Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 133(1): 48-67, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37254794

RESUMEN

BACKGROUND: Left ventricular noncompaction (LVNC) is a prevalent cardiomyopathy associated with excessive trabeculation and thin compact myocardium. Patients with LVNC are vulnerable to cardiac dysfunction and at high risk of sudden death. Although sporadic and inherited mutations in cardiac genes are implicated in LVNC, understanding of the mechanisms responsible for human LVNC is limited. METHODS: We screened the complete exome sequence database of the Pediatrics Cardiac Genomics Consortium and identified a cohort with a de novo CHD4 (chromodomain helicase DNA-binding protein 4) proband, CHD4M202I, with congenital heart defects. We engineered a humanized mouse model of CHD4M202I (mouse CHD4M195I). Histological analysis, immunohistochemistry, flow cytometry, transmission electron microscopy, and echocardiography were used to analyze cardiac anatomy and function. Ex vivo culture, immunopurification coupled with mass spectrometry, transcriptional profiling, and chromatin immunoprecipitation were performed to deduce the mechanism of CHD4M195I-mediated ventricular wall defects. RESULTS: CHD4M195I/M195I mice developed biventricular hypertrabeculation and noncompaction and died at birth. Proliferation of cardiomyocytes was significantly increased in CHD4M195I hearts, and the excessive trabeculation was associated with accumulation of ECM (extracellular matrix) proteins and a reduction of ADAMTS1 (ADAM metallopeptidase with thrombospondin type 1 motif 1), an ECM protease. We rescued the hyperproliferation and hypertrabeculation defects in CHD4M195I hearts by administration of ADAMTS1. Mechanistically, the CHD4M195I protein showed augmented affinity to endocardial BRG1 (SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily A, member 4). This enhanced affinity resulted in the failure of derepression of Adamts1 transcription such that ADAMTS1-mediated trabeculation termination was impaired. CONCLUSIONS: Our study reveals how a single mutation in the chromatin remodeler CHD4, in mice or humans, modulates ventricular chamber maturation and that cardiac defects associated with the missense mutation CHD4M195I can be attenuated by the administration of ADAMTS1.


Asunto(s)
No Compactación Aislada del Miocardio Ventricular , Mutación Missense , Humanos , Animales , Niño , Ratones , Ventrículos Cardíacos , Causalidad , Mutación , Miocitos Cardíacos , Cromatina , No Compactación Aislada del Miocardio Ventricular/genética , Proteína ADAMTS1/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética
2.
Cell Mol Life Sci ; 81(1): 158, 2024 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-38556571

RESUMEN

Mutations in cysteine and glycine-rich protein 3 (CSRP3)/muscle LIM protein (MLP), a key regulator of striated muscle function, have been linked to hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) in patients. However, the roles of CSRP3 in heart development and regeneration are not completely understood. In this study, we characterized a novel zebrafish gene-trap line, gSAIzGFFM218A, which harbors an insertion in the csrp3 genomic locus, heterozygous fish served as a csrp3 expression reporter line and homozygous fish served as a csrp3 mutant line. We discovered that csrp3 is specifically expressed in larval ventricular cardiomyocytes (CMs) and that csrp3 deficiency leads to excessive trabeculation, a common feature of CSRP3-related HCM and DCM. We further revealed that csrp3 expression increased in response to different cardiac injuries and was regulated by several signaling pathways vital for heart regeneration. Csrp3 deficiency impeded zebrafish heart regeneration by impairing CM dedifferentiation, hindering sarcomere reassembly, and reducing CM proliferation while aggravating apoptosis. Csrp3 overexpression promoted CM proliferation after injury and ameliorated the impairment of ventricle regeneration caused by pharmacological inhibition of multiple signaling pathways. Our study highlights the critical role of Csrp3 in both zebrafish heart development and regeneration, and provides a valuable animal model for further functional exploration that will shed light on the molecular pathogenesis of CSRP3-related human cardiac diseases.


Asunto(s)
Cardiomiopatía Hipertrófica , Proteínas con Dominio LIM , Pez Cebra , Animales , Humanos , Pez Cebra/genética , Pez Cebra/metabolismo , Cisteína/genética , Cisteína/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Miocitos Cardíacos/metabolismo
3.
J Physiol ; 602(4): 597-617, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38345870

RESUMEN

Cardiac trabeculae are uneven ventricular muscular structures that develop during early embryonic heart development at the outer curvature of the ventricle. Their biomechanical function is not completely understood, and while their formation is known to be mechanosensitive, it is unclear whether ventricular tissue internal stresses play an important role in their formation. Here, we performed imaging and image-based cardiac biomechanics simulations on zebrafish embryonic ventricles to investigate these issues. Microscopy-based ventricular strain measurements show that the appearance of trabeculae coincided with enhanced deformability of the ventricular wall. Image-based biomechanical simulations reveal that the presence of trabeculae reduces ventricular tissue internal stresses, likely acting as structural support in response to the geometry of the ventricle. Passive ventricular pressure-loading experiments further reveal that the formation of trabeculae is associated with a spatial homogenization of ventricular tissue stiffnesses in healthy hearts, but gata1 morphants with a disrupted trabeculation process retain a spatial stiffness heterogeneity. Our findings thus suggest that modulating ventricular wall deformability, stresses, and stiffness are among the biomechanical functions of trabeculae. Further, experiments with gata1 morphants reveal that a reduction in fluid pressures and consequently ventricular tissue internal stresses can disrupt trabeculation, but a subsequent restoration of ventricular tissue internal stresses via vasopressin rescues trabeculation, demonstrating that tissue stresses are important to trabeculae formation. Overall, we find that tissue biomechanics is important to the formation and function of embryonic heart trabeculation. KEY POINTS: Trabeculations are fascinating and important cardiac structures and their abnormalities are linked to embryonic demise. However, their function in the heart and their mechanobiological formation processes are not completely understood. Our imaging and modelling show that tissue biomechanics is the key here. We find that trabeculations enhance cardiac wall deformability, reduce fluid pressure stresses, homogenize wall stiffness, and have alignments that are optimal for providing load-bearing structural support for the heart. We further discover that high ventricular tissue internal stresses consequent to high fluid pressures are needed for trabeculation formation through a rescue experiment, demonstrating that myocardial tissue stresses are as important as fluid flow wall shear stresses for trabeculation formation.


Asunto(s)
Miocitos Cardíacos , Pez Cebra , Animales , Fenómenos Biomecánicos , Transducción de Señal/fisiología , Miocardio , Corazón , Ventrículos Cardíacos
4.
J Anat ; 244(6): 1040-1053, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38284175

RESUMEN

That the highly trabeculated ventricular walls of the developing embryos transform to the arrangement during the fetal stages, when the mural architecture is dominated by the thickness of the compact myocardium, has been explained by the coalescence of trabeculations, often erroneously described as 'compaction'. Recent data, however, support differential rates of growth of the trabecular and compact layers as the major driver of change. Here, these processes were assessed quantitatively and visualized in standardized views. We used a larger dataset than has previously been available of mouse hearts, covering the period from embryonic day 10.5 to postnatal day 3, supported by images from human hearts. The volume of the trabecular layer increased throughout development, in contrast to what would be expected had there been 'compaction'. During the transition from embryonic to fetal life, the rapid growth of the compact layer diminished the proportion of trabeculations. Similarly, great expansion of the central cavity reduced the proportion of the total cavity made up of intertrabecular recesses. Illustrations of the hearts with the median value of left ventricular trabeculation confirm a pronounced growth of the compact wall, with prominence of the central cavity. This corresponds, in morphological terms, to a reduction in the extent of the trabecular layer. Similar observations were made in the human hearts. We conclude that it is a period of comparatively slow growth of the trabecular layer, rather than so-called compaction, that is the major determinant of the changing morphology of the ventricular walls of both mouse and human hearts.


Asunto(s)
Ventrículos Cardíacos , Animales , Humanos , Ratones , Ventrículos Cardíacos/anatomía & histología , Ventrículos Cardíacos/embriología , Edad Gestacional
5.
Adv Exp Med Biol ; 1441: 253-268, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884716

RESUMEN

Mammalian cardiac development is a complex, multistage process. Though traditional lineage tracing studies have characterized the broad trajectories of cardiac progenitors, the advent and rapid optimization of single-cell RNA sequencing methods have yielded an ever-expanding toolkit for characterizing heterogeneous cell populations in the developing heart. Importantly, they have allowed for a robust profiling of the spatiotemporal transcriptomic landscape of the human and mouse heart, revealing the diversity of cardiac cells-myocyte and non-myocyte-over the course of development. These studies have yielded insights into novel cardiac progenitor populations, chamber-specific developmental signatures, the gene regulatory networks governing cardiac development, and, thus, the etiologies of congenital heart diseases. Furthermore, single-cell RNA sequencing has allowed for the exquisite characterization of distinct cardiac populations such as the hard-to-capture cardiac conduction system and the intracardiac immune population. Therefore, single-cell profiling has also resulted in new insights into the regulation of cardiac regeneration and injury repair. Single-cell multiomics approaches combining transcriptomics, genomics, and epigenomics may uncover an even more comprehensive atlas of human cardiac biology. Single-cell analyses of the developing and adult mammalian heart offer an unprecedented look into the fundamental mechanisms of cardiac development and the complex diseases that may arise from it.


Asunto(s)
Corazón , Análisis de la Célula Individual , Animales , Humanos , Ratones , Diferenciación Celular/genética , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Corazón/embriología , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/citología , Organogénesis/genética , Regeneración/genética , Análisis de la Célula Individual/métodos , Transcriptoma/genética
6.
Semin Cell Dev Biol ; 118: 144-149, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33994094

RESUMEN

Heart malformation is the leading cause of human birth defects, and many of the congenital heart diseases (CHDs) originate from genetic defects that impact cardiac development and maturation. During development, the vertebrate heart undergoes a series of complex morphogenetic processes that increase its ability to pump blood. One of these processes leads to the formation of the sheet-like muscular projections called trabeculae. Trabeculae increase cardiac output and permit nutrition and oxygen uptake in the embryonic myocardium prior to coronary vascularization without increasing heart size. Cardiac trabeculation is also crucial for the development of the intraventricular fast conduction system. Alterations in cardiac trabecular development can manifest as a variety of congenital defects such as left ventricular noncompaction. In this review, we discuss the latest advances in understanding the molecular and cellular mechanisms underlying cardiac trabecular development.


Asunto(s)
Miocardio/patología , Miocitos Cardíacos/metabolismo , Humanos , Miocitos Cardíacos/citología
7.
J Biol Chem ; 298(1): 101449, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34838591

RESUMEN

The G-quadruplex (G4) resolvase RNA helicase associated with AU-rich element (RHAU) possesses the ability to unwind G4 structures in both DNA and RNA molecules. Previously, we revealed that RHAU plays a critical role in embryonic heart development and postnatal heart function through modulating mRNA translation and stability. However, whether RHAU functions to resolve DNA G4 in the regulation of cardiac physiology is still elusive. Here, we identified a phenotype of noncompaction cardiomyopathy in cardiomyocyte-specific Rhau deletion mice, including such symptoms as spongiform cardiomyopathy, heart dilation, and death at young ages. We also observed reduced cardiomyocyte proliferation and advanced sarcomere maturation in Rhau mutant mice. Further studies demonstrated that RHAU regulates the expression levels of several genes associated with ventricular trabeculation and compaction, including the Nkx2-5 and Hey2 that encode cardiac transcription factors of NKX2-5 and Hey2, and the myosin heavy chain 7 (Myh7) whose protein product is MYH7. While RHAU modulates Nkx2-5 mRNA and Hey2 mRNA at the post-transcriptional level, we uncovered that RHAU facilitates the transcription of Myh7 through unwinding of the G4 structures in its promoter. These findings demonstrated that RHAU regulates ventricular chamber development through both transcriptional and post-transcriptional mechanisms. These results contribute to a knowledge base that will help to understand the pathogenesis of diseases such as noncompaction cardiomyopathy.


Asunto(s)
ARN Helicasas DEAD-box , G-Cuádruplex , Miocitos Cardíacos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , ADN/metabolismo , Ventrículos Cardíacos , Proteína Homeótica Nkx-2.5/genética , Proteína Homeótica Nkx-2.5/metabolismo , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Procesamiento Proteico-Postraduccional , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
J Cell Sci ; 134(21)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34622921

RESUMEN

Cardiac looping and trabeculation are key processes during cardiac chamber maturation. However, the underlying mechanisms remain incompletely understood. Here, we report the isolation, cloning and characterization of the proprotein convertase furina from the cardiovascular mutant loft in zebrafish. loft is an ethylnitrosourea-induced mutant and has evident defects in the cardiac outflow tract, heart looping and trabeculation, the craniofacial region and pharyngeal arch arteries. Positional cloning revealed that furina mRNA was barely detectable in loft mutants, and loft failed to complement the TALEN-induced furina mutant pku338, confirming that furina is responsible for the loft mutant phenotypes. Mechanistic studies demonstrated that Notch reporter Tg(tp1:mCherry) signals were largely eliminated in mutant hearts, and overexpression of the Notch intracellular domain partially rescued the mutant phenotypes, probably due to the lack of Furina-mediated cleavage processing of Notch1b proteins, the only Notch receptor expressed in the heart. Together, our data suggest a potential post-translational modification of Notch1b proteins via the proprotein convertase Furina in the heart, and unveil the function of the Furina-Notch1b axis in cardiac looping and trabeculation in zebrafish, and possibly in other organisms.


Asunto(s)
Proproteína Convertasas , Proteínas de Pez Cebra , Pez Cebra , Animales , Corazón , Organogénesis/genética , Receptores Notch/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
9.
Morphologie ; 107(356): 147-150, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35787342

RESUMEN

Knowledge of anatomical variations of the heart are important to cardiac surgeons, cardiologists, and radiologist. During routine dissection of a 77-year-old male cadaver, we observed an unusual origin of a papillary muscle of the right ventricle arising from the atrioventricular aspect of the moderator band. This papillary muscle was 6.7mm long and 2.6mm wide. It gave rise to two chordae tendineae: one to the inferior (posterior) papillary muscle of the right ventricle and one directly to the inferior (posterior) leaflet of the tricuspid valve. Variants of the internal anatomy of the heart as exemplified in the present case report should be born in mind during image interpretation and invasive procedures of the right ventricle of the heart.


Asunto(s)
Ventrículos Cardíacos , Músculos Papilares , Masculino , Humanos , Anciano , Músculos Papilares/diagnóstico por imagen , Músculos Papilares/anatomía & histología , Músculos Papilares/fisiología , Ventrículos Cardíacos/diagnóstico por imagen , Cuerdas Tendinosas/fisiología , Cuerdas Tendinosas/cirugía , Válvula Tricúspide/fisiología , Válvula Tricúspide/cirugía , Cadáver
10.
Development ; 146(9)2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30992276

RESUMEN

Tissue morphogenesis requires changes in cell-cell adhesion as well as in cell shape and polarity. Cardiac trabeculation is a morphogenetic process essential for forming a functional ventricular wall. Here, we show that zebrafish hearts lacking Crb2a, a component of the Crumbs polarity complex, display compact wall integrity defects and fail to form trabeculae. Crb2a localization is very dynamic at a time when other cardiomyocyte junctional proteins also relocalize. Before the initiation of cardiomyocyte delamination to form the trabecular layer, Crb2a is expressed in all ventricular cardiomyocytes and colocalizes with the junctional protein ZO-1. Subsequently, Crb2a becomes localized all along the apical membrane of compact layer cardiomyocytes and is downregulated in the delaminating cardiomyocytes. We show that blood flow and Nrg/ErbB2 signaling regulate Crb2a localization dynamics. crb2a-/- display a multilayered wall with polarized cardiomyocytes: a unique phenotype. Our data further indicate that Crb2a regulates cardiac trabeculation by controlling the localization of tight and adherens junction proteins in cardiomyocytes. Importantly, transplantation data show that Crb2a controls CM behavior in a cell-autonomous manner in the sense that crb2a-/- cardiomyocytes transplanted into wild-type animals were always found in the trabecular layer. In summary, our study reveals a crucial role for Crb2a during cardiac development.


Asunto(s)
Proteínas de la Membrana/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas de Pez Cebra/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Animales , Adhesión Celular/genética , Adhesión Celular/fisiología , Polaridad Celular/genética , Polaridad Celular/fisiología , Proteínas de la Membrana/genética , Miocitos Cardíacos/citología , Pez Cebra , Proteínas de Pez Cebra/genética , Proteína de la Zonula Occludens-1/genética
11.
J Anat ; 241(1): 173-190, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35128670

RESUMEN

Shrews occupy the lower extreme of the seven orders of magnitude mammals range in size. Their hearts are large relative to body weight and heart rate can exceed a thousand beats a minute. It is not known whether traits typical of mammal hearts scale to these extremes. We assessed the heart of three species of shrew (genus Sorex) following the sequential segmental analysis developed for human hearts. Using micro-computed tomography, we describe the overall structure and find, in agreement with previous studies, a large and elongate ventricle. The atrial and ventricular septums and the atrioventricular (AV) and arterial valves are typically mammalian. The ventricular walls comprise mostly compact myocardium and especially the right ventricle has few trabeculations on the luminal side. A developmental process of compaction is thought to reduce trabeculations in mammals, but in embryonic shrews the volume of trabeculations increase for every gestational stage, only slower than the compact volume. By expression of Hcn4, we identify a sinus node and an AV conduction axis which is continuous with the ventricular septal crest. Outstanding traits include pulmonary venous sleeve myocardium that reaches farther into the lungs than in any other mammals. Typical proportions of coronary arteries-to-aorta do not scale and the shrew coronary arteries are proportionally enormous, presumably to avoid the high resistance to blood flow of narrow vessels. In conclusion, most cardiac traits do scale to the miniscule shrews. The shrew heart, nevertheless, stands out by its relative size, elongation, proportionally large coronary vessels, and extent of pulmonary venous myocardium.


Asunto(s)
Ventrículos Cardíacos , Corazón , Animales , Vasos Coronarios/anatomía & histología , Corazón/anatomía & histología , Atrios Cardíacos , Frecuencia Cardíaca , Humanos , Musarañas , Microtomografía por Rayos X
12.
J Mol Cell Cardiol ; 156: 45-56, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33773996

RESUMEN

CRELD1 (Cysteine-Rich with EGF-Like Domains 1) is a risk gene for non-syndromic atrioventricular septal defects in human patients. In a mouse model, Creld1 has been shown to be essential for heart development, particularly in septum and valve formation. However, due to the embryonic lethality of global Creld1 knockout (KO) mice, its cell type-specific function during peri- and postnatal stages remains unknown. Here, we generated conditional Creld1 KO mice lacking Creld1 either in the endocardium (KOTie2) or the myocardium (KOMyHC). Using a combination of cardiac phenotyping, histology, immunohistochemistry, RNA-sequencing, and flow cytometry, we demonstrate that Creld1 function in the endocardium is dispensable for heart development. Lack of myocardial Creld1 causes extracellular matrix remodeling and trabeculation defects by modulation of the Notch1 signaling pathway. Hence, KOMyHC mice die early postnatally due to myocardial hypoplasia. Our results reveal that Creld1 not only controls the formation of septa and valves at an early stage during heart development, but also cardiac maturation and function at a later stage. These findings underline the central role of Creld1 in mammalian heart development and function.


Asunto(s)
Moléculas de Adhesión Celular/genética , Proteínas de la Matriz Extracelular/genética , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Corazón/fisiología , Miocardio/metabolismo , Organogénesis/genética , Animales , Biomarcadores , Moléculas de Adhesión Celular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Ratones Noqueados , Análisis de la Célula Individual
13.
Development ; 145(14)2018 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-30061167

RESUMEN

Cardiomyocyte proliferation is crucial for cardiac growth, patterning and regeneration; however, few studies have investigated the behavior of dividing cardiomyocytes in vivo Here, we use time-lapse imaging of beating hearts in combination with the FUCCI system to monitor the behavior of proliferating cardiomyocytes in developing zebrafish. Confirming in vitro observations, sarcomere disassembly, as well as changes in cell shape and volume, precede cardiomyocyte cytokinesis. Notably, cardiomyocytes in zebrafish embryos and young larvae mostly divide parallel to the myocardial wall in both the compact and trabecular layers, and cardiomyocyte proliferation is more frequent in the trabecular layer. While analyzing known regulators of cardiomyocyte proliferation, we observed that the Nrg/ErbB2 and TGFß signaling pathways differentially affect compact and trabecular layer cardiomyocytes, indicating that distinct mechanisms drive proliferation in these two layers. In summary, our data indicate that, in zebrafish, cardiomyocyte proliferation is essential for trabecular growth, but not initiation, and set the stage to further investigate the cellular and molecular mechanisms driving cardiomyocyte proliferation in vivo.


Asunto(s)
Miocitos Cardíacos/citología , Organogénesis , Pez Cebra/crecimiento & desarrollo , Animales , División Celular , Proliferación Celular , Forma de la Célula , Tamaño de la Célula , Regulación del Desarrollo de la Expresión Génica , Corazón/crecimiento & desarrollo , Ligandos , Miocitos Cardíacos/metabolismo , Sarcómeros/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
14.
Development ; 145(10)2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29773645

RESUMEN

Cardiac trabeculation is a highly regulated process that starts with the delamination of compact layer cardiomyocytes. The Hippo signaling pathway has been implicated in cardiac development but many questions remain. We have investigated the role of Wwtr1, a nuclear effector of the Hippo pathway, in zebrafish and find that its loss leads to reduced cardiac trabeculation. However, in mosaic animals, wwtr1-/- cardiomyocytes contribute more frequently than wwtr1+/- cardiomyocytes to the trabecular layer of wild-type hearts. To investigate this paradox, we examined the myocardial wall at early stages and found that compact layer cardiomyocytes in wwtr1-/- hearts exhibit disorganized cortical actin structure and abnormal cell-cell junctions. Accordingly, wild-type cardiomyocytes in mosaic mutant hearts contribute less frequently to the trabecular layer than when present in mosaic wild-type hearts, indicating that wwtr1-/- hearts are not able to support trabeculation. We also found that Nrg/Erbb2 signaling, which is required for trabeculation, could promote Wwtr1 nuclear export in cardiomyocytes. Altogether, these data suggest that Wwtr1 establishes the compact wall architecture necessary for trabeculation, and that Nrg/Erbb2 signaling negatively regulates its nuclear localization and therefore its activity.


Asunto(s)
Corazón/embriología , Corazón/crecimiento & desarrollo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Miocitos Cardíacos/citología , Organogénesis/fisiología , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Proliferación Celular/fisiología , Uniones Intercelulares/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Morfolinos/genética , Cadenas Pesadas de Miosina/genética , Neurregulinas/metabolismo , Organogénesis/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor ErbB-2/metabolismo , Serina-Treonina Quinasa 3 , Transducción de Señal/fisiología , Transactivadores/metabolismo , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Troponina T/genética , Proteínas Señalizadoras YAP , Pez Cebra , Proteínas de Pez Cebra/genética
15.
Curr Top Membr ; 87: 131-151, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34696883

RESUMEN

Living cells are exposed to multiple mechanical stimuli from the extracellular matrix or from surrounding cells. Mechanoreceptors are molecules that display status changes in response to mechanical stimulation, transforming physical cues into biological responses to help the cells adapt to dynamic changes of the microenvironment. Mechanical stimuli are responsible for shaping the tridimensional development and patterning of the organs in early embryonic stages. The development of the heart is one of the first morphogenetic events that occur in embryos. As the circulation is established, the vascular system is exposed to constant shear stress, which is the force created by the movement of blood. Both spatial and temporal variations in shear stress differentially modulate critical steps in heart development, such as trabeculation and compaction of the ventricular wall and the formation of the heart valves. Zebrafish embryos are small, transparent, have a short developmental period and allow for real-time visualization of a variety of fluorescently labeled proteins to recapitulate developmental dynamics. In this review, we will highlight the application of zebrafish models as a genetically tractable model for investigating cardiovascular development and regeneration. We will introduce our approaches to manipulate mechanical forces during critical stages of zebrafish heart development and in a model of vascular regeneration, as well as advances in imaging technologies to capture these processes at high resolution. Finally, we will discuss the role of molecules of the Plexin family and Piezo cation channels as major mechanosensors recently implicated in cardiac morphogenesis.


Asunto(s)
Mecanotransducción Celular , Pez Cebra , Animales , Modelos Animales , Morfogénesis , Estrés Mecánico
16.
Dev Biol ; 446(2): 142-150, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30611731

RESUMEN

Ventricular trabeculation is one essential step for generating a functionally competent ventricular wall, while how the early trabeculae carneae forms and subsequently develops into mature chambers is poorly understood. We found that in zebrafish zfpm1-/- juvenile, cardiac function is significantly compromised, with hearts exhibiting deformed trabecular meshwork. To elucidate the mechanisms of Zfpm1 function in cardiac trabeculation, we analyzed zfpm1 mutant hearts more closely and found that loss of Zfpm1 activity resulted in over-activation of Neuregulin-ErbB signalling and abnormally elevated cardiomyocyte proliferation during cardiac trabeculae growth and modeling stages. These results implicate Zfpm1 plays a pivotal role in coordinating trabeculae patterning and growth.


Asunto(s)
Miocardio/metabolismo , Neurregulinas/genética , Receptor ErbB-2/genética , Transducción de Señal/genética , Proteínas de Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Proliferación Celular/genética , Células Cultivadas , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Corazón/embriología , Microscopía Confocal , Miocardio/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Neurregulinas/metabolismo , Receptor ErbB-2/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
17.
Dev Biol ; 442(1): 127-137, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30012423

RESUMEN

Development of the embryonic heart involves an intricate network of biochemical and genetic cues to ensure its proper growth and morphogenesis. However, studies from avian and teleost models reveal that biomechanical force, namely hemodynamic loading (blood pressure and shear stress), plays a significant role in regulating heart development. To study how hemodynamic loading impacts development of the mammalian embryonic heart, we utilized mouse embryo culture and manipulation techniques and performed optical projection tomography imaging followed by morphometric analysis to determine how reduced-loading affects heart volume, myocardial thickness, trabeculation and looping. Our results reveal that hemodynamic loading can regulate these features at different thresholds. Intermediate levels of hemodynamic loading are sufficient to promote proper myocardial growth and heart size, but insufficient to promote looping and trabeculation. Whereas, low levels of hemodynamic loading fails to promote proper growth of the myocardium and heart size. These results reveal that the regulation of heart development by biomechanical force is conserved across many vertebrate classes, and this study begins to elucidate how these specific forces regulate development of the mammalian heart.


Asunto(s)
Corazón/embriología , Hemodinámica/fisiología , Animales , Fenómenos Biomecánicos/fisiología , Ratones/embriología , Morfogénesis/fisiología , Miocardio/patología , Organogénesis , Estrés Mecánico
18.
J Card Fail ; 25(12): 1004-1008, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31626950

RESUMEN

BACKGROUND: Wolff-Parkinson-White (WPW) has been associated with left ventricular noncompaction (LVNC) in children. Little is known about the prevalence of this association, clinical outcomes, and treatment options. METHODS: Retrospective review of subjects with LVNC. LVNC was defined by established criteria; those with congenital heart disease were excluded. Electrocardiograms (ECGs) were reviewed for presence of pre-excitation. Outcomes were compared between those with isolated LVNC and those with WPW and LVNC. RESULTS: A total of 348 patients with LVNC were identified. Thirty-eight (11%) were found to have WPW pattern on ECG, and 84% of those with WPW and LVNC had cardiac dysfunction. In Kaplan-Meier analysis, there was significantly lower freedom from significant dysfunction (ejection fraction ≤ 40%) among those with WPW and LVNC (P < .001). Further analysis showed a higher risk of developing significant dysfunction in patients with WPW and LVNC versus LVNC alone (hazard ratio 4.64 [2.79, 9.90]). Twelve patients underwent an ablation procedure with an acute success rate of 83%. Four patients with cardiac dysfunction were successfully ablated, 3 having improvement in function. CONCLUSION: WPW is common among children with LVNC and is associated with cardiac dysfunction. Ablation therapy can be safely and effectively performed and may result in improvement in function.


Asunto(s)
No Compactación Aislada del Miocardio Ventricular/diagnóstico , No Compactación Aislada del Miocardio Ventricular/epidemiología , Síndrome de Wolff-Parkinson-White/diagnóstico , Síndrome de Wolff-Parkinson-White/epidemiología , Adolescente , Niño , Preescolar , Estudios de Cohortes , Electrocardiografía/métodos , Femenino , Estudios de Seguimiento , Humanos , Lactante , No Compactación Aislada del Miocardio Ventricular/fisiopatología , Masculino , Estudios Retrospectivos , Síndrome de Wolff-Parkinson-White/fisiopatología
19.
J Exp Biol ; 222(Pt 18)2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31439654

RESUMEN

The ejection fraction of the trabeculated cardiac ventricle of reptiles has not previously been measured. Here, we used the gold standard clinical methodology - electrocardiogram-gated flow magnetic resonance imaging (MRI) - to validate stroke volume measurements and end diastolic ventricular blood volume. This produced an estimate of ejection fraction in our study species, the red footed tortoise Chelonoidis carbonarius (n=5), under isoflurane anaesthesia of 88±11%. After reduction of the prevailing right-to-left intraventricular shunt through the action of atropine, the ejection fraction was 96±6%. This methodology opens new avenues for studying the complex hearts of ectotherms, and validating hypotheses on the function of a more highly trabeculated heart than that of endotherms, which have lower ejection fractions.


Asunto(s)
Volumen Sistólico/fisiología , Tortugas/fisiología , Anestésicos por Inhalación/administración & dosificación , Animales , Atropina/administración & dosificación , Electrocardiografía/métodos , Electrocardiografía/veterinaria , Femenino , Ventrículos Cardíacos , Isoflurano/administración & dosificación , Imagen por Resonancia Magnética/métodos , Imagen por Resonancia Magnética/veterinaria
20.
Echocardiography ; 36(4): 813-814, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30803005

RESUMEN

A 22-day-old boy born to a 21-year-old gravida 1, para 1 mother was admitted to our hospital for routine newborn examination. On physical examination, any clinical abnormality or malformations were not observed except 1/6 systolic murmur. A well-demarcated membranous structure was confirmed by echocardiography which was starting from left ventricular apex, extending to the mitral chords including papillary muscles in apical four-chamber and parasternal long-axis examination. This structure was considered as a variant of left ventricular noncompaction. The patient is still being followed up in our pediatric cardiology department.


Asunto(s)
Ecocardiografía/métodos , Ventrículos Cardíacos/diagnóstico por imagen , No Compactación Aislada del Miocardio Ventricular/diagnóstico por imagen , Humanos , Recién Nacido , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA