Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Adv Exp Med Biol ; 1441: 535-549, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884730

RESUMEN

Ventricular septation is a complex process which involves the major genes of cardiac development, acting on myocardial cells from first and second heart fields, and on mesenchymal cells from endocardial cushions. These genes, coding for transcription factors, interact with each other, and their differential expression conditions the severity of the phenotype. In this chapter, we will describe the formation of the ventricular septum in the normal heart, as well as the molecular mechanisms leading to the four main anatomic types of ventricular septal defects: outlet, inlet, muscular, and central perimembranous, resulting from failure of development of the different parts of the ventricular septum. Experiments on animal models, particularly transgenic mouse lines, have helped us to decipher the molecular determinants of ventricular septation. However, a precise description of the anatomic phenotypes found in these models is mandatory to achieve a better comprehension of the complex mechanisms responsible for the various types of VSDs.


Asunto(s)
Modelos Animales de Enfermedad , Defectos del Tabique Interventricular , Animales , Humanos , Ratones , Regulación del Desarrollo de la Expresión Génica , Defectos del Tabique Interventricular/genética , Defectos del Tabique Interventricular/patología , Defectos del Tabique Interventricular/metabolismo , Ratones Transgénicos , Transducción de Señal/genética , Tabique Interventricular/patología , Tabique Interventricular/metabolismo , Tabique Interventricular/embriología
2.
Adv Exp Med Biol ; 1441: 145-153, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884709

RESUMEN

The development of the inflow tract is undoubtedly one of the most complex remodeling events in the formation of the four-chambered heart. It involves the creation of two separate atrial chambers, the formation of an atrial/atrioventricular (AV) septal complex, the incorporation of the caval veins and coronary sinus into the right atrium, and the remodeling events that result in pulmonary venous return draining into the left atrium. In these processes, the atrioventricular mesenchymal complex, consisting of the major atrioventricular (AV) cushions, the mesenchymal cap on the primary atrial septum (pAS), and the dorsal mesenchymal protrusion (DMP), plays a crucial role.


Asunto(s)
Atrios Cardíacos , Animales , Humanos , Seno Coronario/embriología , Seno Coronario/anomalías , Corazón/embriología , Mesodermo/embriología , Venas Pulmonares/anomalías
3.
Adv Exp Med Biol ; 1441: 295-311, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884718

RESUMEN

Cardiac development is a fine-tuned process governed by complex transcriptional networks, in which transcription factors (TFs) interact with other regulatory layers. In this chapter, we introduce the core cardiac TFs including Gata, Hand, Nkx2, Mef2, Srf, and Tbx. These factors regulate each other's expression and can also act in a combinatorial manner on their downstream targets. Their disruption leads to various cardiac phenotypes in mice, and mutations in humans have been associated with congenital heart defects. In the second part of the chapter, we discuss different levels of regulation including cis-regulatory elements, chromatin structure, and microRNAs, which can interact with transcription factors, modulate their function, or are downstream targets. Finally, examples of disturbances of the cardiac regulatory network leading to congenital heart diseases in human are provided.


Asunto(s)
Redes Reguladoras de Genes , Cardiopatías Congénitas , Factores de Transcripción , Animales , Humanos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Corazón/fisiología , Miocardio/metabolismo
4.
Int J Mol Sci ; 25(3)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38339013

RESUMEN

The use of next-generation sequencing has provided new insights into the causes and mechanisms of congenital heart disease (CHD). Examinations of the whole exome sequence have detected detrimental gene variations modifying single or contiguous nucleotides, which are characterised as pathogenic based on statistical assessments of families and correlations with congenital heart disease, elevated expression during heart development, and reductions in harmful protein-coding mutations in the general population. Patients with CHD and extracardiac abnormalities are enriched for gene classes meeting these criteria, supporting a common set of pathways in the organogenesis of CHDs. Single-cell transcriptomics data have revealed the expression of genes associated with CHD in specific cell types, and emerging evidence suggests that genetic mutations disrupt multicellular genes essential for cardiogenesis. Metrics and units are being tracked in whole-genome sequencing studies.


Asunto(s)
Cardiopatías Congénitas , Humanos , Cardiopatías Congénitas/genética , Mutación , Exoma , Secuenciación Completa del Genoma , Genómica , Variaciones en el Número de Copia de ADN
5.
Dev Biol ; 473: 119-129, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33607112

RESUMEN

Retinoic acid (RA) signaling is required to restrict heart size through limiting the posterior boundary of the vertebrate cardiac progenitor field within the anterior lateral plate mesoderm (ALPM). However, we still do not fully understand how different cardiac progenitor populations that contribute to the developing heart, including earlier-differentiating first heart field (FHF), later-differentiating second heart field (SHF), and neural crest-derived progenitors, are each affected in RA-deficient embryos. Here, we quantified the number of cardiac progenitors and differentiating cardiomyocytes (CMs) in RA-deficient zebrafish embryos. While Nkx2.5+ cells were increased overall in the nascent hearts of RA-deficient embryos, unexpectedly, we found that the major effect within this population was a significant expansion in the number of differentiating FHF CMs. In contrast to the expansion of the FHF, there was a progressive decrease in SHF progenitors at the arterial pole as the heart tube elongated. Temporal differentiation assays and immunostaining in RA-deficient embryos showed that the outflow tracts (OFTs) of the hearts were significantly smaller, containing fewer differentiated SHF-derived ventricular CMs and a complete absence of SHF-derived smooth muscle at later stages. At the venous pole of the heart, pacemaker cells of the sinoatrial node also failed to differentiate in RA-deficient embryos. Interestingly, genetic lineage tracing showed that the number of neural-crest derived CMs was not altered within the enlarged hearts of RA-deficient zebrafish embryos. Altogether, our data show that the enlarged hearts in RA-deficient zebrafish embryos are comprised of an expansion in earlier differentiating FHF-derived CMs coupled with a progressive depletion of the SHF, suggesting RA signaling determines the relative ratios of earlier- and later-differentiation cardiac progenitors within an expanded cardiac progenitor pool.


Asunto(s)
Corazón/embriología , Mesodermo/metabolismo , Tretinoina/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica/genética , Corazón/fisiología , Ventrículos Cardíacos/metabolismo , Mesodermo/fisiología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Organogénesis/genética , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Tretinoina/fisiología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
6.
Genesis ; 59(11): e23458, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34665508

RESUMEN

The influence of retinoic acid (RA) signaling on vertebrate development has a well-studied history. Cumulatively, we now understand that RA signaling has a conserved requirement early in development restricting cardiac progenitors within the anterior lateral plate mesoderm of vertebrate embryos. Moreover, genetic and pharmacological manipulations of RA signaling in vertebrate models have shown that proper heart development is achieved through the deployment of positive and negative feedback mechanisms, which maintain appropriate RA levels. In this brief review, we present a chronological overview of key work that has led to a current model of the critical role for early RA signaling in limiting the generation of cardiac progenitors within vertebrate embryos. Furthermore, we integrate the previous work in mice and our recent findings using zebrafish, which together show that RA signaling has remarkably conserved influences on the later-differentiating progenitor populations at the arterial and venous poles. We discuss how recognizing the significant conservation of RA signaling on the differentiation of these progenitor populations offers new perspectives and may impact future work dedicated to examining vertebrate heart development.


Asunto(s)
Corazón/embriología , Mioblastos Cardíacos/metabolismo , Transducción de Señal , Tretinoina/metabolismo , Animales , Tipificación del Cuerpo , Mioblastos Cardíacos/citología , Vertebrados
7.
Development ; 142(11): 2037-47, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25953344

RESUMEN

Organ growth occurs through the integration of external growth signals during the G1 phase of the cell cycle to initiate DNA replication. Although numerous growth factor signals have been shown to be required for the proliferation of cardiomyocytes, genetic studies have only identified a very limited number of transcription factors that act to regulate the entry of cardiomyocytes into S phase. Here, we report that the cardiac para-zinc-finger protein CASZ1 is expressed in murine cardiomyocytes. Genetic fate mapping with an inducible Casz1 allele demonstrates that CASZ1-expressing cells give rise to cardiomyocytes in the first and second heart fields. We show through the generation of a cardiac conditional null mutation that Casz1 is essential for the proliferation of cardiomyocytes in both heart fields and that loss of Casz1 leads to a decrease in cardiomyocyte cell number. We further report that the loss of Casz1 leads to a prolonged or arrested S phase, a decrease in DNA synthesis, an increase in phospho-RB and a concomitant decrease in the cardiac mitotic index. Taken together, these studies establish a role for CASZ1 in mammalian cardiomyocyte cell cycle progression in both the first and second heart fields.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Fase G1 , Corazón/embriología , Mamíferos/embriología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fase S , Factores de Transcripción/metabolismo , Animales , Linaje de la Célula , Proliferación Celular , Embrión de Mamíferos/metabolismo , Femenino , Integrasas/metabolismo , Masculino , Ratones , Miocardio/citología , Miocardio/metabolismo , Miocardio/ultraestructura
8.
Development ; 141(16): 3112-22, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25038045

RESUMEN

Embryonic heart formation requires the production of an appropriate number of cardiomyocytes; likewise, cardiac regeneration following injury relies upon the recovery of lost cardiomyocytes. The basic helix-loop-helix (bHLH) transcription factor Hand2 has been implicated in promoting cardiomyocyte formation. It is unclear, however, whether Hand2 plays an instructive or permissive role during this process. Here, we find that overexpression of hand2 in the early zebrafish embryo is able to enhance cardiomyocyte production, resulting in an enlarged heart with a striking increase in the size of the outflow tract. Our evidence indicates that these increases are dependent on the interactions of Hand2 in multimeric complexes and are independent of direct DNA binding by Hand2. Proliferation assays reveal that hand2 can impact cardiomyocyte production by promoting division of late-differentiating cardiac progenitors within the second heart field. Additionally, our data suggest that hand2 can influence cardiomyocyte production by altering the patterning of the anterior lateral plate mesoderm, potentially favoring formation of the first heart field at the expense of hematopoietic and vascular lineages. The potency of hand2 during embryonic cardiogenesis suggested that hand2 could also impact cardiac regeneration in adult zebrafish; indeed, we find that overexpression of hand2 can augment the regenerative proliferation of cardiomyocytes in response to injury. Together, our studies demonstrate that hand2 can drive cardiomyocyte production in multiple contexts and through multiple mechanisms. These results contribute to our understanding of the potential origins of congenital heart disease and inform future strategies in regenerative medicine.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Miocitos Cardíacos/citología , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Secuencia de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proliferación Celular , ADN/química , Perfilación de la Expresión Génica , Genotipo , Hibridación in Situ , Hibridación Fluorescente in Situ , Ratones , Datos de Secuencia Molecular , Regeneración , Homología de Secuencia de Aminoácido , Transgenes , Proteínas de Pez Cebra/genética
9.
Development ; 140(20): 4165-76, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24026118

RESUMEN

In mammals, cardiac development proceeds from the formation of the linear heart tube, through complex looping and septation, all the while increasing in mass to provide the oxygen delivery demands of embryonic growth. The developing heart must orchestrate regional differences in cardiomyocyte proliferation to control cardiac morphogenesis. During ventricular wall formation, the compact myocardium proliferates more vigorously than the trabecular myocardium, but the mechanisms controlling such regional differences among cardiomyocyte populations are not understood. Control of definitive cardiomyocyte proliferation is of great importance for application to regenerative cell-based therapies. We have used murine and human pluripotent stem cell systems to demonstrate that, during in vitro cellular differentiation, early ventricular cardiac myocytes display a robust proliferative response to ß-catenin-mediated signaling and conversely accelerate differentiation in response to inhibition of this pathway. Using gain- and loss-of-function murine genetic models, we show that ß-catenin controls ventricular myocyte proliferation during development and the perinatal period. We further demonstrate that the differential activation of the Wnt/ß-catenin signaling pathway accounts for the observed differences in the proliferation rates of the compact versus the trabecular myocardium during normal cardiac development. Collectively, these results provide a mechanistic explanation for the differences in localized proliferation rates of cardiac myocytes and point to a practical method for the generation of the large numbers of stem cell-derived cardiac myocytes necessary for clinical applications.


Asunto(s)
Ventrículos Cardíacos/citología , Miocitos Cardíacos/citología , Células Madre Pluripotentes/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Activación Enzimática , Regulación del Desarrollo de la Expresión Génica , Ventrículos Cardíacos/embriología , Humanos , Ratones , Morfogénesis , Miocitos Cardíacos/metabolismo
10.
Heliyon ; 9(3): e14230, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36923876

RESUMEN

Previously, a single source of progenitor cells was thought to be responsible for the formation of the cardiac muscle. However, the second heart field has recently been identified as an additional source of myocardial progenitor cells. The chicken embryo, which develops in the egg, outside the mother can easily be manipulated in vivo and in vitro. Hence, it was an excellent model for establishing the concept of the second heart field. Here, our review will focus on the chicken model, specifically its role in understanding the second heart field. In addition to discussing historical aspects, we provide an overview of recent findings that have helped to define the chicken second heart field progenitor cells. A better understanding of the second heart field development will provide important insights into the congenital malformations affecting cardiac muscle formation and function.

11.
Methods Mol Biol ; 2429: 257-267, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35507167

RESUMEN

Myocardial infarction (MI) can lead to irreversible loss of cardiomyocytes (CMs), primarily localized to the left ventricle (LV) of the heart. The CMs of the LV are predominantly derived from first heart field (FHF) progenitors, whereas the majority of CMs within the right ventricle originate from the second heart field (SHF) during early cardiogenesis. Human embryonic stem cells (hESCs) serve as a valuable source of CMs for understanding early cardiac development and lineage commitment of CMs within these two heart fields that ultimately enable the development of more effective candidates for cell therapy. An ideal candidate may be FHF CMs that share the same ontogeny with the LV CMs that die after MI. We previously generated a double reporter hESC line that utilizes two important cardiac transcription factors, TBX5 and NKX2-5. TBX5 marks FHF progenitors and CMs, while NKX2-5 is expressed in nearly all myocytes of the developing heart. Here, we describe a step-by-step approach to efficiently generate FHF and SHF CMs using this double reporter hESC line. In addition, this approach can be applied to any non-genetically modified hESC lines to enrich FHF and SHF CMs. Obtaining enriched populations of these two CM subtypes provides a platform for downstream comparative analyses and in vitro studies to facilitate a deeper understanding of cardiovascular lineage commitment and the development of more effective candidates for cell therapy to treat diseases or defects that affect specific regions of the heart.


Asunto(s)
Células Madre Embrionarias Humanas , Infarto del Miocardio , Diferenciación Celular , Corazón , Ventrículos Cardíacos/metabolismo , Humanos , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo
12.
J Cardiovasc Dev Dis ; 8(4)2021 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-33918756

RESUMEN

The vertebrate heart integrates cells from the early-differentiating first heart field (FHF) and the later-differentiating second heart field (SHF), both emerging from the lateral plate mesoderm. In mammals, this process forms the basis for the development of the left and right ventricle chambers and subsequent chamber septation. The single ventricle-forming zebrafish heart also integrates FHF and SHF lineages during embryogenesis, yet the contributions of these two myocardial lineages to the adult zebrafish heart remain incompletely understood. Here, we characterize the myocardial labeling of FHF descendants in both the developing and adult zebrafish ventricle. Expanding previous findings, late gastrulation-stage labeling using drl-driven CreERT2 recombinase with a myocardium-specific, myl7-controlled, loxP reporter results in the predominant labeling of FHF-derived outer curvature and the right side of the embryonic ventricle. Raised to adulthood, such lineage-labeled hearts retain broad areas of FHF cardiomyocytes in a region of the ventricle that is positioned at the opposite side to the atrium and encompasses the apex. Our data add to the increasing evidence for a persisting cell-based compartmentalization of the adult zebrafish ventricle even in the absence of any physical boundary.

14.
J Cardiovasc Dev Dis ; 4(4)2017 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-29367549

RESUMEN

In this review, we will focus on the growth and morphogenesis of the developing heart, an aspect of cardiovascular development to which Antoon Moorman and colleagues have extensively contributed. Over the last decades, genetic studies and characterization of regionally regulated gene programs have provided abundant novel insights into heart development essential to understand the basis of congenital heart disease. Heart morphogenesis, however, is inherently a complex and dynamic three-dimensional process and we are far from understanding its cellular basis. Here, we discuss recent advances in studying heart morphogenesis and regionalization under the light of the pioneering work of Moorman and colleagues, which allowed the reinterpretation of regional gene expression patterns under a new morphogenetic framework. Two aspects of early heart formation will be discussed in particular: (1) the initial formation of the heart tube and (2) the formation of the cardiac chambers by the ballooning process. Finally, we emphasize that in addition to analyses based on fixed samples, new approaches including clonal analysis, single-cell sequencing, live-imaging and quantitative analysis of the data generated will likely lead to novel insights in understanding early heart tube regionalization and morphogenesis in the near future.

15.
Elife ; 62017 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-29202929

RESUMEN

During vertebrate heart development, two progenitor populations, first and second heart fields (FHF, SHF), sequentially contribute to longitudinal subdivisions of the heart tube (HT), with the FHF contributing the left ventricle and part of the atria, and the SHF the rest of the heart. Here, we study the dynamics of cardiac differentiation and morphogenesis by tracking individual cells in live analysis of mouse embryos. We report that during an initial phase, FHF precursors differentiate rapidly to form a cardiac crescent, while limited morphogenesis takes place. In a second phase, no differentiation occurs while extensive morphogenesis, including splanchnic mesoderm sliding over the endoderm, results in HT formation. In a third phase, cardiac precursor differentiation resumes and contributes to SHF-derived regions and the dorsal closure of the HT. These results reveal tissue-level coordination between morphogenesis and differentiation during HT formation and provide a new framework to understand heart development.


Asunto(s)
Corazón/embriología , Microscopía Intravital , Morfogénesis , Animales , Diferenciación Celular , Ratones , Análisis Espacio-Temporal
16.
Semin Fetal Neonatal Med ; 18(5): 237-44, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23886508

RESUMEN

Heart development is a complex process during which the heart needs to transform from a single tube towards a fully septated heart with four chambers and a separated outflow tract. Several major events contribute to this process, that largely overlap in time. Abnormal heart development results in congenital heart disease, which has an estimated incidence of 1% of liveborn children. Eighty percent of cases of congenital heart disease are considered to have a multifactoral developmental background, whereas knowledge of monogenetic causes for congenital heart disease is still limited. This review focuses on several novel findings in cardiac development that might enhance our knowledge of aetiology and support refinement of prenatal diagnosis of congenital heart disease.


Asunto(s)
Desarrollo Fetal , Corazón Fetal/fisiopatología , Cardiopatías/fisiopatología , Modelos Cardiovasculares , Animales , Modelos Animales de Enfermedad , Femenino , Corazón Fetal/fisiología , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/etiología , Cardiopatías Congénitas/fisiopatología , Cardiopatías/diagnóstico , Cardiopatías/embriología , Cardiopatías/etiología , Humanos , Recién Nacido , Masculino , Embarazo , Diagnóstico Prenatal
17.
Arch Cardiovasc Dis ; 106(11): 612-23, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24138816

RESUMEN

Accurate knowledge of normal cardiac development is essential for properly understanding the morphogenesis of congenital cardiac malformations that represent the most common congenital anomaly in newborns. The heart is the first organ to function during embryonic development and is fully formed at 8 weeks of gestation. Recent studies stemming from molecular genetics have allowed specification of the role of cellular precursors in the field of heart development. In this article we review the different steps of heart development, focusing on the processes of alignment and septation. We also show, as often as possible, the links between abnormalities of cardiac development and the main congenital heart defects. The development of animal models has permitted the unraveling of many mechanisms that potentially lead to cardiac malformations. A next step towards a better knowledge of cardiac development could be multiscale cardiac modelling.


Asunto(s)
Corazón/embriología , Animales , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Humanos , Modelos Animales , Morfogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA