Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Dev Cell ; 58(24): 2881-2895.e7, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-37967560

RESUMEN

Generating organs from stem cells through blastocyst complementation is a promising approach to meet the clinical need for transplants. In order to generate rejection-free organs, complementation of both parenchymal and vascular cells must be achieved, as endothelial cells play a key role in graft rejection. Here, we used a lineage-specific cell ablation system to produce mouse embryos unable to form both the cardiac and vascular systems. By mouse intraspecies blastocyst complementation, we rescued heart and vascular system development separately and in combination, obtaining complemented hearts with cardiomyocytes and endothelial cells of exogenous origin. Complemented chimeras were viable and reached adult stage, showing normal cardiac function and no signs of histopathological defects in the heart. Furthermore, we implemented the cell ablation system for rat-to-mouse blastocyst complementation, obtaining xenogeneic hearts whose cardiomyocytes were completely of rat origin. These results represent an advance in the experimentation towards the in vivo generation of transplantable organs.


Asunto(s)
Sistema Cardiovascular , Corazón , Células Madre Pluripotentes , Animales , Ratones , Ratas , Blastocisto , Células Endoteliales , Miocitos Cardíacos , Corazón/embriología , Sistema Cardiovascular/embriología
2.
Circulation ; 147(1): 47-65, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36325906

RESUMEN

BACKGROUND: The complex genetics underlying human cardiac disease is evidenced by its heterogenous manifestation, multigenic basis, and sporadic occurrence. These features have hampered disease modeling and mechanistic understanding. Here, we show that 2 structural cardiac diseases, left ventricular noncompaction (LVNC) and bicuspid aortic valve, can be caused by a set of inherited heterozygous gene mutations affecting the NOTCH ligand regulator MIB1 (MINDBOMB1) and cosegregating genes. METHODS: We used CRISPR-Cas9 gene editing to generate mice harboring a nonsense or a missense MIB1 mutation that are both found in LVNC families. We also generated mice separately carrying these MIB1 mutations plus 5 additional cosegregating variants in the ASXL3, APCDD1, TMX3, CEP192, and BCL7A genes identified in these LVNC families by whole exome sequencing. Histological, developmental, and functional analyses of these mouse models were carried out by echocardiography and cardiac magnetic resonance imaging, together with gene expression profiling by RNA sequencing of both selected engineered mouse models and human induced pluripotent stem cell-derived cardiomyocytes. Potential biochemical interactions were assayed in vitro by coimmunoprecipitation and Western blot. RESULTS: Mice homozygous for the MIB1 nonsense mutation did not survive, and the mutation caused LVNC only in heteroallelic combination with a conditional allele inactivated in the myocardium. The heterozygous MIB1 missense allele leads to bicuspid aortic valve in a NOTCH-sensitized genetic background. These data suggest that development of LVNC is influenced by genetic modifiers present in affected families, whereas valve defects are highly sensitive to NOTCH haploinsufficiency. Whole exome sequencing of LVNC families revealed single-nucleotide gene variants of ASXL3, APCDD1, TMX3, CEP192, and BCL7A cosegregating with the MIB1 mutations and LVNC. In experiments with mice harboring the orthologous variants on the corresponding Mib1 backgrounds, triple heterozygous Mib1 Apcdd1 Asxl3 mice showed LVNC, whereas quadruple heterozygous Mib1 Cep192 Tmx3;Bcl7a mice developed bicuspid aortic valve and other valve-associated defects. Biochemical analysis suggested interactions between CEP192, BCL7A, and NOTCH. Gene expression profiling of mutant mouse hearts and human induced pluripotent stem cell-derived cardiomyocytes revealed increased cardiomyocyte proliferation and defective morphological and metabolic maturation. CONCLUSIONS: These findings reveal a shared genetic substrate underlying LVNC and bicuspid aortic valve in which MIB1-NOTCH variants plays a crucial role in heterozygous combination with cosegregating genetic modifiers.


Asunto(s)
Enfermedad de la Válvula Aórtica Bicúspide , Cardiomiopatías , Cardiopatías Congénitas , Células Madre Pluripotentes Inducidas , Humanos , Animales , Ratones , Cardiopatías Congénitas/complicaciones , Cardiomiopatías/etiología , Miocitos Cardíacos , Válvula Aórtica/diagnóstico por imagen , Factores de Transcripción , Proteínas Cromosómicas no Histona
3.
Int J Mol Sci ; 23(7)2022 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-35408974

RESUMEN

The presence of cartilage tissue in the embryonic and adult hearts of different vertebrate species is a well-recorded fact. However, while the embryonic neural crest has been historically considered as the main source of cardiac cartilage, recently reported results on the wide connective potential of epicardial lineage cells suggest they could also differentiate into chondrocytes. In this work, we describe the formation of cardiac cartilage clusters from proepicardial cells, both in vivo and in vitro. Our findings report, for the first time, cartilage formation from epicardial progenitor cells, and strongly support the concept of proepicardial cells as multipotent connective progenitors. These results are relevant to our understanding of cardiac cell complexity and the responses of cardiac connective tissues to pathologic stimuli.


Asunto(s)
Cresta Neural , Pericardio , Diferenciación Celular/fisiología , Condrocitos , Células Madre Embrionarias
4.
Biochem Mol Biol Educ ; 49(5): 782-788, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34156745

RESUMEN

A high variety of experimental model organisms have been used in developmental biology practical lectures. The work with developing embryos is crucial to make students aware of the multiple biological phenomena underlying normal animal embryogenesis and morphogenesis and represent a unique experimental platform to analyze the impact of molecular signaling in the regulation of all these processes. In particular, Biochemistry undergraduate students enjoy both practical and theoretical lectures on the molecular mechanisms of embryonic development, as that allows them for the integration of crucial molecular concepts (e.g. signaling and signal transduction mechanisms; molecular patterning of development) into the dynamic and progressive context of animal embryonic ontogenesis. Accordingly, it is important to carefully design practical laboratory lectures in developmental biology, as these are a unique pedagogical tools fostering the interests of the students in this subject. This study describes the design, implementation, and evaluation of a two-session laboratory practical activity performed by Biochemistry undergraduate students at University of Málaga (Spain). In this practical activity, which takes advantage of the unique characteristics of the chick embryo, students learn how the vertebrate heart forms from the fusion of two bilateral-symmetric cardiac progenitor pools under the guidance of the underlying endoderm. This cheap and easy practical laboratory activity provides relevant visual information on how experimental manipulations can severely influence anatomical form during organ development, as well as an excellent experimental setting to test molecular regulation of morphogenesis in an ex vivo (ex ovo) context.


Asunto(s)
Bioquímica , Cardias , Animales , Embrión de Pollo , Biología Evolutiva , Femenino , Humanos , Aprendizaje , Embarazo , Estudiantes
5.
J Anat ; 238(2): 508-514, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32920869

RESUMEN

Fsp1 (a.k.a S100A4 or Metastatin) is an intracellular and secreted protein widely regarded as a fibroblast marker. Recent studies have nonetheless shown that Fsp1 is also expressed by other cell types, including small subsets of endothelial cells. Since no detailed and systematic description of Fsp1 spatio-temporal expression pattern in cardiac vascular cells is available in the literature, we have used a transgenic murine line (Fsp1-GFP) to study Fsp1 expression in the developing and postnatal cardiac vasculature and endocardium. Our work shows that Fsp1 is expressed in the endocardium and mesenchyme of atrioventricular valve primordia, as well as in some coronary venous and lymphatic endothelial cells. Fsp1 expression in cardiac venous and lymphatic endothelium is progressively restricted to the leaflets of cardiac venous and lymphatic valves. Our results suggest that Fsp1 could play a role in the development of atrioventricular valves and participate in the patterning and morphogenesis of cardiac venous and lymphatic vessel valves.


Asunto(s)
Vasos Coronarios/embriología , Embrión de Mamíferos/metabolismo , Endocardio/embriología , Proteína de Unión al Calcio S100A4/metabolismo , Animales , Vasos Coronarios/metabolismo , Endocardio/metabolismo , Endotelio Linfático/metabolismo , Femenino , Ratones , Ratones Transgénicos , Embarazo , Válvulas Venosas/metabolismo
6.
J Exp Zool B Mol Dev Evol ; 332(6): 179-186, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31298492

RESUMEN

Cyclopia is a congenital anomaly characterized by the presence of a single or partially divided eye in a single orbit at the body midline. This condition is usually associated with other severe facial malformations, such as the absence of the nose and, on rare occasions, the presence of a proboscis located above the ocular structures. The developmental origin of cyclopia in vertebrates is the failure of the embryonic prosencephalon to divide properly during the formation of the two bilateral eyes. Although the developmental origin of the cyclopia-associated proboscis is not clear, it has been suggested that this unique structure results from the disrupted morphogenesis of the olfactory placodes, the main organizers of the developing nose. In this study, we report a spontaneous congenital case of cyclopia with a proboscis-like appendage in a chick embryo. By means of both conventional histology and immunohistochemical methods, we have analyzed this anomaly in detail to suggest an alternative identity for the anatomical embryonic features of cyclopic vertebrate embryos displaying a proboscis. Our findings are discussed in the context of previously reported cases of cyclopia, and provide additional insight into this complex congenital malformation.


Asunto(s)
Embrión de Pollo/anomalías , Holoprosencefalia/veterinaria , Animales , Holoprosencefalia/embriología , Holoprosencefalia/patología , Inmunohistoquímica
8.
Heart Fail Rev ; 24(1): 133-142, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30421074

RESUMEN

Ischemic cardiomyopathy is the cardiovascular condition with the highest impact on the Western population. In mammals (humans included), prolonged ischemia in the ventricular walls causes the death of cardiomyocytes (myocardial infarction, MI). The loss of myocardial mass is soon compensated by the formation of a reparative, non-contractile fibrotic scar that ultimately affects heart performance. Despite the enormous clinical relevance of MI, no effective therapy is available for the long-term treatment of this condition. Moreover, since the human heart is not able to undergo spontaneous regeneration, many researchers aim at designing cell-based therapies that allow for the substitution of dead cardiomyocytes by new, functional ones. So far, the majority of such strategies rely on the injection of different progenitor/stem cells to the infarcted heart. These cardiovascular progenitors, which are expected to differentiate into cardiomyocytes de novo, seldom give rise to new cardiac muscle. In this context, the most important challenge in the field is to fully disclose the molecular and cellular mechanisms that could promote active myocardial regeneration after cardiac damage. Accordingly, we suggest that such strategy should be inspired by the unique regenerative and reparative responses displayed by non-human animal models, from the restricted postnatal myocardial regeneration abilities of the murine heart to the full ventricular regeneration of some bony fishes (e.g., zebrafish). In this review article, we will discuss about current scientific approaches to study cardiac reparative and regenerative phenomena using animal models.


Asunto(s)
Infarto del Miocardio/terapia , Miocitos Cardíacos/fisiología , Regeneración/fisiología , Trasplante de Células Madre , Células Madre/fisiología , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Fibrosis/prevención & control , Humanos , Ratones , Miocitos Cardíacos/patología , Pez Cebra
9.
Arterioscler Thromb Vasc Biol ; 38(9): 2160-2173, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29930004

RESUMEN

Objective- Cardiac progenitor cells reside in the heart in adulthood, although their physiological relevance remains unknown. Here, we demonstrate that after myocardial infarction, adult Bmi1+ (B lymphoma Mo-MLV insertion region 1 homolog [PCGF4]) cardiac cells are a key progenitor-like population in cardiac neovascularization during ventricular remodeling. Approach and Results- These cells, which have a strong in vivo differentiation bias, are a mixture of endothelial- and mesenchymal-related cells with in vitro spontaneous endothelial cell differentiation capacity. Genetic lineage tracing analysis showed that heart-resident Bmi1+ progenitor cells proliferate after acute myocardial infarction and differentiate to generate de novo cardiac vasculature. In a mouse model of induced myocardial infarction, genetic ablation of these cells substantially deteriorated both heart angiogenesis and the ejection fraction, resulting in an ischemic-dilated cardiac phenotype. Conclusions- These findings imply that endothelial-related Bmi1+ progenitor cells are necessary for injury-induced neovascularization in adult mouse heart and highlight these cells as a suitable therapeutic target for preventing dysfunctional left ventricular remodeling after injury.


Asunto(s)
Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Neovascularización Patológica , Complejo Represivo Polycomb 1/fisiología , Células Madre/patología , Células Madre/fisiología , Remodelación Ventricular , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factores de Transcripción/metabolismo
10.
Cell Death Dis ; 9(3): 399, 2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29540665

RESUMEN

During mammalian heart development, restricted myocardial Bmp2 expression is a key patterning signal for atrioventricular canal specification and the epithelial-mesenchyme transition that gives rise to the valves. Using a mouse transgenic line conditionally expressing Bmp2, we show that widespread Bmp2 expression in the myocardium leads to valve and chamber dysmorphogenesis and embryonic death by E15.5. Transgenic embryos show thickened valves, ventricular septal defect, enlarged trabeculae and dilated ventricles, with an endocardium able to undergo EMT both in vivo and in vitro. Gene profiling and marker analysis indicate that cellular proliferation is increased in transgenic embryos, whereas chamber maturation and patterning are impaired. Similarly, forced Bmp2 expression stimulates proliferation and blocks cardiomyocyte differentiation of embryoid bodies. These data show that widespread myocardial Bmp2 expression directs ectopic valve primordium formation and maintains ventricular myocardium and cardiac progenitors in a primitive, proliferative state, identifying the potential of Bmp2 in the expansion of immature cardiomyocytes.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Proliferación Celular , Transición Epitelial-Mesenquimal , Miocardio/metabolismo , Miocitos Cardíacos/citología , Animales , Proteína Morfogenética Ósea 2/genética , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Transducción de Señal
11.
Cardiovasc Res ; 109(2): 204-16, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26811390

RESUMEN

Congenital coronary artery anomalies are of major significance in clinical cardiology and cardiac surgery due to their association with myocardial ischaemia and sudden death. Such anomalies are detectable by imaging modalities and, according to various definitions, their prevalence ranges from 0.21 to 5.79%. This consensus document from the Development, Anatomy and Pathology Working Group of the European Society of Cardiology aims to provide: (i) a definition of normality that refers to essential anatomical and embryological features of coronary vessels, based on the integrated analysis of studies of normal and abnormal coronary embryogenesis and pathophysiology; (ii) an animal model-based systematic survey of the molecular and cellular mechanisms that regulate coronary blood vessel development; (iii) an organization of the wide spectrum of coronary artery anomalies, according to a comprehensive anatomical and embryological classification scheme; (iv) current knowledge of the pathophysiological mechanisms underlying symptoms and signs of coronary artery anomalies, with diagnostic and therapeutic implications. This document identifies the mosaic-like embryonic development of the coronary vascular system, as coronary cell types differentiate from multiple cell sources through an intricate network of molecular signals and haemodynamic cues, as the necessary framework for understanding the complex spectrum of coronary artery anomalies observed in human patients.


Asunto(s)
Enfermedad de la Arteria Coronaria/congénito , Anomalías de los Vasos Coronarios , Vasos Coronarios , Corazón/anatomía & histología , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/patología , Animales , Cardiología/métodos , Enfermedad de la Arteria Coronaria/patología , Anomalías de los Vasos Coronarios/embriología , Anomalías de los Vasos Coronarios/patología , Anomalías de los Vasos Coronarios/fisiopatología , Vasos Coronarios/anatomía & histología , Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/patología , Corazón/fisiología , Humanos
12.
Dev Dyn ; 245(4): 445-59, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26708418

RESUMEN

BACKGROUND: Coronary artery (CA) stems connect the ventricular coronary tree with the aorta. Defects in proximal CA patterning are a cause of sudden cardiac death. In mice lacking Tbx1, common arterial trunk is associated with an abnormal trajectory of the proximal left CA. Here we investigate CA stem development in wild-type and Tbx1 null embryos. RESULTS: Genetic lineage tracing reveals that limited outgrowth of aortic endothelium contributes to proximal CA stems. Immunohistochemistry and fluorescent tracer injections identify a periarterial vascular plexus present at the onset of CA stem development. Transplantation experiments in avian embryos indicate that the periarterial plexus originates in mesenchyme distal to the outflow tract. Tbx1 is required for the patterning but not timing of CA stem development and a Tbx1 reporter allele is expressed in myocardium adjacent to the left but not right CA stem. This expression domain is maintained in Sema3c(-/-) hearts with a common arterial trunk and leftward positioned CA. Ectopic myocardial differentiation is observed on the left side of the Tbx1(-/-) common arterial trunk. CONCLUSIONS: A periarterial plexus bridges limited outgrowth of the aortic endothelium with the ventricular plexus during CA stem development. Molecular differences associated with left and right CA stems provide new insights into the etiology of CA patterning defects.


Asunto(s)
Aorta/embriología , Vasos Coronarios/embriología , Endotelio Vascular/embriología , Corazón/embriología , Células Madre/metabolismo , Proteínas de Dominio T Box/deficiencia , Animales , Aorta/patología , Embrión de Pollo , Vasos Coronarios/patología , Endotelio Vascular/patología , Ratones , Ratones Mutantes , Células Madre/patología
13.
Circ Res ; 109(12): 1429-42, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22158650

RESUMEN

The epicardium, the tissue layer covering the cardiac muscle (myocardium), develops from the proepicardium, a mass of coelomic progenitors located at the venous pole of the embryonic heart. Proepicardium cells attach to and spread over the myocardium to form the primitive epicardial epithelium. The epicardium subsequently undergoes an epithelial-to-mesenchymal transition to give rise to a population of epicardium-derived cells, which in turn invade the heart and progressively differentiate into various cell types, including cells of coronary blood vessels and cardiac interstitial cells. Epicardial cells and epicardium-derived cells signal to the adjacent cardiac muscle in a paracrine fashion, promoting its proliferation and expansion. Recently, high expectations have been raised about the epicardium as a candidate source of cells for the repair of the damaged heart. Because of its developmental importance and therapeutic potential, current research on this topic focuses on the complex signals that control epicardial biology. This review describes the signaling pathways involved in the different stages of epicardial development and discusses the potential of epicardial signals as targets for the development of therapies to repair the diseased heart.


Asunto(s)
Vasos Coronarios/embriología , Pericardio/embriología , Transducción de Señal/fisiología , Animales , Diferenciación Celular/fisiología , Vasos Coronarios/citología , Cardiopatías/fisiopatología , Cardiopatías/terapia , Humanos , Modelos Animales , Pericardio/citología
14.
PLoS One ; 6(9): e24175, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21949696

RESUMEN

During embryonic vasculogenesis, endothelial precursor cells of mesodermal origin known as angioblasts assemble into a characteristic network pattern. Although a considerable amount of markers and signals involved in this process have been identified, the mechanisms underlying the coalescence of angioblasts into this reticular pattern remain unclear. Various recent studies hypothesize that autocrine regulation of the chemoattractant vascular endothelial growth factor (VEGF) is responsible for the formation of vascular networks in vitro. However, the autocrine regulation hypothesis does not fit well with reported data on in vivo early vascular development. In this study, we propose a mathematical model based on the alternative assumption that endodermal VEGF signalling activity, having a paracrine effect on adjacent angioblasts, is mediated by its binding to the extracellular matrix (ECM). Detailed morphometric analysis of simulated networks and images obtained from in vivo quail embryos reveals the model mimics the vascular patterns with high accuracy. These results show that paracrine signalling can result in the formation of fine-grained cellular networks when mediated by angioblast-produced ECM. This lends additional support to the theory that patterning during early vascular development in the vertebrate embryo is regulated by paracrine signalling.


Asunto(s)
Matriz Extracelular/metabolismo , Mesodermo/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Comunicación Paracrina/fisiología , Algoritmos , Animales , Simulación por Computador , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Mesodermo/embriología , Mesodermo/metabolismo , Modelos Biológicos , Unión Proteica , Codorniz , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Circ Res ; 108(7): 824-36, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21311046

RESUMEN

RATIONALE: The proepicardium is a transient structure comprising epicardial progenitor cells located at the posterior limit of the embryonic cardiac inflow. A network of signals regulates proepicardial cell fate and defines myocardial and nonmyocardial domains at the venous pole of the heart. During cardiac development, epicardial-derived cells also contribute to coronary vessel morphogenesis. OBJECTIVE: To study Notch function during proepicardium development and coronary vessel formation in the mouse. METHODS AND RESULTS: Using in situ hybridization, RT-PCR, and immunohistochemistry, we find that Notch pathway elements are differentially activated throughout the proepicardial-epicardial-coronary transition. Analysis of RBPJk-targeted embryos indicates that Notch ablation causes ectopic procardiogenic signaling in the proepicardium that in turn promotes myocardial differentiation in adjacent mesodermal progenitors, resulting in a premature muscularization of the sinus venosus horns. Epicardium-specific Notch1 ablation using a Wt1-Cre driver line disrupts coronary artery differentiation, reduces myocardium wall thickness and myocyte proliferation, and reduces Raldh2 expression. Ectopic Notch1 activation disrupts epicardium development and causes thinning of ventricular walls. CONCLUSIONS: Epicardial Notch modulates cell differentiation in the proepicardium and adjacent pericardial mesoderm. Notch1 is later required for arterial endothelium commitment and differentiation and for vessel wall maturation during coronary vessel development and myocardium growth.


Asunto(s)
Circulación Sanguínea/fisiología , Vasos Coronarios/embriología , Morfogénesis/fisiología , Pericardio/embriología , Receptores Notch/fisiología , Transducción de Señal/fisiología , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/fisiología , Animales , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Vasos Coronarios/citología , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/fisiología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Modelos Animales , Mutación , Pericardio/citología , Receptor Notch1/genética , Receptor Notch1/fisiología , Receptores Notch/genética
16.
Development ; 138(6): 1093-7, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21343363

RESUMEN

Epicardial-derived signals are key regulators of cardiac embryonic development. An important part of these signals is known to relate to a retinoic acid (RA) receptor-dependent mechanism. RA is a potent morphogen synthesised by Raldh enzymes, Raldh2 being the predominant one in mesodermal tissues. Despite the importance of epicardial retinoid signalling in the heart, the molecular mechanisms controlling cardiac Raldh2 transcription remain unknown. In the current study, we show that Wt1-null epicardial cells display decreased expression of Raldh2 both in vivo and in vitro. Using a RA-responsive reporter, we have confirmed that Wt1-null epicardial cells actually show reduced synthesis of RA. We also demonstrate that Raldh2 is a direct transcriptional target of Wt1 in epicardial cells. A secondary objective of this study was to identify the status of RA-related receptors previously reported to be critical to epicardial biology (PDGFRα,ß; RXRα). PDGFRα and PDGFRß mRNA and protein levels are downregulated in the absence of Wt1, but only Pdgfra expression is rescued by the addition of RA to Wt1-null epicardial cells. RXRα mRNA levels are not affected in Wt1-null epicardial cells. Taken together, our results indicate that Wt1 critically regulates epicardial RA signalling via direct activation of the Raldh2 gene, and identify a role for Wt1 in the regulation of morphogen receptors involved in the proliferation, migration, and differentiation of epicardial and epicardially-derived cells (EPDC).


Asunto(s)
Aldehído Oxidorreductasas/genética , Pericardio/embriología , Tretinoina/metabolismo , Proteínas WT1/fisiología , Aldehído Oxidorreductasas/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Corazón/embriología , Ratones , Ratones Transgénicos , Pericardio/metabolismo , Transducción de Señal/genética , Activación Transcripcional/efectos de los fármacos , Tretinoina/farmacología , Tretinoina/fisiología
17.
J Clin Invest ; 120(10): 3493-507, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20890042

RESUMEN

Cardiac valve formation is crucial for embryonic and adult heart function. Valve malformations constitute the most common congenital cardiac defect, but little is known about the molecular mechanisms regulating valve formation and homeostasis. Here, we show that endocardial Notch1 and myocardial Bmp2 signal integration establish a valve-forming field between 2 chamber developmental domains. Patterning occurs through the activation of endocardial epithelial-to-mesenchymal transition (EMT) exclusively in prospective valve territories. Mice with constitutive endocardial Notch1 activity ectopically express Hey1 and Heyl. They also display an activated mesenchymal gene program in ventricles and a partial (noninvasive) EMT in vitro that becomes invasive upon BMP2 treatment. Snail1, TGF-ß2, or Notch1 inhibition reduces BMP2-induced ventricular transformation and invasion, whereas BMP2 treatment inhibits endothelial Gsk3ß, stabilizing Snail1 and promoting invasiveness. Integration of Notch and Bmp2 signals is consistent with Notch1 signaling being attenuated after myocardial Bmp2 deletion. Notch1 activation in myocardium extends Hey1 expression to nonchamber myocardium, represses Bmp2, and impairs EMT. In contrast, Notch deletion abrogates endocardial Hey gene transcription and extends Bmp2 expression to the ventricular endocardium. This embryonic Notch1-Bmp2-Snail1 relationship may be relevant in adult valve disease, in which decreased NOTCH signaling causes valve mesenchyme cell formation, fibrosis, and calcification.


Asunto(s)
Proteína Morfogenética Ósea 2/fisiología , Válvulas Cardíacas/embriología , Mesodermo/metabolismo , Receptor Notch1/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/análisis , Proteínas de Ciclo Celular/análisis , Células Epiteliales/patología , Regulación del Desarrollo de la Expresión Génica , Humanos , Mesodermo/patología , Ratones , Proteínas Represoras/análisis , Transducción de Señal , Factores de Transcripción de la Familia Snail , Factores de Transcripción/fisiología , Factor de Crecimiento Transformador beta2/fisiología
18.
Evol Dev ; 10(2): 210-6, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18315814

RESUMEN

The epicardium is the outer layer of the vertebrate heart. Both the embryonic epicardium and its derived mesenchyme are critical to heart development, contributing to the coronary vasculature and modulating the proliferation of the ventricular myocardium. The embryonic epicardium arises from an extracardiac, originally paired progenitor tissue called the proepicardium, a proliferation of coelomic cells found at the limit between the liver and the sinus venosus. Proepicardial cells attach to and spread over the cardiac surface giving rise to the epicardium. Invertebrate hearts always lack of epicardium, and no hypothesis has been proposed about the origin of this tissue and its proepicardial progenitor in vertebrates. We herein describe the epicardial development in a representative of the most basal living lineage of vertebrates, the agnathan Petromyzon marinus (lamprey). The epicardium in lampreys develops by migration of coelomic cells clustered in a paired structure at the roof of the coelomic cavity, between the pronephros and the gut. Later on, these outgrowths differentiate into the pronephric external glomerulus (PEG), a structure composed of capillary networks, mesangial cells, and podocytes. This observation is consistent with the conclusion that the primordia of the most anterior pair of PEG in agnathans have been retained and transformed into the proepicardium in gnathostomes. Glomerular progenitor cells are highly vasculogenic and probably allowed for the vascularization of a cardiac tube primarily devoid of coronary vessels. This new hypothesis accounts for the striking epicardial expression of Wt1 and Pod1, two transcription factors essential for development of the excretory system.


Asunto(s)
Evolución Biológica , Pericardio/embriología , Petromyzon/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cazón/embriología , Cazón/crecimiento & desarrollo , Riñón/embriología , Riñón/crecimiento & desarrollo , Larva/crecimiento & desarrollo , Pericardio/crecimiento & desarrollo , Petromyzon/genética , Petromyzon/crecimiento & desarrollo , Codorniz/embriología , Codorniz/genética , Codorniz/crecimiento & desarrollo , Especificidad de la Especie , Proteínas WT1/genética
19.
Dev Cell ; 12(3): 415-29, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17336907

RESUMEN

Ventricular chamber morphogenesis, first manifested by trabeculae formation, is crucial for cardiac function and embryonic viability and depends on cellular interactions between the endocardium and myocardium. We show that ventricular Notch1 activity is highest at presumptive trabecular endocardium. RBPJk and Notch1 mutants show impaired trabeculation and marker expression, attenuated EphrinB2, NRG1, and BMP10 expression and signaling, and decreased myocardial proliferation. Functional and molecular analyses show that Notch inhibition prevents EphrinB2 expression, and that EphrinB2 is a direct Notch target acting upstream of NRG1 in the ventricles. However, BMP10 levels are found to be independent of both EphrinB2 and NRG1 during trabeculation. Accordingly, exogenous BMP10 rescues the myocardial proliferative defect of in vitro-cultured RBPJk mutants, while exogenous NRG1 rescues differentiation in parallel. We suggest that during trabeculation Notch independently regulates cardiomyocyte proliferation and differentiation, two exquisitely balanced processes whose perturbation may result in congenital heart disease.


Asunto(s)
Diferenciación Celular/fisiología , Corazón/embriología , Mioblastos Cardíacos/metabolismo , Miocitos Cardíacos/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular , Efrina-B2/genética , Efrina-B2/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/metabolismo , Ratones , Mutación/genética , Mioblastos Cardíacos/citología , Miocitos Cardíacos/citología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neurregulina-1 , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores Notch/genética
20.
Dev Biol ; 295(2): 507-22, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16753139

RESUMEN

Proepicardial cells give rise to epicardium, coronary vasculature and cardiac fibroblasts. The proepicardium is derived from the mesodermal lining of the prospective pericardial cavity that simultaneously contributes myocardium to the venous pole of the elongating primitive heart tube. Using proepicardial explant cultures, we show that proepicardial cells have the potential to differentiate into cardiac muscle cells, reflecting the multipotency of this pericardial mesoderm. The differentiation into the myocardial or epicardial lineage is mediated by the cooperative action of BMP and FGF signaling. BMP2 is expressed in the distal IFT myocardium and stimulates cardiomyocyte formation. FGF2 is expressed in the proepicardium and stimulates differentiation into the epicardial lineage. In the base of the proepicardium, coexpression of BMP2 and FGF2 inhibits both myocardial and epicardial differentiation. We conclude that the epicardial/myocardial lineage decisions are mediated by an extrinsic, inductive mechanism, which is determined by the position of the cells in the pericardial mesoderm.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular , Linaje de la Célula , Factor 2 de Crecimiento de Fibroblastos/fisiología , Mesodermo/citología , Células Madre Multipotentes/citología , Miocardio/citología , Factor de Crecimiento Transformador beta/fisiología , Animales , Proteína Morfogenética Ósea 2 , Células Cultivadas , Embrión de Pollo , Miocitos Cardíacos/citología , Pericardio/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA