Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Angiogenesis ; 27(3): 561-582, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38775849

RESUMEN

Coronary microvascular disease (CMD) and its progression towards major adverse coronary events pose a significant health challenge. Accurate in vitro investigation of CMD requires a robust cell model that faithfully represents the cells within the cardiac microvasculature. Human pluripotent stem cell-derived endothelial cells (hPSC-ECs) offer great potential; however, they are traditionally derived via differentiation protocols that are not readily scalable and are not specified towards the microvasculature. Here, we report the development and comprehensive characterisation of a scalable 3D protocol enabling the generation of phenotypically stable cardiac hPSC-microvascular-like ECs (hPSC-CMVECs) and cardiac pericyte-like cells. These were derived by growing vascular organoids within 3D stirred tank bioreactors and subjecting the emerging 3D hPSC-ECs to high-concentration VEGF-A treatment (3DV). Not only did this promote phenotypic stability of the 3DV hPSC-ECs; single cell-RNA sequencing (scRNA-seq) revealed the pronounced expression of cardiac endothelial- and microvascular-associated genes. Further, the generated mural cells attained from the vascular organoid exhibited markers characteristic of cardiac pericytes. Thus, we present a suitable cell model for investigating the cardiac microvasculature as well as the endothelial-dependent and -independent mechanisms of CMD. Moreover, owing to their phenotypic stability, cardiac specificity, and high angiogenic potential, the cells described within would also be well suited for cardiac tissue engineering applications.


Asunto(s)
Diferenciación Celular , Células Endoteliales , Microvasos , Células Madre Pluripotentes , Humanos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/citología , Microvasos/citología , Microvasos/metabolismo , Pericitos/citología , Pericitos/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/farmacología , Organoides/citología , Organoides/irrigación sanguínea , Organoides/metabolismo
2.
PLoS Comput Biol ; 18(4): e1010030, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35363778

RESUMEN

Application of epicardial patches constructed from human-induced pluripotent stem cell- derived cardiomyocytes (hiPSC-CMs) has been proposed as a long-term therapy to treat scarred hearts post myocardial infarction (MI). Understanding electrical interaction between engineered heart tissue patches (EHT) and host myocardium represents a key step toward a successful patch engraftment. EHT retain different electrical properties with respect to the host heart tissue due to the hiPSC-CMs immature phenotype, which may lead to increased arrhythmia risk. We developed a modelling framework to examine the influence of patch design on electrical activation at the engraftment site. We performed an in silico investigation of different patch design approaches to restore pre-MI activation properties and evaluated the associated arrhythmic risk. We developed an in silico cardiac electrophysiology model of a transmural cross section of host myocardium. The model featured an infarct region, an epicardial patch spanning the infarct region and a bath region. The patch is modelled as a layer of hiPSC-CM, combined with a layer of conductive polymer (CP). Tissue and patch geometrical dimensions and conductivities were incorporated through 10 modifiable model parameters. We validated our model against 4 independent experimental studies and showed that it can qualitatively reproduce their findings. We performed a global sensitivity analysis (GSA) to isolate the most important parameters, showing that the stimulus propagation is mainly governed by the scar depth, radius and conductivity when the scar is not transmural, and by the EHT patch conductivity when the scar is transmural. We assessed the relevance of small animal studies to humans by comparing simulations of rat, rabbit and human myocardium. We found that stimulus propagation paths and GSA sensitivity indices are consistent across species. We explored which EHT design variables have the potential to restore physiological propagation. Simulations predict that increasing EHT conductivity from 0.28 to 1-1.1 S/m recovered physiological activation in rat, rabbit and human. Finally, we assessed arrhythmia risk related to increasing EHT conductivity and tested increasing the EHT Na+ channel density as an alternative strategy to match healthy activation. Our results revealed a greater arrhythmia risk linked to increased EHT conductivity compared to increased Na+ channel density. We demonstrated that our modeling framework could capture the interaction between host and EHT patches observed in in vitro experiments. We showed that large (patch and tissue dimensions) and small (cardiac myocyte electrophysiology) scale differences between small animals and humans do not alter EHT patch effect on infarcted tissue. Our model revealed that only when the scar is transmural do EHT properties impact activation times and isolated the EHT conductivity as the main parameter influencing propagation. We predicted that restoring physiological activation by tuning EHT conductivity is possible but may promote arrhythmic behavior. Finally, our model suggests that acting on hiPSC-CMs low action potential upstroke velocity and lack of IK1 may restore pre-MI activation while not promoting arrhythmia.


Asunto(s)
Células Madre Pluripotentes Inducidas , Infarto del Miocardio , Animales , Arritmias Cardíacas/patología , Cicatriz/patología , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Miocardio/patología , Miocitos Cardíacos , Conejos , Ratas
3.
Circulation ; 143(8): 821-836, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33297741

RESUMEN

BACKGROUND: Ischemic heart disease is a leading cause of heart failure and despite advanced therapeutic options, morbidity and mortality rates remain high. Although acute inflammation in response to myocardial cell death has been extensively studied, subsequent adaptive immune activity and anti-heart autoimmunity may also contribute to the development of heart failure. After ischemic injury to the myocardium, dendritic cells (DC) respond to cardiomyocyte necrosis, present cardiac antigen to T cells, and potentially initiate a persistent autoimmune response against the heart. Cross-priming DC have the ability to activate both CD4+ helper and CD8+ cytotoxic T cells in response to necrotic cells and may thus be crucial players in exacerbating autoimmunity targeting the heart. This study investigates a role for cross-priming DC in post-myocardial infarction immunopathology through presentation of self-antigen from necrotic cardiac cells to cytotoxic CD8+ T cells. METHODS: We induced type 2 myocardial infarction-like ischemic injury in the heart by treatment with a single high dose of the ß-adrenergic agonist isoproterenol. We characterized the DC population in the heart and mediastinal lymph nodes and analyzed long-term cardiac immunopathology and functional decline in wild type and Clec9a-depleted mice lacking DC cross-priming function. RESULTS: A diverse DC population, including cross-priming DC, is present in the heart and activated after ischemic injury. Clec9a-/- mice deficient in DC cross-priming are protected from persistent immune-mediated myocardial damage and decline of cardiac function, likely because of dampened activation of cytotoxic CD8+ T cells. CONCLUSION: Activation of cytotoxic CD8+ T cells by cross-priming DC contributes to exacerbation of postischemic inflammatory damage of the myocardium and corresponding decline in cardiac function. Importantly, this provides novel therapeutic targets to prevent postischemic immunopathology and heart failure.


Asunto(s)
Reactividad Cruzada , Células Dendríticas/inmunología , Miocardio/patología , Animales , Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Insuficiencia Cardíaca/patología , Humanos , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/inmunología , Miocardio/metabolismo , Receptores de Quimiocina/metabolismo , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética
4.
J Cell Mol Med ; 25(1): 229-243, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33249764

RESUMEN

Heart failure is the common final pathway of several cardiovascular conditions and a major cause of morbidity and mortality worldwide. Aberrant activation of the adaptive immune system in response to myocardial necrosis has recently been implicated in the development of heart failure. The ß-adrenergic agonist isoproterenol hydrochloride is used for its cardiac effects in a variety of different dosing regimens with high doses causing acute cardiomyocyte necrosis. To assess whether isoproterenol-induced cardiomyocyte necrosis triggers an adaptive immune response against the heart, we treated C57BL/6J mice with a single intraperitoneal injection of isoproterenol. We confirmed tissue damage reminiscent of human type 2 myocardial infarction. This is followed by an adaptive immune response targeting the heart as demonstrated by the activation of T cells, the presence of anti-heart auto-antibodies in the serum as late as 12 weeks after initial challenge and IgG deposition in the myocardium. All of these are hallmark signs of an established autoimmune response. Adoptive transfer of splenocytes from isoproterenol-treated mice induces left ventricular dilation and impairs cardiac function in healthy recipients. In summary, a single administration of a high dose of isoproterenol is a suitable high-throughput model for future studies of the pathological mechanisms of anti-heart autoimmunity and to test potential immunomodulatory therapeutic approaches.


Asunto(s)
Inmunidad Adaptativa , Infarto del Miocardio/inmunología , Miocardio/patología , Traslado Adoptivo , Animales , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Femenino , Fibrosis , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Isoproterenol , Antígenos Comunes de Leucocito/metabolismo , Masculino , Ratones Endogámicos C57BL , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Necrosis , Especificidad de Órganos , Bazo/inmunología , Sístole , Linfocitos T Colaboradores-Inductores/inmunología , Vasodilatación
5.
Circ Res ; 125(10): e75-e92, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31533542

RESUMEN

It is now well recognized that many lifesaving oncology drugs may adversely affect the heart and cardiovascular system, including causing irreversible cardiac injury that can result in reduced quality of life. These effects, which may manifest in the short term or long term, are mechanistically not well understood. Research is hampered by the reliance on whole-animal models of cardiotoxicity that may fail to reflect the fundamental biology or cardiotoxic responses of the human myocardium. The emergence of human induced pluripotent stem cell-derived cardiomyocytes as an in vitro research tool holds great promise for understanding drug-induced cardiotoxicity of oncological drugs that may manifest as contractile and electrophysiological dysfunction, as well as structural abnormalities, making it possible to deliver novel drugs free from cardiac liabilities and guide personalized therapy. This article briefly reviews the challenges of cardio-oncology, the strengths and limitations of using human induced pluripotent stem cell-derived cardiomyocytes to represent clinical findings in the nonclinical research space, and future directions for their further use.


Asunto(s)
American Heart Association , Antineoplásicos/toxicidad , Cardiotoxicidad/genética , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Animales , Cardiotoxicidad/metabolismo , Cardiotoxicidad/patología , Evaluación Preclínica de Medicamentos/métodos , Humanos , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/fisiología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Estados Unidos/epidemiología
6.
Europace ; 23(2): 305-312, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33083839

RESUMEN

AIMS: Rate adaptation of the action potential ensures spatial heterogeneities in conduction across the myocardium are minimized at different heart rates providing a protective mechanism against ventricular fibrillation (VF) and sudden cardiac death (SCD), which can be quantified by the ventricular conduction stability (V-CoS) test previously described. We tested the hypothesis that patients with a history of aborted SCD due to an underlying channelopathy or cardiomyopathy have a reduced capacity to maintain uniform activation following exercise. METHODS AND RESULTS: Sixty individuals, with (n = 28) and without (n = 32) previous aborted-SCD event underwent electro-cardiographic imaging recordings following exercise treadmill test. These included 25 Brugada syndrome, 13 hypertrophic cardiomyopathy, 12 idiopathic VF, and 10 healthy controls. Data were inputted into the V-CoS programme to calculate a V-CoS score that indicate the percentage of ventricle that showed no significant change in ventricular activation, with a lower score indicating the development of greater conduction heterogeneity. The SCD group, compared to those without, had a lower median (interquartile range) V-CoS score at peak exertion [92.8% (89.8-96.3%) vs. 97.3% (94.9-99.1%); P < 0.01] and 2 min into recovery [95.2% (91.1-97.2%) vs. 98.9% (96.9-99.5%); P < 0.01]. No significant difference was observable later into recovery at 5 or 10 min. Using the lowest median V-CoS scores obtained during the entire recovery period post-exertion, SCD survivors had a significantly lower score than those without for each of the different underlying aetiologies. CONCLUSION: Data from this pilot study demonstrate the potential use of this technique in risk stratification for the inherited cardiac conditions.


Asunto(s)
Muerte Súbita Cardíaca , Fibrilación Ventricular , Muerte Súbita Cardíaca/etiología , Corazón , Humanos , Proyectos Piloto , Factores de Riesgo , Sobrevivientes , Fibrilación Ventricular/diagnóstico
7.
Int J Mol Sci ; 22(24)2021 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-34948064

RESUMEN

The dextro-transposition of the great arteries (d-TGA) is one of the most common congenital heart diseases. To identify biological processes that could be related to the development of d-TGA, we established induced pluripotent stem cell (iPSC) lines from two patients with d-TGA and from two healthy subjects (as controls) and differentiated them into endothelial cells (iPSC-ECs). iPSC-EC transcriptome profiling and bioinformatics analysis revealed differences in the expression level of genes involved in circulatory system and animal organ development. iPSC-ECs from patients with d-TGA showed impaired ability to develop tubular structures in an in vitro capillary-like tube formation assay, and interactome studies revealed downregulation of biological processes related to Notch signaling, circulatory system development and angiogenesis, pointing to alterations in vascular structure development. Our study provides an iPSC-based cellular model to investigate the etiology of d-TGA.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Células Madre Pluripotentes Inducidas/citología , Receptores Notch/genética , Transposición de los Grandes Vasos/patología , Estudios de Casos y Controles , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Redes Reguladoras de Genes , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Modelos Biológicos , Análisis de Secuencia de ARN , Transducción de Señal , Transposición de los Grandes Vasos/genética
8.
J Mol Cell Cardiol ; 141: 11-16, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32201175

RESUMEN

Determining transmural mechanical properties in the heart provides a foundation to understand physiological and pathophysiological cardiac mechanics. Although work on mechanical characterisation has begun in isolated cells and permeabilised samples, the mechanical profile of living individual cardiac layers has not been examined. Myocardial slices are 300 µm-thin sections of heart tissue with preserved cellular stoichiometry, extracellular matrix, and structural architecture. This allows for cardiac mechanics assays in the context of an intact in vitro organotypic preparation. In slices obtained from the subendocardium, midmyocardium and subepicardium of rats, a distinct pattern in transmural contractility is found that is different from that observed in other models. Slices from the epicardium and midmyocardium had a higher active tension and passive tension than the endocardium upon stretch. Differences in total myocyte area coverage, and aspect ratio between layers underlined the functional readouts, while no differences were found in total sarcomeric protein and phosphoprotein between layers. Such intrinsic heterogeneity may orchestrate the normal pumping of the heart in the presence of transmural strain and sarcomere length gradients in the in vivo heart.


Asunto(s)
Miocardio/metabolismo , Animales , Fenómenos Biomecánicos , Proteínas Portadoras/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Ratas Sprague-Dawley , Sarcómeros/metabolismo , Troponina/metabolismo
9.
Pflugers Arch ; 472(10): 1435-1446, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32870378

RESUMEN

We describe a human and large animal Langendorff experimental apparatus for live electrophysiological studies and measure the electrophysiological changes due to gap junction uncoupling in human and porcine hearts. The resultant ex vivo intact human and porcine model can bridge the translational gap between smaller simple laboratory models and clinical research. In particular, electrophysiological models would benefit from the greater myocardial mass of a large heart due to its effects on far-field signal, electrode contact issues and motion artefacts, consequently more closely mimicking the clinical setting. Porcine (n = 9) and human (n = 4) donor hearts were perfused on a custom-designed Langendorff apparatus. Epicardial electrograms were collected at 16 sites across the left atrium and left ventricle. A total of 1 mM of carbenoxolone was administered at 5 ml/min to induce cellular uncoupling, and then recordings were repeated at the same sites. Changes in electrogram characteristics were analysed. We demonstrate the viability of a controlled ex vivo model of intact porcine and human hearts for electrophysiology with pharmacological modulation. Carbenoxolone reduces cellular coupling and changes contact electrogram features. The time from stimulus artefact to (-dV/dt)max increased between baseline and carbenoxolone (47.9 ± 4.1-67.2 ± 2.7 ms) indicating conduction slowing. The features with the largest percentage change between baseline and carbenoxolone were fractionation + 185.3%, endpoint amplitude - 106.9%, S-endpoint gradient + 54.9%, S point - 39.4%, RS ratio + 38.6% and (-dV/dt)max - 20.9%. The physiological relevance of this methodological tool is that it provides a model to further investigate pharmacologically induced pro-arrhythmic substrates.


Asunto(s)
Corazón/fisiología , Preparación de Corazón Aislado/métodos , Adulto , Animales , Carbenoxolona/farmacología , Electrocardiografía/métodos , Acoplamiento Excitación-Contracción , Femenino , Corazón/efectos de los fármacos , Humanos , Preparación de Corazón Aislado/instrumentación , Masculino , Miocardio/metabolismo , Porcinos
10.
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
11.
Hum Mol Genet ; 26(16): 3031-3045, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28521042

RESUMEN

Hypoplastic left heart syndrome (HLHS) is among the most severe forms of congenital heart disease. Although the consensus view is that reduced flow through the left heart during development is a key factor in the development of the condition, the molecular mechanisms leading to hypoplasia of left heart structures are unknown. We have generated induced pluripotent stem cells (iPSC) from five HLHS patients and two unaffected controls, differentiated these to cardiomyocytes and identified reproducible in vitro cellular and functional correlates of the HLHS phenotype. Our data indicate that HLHS-iPSC have a reduced ability to give rise to mesodermal, cardiac progenitors and mature cardiomyocytes and an enhanced ability to differentiate to smooth muscle cells. HLHS-iPSC-derived cardiomyocytes are characterised by a lower beating rate, disorganised sarcomeres and sarcoplasmic reticulum and a blunted response to isoprenaline. Whole exome sequencing of HLHS fibroblasts identified deleterious variants in NOTCH receptors and other genes involved in the NOTCH signalling pathway. Our data indicate that the expression of NOTCH receptors was significantly downregulated in HLHS-iPSC-derived cardiomyocytes alongside NOTCH target genes confirming downregulation of NOTCH signalling activity. Activation of NOTCH signalling via addition of Jagged peptide ligand during the differentiation of HLHS-iPSC restored their cardiomyocyte differentiation capacity and beating rate and suppressed the smooth muscle cell formation. Together, our data provide firm evidence for involvement of NOTCH signalling in HLHS pathogenesis, reveal novel genetic insights important for HLHS pathology and shed new insights into the role of this pathway during human cardiac development.


Asunto(s)
Síndrome del Corazón Izquierdo Hipoplásico/metabolismo , Síndrome del Corazón Izquierdo Hipoplásico/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Receptor Notch1/metabolismo , Estudios de Casos y Controles , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Recién Nacido/metabolismo , Masculino , Miocitos del Músculo Liso/metabolismo , Organogénesis , Transducción de Señal/fisiología
12.
J Muscle Res Cell Motil ; 40(2): 151-157, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31222588

RESUMEN

Tissue engineering is a complex field where the elements of biology and engineering are combined in an attempt to recapitulate the native environment of the body. Tissue engineering has shown one thing categorically; that the human body is extremely complex and it is truly a difficult task to generate this in the lab. There have been varied attempts at trying to generate a model for the heart with numerous cell types and different scaffolds or materials. The common underlying theme in these approaches is to combine together matrix material and different cell types to make something similar to heart tissue. Multi-cellularity is an essential aspect of the heart and therefore critical to any approach which would try to mimic such a complex tissue. The heart is made up of many cell types that combine to form complex structures like: deformable chambers, a tri-layered heart muscle, and vessels. Thus, in this review we will summarise how tissue engineering has progressed in modelling the heart and what gaps still exist in this dynamic field.


Asunto(s)
Matriz Extracelular/metabolismo , Miocardio/citología , Miocardio/metabolismo , Ingeniería de Tejidos , Animales , Humanos
13.
Europace ; 21(9): 1422-1431, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-30820561

RESUMEN

AIMS: Abnormal rate adaptation of the action potential is proarrhythmic but is difficult to measure with current electro-anatomical mapping techniques. We developed a method to rapidly quantify spatial discordance in whole heart activation in response to rate cycle length changes. We test the hypothesis that patients with underlying channelopathies or history of aborted sudden cardiac death (SCD) have a reduced capacity to maintain uniform activation following exercise. METHODS AND RESULTS: Electrocardiographical imaging (ECGI) reconstructs >1200 electrograms (EGMs) over the ventricles from a single beat, providing epicardial whole heart activation maps. Thirty-one individuals [11 SCD survivors; 10 Brugada syndrome (BrS) without SCD; and 10 controls] with structurally normal hearts underwent ECGI vest recordings following exercise treadmill. For each patient, we calculated the relative change in EGM local activation times (LATs) between a baseline and post-exertion phase using custom written software. A ventricular conduction stability (V-CoS) score calculated to indicate the percentage of ventricle that showed no significant change in relative LAT (<10 ms). A lower score reflected greater conduction heterogeneity. Mean variability (standard deviation) of V-CoS score over 10 consecutive beats was small (0.9 ± 0.5%), with good inter-operator reproducibility of V-CoS scores. Sudden cardiac death survivors, compared to BrS and controls, had the lowest V-CoS scores post-exertion (P = 0.011) but were no different at baseline (P = 0.50). CONCLUSION: We present a method to rapidly quantify changes in global activation which provides a measure of conduction heterogeneity and proof of concept by demonstrating SCD survivors have a reduced capacity to maintain uniform activation following exercise.


Asunto(s)
Mapeo del Potencial de Superficie Corporal/métodos , Síndrome de Brugada/fisiopatología , Muerte Súbita Cardíaca , Sistema de Conducción Cardíaco/fisiopatología , Ventrículos Cardíacos/fisiopatología , Corazón/fisiopatología , Estrés Fisiológico/fisiología , Fibrilación Ventricular/fisiopatología , Potenciales de Acción/fisiología , Adulto , Síndrome de Brugada/diagnóstico por imagen , Estudios de Casos y Controles , Electrocardiografía/métodos , Prueba de Esfuerzo , Femenino , Corazón/diagnóstico por imagen , Sistema de Conducción Cardíaco/diagnóstico por imagen , Ventrículos Cardíacos/diagnóstico por imagen , Humanos , Procesamiento de Imagen Asistido por Computador , Imagenología Tridimensional , Masculino , Persona de Mediana Edad , Procesamiento de Señales Asistido por Computador , Sobrevivientes , Pruebas de Mesa Inclinada , Tomografía Computarizada por Rayos X , Fibrilación Ventricular/diagnóstico por imagen , Dispositivos Electrónicos Vestibles
14.
Pflugers Arch ; 470(3): 559-570, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29297096

RESUMEN

Estrogen and ß2-adrenergic receptors (ß2AR) play important roles in the processes that protect the heart. Here, we investigated how ovariectomy influenced the ß2AR downstream pathways in the context of catecholaminergic stress. In vivo and in vitro stress models were developed in female Sprague-Dawley (SD) rats by epinephrine (Epi) treatments. The cardiac function was evaluated at in vivo and in vitro levels in terms of contraction, rhythm, and injury. We found that myocardial contractility was not significantly different between Sham and ovariectomized (OVX) group rats in the normal state. However, Epi pretreatment decreased the contractility and increased abnormal rhythms especially in OVX group, which were attributed to lack of estrogen. Inhibition of the ß2AR-Gi-PI3K/p38MAPK pathway with ICI118,551, PTX or LY294002 increased contractility and aggravated Epi-induced injury on cardiomyocytes, decreased p38MAPK phosphorylation, and only increased arrhythmia in Sham group. These results indicated that OVX exacerbated cardiac injury and abnormal rhythms through ß2AR-Gi-PI3K and ß2AR-Gi-p38MAPK pathways, respectively. In normal state, the levels of activated Gi were similar in both groups, but those of cAMP and activated Gs were higher in OVX group. Epi treatment increased activated Gi (especially in Sham group) and activated Gs and cAMP in Sham group but decreased it in OVX group. These results suggested that estrogen increased the Gi activity in normal and stress states and Gs activity in stress state. These results indicated that lack of estrogen impaired the ß2AR-Gs/Gi coupling during stress which compromised cardiac contractility and increased abnormal rhythms.


Asunto(s)
Arritmias Cardíacas/metabolismo , Estrógenos/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Estrés Psicológico/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/farmacología , Animales , Arritmias Cardíacas/etiología , Células Cultivadas , Epinefrina/farmacología , Femenino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/complicaciones , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Adv Funct Mater ; 28(21): 1800618, 2018 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-29875619

RESUMEN

An auxetic conductive cardiac patch (AuxCP) for the treatment of myocardial infarction (MI) is introduced. The auxetic design gives the patch a negative Poisson's ratio, providing it with the ability to conform to the demanding mechanics of the heart. The conductivity allows the patch to interface with electroresponsive tissues such as the heart. Excimer laser microablation is used to micropattern a re-entrant honeycomb (bow-tie) design into a chitosan-polyaniline composite. It is shown that the bow-tie design can produce patches with a wide range in mechanical strength and anisotropy, which can be tuned to match native heart tissue. Further, the auxetic patches are conductive and cytocompatible with murine neonatal cardiomyocytes in vitro. Ex vivo studies demonstrate that the auxetic patches have no detrimental effect on the electrophysiology of both healthy and MI rat hearts and conform better to native heart movements than unpatterned patches of the same material. Finally, the AuxCP applied in a rat MI model results in no detrimental effect on cardiac function and negligible fibrotic response after two weeks in vivo. This approach represents a versatile and robust platform for cardiac biomaterial design and could therefore lead to a promising treatment for MI.

16.
J Cardiovasc Electrophysiol ; 29(1): 115-126, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29091329

RESUMEN

BACKGROUND: Models of cardiac arrhythmogenesis predict that nonuniformity in repolarization and/or depolarization promotes ventricular fibrillation and is modulated by autonomic tone, but this is difficult to evaluate in patients. We hypothesize that such spatial heterogeneities would be detected by noninvasive ECG imaging (ECGi) in sudden cardiac death (SCD) survivors with structurally normal hearts under physiological stress. METHODS: ECGi was applied to 11 SCD survivors, 10 low-risk Brugada syndrome patients (BrS), and 10 controls undergoing exercise treadmill testing. ECGi provides whole heart activation maps and >1,200 unipolar electrograms over the ventricular surface from which global dispersion of activation recovery interval (ARI) and regional delay in conduction were determined. These were used as surrogates for spatial heterogeneities in repolarization and depolarization. Surface ECG markers of dispersion (QT and Tpeak-end intervals) were also calculated for all patients for comparison. RESULTS: Following exertion, the SCD group demonstrated the largest increase in ARI dispersion compared to BrS and control groups (13 ± 8 ms vs. 4 ± 7 ms vs. 4 ± 5 ms; P = 0.009), with baseline dispersion being similar in all groups. In comparison, surface ECG markers of dispersion of repolarization were unable to discriminate between the groups at baseline or following exertion. Spatial heterogeneities in conduction were also present following exercise but were not significantly different between SCD survivors and the other groups. CONCLUSION: Increased dispersion of repolarization is apparent during physiological stress in SCD survivors and is detectable with ECGi but not with standard ECG parameters. The electrophysiological substrate revealed by ECGi could be the basis of alternative risk-stratification techniques.


Asunto(s)
Potenciales de Acción , Mapeo del Potencial de Superficie Corporal , Muerte Súbita Cardíaca/etiología , Prueba de Esfuerzo , Ejercicio Físico , Sistema de Conducción Cardíaco/fisiopatología , Estrés Fisiológico , Fibrilación Ventricular/diagnóstico , Adulto , Anciano , Muerte Súbita Cardíaca/prevención & control , Técnicas Electrofisiológicas Cardíacas , Femenino , Frecuencia Cardíaca , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Fibrilación Ventricular/complicaciones , Fibrilación Ventricular/mortalidad , Fibrilación Ventricular/fisiopatología
17.
Clin Chem ; 63(5): 990-996, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28377413

RESUMEN

BACKGROUND: Myocardial infarction is diagnosed when biomarkers of cardiac necrosis exceed the 99th centile, although guidelines advocate even lower concentrations for early rule-out. We examined how many myocytes and how much myocardium these concentrations represent. We also examined if dietary troponin can confound the rule-out algorithm. METHODS: Individual rat cardiac myocytes, rat myocardium, ovine myocardium, or human myocardium were spiked into 400-µL aliquots of human serum. Blood was drawn from a volunteer after ingestion of ovine myocardium. High-sensitivity assays were used to measure cardiac troponin T (cTnT; Roche, Elecsys), cTnI (Abbott, Architect), and cardiac myosin-binding protein C (cMyC; EMD Millipore, Erenna®). RESULTS: The cMyC assay could only detect the human protein. For each rat cardiac myocyte added to 400 µL of human serum, cTnT and cTnI increased by 19.0 ng/L (95% CI, 16.8-21.2) and 18.9 ng/L (95% CI, 14.7-23.1), respectively. Under identical conditions cTnT, cTnI, and cMyC increased by 3.9 ng/L (95% CI, 3.6-4.3), 4.3 ng/L (95% CI, 3.8-4.7), and 41.0 ng/L (95% CI, 38.0-44.0) per µg of human myocardium. There was no detectable change in cTnI or cTnT concentration after ingestion of sufficient ovine myocardium to increase cTnT and cTnI to approximately 1 × 108 times their lower limits of quantification. CONCLUSIONS: Based on pragmatic assumptions regarding cTn and cMyC release efficiency, circulating species, and volume of distribution, 99th centile concentrations may be exceeded by necrosis of 40 mg of myocardium. This volume is much too small to detect by noninvasive imaging.


Asunto(s)
Biomarcadores/metabolismo , Infarto del Miocardio/diagnóstico , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Biomarcadores/química , Ingestión de Alimentos , Humanos , Infarto del Miocardio/sangre , Miocitos Cardíacos/citología , Ratas , Ovinos , Troponina I/sangre
18.
Stem Cells ; 34(1): 34-43, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26352327

RESUMEN

Heart failure is still a major cause of hospitalization and mortality in developed countries. Many clinical trials have tested the use of multipotent stem cells as a cardiac regenerative medicine. The benefit for the patients of this therapeutic intervention has remained limited. Herein, we review the pluripotent stem cells as a cell source for cardiac regeneration. We more specifically address the various challenges of this cell therapy approach. We question the cell delivery systems, the immune tolerance of allogenic cells, the potential proarrhythmic effects, various drug mediated interventions to facilitate cell grafting and, finally, we describe the pathological conditions that may benefit from such an innovative approach. As members of a transatlantic consortium of excellence of basic science researchers and clinicians, we propose some guidelines to be applied to cell types and modes of delivery in order to translate pluripotent stem cell cardiac derivatives into safe and effective clinical trials.


Asunto(s)
Insuficiencia Cardíaca/terapia , Miocardio/citología , Células Madre Pluripotentes/citología , Trasplante de Células Madre , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/fisiopatología , Humanos
19.
Postgrad Med J ; 92(1092): 581-6, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27531963

RESUMEN

OBJECTIVES: The 'gender gap' in academic medicine remains significant and predominantly favours males. This study investigates gender disparities in research performance in an Academic Health Science Centre, while considering factors such as mentoring and scientific collaboration. MATERIALS AND METHODS: Professorial registry-based electronic survey (n=215) using bibliometric data, a mentoring perception survey and social network analysis. Survey outcomes were aggregated with measures of research performance (publications, citations and h-index) and measures of scientific collaboration (authorship position, centrality and social capital). Univariate and multivariate regression models were constructed to evaluate inter-relationships and identify gender differences. RESULTS: One hundred and four professors responded (48% response rate). Males had a significantly higher number of previous publications than females (mean 131.07 (111.13) vs 79.60 (66.52), p=0.049). The distribution of mentoring survey scores between males and females was similar for the quality and frequency of shared core, mentor-specific and mentee-specific skills. In multivariate analysis including gender as a variable, the quality of managing the relationship, frequency of providing corrective feedback and frequency of building trust had a statistically significant positive influence on number of publications (all p<0.05). CONCLUSIONS: This is the first study in healthcare research to investigate the relationship between mentoring perception, scientific collaboration and research performance in the context of gender. It presents a series of initiatives that proved effective in marginalising the gender gap. These include the Athena Scientific Women's Academic Network charter, new recruitment and advertisement strategies, setting up a 'Research and Family Life' forum, establishing mentoring circles for women and projecting female role models.


Asunto(s)
Investigación Biomédica/estadística & datos numéricos , Conducta Cooperativa , Docentes Médicos/estadística & datos numéricos , Tutoría/estadística & datos numéricos , Sexismo , Centros Médicos Académicos , Bibliometría , Femenino , Humanos , Masculino , Análisis Multivariante , Percepción , Edición , Análisis de Regresión , Apoyo Social , Encuestas y Cuestionarios
20.
J Mol Cell Cardiol ; 67: 38-48, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24345421

RESUMEN

The purpose of this study was to investigate whether caveolin-3 (Cav3) regulates localization of ß2-adrenergic receptor (ß2AR) and its cAMP signaling in healthy or failing cardiomyocytes. We co-expressed wildtype Cav3 or its dominant-negative mutant (Cav3DN) together with the Förster resonance energy transfer (FRET)-based cAMP sensor Epac2-camps in adult rat ventricular myocytes (ARVMs). FRET and scanning ion conductance microscopy were used to locally stimulate ß2AR and to measure cytosolic cAMP. Cav3 overexpression increased the number of caveolae and decreased the magnitude of ß2AR-cAMP signal. Conversely, Cav3DN expression resulted in an increased ß2AR-cAMP response without altering the whole-cell L-type calcium current. Following local stimulation of Cav3DN-expressing ARVMs, ß2AR response could only be generated in T-tubules. However, the normally compartmentalized ß2AR-cAMP signal became diffuse, similar to the situation observed in heart failure. Finally, overexpression of Cav3 in failing myocytes led to partial ß2AR redistribution back into the T-tubules. In conclusion, Cav3 plays a crucial role for the localization of ß2AR and compartmentation of ß2AR-cAMP signaling to the T-tubules of healthy ARVMs, and overexpression of Cav3 in failing myocytes can partially restore the disrupted localization of these receptors.


Asunto(s)
Caveolina 3/metabolismo , Simulación por Computador , AMP Cíclico/metabolismo , Miocitos Cardíacos/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal , Animales , Western Blotting , Caveolina 3/genética , Síndromes Compartimentales/fisiopatología , Expresión Génica , Insuficiencia Cardíaca/fisiopatología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA