewer myofibrils and mitochondria than cardiomyocytes. Our aim is to uncover regional variations of cardiac conduction fibers through histological and morphometric study in a porcine and human model. We analyzed five male adult human hearts and five male pig hearts. The left ventricles were dissected and sectioned in the axial plane into three parts: basal, middle third and apex regions. Cardiac conduction fibers study was carried out using hematoxylin-eosin and Masson's trichrome staining, and cardiac conduction cells and their junctions were identified using desmin, and a PAS method. Cardiac conduction fibers were difficult to pinpoint in humans, mostly showing a darker color or equal to cardiomyocytes. Cardiac conduction fibers in humans were in the subendocardium and in pigs in the myocardium and subendocardium. Cardiac conduction fibers were located mainly in the septal region in both humans and pigs. In our morphometric analysis, we were able to determine that cardiac conduction cells in humans (18.52 +/- 5.41 µm) and pigs (21.32 +/- 6.45 µm) were large, compared to cardiomyocytes. Conduction fiber-myocardial junctions were present in 10% in humans and 24.2% in pigs. The performance of immunohistochemical methods made it possible to improve the identification of cardiac conduction cells in the species studied. Study of cardiac conduction fibers and cells and their myocardial junctions is vital to gain insight into their normal distribution in the species analyzed, and thus advance the use of pigs in experimental models of the cardiac conduction system in humans.


Asunto(s)
Sistema de Conducción Cardíaco/fisiología , Ventrículos Cardíacos/citología , Corazón/fisiología , Miocardio/citología , Sus scrofa/fisiología , Animales , Sistema de Conducción Cardíaco/citología , Humanos , Masculino , Coloración y Etiquetado/veterinaria
3.
Biochem Biophys Res Commun ; 533(3): 376-382, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-32962862

RESUMEN

Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) resemble fetal cardiomyocytes and electrical stimulation (ES) has been explored to mature the differentiated cells. Here, we hypothesize that ES applied at the beginning of the differentiation process, triggers both differentiation of the hiPSC-CMs into a specialized conduction system (CS) phenotype and cell maturation. We applied ES for 15 days starting on day 0 of the differentiation process and found an increased expression of transcription factors and proteins associated with the development and function of CS including Irx3, Nkx2.5 and contactin 2, Hcn4 and Scn5a, respectively. We also found activation of intercalated disc proteins (Nrap and ß-catenin). We detected ES-induced CM maturation as indicated by increased Tnni1 and Tnni3 expression. Confocal micrographs showed a shift towards expression of the gap junction protein connexin 40 in ES hiPSC-CM compared to the more dominant expression of connexin 43 in controls. Finally, analysis of functional parameters revealed that ES hiPSC-CMs exhibited faster action potential (AP) depolarization, longer intracellular Ca2+ transients, and slower AP duration at 90% of repolarization, resembling fast conducting fibers. Altogether, we provided evidence that ES during the differentiation of hiPSC to cardiomyocytes lead to development of cardiac conduction-like cells with more mature cytoarchitecture. Thus, hiPSC-CMs exposed to ES during differentiation can be instrumental to develop CS cells for cardiac disease modelling, screening individual drugs on a precison medicine type platform and support the development of novel therapeutics for arrhythmias.


Asunto(s)
Potenciales de Acción/fisiología , Calcio/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Miocitos Cardíacos/fisiología , Biomarcadores/metabolismo , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Conexinas/genética , Conexinas/metabolismo , Contactina 2/genética , Contactina 2/metabolismo , Estimulación Eléctrica , Expresión Génica , Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/fisiología , Proteína Homeótica Nkx-2.5/genética , Proteína Homeótica Nkx-2.5/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Células Madre Pluripotentes Inducidas/citología , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocitos Cardíacos/citología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Canales de Potasio/genética , Canales de Potasio/metabolismo , Cultivo Primario de Células , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Troponina I/genética , Troponina I/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Proteína alfa-5 de Unión Comunicante
4.
Circ Res ; 125(4): 379-397, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31284824

RESUMEN

RATIONALE: The cardiac conduction system (CCS) consists of distinct components including the sinoatrial node, atrioventricular node, His bundle, bundle branches, and Purkinje fibers. Despite an essential role for the CCS in heart development and function, the CCS has remained challenging to interrogate because of inherent obstacles including small cell numbers, large cell-type heterogeneity, complex anatomy, and difficulty in isolation. Single-cell RNA-sequencing allows for genome-wide analysis of gene expression at single-cell resolution. OBJECTIVE: Assess the transcriptional landscape of the entire CCS at single-cell resolution by single-cell RNA-sequencing within the developing mouse heart. METHODS AND RESULTS: Wild-type, embryonic day 16.5 mouse hearts (n=6 per zone) were harvested and 3 zones of microdissection were isolated, including: Zone I-sinoatrial node region; Zone II-atrioventricular node/His region; and Zone III-bundle branch/Purkinje fiber region. Tissue was digested into single-cell suspensions, cells isolated, mRNA reverse transcribed, and barcoded before high-throughput sequencing and bioinformatics analyses. Single-cell RNA-sequencing was performed on over 22 000 cells, and all major cell types of the murine heart were successfully captured including bona fide clusters of cells consistent with each major component of the CCS. Unsupervised weighted gene coexpression network analysis led to the discovery of a host of novel CCS genes, a subset of which were validated using fluorescent in situ hybridization as well as whole-mount immunolabeling with volume imaging (iDISCO+) in 3 dimensions on intact mouse hearts. Further, subcluster analysis unveiled isolation of distinct CCS cell subtypes, including the clinically relevant but poorly characterized transitional cells that bridge the CCS and surrounding myocardium. CONCLUSIONS: Our study represents the first comprehensive assessment of the transcriptional profiles from the entire CCS at single-cell resolution and provides a characterization in the context of development and disease.


Asunto(s)
Sistema de Conducción Cardíaco/metabolismo , Transcriptoma , Animales , Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/embriología , Ratones , RNA-Seq , Análisis de la Célula Individual
5.
Mol Med Rep ; 19(5): 3584-3592, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30864738

RESUMEN

Biological pacemakers that combine cell­based and gene­based therapies are a promising treatment for sick sinus syndrome or severe atrioventricular block. The current study aimed to induce differentiation of adipose tissue­derived stem cells (ADSCs) into cardiac pacemaker cells through co­expression of the transcription factors insulin gene enhancer binding protein 1 (ISL­1) and T­box18 (Tbx18). ADSCs were transfected with green fluorescent protein, ISL­1, Tbx18 or ISL­1+Tbx18 fluorescent protein lentiviral vectors, and subsequently co­cultured with neonatal rat ventricular cardiomyocytes in vitro for 7 days. The potential for regulating the differentiation of ADSCs into pacemaker­like cells was evaluated by cell morphology, beating rate, reverse transcription­quantitative polymerase chain reaction, western blotting, immunofluorescence and electrophysiological activity. ADSCs were successfully transformed into spontaneously beating cells that exhibited a behavior similar to that of co­cultured pacemaker cells. This effect was significantly increased in the combined ISL­1 and Tbx18 group. These results provide a potential strategy for enriching the cardiac pacemaker cell population from ADSCs.


Asunto(s)
Diferenciación Celular/genética , Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/metabolismo , Factores de Transcripción/genética , Tejido Adiposo/citología , Animales , Biomarcadores , Reprogramación Celular , Fenómenos Electrofisiológicos , Expresión Génica , Humanos , Proteínas con Homeodominio LIM/genética , Masculino , Ratas , Células Madre/citología , Proteínas de Dominio T Box/genética , Transducción Genética
6.
Physiol Rep ; 7(1): e13860, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30604919

RESUMEN

Two hypotheses have been proposed to explain propagation of the action potential in heart. According to the gap junction hypothesis local short-circuit currents pass from the proximal depolarized cell to the distal inactive cell via gap junctions and are responsible for the depolarization of the distal cell. In the ephapse hypothesis the depolarization of the proximal cell generates an electrical field in the narrow cleft between cells resulting in depolarization beyond threshold of the distal cell. Measurements of length constant, free diffusion of 42 K, local currents between cells, existence of high-conductance gap junctions led to the conclusion that heart muscle is a functional syncytium. Propagation of the action potential, however, is not uniform but anisotropic and discontinuous; it can be also unidirectional. These findings are strong arguments in favor of the gap junction thesis. They do not exclude, as predicted by theoretical calculations, that in conditions of an abnormal fall in gap junction conductance ephaptic conduction takes over. In this last case, definitive experimental confirmation is still required. See also: https://doi.org/10.14814/phy2.13861 & https://doi.org/10.14814/phy2.13862.


Asunto(s)
Sistema de Conducción Cardíaco/fisiología , Potenciales de Acción , Animales , Electrofisiología Cardíaca/historia , Uniones Comunicantes/fisiología , Células Gigantes/fisiología , Sistema de Conducción Cardíaco/citología , Historia del Siglo XIX , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/ultraestructura
7.
Bull Math Biol ; 81(1): 7-38, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30291590

RESUMEN

Distinct electrophysiological phenotypes are exhibited by biological cells that have differentiated into particular cell types. The usual approach when simulating the cardiac electrophysiology of tissue that includes different cell types is to model the different cell types as occupying spatially distinct yet coupled regions. Instead, we model the electrophysiology of well-mixed cells by using homogenisation to derive an extension to the commonly used monodomain or bidomain equations. These new equations permit spatial variations in the distribution of the different subtypes of cells and will reduce the computational demands of solving the governing equations. We validate the homogenisation computationally, and then use the new model to explain some experimental observations from stem cell-derived cardiomyocyte monolayers.


Asunto(s)
Modelos Cardiovasculares , Miocitos Cardíacos/fisiología , Potenciales de Acción/fisiología , Simulación por Computador , Diástole/fisiología , Fenómenos Electrofisiológicos , Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/fisiología , Humanos , Conceptos Matemáticos , Miocitos Cardíacos/clasificación , Fenotipo , Células Madre/clasificación , Células Madre/fisiología
8.
Int J Mol Med ; 43(2): 879-889, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30483766

RESUMEN

Hybrid approaches combining gene­ and cell­based therapies to make biological pacemakers are a promising therapeutic avenue for bradyarrhythmia. The present study aimed to direct adipose tissue­derived stem cells (ADSCs) to differentiate specifically into cardiac pacemaker cells by overexpressing a single transcription factor, insulin gene enhancer binding protein 1 (ISL­1). In the present study, the ADSCs were transfected with ISL­1 or mCherry fluorescent protein lentiviral vectors and co­cultured with neonatal rat ventricular cardiomyocytes (NRVMs) in vitro for 5­7 days. The feasibility of regulating the differentiation of ADSCs into pacemaker­like cells by overexpressing ISL­1 was evaluated by observation of cell morphology and beating rate, reverse transcription­quantitative polymerase chain reaction analysis, western blotting, immunofluorescence and analysis of electrophysiological activity. In conclusion, these data indicated that the overexpression of ISL­1 in ADSCs may enhance the pacemaker phenotype and automaticity in vitro, features which were significantly increased following co­culture induction.


Asunto(s)
Tejido Adiposo/citología , Sistema de Conducción Cardíaco/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Biomarcadores , Células Cultivadas , Técnicas de Cocultivo , Fenómenos Electrofisiológicos , Técnica del Anticuerpo Fluorescente , Expresión Génica , Inmunofenotipificación , Masculino , Ratas , Transfección
9.
Eur J Pharmacol ; 833: 349-356, 2018 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-29940156

RESUMEN

Heart failure (HF) is a clinical syndrome characterized by ventricular contractile dysfunction. About 50% of death in patients with HF are due to fetal ventricular arrhythmias including ventricular tachycardia and ventricular fibrillation. Understanding ventricular arrhythmic substrates and discovering effective antiarrhythmic interventions are extremely important for improving the prognosis of patients with HF and reducing its mortality. In this review, we discussed ventricular arrhythmic substrates and current clinical therapeutics for ventricular arrhythmias in HF. Base on the fact that classic antiarrhythmic drugs have the limited efficacy, side effects, and proarrhythmic potentials, we also updated some therapeutic strategies for the development of potential new antiarrhythmic interventions for patients with HF.


Asunto(s)
Antiarrítmicos/farmacología , Sistema de Conducción Cardíaco/efectos de los fármacos , Insuficiencia Cardíaca/tratamiento farmacológico , Taquicardia Ventricular/tratamiento farmacológico , Fibrilación Ventricular/tratamiento farmacológico , Antiarrítmicos/uso terapéutico , Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/fisiopatología , Insuficiencia Cardíaca/mortalidad , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Humanos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Pronóstico , Taquicardia Ventricular/mortalidad , Taquicardia Ventricular/fisiopatología , Resultado del Tratamiento , Fibrilación Ventricular/mortalidad , Fibrilación Ventricular/fisiopatología
10.
Nat Commun ; 9(1): 2526, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29955040

RESUMEN

Long-range chromosomal interactions bring distal regulatory elements and promoters together to regulate gene expression in biological processes. By performing promoter capture Hi-C (PCHi-C) on human embryonic stem cell-derived cardiomyocytes (hESC-CMs), we show that such promoter interactions are a key mechanism by which enhancers contact their target genes after hESC-CM differentiation from hESCs. We also show that the promoter interactome of hESC-CMs is associated with expression quantitative trait loci (eQTLs) in cardiac left ventricular tissue; captures the dynamic process of genome reorganisation after hESC-CM differentiation; overlaps genome-wide association study (GWAS) regions associated with heart rate; and identifies new candidate genes in such regions. These findings indicate that regulatory elements in hESC-CMs identified by our approach control gene expression involved in ventricular conduction and rhythm of the heart. The study of promoter interactions in other hESC-derived cell types may be of utility in functional investigation of GWAS-associated regions.


Asunto(s)
Actinina/genética , Calpaína/genética , Redes Reguladoras de Genes , Células Madre Embrionarias Humanas/metabolismo , Miocitos Cardíacos/metabolismo , Regiones Promotoras Genéticas , Actinina/metabolismo , Calpaína/metabolismo , Diferenciación Celular , Línea Celular , Elementos de Facilitación Genéticos , Genoma Humano , Estudio de Asociación del Genoma Completo , Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/metabolismo , Frecuencia Cardíaca/fisiología , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Histonas/genética , Histonas/metabolismo , Células Madre Embrionarias Humanas/citología , Humanos , Miocitos Cardíacos/citología , Mapeo de Interacción de Proteínas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sitios de Carácter Cuantitativo
11.
Pediatr Cardiol ; 39(6): 1107-1114, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29774393

RESUMEN

The components of the cardiac conduction system (CCS) generate and propagate the electrical impulse that initiates cardiac contraction. These interconnected components share properties, such as automaticity, that set them apart from the working myocardium of the atria and ventricles. A variety of tools and approaches have been used to define the CCS lineages. These include genetic labeling of cells expressing lineage markers and fate mapping of dye labeled cells, which we will discuss in this review. We conclude that there is not a single CCS lineage, but instead early cell fate decisions segregate the lineages of the CCS components while they remain interconnected. The latter is relevant for development of therapies for conduction system disease that focus on reprogramming cardiomyocytes or instruction of pluripotent stem cells.


Asunto(s)
Sistema de Conducción Cardíaco/embriología , Miocardio/citología , Animales , Diferenciación Celular , Sistema de Conducción Cardíaco/citología , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/embriología , Humanos , Miocitos Cardíacos
12.
Toxicol Lett ; 294: 61-72, 2018 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-29758359

RESUMEN

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are emerging tools for applications such as drug discovery and screening for pro-arrhythmogenicity and cardiotoxicity as leading causes for drug attrition. Understanding the electrophysiology (EP) of hPSC-CMs is essential but conventional manual patch-clamping is highly laborious and low-throughput. Here we adapted hPSC-CMs derived from two human embryonic stem cell (hESC) lines, HES2 and H7, for a 16-channel automated planar-recording approach for single-cell EP characterization. Automated current- and voltage-clamping, with an overall success rate of 55.0 ±â€¯11.3%, indicated that 90% of hPSC-CMs displayed ventricular-like action potential (AP) and the ventricular cardiomyocytes (VCMs) derived from the two hESC lines expressed similar levels of INa, ICaL, Ikr and If and similarly lacked Ito and IK1. These well-characterized hPSC-VCMs could also be readily adapted for automated assays of pro-arrhythmic drug screening. As an example, we showed that flecainide (FLE) induced INa blockade, leftward steady-state inactivation shift, slowed recovery from inactivation in our hPSC-VCMs. Since single-cell EP assay is insufficient to predict drug-induced reentrant arrhythmias, hPSC-VCMs were further reassembled into 2D human ventricular cardiac monolayers (hvCMLs) for multi-cellular electrophysiological assessments. Indeed, FLE significantly slowed the conduction velocity while causing AP prolongation. Our RNA-seq data suggested that cell-cell interaction enhanced the maturity of hPSC-VCMs. Taken collectively, a combinatorial approach using single-cell EP and hvCMLs is needed to comprehensively assess drug-induced arrhythmogenicity.


Asunto(s)
Evaluación Preclínica de Medicamentos , Flecainida/efectos adversos , Ventrículos Cardíacos/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Miocitos Cardíacos/efectos de los fármacos , Bloqueadores del Canal de Sodio Activado por Voltaje/efectos adversos , Canales de Sodio Activados por Voltaje/metabolismo , Potenciales de Acción/efectos de los fármacos , Automatización de Laboratorios , Diferenciación Celular , Línea Celular , Células Cultivadas , Fenómenos Electrofisiológicos/efectos de los fármacos , Estudios de Factibilidad , Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Reproducibilidad de los Resultados , Análisis de la Célula Individual , Canales de Sodio Activados por Voltaje/química
13.
IEEE Trans Biomed Eng ; 65(1): 123-130, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28436840

RESUMEN

OBJECTIVE: A flexible, efficient, and verifiable pacemaker cell model is essential to the design of real-time virtual hearts that can be used for closed-loop validation of cardiac devices. A new parametric model of pacemaker action potential is developed to address this need. METHODS: The action potential phases are modeled using hybrid automaton with one piecewise-linear continuous variable. The model can capture rate-dependent dynamics, such as action potential duration restitution, conduction velocity restitution, and overdrive suppression by incorporating nonlinear update functions. Simulated dynamics of the model compared well with previous models and clinical data. CONCLUSION: The results show that the parametric model can reproduce the electrophysiological dynamics of a variety of pacemaker cells, such as sinoatrial node, atrioventricular node, and the His-Purkinje system, under varying cardiac conditions. SIGNIFICANCE: This is an important contribution toward closed-loop validation of cardiac devices using real-time heart models.


Asunto(s)
Potenciales de Acción/fisiología , Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/fisiología , Modelos Cardiovasculares , Humanos
14.
Sci Rep ; 7(1): 7188, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28775383

RESUMEN

Cardiac arrhythmias and conduction disturbances are accompanied by structural remodelling of the specialised cardiomyocytes known collectively as the cardiac conduction system. Here, using contrast enhanced micro-computed tomography, we present, in attitudinally appropriate fashion, the first 3-dimensional representations of the cardiac conduction system within the intact human heart. We show that cardiomyocyte orientation can be extracted from these datasets at spatial resolutions approaching the single cell. These data show that commonly accepted anatomical representations are oversimplified. We have incorporated the high-resolution anatomical data into mathematical simulations of cardiac electrical depolarisation. The data presented should have multidisciplinary impact. Since the rate of depolarisation is dictated by cardiac microstructure, and the precise orientation of the cardiomyocytes, our data should improve the fidelity of mathematical models. By showing the precise 3-dimensional relationships between the cardiac conduction system and surrounding structures, we provide new insights relevant to valvar replacement surgery and ablation therapies. We also offer a practical method for investigation of remodelling in disease, and thus, virtual pathology and archiving. Such data presented as 3D images or 3D printed models, will inform discussions between medical teams and their patients, and aid the education of medical and surgical trainees.


Asunto(s)
Sistema de Conducción Cardíaco/anatomía & histología , Sistema de Conducción Cardíaco/diagnóstico por imagen , Imagenología Tridimensional , Modelos Anatómicos , Modelos Teóricos , Fascículo Atrioventricular , Medios de Contraste , Sistema de Conducción Cardíaco/citología , Humanos , Aumento de la Imagen , Ramos Subendocárdicos , Nodo Sinoatrial/anatomía & histología , Nodo Sinoatrial/citología , Nodo Sinoatrial/diagnóstico por imagen , Microtomografía por Rayos X/métodos
15.
Mol Cell Biochem ; 433(1-2): 61-77, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28382491

RESUMEN

A cell-sourced biological pacemaker is a promising therapeutic approach for sick sinus syndrome (SSS) or severe atrial ventricular block (AVB). Adipose tissue-derived stem cells (ATSCs), which are optimal candidate cells for possible use in regenerative therapy for acute or chronic myocardial injury, have the potential to differentiate into spontaneous beating cardiomyocytes. However, the pacemaker characteristics of the beating cells need to be confirmed, and little is known about the underlying differential mechanism. In this study, we found that brown adipose tissue-derived stem cells (BATSCs) in mice could differentiate into spontaneous beating cells in 15% FBS Dulbecco's modified Eagle's medium (DMEM) without additional treatment. Subsequently, we provide additional evidence, including data regarding ultrastructure, protein expression, electrophysiology, and pharmacology, to support the differentiation of BATSCs into a cardiac pacemaker phenotype during the course of early cultivation. Furthermore, we found that silencing Tbx18, a key transcription factor in the development of pacemaker cells, terminated the differentiation of BATSCs into a pacemaker phenotype, suggesting that Tbx18 is required to direct BATSCs toward a cardiac pacemaker fate. The expression of Tbx3 and shox2, the other two important transcription factors in the development of pacemaker cells, was decreased by silencing Tbx18, which suggests that Tbx18 mediates the differentiation of BATSCs into a pacemaker phenotype via these two downstream transcription factors.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Diferenciación Celular , Sistema de Conducción Cardíaco/metabolismo , Células Madre/metabolismo , Proteínas de Dominio T Box/metabolismo , Tejido Adiposo Pardo/citología , Animales , Sistema de Conducción Cardíaco/citología , Ratones , Células Madre/citología , Proteínas de Dominio T Box/genética
16.
Am J Physiol Regul Integr Comp Physiol ; 312(2): R165-R171, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27903513

RESUMEN

Temperature-induced changes in cardiac output (Q̇) in fish are largely dependent on thermal modulation of heart rate (fH), and at high temperatures Q̇ collapses due to heat-dependent depression of fH This study tests the hypothesis that firing rate of sinoatrial pacemaker cells sets the upper thermal limit of fH in vivo. To this end, temperature dependence of action potential (AP) frequency of enzymatically isolated pacemaker cells (pacemaker rate, fPM), spontaneous beating rate of isolated sinoatrial preparations (fSA), and in vivo fH of the cold-acclimated (4°C) brown trout (Salmo trutta fario) were compared under acute thermal challenges. With rising temperature, fPM steadily increased because of the acceleration of diastolic depolarization and shortening of AP duration up to the break point temperature (TBP) of 24.0 ± 0.37°C, at which point the electrical activity abruptly ceased. The maximum fPM at TBP was much higher [193 ± 21.0 beats per minute (bpm)] than the peak fSA (94.3 ± 6.0 bpm at 24.1°C) or peak fH (76.7 ± 2.4 at 15.7 ± 0.82°C) (P < 0.05). These findings strongly suggest that the frequency generator of the sinoatrial pacemaker cells does not limit fH at high temperatures in the brown trout in vivo.


Asunto(s)
Potenciales de Acción/fisiología , Relojes Biológicos/fisiología , Regulación de la Temperatura Corporal/fisiología , Frecuencia Cardíaca/fisiología , Miocitos Cardíacos/fisiología , Trucha/fisiología , Animales , Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/fisiología
17.
Adv Exp Med Biol ; 913: 241-251, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27796892

RESUMEN

The heart is an electrically conducting organ with networked bioelectric currents that transverse a large segment of interstitial space interspersed with the muscular parenchyma. Non-excitable connective cells in the interstitial space contributed importantly to many structural, biochemical, and physiological activities of cardiac homeostasis. However, contribution of interstitial cells in the cardiac niche has long been neglected. Telocyte is recently recognized as a distinct class of interstitial cell that resides in a wide array of tissues including in the epicardium, myocardium, and endocardium of the heart. They are increasingly described to conduct ionic currents that may have significant implications in bioelectric signaling. In this review, we highlight the significance of telocytes in such connectivity and conductivity within the interstitial bioelectric network in tissue homeostasis.


Asunto(s)
Sistema de Conducción Cardíaco/fisiología , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Miocardio/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Telocitos/fisiología , Antígenos CD34/genética , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Endocardio/citología , Endocardio/fisiología , Expresión Génica , Sistema de Conducción Cardíaco/citología , Homeostasis/fisiología , Humanos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Miocardio/citología , Técnicas de Placa-Clamp , Pericardio/citología , Pericardio/fisiología , Canales de Potasio de Rectificación Interna/genética , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Telocitos/citología
18.
PLoS One ; 11(10): e0164093, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27716829

RESUMEN

The specialised conducting tissues present in the ventricles are responsible for the fast distribution of the electrical impulse from the atrio-ventricular node to regions in the subendocardial myocardium. Characterisation of anatomical features of the specialised conducting tissues in the ventricles is highly challenging, in particular its most distal section, which is connected to the working myocardium via Purkinje-myocardial junctions. The goal of this work is to characterise the architecture of the distal section of the Purkinje network by differentiating Purkinje cells from surrounding tissue, performing a segmentation of Purkinje fibres at cellular scale, and mathematically describing its morphology and interconnections. Purkinje cells from rabbit hearts were visualised by confocal microscopy using wheat germ agglutinin labelling. A total of 16 3D stacks including labeled Purkinje cells were collected, and semi-automatically segmented. State-of-the-art graph metrics were applied to estimate regional and global features of the Purkinje network complexity. Two types of cell types, tubular and star-like, were characterised from 3D segmentations. The analysis of 3D imaging data confirms the previously suggested presence of two types of Purkinje-myocardium connections, a 2D interconnection sheet and a funnel one, in which the narrow side of a Purkinje fibre connect progressively to muscle fibres. The complex network analysis of interconnected Purkinje cells showed no small-world connectivity or assortativity properties. These results might help building more realistic computational PK systems at high resolution levels including different cell configurations and shapes. Better knowledge on the organisation of the network might help in understanding the effects that several treatments such as radio-frequency ablation might have when the PK system is disrupted locally.


Asunto(s)
Sistema de Conducción Cardíaco/anatomía & histología , Sistema de Conducción Cardíaco/citología , Ventrículos Cardíacos/anatomía & histología , Ventrículos Cardíacos/citología , Animales , Imagenología Tridimensional/métodos , Microscopía Confocal/métodos , Miocardio/citología , Células de Purkinje/citología , Conejos
19.
Stem Cells ; 34(11): 2670-2680, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27434649

RESUMEN

Insights into the expression of pacemaker-specific markers in human induced pluripotent stem cell (hiPSC)-derived cardiomyocyte subtypes can facilitate the enrichment and track differentiation and maturation of hiPSC-derived pacemaker-like cardiomyocytes. To date, no study has directly assessed gene expression in each pacemaker-, atria-, and ventricular-like cardiomyocyte subtype derived from hiPSCs since currently the subtypes of these immature cardiomyocytes can only be identified by action potential profiles. Traditional acquisition of action potentials using patch-clamp recordings renders the cells unviable for subsequent analysis. We circumvented these issues by acquiring the action potential profile of a single cell optically followed by assessment of protein expression through immunostaining in that same cell. Our same-single-cell analysis for the first time revealed expression of proposed pacemaker-specific markers-hyperpolarization-activated cyclic nucleotide-modulated (HCN)4 channel and Islet (Isl)1-at the protein level in all three hiPSC-derived cardiomyocyte subtypes. HCN4 expression was found to be higher in pacemaker-like hiPSC-derived cardiomyocytes than atrial- and ventricular-like subtypes but its downregulation over time in all subtypes diminished the differences. Isl1 expression in pacemaker-like hiPSC-derived cardiomyocytes was initially not statistically different than the contractile subtypes but did become statistically higher than ventricular-like cells with time. Our observations suggest that although HCN4 and Isl1 are differentially expressed in hiPSC-derived pacemaker-like relative to ventricular-like cardiomyocytes, these markers alone are insufficient in identifying hiPSC-derived pacemaker-like cardiomyocytes. Stem Cells 2016;34:2670-2680.


Asunto(s)
Potenciales de Acción/fisiología , Atrios Cardíacos/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Ventrículos Cardíacos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Linaje de la Célula/genética , Electrofisiología , Expresión Génica , Atrios Cardíacos/citología , Sistema de Conducción Cardíaco/citología , Ventrículos Cardíacos/citología , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/citología , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocitos Cardíacos/citología , Especificidad de Órganos , Canales de Potasio/genética , Canales de Potasio/metabolismo , Análisis de la Célula Individual/métodos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Prog Biophys Mol Biol ; 120(1-3): 164-78, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26743207

RESUMEN

INTRODUCTION: Despite a century of extensive study on the human sinoatrial node (SAN), the structure-to-function features of specialized SAN conduction pathways (SACP) are still unknown and debated. We report a new method for direct analysis of the SAN microstructure in optically-mapped human hearts with and without clinical history of SAN dysfunction. METHODS: Two explanted donor human hearts were coronary-perfused and optically-mapped. Structural analyses of histological sections parallel to epicardium (∼13-21 µm intervals) were integrated with optical maps to create 3D computational reconstructions of the SAN complex. High-resolution fiber fields were obtained using 3D Eigen-analysis of the structure tensor, and used to analyze SACP microstructure with a fiber-tracking approach. RESULTS: Optical mapping revealed normal SAN activation of the atria through a lateral SACP proximal to the crista terminalis in Heart #1 but persistent SAN exit block in diseased Heart #2. 3D structural analysis displayed a functionally-observed SAN border composed of fibrosis, fat, and/or discontinuous fibers between SAN and atria, which was only crossed by several branching myofiber tracts in SACP regions. Computational 3D fiber-tracking revealed that myofiber tracts of SACPs created continuous connections between SAN #1 and atria, but in SAN #2, SACP region myofiber tracts were discontinuous due to fibrosis and fat. CONCLUSIONS: We developed a new integrative functional, structural and computational approach that allowed for the resolution of the specialized 3D microstructure of human SACPs for the first time. Application of this integrated approach will shed new light on the role of the specialized SAN microanatomy in maintaining sinus rhythm.


Asunto(s)
Sistema de Conducción Cardíaco/anatomía & histología , Modelos Anatómicos , Nodo Sinoatrial/anatomía & histología , Nodo Sinoatrial/fisiología , Relojes Biológicos , Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/fisiología , Humanos , Nodo Sinoatrial/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...