Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
PLoS One ; 16(10): e0259426, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34714866

RESUMEN

ERBB family members and their ligands play an essential role in embryonic heart development and adult heart physiology. Among them, ERBB3 is a binding partner of ERBB2; the ERBB2/3 complex mediates downstream signaling for cell proliferation. ERBB3 has seven consensus binding sites to the p85 regulatory subunit of PI3K, which activates the downstream AKT pathway, leading to the proliferation of various cells. This study generated a human ERBB3 knock-in mouse expressing a mutant ERBB3 whose seven YXXM p85 binding sites were replaced with YXXA. Erbb3 knock-in embryos exhibited lethality between E12.5 to E13.5, and showed a decrease in mesenchymal cell numbers and density in AV cushions. We determined that the proliferation of mesenchymal cells in the atrioventricular (AV) cushion in Erbb3 knock-in mutant embryos was temporarily reduced due to the decrease of AKT and ERK1/2 phosphorylation. Overall, our results suggest that AKT/ERK activation by the ERBB3-dependent PI3K signaling is crucial for AV cushion morphogenesis during embryonic heart development.


Asunto(s)
Defectos de la Almohadilla Endocárdica/genética , Cojinetes Endocárdicos/metabolismo , Receptor ErbB-3/metabolismo , Animales , Sitios de Unión , Células CHO , Cricetinae , Cricetulus , Defectos de la Almohadilla Endocárdica/metabolismo , Cojinetes Endocárdicos/embriología , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-3/química , Receptor ErbB-3/genética , Transducción de Señal
2.
Cardiovasc Res ; 104(1): 49-60, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25100766

RESUMEN

AIMS: Valvular heart disease is responsible for considerable morbidity and mortality. Cardiac valves develop as the heart contracts, and they function throughout the lifetime of the organism to prevent retrograde blood flow. Their precise morphogenesis is crucial for cardiac function. Zebrafish is an ideal model to investigate cardiac valve development as it allows these studies to be carried out in vivo through non-invasive imaging. Accumulating evidence suggests a role for contractility and intracardiac flow dynamics in cardiac valve development. However, these two factors have proved difficult to uncouple, especially since altering myocardial function affects the intracardiac flow pattern. METHODS AND RESULTS: Here, we describe novel zebrafish models of developmental valve defects. We identified two mutant alleles of myosin heavy chain 6 that can be raised to adulthood despite having only one functional chamber-the ventricle. The adult mutant ventricle undergoes remodelling, and the atrioventricular (AV) valves fail to form four cuspids. In parallel, we characterized a novel mutant allele of southpaw, a nodal-related gene involved in the establishment of left-right asymmetry, which exhibits randomized heart and endoderm positioning. We first observed that in southpaw mutants the relative position of the two cardiac chambers is altered, affecting the geometry of the heart, while myocardial function appears unaffected. Mutant hearts that loop properly or exhibit situs inversus develop normally, whereas midline, unlooped hearts exhibit defects in their transvalvular flow pattern during AV valve development as well as defects in valve morphogenesis. CONCLUSION: Our data indicate that intracardiac flow dynamics regulate valve morphogenesis independently of myocardial contractility.


Asunto(s)
Circulación Coronaria , Defectos de la Almohadilla Endocárdica/embriología , Cojinetes Endocárdicos/embriología , Válvulas Cardíacas/anomalías , Hemodinámica , Mecanotransducción Celular , Animales , Animales Modificados Genéticamente , Función Atrial , Defectos de la Almohadilla Endocárdica/genética , Defectos de la Almohadilla Endocárdica/metabolismo , Defectos de la Almohadilla Endocárdica/fisiopatología , Cojinetes Endocárdicos/metabolismo , Cojinetes Endocárdicos/fisiopatología , Genotipo , Válvulas Cardíacas/metabolismo , Válvulas Cardíacas/fisiopatología , Morfogénesis , Mutación , Contracción Miocárdica , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Fenotipo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
3.
Cardiovasc Res ; 99(3): 452-60, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23723064

RESUMEN

AIMS: Anomalies of the arterial valves, principally bicuspid aortic valve (BAV), are the most common congenital anomalies. The cellular mechanisms that underlie arterial valve development are poorly understood. While it is known that the valve leaflets derive from the outflow cushions, which are populated by cells derived from the endothelium and neural crest cells (NCCs), the mechanism by which these cushions are sculpted to form the leaflets of the arterial valves remains unresolved. We set out to investigate how NCCs participate in arterial valve formation, reasoning that disrupting NCC within the developing outflow cushions would result in arterial valve anomalies, in the process elucidating the normal mechanism of arterial valve leaflet formation. METHODS AND RESULTS: By disrupting Rho kinase signalling specifically in NCC using transgenic mice and primary cultures, we show that NCC condensation within the cardiac jelly is required for correct positioning of the outflow cushions. Moreover, we show that this process is essential for normal patterning of the arterial valve leaflets with disruption leading to a spectrum of valve leaflet patterning anomalies, abnormal positioning of the orifices of the coronary arteries, and abnormalities of the arterial wall. CONCLUSION: NCCs are required at earlier stages of arterial valve development than previously recognized, playing essential roles in positioning the cushions, and patterning the valve leaflets. Abnormalities in the process of NCC condensation at early stages of outflow cushion formation may provide a common mechanism underlying BAV, and also explain the link with arterial wall anomalies and outflow malalignment defects.


Asunto(s)
Válvula Aórtica/embriología , Cojinetes Endocárdicos/citología , Cresta Neural/citología , Animales , Válvula Aórtica/anomalías , Válvula Aórtica/citología , Válvula Aórtica/metabolismo , Enfermedad de la Válvula Aórtica Bicúspide , Tipificación del Cuerpo , Adhesión Celular , Comunicación Celular , Células Cultivadas , Anomalías de los Vasos Coronarios/embriología , Anomalías de los Vasos Coronarios/metabolismo , Vasos Coronarios/embriología , Vasos Coronarios/metabolismo , Modelos Animales de Enfermedad , Defectos de la Almohadilla Endocárdica/embriología , Defectos de la Almohadilla Endocárdica/metabolismo , Cojinetes Endocárdicos/embriología , Cojinetes Endocárdicos/metabolismo , Enfermedades de las Válvulas Cardíacas/embriología , Enfermedades de las Válvulas Cardíacas/etiología , Enfermedades de las Válvulas Cardíacas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Modelos Cardiovasculares , Cresta Neural/anomalías , Cresta Neural/metabolismo , Transducción de Señal , Quinasas Asociadas a rho/deficiencia , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
4.
Development ; 139(2): 385-95, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22186728

RESUMEN

The progress of molecular genetics has enabled us to identify the genes responsible for congenital heart malformations. However, recent studies suggest that congenital heart diseases are induced not only by mutations in certain genes, but also by abnormal maternal factors. A high concentration of maternal retinoic acid (RA), the active derivative of vitamin A, is well known as a teratogenic agent that can cause developmental defects. Our previous studies have shown that the maternal administration of RA to mice within a narrow developmental window induces outflow tract (OFT) septum defects, a condition that closely resembles human transposition of the great arteries (TGA), although the responsible factors and pathogenic mechanisms of the TGA induced by RA remain unknown. We herein demonstrate that the expression of Tbx2 in the OFT myocardium is responsive to RA, and its downregulation is associated with abnormal OFT development. We found that RA could directly downregulate the Tbx2 expression through a functional retinoic acid response element (RARE) in the Tbx2 promoter region, which is also required for the initiation of Tbx2 transcription during OFT development. Tgfb2 expression was also downregulated in the RA-treated OFT region and was upregulated by Tbx2 in a culture system. Moreover, defective epithelial-mesenchymal transition caused by the excess RA was rescued by the addition of Tgfß2 in an organ culture system. These data suggest that RA signaling participates in the Tbx2 transcriptional mechanism during OFT development and that the Tbx2-Tgfß2 cascade is one of the key pathways involved in inducing the TGA phenotype.


Asunto(s)
Defectos de la Almohadilla Endocárdica/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Intercambio Materno-Fetal/fisiología , Miocardio/metabolismo , Transducción de Señal/fisiología , Proteínas de Dominio T Box/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Tretinoina/metabolismo , Animales , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Defectos de la Almohadilla Endocárdica/etiología , Femenino , Galactósidos , Inmunohistoquímica , Hibridación in Situ , Indoles , Luciferasas , Ratones , Análisis por Micromatrices , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tretinoina/fisiología
5.
Dev Biol ; 358(2): 368-78, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21839733

RESUMEN

Members of the GATA family of transcription factors are critical regulators of heart development and mutations in 2 of them, GATA4 and GATA6 are associated with outflow tract and septal defects in human. The heart expresses 3 GATA factors, GATA4, 5 and 6 in a partially overlapping pattern. Here, we report that compound Gata4/Gata5 and Gata5/Gata6 mutants die embryonically or perinatally due to severe congenital heart defects. Almost all Gata4(+/-)Gata5(+/-) mutant embryos have double outlet right ventricles (DORV), large ventricular septal defects (VSD) as well as hypertrophied mitral and tricuspid valves. Only 25% of double compound Gata4/Gata5 heterozygotes survive to adulthood and these mice have aortic stenosis. Compound loss of a Gata5 and a Gata6 allele also leads to DORVs associated with subaortic VSDs. Expression of several transcription factors important for endocardial and myocardial cell differentiation, such as Tbx20, Mef2c, Hey1 and Hand2, was reduced in compound heterozygote embryos. These findings suggest the existence of important genetic interactions between Gata5 and the 2 other cardiac GATA factors in endocardial cushion formation and outflow tract morphogenesis. The data identify GATA5 as a potential genetic modifier of congenital heart disease and provide insight for elucidating the genetic basis of an important class of human birth defects.


Asunto(s)
Corazón Fetal/embriología , Corazón Fetal/metabolismo , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA5/metabolismo , Factor de Transcripción GATA6/metabolismo , Animales , Estenosis de la Válvula Aórtica/embriología , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/metabolismo , Defectos de la Almohadilla Endocárdica/embriología , Defectos de la Almohadilla Endocárdica/genética , Defectos de la Almohadilla Endocárdica/metabolismo , Femenino , Factor de Transcripción GATA4/deficiencia , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA5/deficiencia , Factor de Transcripción GATA5/genética , Factor de Transcripción GATA6/deficiencia , Factor de Transcripción GATA6/genética , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Defectos del Tabique Interventricular/embriología , Defectos del Tabique Interventricular/genética , Defectos del Tabique Interventricular/metabolismo , Heterocigoto , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo
6.
Circ Res ; 107(10): 1209-19, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20847311

RESUMEN

RATIONALE: Heart valves develop from precursor structures called cardiac cushions, an endothelial-lined cardiac jelly that resides in the inner side of the heart tube. The cushions are then invaded by cells from different sources, undergo a series of complicated and poorly understood remodeling processes, and give rise to valves. Disruption of the fibroblast growth factor (FGF) signaling axis impairs morphogenesis of the outflow tract (OFT). Yet, whether FGF signaling regulates OFT valve formation is unknown. OBJECTIVE: To study how OFT valve formation is regulated and how aberrant cell signaling causes valve defects. METHODS AND RESULTS: By using mouse genetic manipulation, cell lineage tracing, ex vivo heart culture, and molecular biology approaches, we demonstrated that FGF signaling in the OFT myocardium upregulated Bmp4 expression, which then enhanced smooth muscle differentiation of neural crest cells (NCCs) in the cushion. FGF signaling also promoted OFT myocardial cell invasion to the cushion. Disrupting FGF signaling interrupted cushion remodeling with reduced NCCs differentiation into smooth muscle and less cardiomyocyte invasion and resulted in malformed OFT valves. CONCLUSIONS: The results demonstrate a novel mechanism by which the FGF-BMP signaling axis regulates formation of OFT valve primordia by controlling smooth muscle differentiation of cushion NCCs.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular , Defectos de la Almohadilla Endocárdica/metabolismo , Cojinetes Endocárdicos/metabolismo , Válvulas Cardíacas/metabolismo , Corazón/embriología , Miocitos del Músculo Liso/metabolismo , Cresta Neural/metabolismo , Transducción de Señal , Animales , Sitios de Unión , Proteína Morfogenética Ósea 4/genética , Diferenciación Celular/genética , Linaje de la Célula , Células Cultivadas , Defectos de la Almohadilla Endocárdica/genética , Defectos de la Almohadilla Endocárdica/patología , Cojinetes Endocárdicos/patología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Genotipo , Válvulas Cardíacas/anomalías , Válvulas Cardíacas/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Morfogénesis , Miocitos del Músculo Liso/patología , Cresta Neural/anomalías , Cresta Neural/patología , Técnicas de Cultivo de Órganos , Fenotipo , Regiones Promotoras Genéticas , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/genética , Factor de Transcripción AP-1/metabolismo , Transfección
7.
Dev Dyn ; 238(12): 3103-10, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19890913

RESUMEN

Defects in cardiac valvulogenesis are a common cause of congenital heart disease, and the study of this process promises to provide mechanistic insights and lead to novel therapeutics. Normal valve development involves multiple signaling pathways, and recently roles have been identified for extracellular matrix components, including glycosaminoglycans. We, therefore, explored the role of the glycosaminoglycan chondroitin sulfate during zebrafish cardiac development. Beginning at 33 hr, there is a distinct zone of chondroitin sulfate expression in the atrioventricular (AV) boundary, in the cardiac jelly between the endocardium and myocardium. This expression is both spatially and temporally restricted, and is undetectable after 48 hr. Chemical as well as genetic inhibition of chondroitin synthesis results in AV canal (AVC) defects, including loss of the atrioventricular constriction, blood regurgitation, and failure of circulation. Lack of chondroitin disrupts a marker of cell migration, results in a loss of myocardial and endothelial markers of valvulogenesis, and misregulates bone morphogenetic protein expression, supporting an early role in AVC development. In summary, we have defined a requirement for chondroitin sulfate expression in the normal patterning of the AV boundary, suggesting that this component of the cardiac jelly provides a necessary signal in this critical transition in vertebrate cardiogenesis.


Asunto(s)
Sulfatos de Condroitina/metabolismo , Cojinetes Endocárdicos/embriología , Corazón/embriología , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Movimiento Celular/fisiología , Embrión no Mamífero , Desarrollo Embrionario/fisiología , Defectos de la Almohadilla Endocárdica/genética , Defectos de la Almohadilla Endocárdica/metabolismo , Cojinetes Endocárdicos/efectos de los fármacos , Cojinetes Endocárdicos/metabolismo , Endocardio/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Miocardio/metabolismo , N-Acetilgalactosaminiltransferasas/antagonistas & inhibidores , N-Acetilgalactosaminiltransferasas/genética , ARN Interferente Pequeño/farmacología , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
8.
Dev Biol ; 302(1): 66-79, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17046737

RESUMEN

Eph proteins are receptor tyrosine kinases that control changes in cell shape and migration during development. We now describe a critical role for EphA3 receptor signaling in heart development as revealed by the phenotype of EphA3 null mice. During heart development mesenchymal outgrowths, the atrioventricular endocardial cushions, form in the atrioventricular canal. This morphogenetic event requires endocardial cushion cells to undergo an epithelial to mesenchymal transformation (EMT), and results in the formation of the atrioventricular valves and membranous portions of the atrial and ventricular septa. We show that EphA3 knockouts have significant defects in the development of their atrial septa and atrioventricular endocardial cushions, and that these cardiac abnormalities lead to the death of approximately 75% of homozygous EphA3(-/-) mutants. We demonstrate that EphA3 and its ligand, ephrin-A1, are expressed in adjacent cells in the developing endocardial cushions. We further demonstrate that EphA3(-/-) atrioventricular endocardial cushions are hypoplastic compared to wildtype and that EphA3(-/-) endocardial cushion explants give rise to fewer migrating mesenchymal cells than wildtype explants. Thus our results indicate that EphA3 plays a crucial role in the development and morphogenesis of the cells that give rise to the atrioventricular valves and septa.


Asunto(s)
Cardiopatías Congénitas/embriología , Corazón/embriología , Organogénesis , Receptor EphA3/genética , Receptor EphA3/metabolismo , Animales , Malformaciones Arteriovenosas/embriología , Malformaciones Arteriovenosas/metabolismo , Malformaciones Arteriovenosas/patología , Electrocardiografía , Defectos de la Almohadilla Endocárdica/embriología , Defectos de la Almohadilla Endocárdica/metabolismo , Defectos de la Almohadilla Endocárdica/patología , Corazón/fisiología , Cardiopatías Congénitas/patología , Cardiopatías Congénitas/fisiopatología , Defectos de los Tabiques Cardíacos/embriología , Defectos de los Tabiques Cardíacos/metabolismo , Defectos de los Tabiques Cardíacos/patología , Tabiques Cardíacos/embriología , Válvulas Cardíacas/embriología , Ratones , Ratones Noqueados , Mutación , Miocardio/metabolismo , Miocardio/patología
9.
Development ; 133(22): 4585-93, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17050629

RESUMEN

The transforming growth factorbeta (Tgfbeta) signaling pathway plays crucial roles in many biological processes. To understand the role(s) of Tgfbeta signaling during cardiogenesis in vivo and to overcome the early lethality of Tgfbr2(-/-) embryos, we applied a Cre/loxp system to specifically inactivate Tgfbr2 in either the myocardium or the endothelium of mouse embryos. Our results show that Tgfbr2 in the myocardium is dispensable for cardiogenesis in most embryos. Contrary to the prediction from results of previous in vitro collagen gel assays, inactivation of Tgfbr2 in the endocardium does not prevent atrioventricular cushion mesenchyme formation, arguing against its essential role in epithelium-mesenchyme transformation in vivo. We further demonstrate that Tgfbeta signaling is required for the proper remodeling of the atrioventricular canal and for cardiac looping, and that perturbation in Tgfbeta signaling causes the double-inlet left ventricle (DILV) defect. Thus, our study provides a unique mouse genetic model for DILV, further characterization of which suggests a potential cellular mechanism for the defect.


Asunto(s)
Diferenciación Celular/fisiología , Defectos de la Almohadilla Endocárdica/embriología , Corazón/embriología , Mesodermo/fisiología , Modelos Animales , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Defectos de la Almohadilla Endocárdica/metabolismo , Técnica del Anticuerpo Fluorescente , Hibridación in Situ , Mesodermo/metabolismo , Ratones , Ratones Mutantes , Microdisección
10.
Birth Defects Res A Clin Mol Teratol ; 76(7): 517-27, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16933305

RESUMEN

BACKGROUND: Formation of the primitive heart is a critical step for establishing a competent circulatory system necessary for continued morphogenesis, and as such has significant potential as a target for environmental insult. The goal of this study was to identify the initial cellular events that precede more superficially observable abnormalities resulting from exposing early chick embryos to trichloroethylene (TCE). METHODS: A whole embryo culture method was used to assess the susceptibility of endocardial epithelial-mesenchymal transformation in the early chick heart to TCE. This method has the benefits of maintaining the anatomical relationships of developing tissues and organs, instantaneously exposing precisely staged embryos to quantifiable levels of TCE in a protein-free medium, and the ability to directly monitor developmental morphology. RESULTS: A minority of embryos (Hamburger and Hamilton [HH] stage 13-14) exposed to TCE (10-80 ppm) were not viable after 24 hr in culture and exhibited a variety of gross malformations in a dose-dependent fashion. However, the majority of treated embryos remained viable and developed into HH stage 17 embryos that were superficially indistinguishable from vehicle-treated controls. Further analysis of the hearts of these superficially normal embryos by whole-mount confocal microscopy revealed selective reduction in the number of atrioventricular canal mesenchymal cells. Additionally, those mesenchymal cells that did develop migrated abnormally as long thin cords of adherent cells. CONCLUSIONS: The regional selectivity of these effects in the chick heart suggests a critical window of susceptibility to TCE in the epithelial-mesenchymal transformation of atrioventricular canal endocardium.


Asunto(s)
Defectos de la Almohadilla Endocárdica/inducido químicamente , Endocardio/efectos de los fármacos , Tricloroetileno/toxicidad , Animales , Embrión de Pollo , Relación Dosis-Respuesta a Droga , Defectos de la Almohadilla Endocárdica/embriología , Defectos de la Almohadilla Endocárdica/metabolismo , Endocardio/embriología , Endocardio/metabolismo , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/embriología , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/metabolismo , Mesodermo/metabolismo , Mesodermo/ultraestructura , Microscopía Confocal , Técnicas de Cultivo de Órganos , Factores de Tiempo
11.
Anat Rec A Discov Mol Cell Evol Biol ; 287(2): 1176-82, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16294330

RESUMEN

The majority of complex congenital heart defects occur in individuals who are afflicted by laterality disease. We hypothesize that the prevalence of valvuloseptal defects in this population is due to defective left-right patterning of the embryonic atrioventricular (AV) canal cushions, which are the progenitor tissue for valve and septal structures in the mature heart. Using embryos of the frog Xenopus laevis, this hypothesis was tested by performing left-right lineage analysis of myocytes and cushion mesenchyme cells of the superior and inferior cushion regions of the AV canal. Lineage analyses were conducted in both wild-type and laterality mutant embryos experimentally induced by misexpression of ALK4, a type I TGF-beta receptor previously shown to modulate left-right axis determination in Xenopus. We find that abnormalities in overall amount and left-right cell lineage composition are present in a majority of ALK4-induced laterality mutant embryos and that much variation in the nature of these abnormalities exists in embryos that exhibit the same overall body situs. We propose that these two parameters of cushion tissue formation-amount and left-right lineage origin-are important for normal processes of valvuloseptal morphogenesis and that defective allocation of cells in the AV canal might be causatively linked to the high incidence of valvuloseptal defects associated with laterality disease.


Asunto(s)
Defectos de la Almohadilla Endocárdica/embriología , Corazón Fetal/embriología , Mesodermo/citología , Miocitos Cardíacos/citología , Situs Inversus/embriología , Receptores de Activinas/genética , Receptores de Activinas/metabolismo , Receptores de Activinas Tipo I , Animales , Tipificación del Cuerpo/genética , Diferenciación Celular , Linaje de la Célula , Defectos de la Almohadilla Endocárdica/genética , Defectos de la Almohadilla Endocárdica/metabolismo , Corazón Fetal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mesodermo/metabolismo , Miocitos Cardíacos/metabolismo , ARN Mensajero/análisis , Situs Inversus/genética , Situs Inversus/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis
12.
Development ; 131(18): 4477-87, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15342473

RESUMEN

We have recently described a new subfamily of Fox genes, Foxp1/2/4, which are transcriptional repressors and are thought to regulate important aspects of development in several tissues, including the lung, brain, thymus and heart. Here, we show that Foxp1 is expressed in the myocardium as well as the endocardium of the developing heart. To further explore the role of Foxp1 in cardiac development, we inactivated Foxp1 through gene targeting in embryonic stem cells. Foxp1 mutant embryos have severe defects in cardiac morphogenesis, including outflow tract septation and cushion defects, a thin ventricular myocardial compact zone caused by defects in myocyte maturation and proliferation, and lack of proper ventricular septation. These defects lead to embryonic death at E14.5 and are similar to those observed in other mouse models of congenital heart disease, including Sox4 and Nfatc1 null embryos. Interestingly, expression of Sox4 in the outflow tract and cushions of Foxp1 null embryos is significantly reduced, while remodeling of the cushions is disrupted, as demonstrated by reduced apoptosis and persistent Nfatc1 expression in the cushion mesenchyme. Our results reveal a crucial role for Foxp1 in three aspects of cardiac development: (1) outflow tract development and septation, (2) tissue remodeling events required for cardiac cushion development, and (3) myocardial maturation and proliferation.


Asunto(s)
Diferenciación Celular , Defectos de la Almohadilla Endocárdica/metabolismo , Corazón/embriología , Corazón/fisiología , Morfogénesis , Células Musculares/patología , Proteínas Represoras/metabolismo , Animales , División Celular , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Regulación hacia Abajo , Pérdida del Embrión/metabolismo , Pérdida del Embrión/patología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Defectos de la Almohadilla Endocárdica/embriología , Defectos de la Almohadilla Endocárdica/patología , Endocardio/metabolismo , Endocardio/patología , Inhibidores Enzimáticos/farmacología , Factores de Transcripción Forkhead , Regulación del Desarrollo de la Expresión Génica , Corazón/efectos de los fármacos , Cardiopatías/congénito , Cardiopatías/metabolismo , Cardiopatías/patología , Proteínas del Grupo de Alta Movilidad/genética , Hibridación in Situ , Ratones , Ratones Noqueados , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Miocardio/metabolismo , Miocardio/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Factores de Transcripción SOXC , Transactivadores/genética
13.
Nat Med ; 10(8): 849-57, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15273746

RESUMEN

Noonan syndrome is a common human autosomal dominant birth defect, characterized by short stature, facial abnormalities, heart defects and possibly increased risk of leukemia. Mutations of Ptpn11 (also known as Shp2), which encodes the protein-tyrosine phosphatase Shp2, occur in approximately 50% of individuals with Noonan syndrome, but their molecular, cellular and developmental effects, and the relationship between Noonan syndrome and leukemia, are unclear. We generated mice expressing the Noonan syndrome-associated mutant D61G. When homozygous, the D61G mutant is embryonic lethal, whereas heterozygotes have decreased viability. Surviving Ptpn11(D61G/+) embryos ( approximately 50%) have short stature, craniofacial abnormalities similar to those in Noonan syndrome, and myeloproliferative disease. Severely affected Ptpn11(D61G/+) embryos ( approximately 50%) have multiple cardiac defects similar to those in mice lacking the Ras-GAP protein neurofibromin. Their endocardial cushions have increased Erk activation, but Erk hyperactivation is cell and pathway specific. Our results clarify the relationship between Noonan syndrome and leukemia and show that a single Ptpn11 gain-of-function mutation evokes all major features of Noonan syndrome by acting on multiple developmental lineages in a gene dosage-dependent and pathway-selective manner.


Asunto(s)
Dosificación de Gen , Mutación/genética , Síndrome de Noonan/genética , Proteínas Tirosina Fosfatasas/genética , Animales , Linaje de la Célula/genética , Anomalías Craneofaciales/genética , Modelos Animales de Enfermedad , Defectos de la Almohadilla Endocárdica/genética , Defectos de la Almohadilla Endocárdica/metabolismo , Citometría de Flujo , Genes Dominantes , Cardiopatías Congénitas/genética , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Mutantes , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Trastornos Mieloproliferativos/genética , Pruebas de Precipitina , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/metabolismo
14.
Mol Genet Metab ; 80(1-2): 27-35, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14567955

RESUMEN

Congenital heart defects, the leading cause of deaths from birth defects, are estimated to occur in close to 1% of live newborns. Among these, abnormal septation of the heart and valve anomalies are the most frequent forms. Despite progress defining several genes involved in normal heart development, we still have a limited understanding of the signaling pathways involved in morphogenesis of the outflow tract (OFT) and, to date, very few genes have been identified that are responsible for defects in humans. Bone Morphogenetic Protein (BMP) signaling pathways are emerging as vital regulators of multiple aspects of cardiogenesis, including the septation of the OFT and valve maturation. Genetic and other in vivo evidence is now supporting the role for BMPs as inducers of endocardial cushion epithelial-to-mesenchymal transformation that was suggested by in vitro explant studies as well as by their patterns of expression in the developing heart. Here, we review briefly the in vitro data, and detail the novel mouse models where perturbed BMP signaling pathways result in impaired OFT septation and semilunar valvulogenesis. We propose that growth of the OFT valve cushions is regulated by the level of BMP signaling, under the control of other signaling pathways.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Defectos de la Almohadilla Endocárdica/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Cardiopatías Congénitas/metabolismo , Válvulas Cardíacas/crecimiento & desarrollo , Miocardio/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Embrión de Mamíferos , Defectos de la Almohadilla Endocárdica/embriología , Defectos de la Almohadilla Endocárdica/patología , Regulación del Desarrollo de la Expresión Génica/fisiología , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/patología , Humanos , Técnicas In Vitro , Recién Nacido , Ratones , Morfogénesis/genética , Morfogénesis/fisiología , Mutación , Miocardio/patología , Transducción de Señal
15.
Clin Lab ; 47(9-10): 441-7, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11596905

RESUMEN

OBJECTIVES: The endogenous production of metabolites of the L-arginine-NO pathway has been found to be altered in patients with left-to-right shunt and pulmonary hypertension. The objective of this study was to analyze the influence of age and of the magnitude of the left-to-right shunt on plasma levels of L-arginine, cyclic guanosine monophosphate (cGMP), nitrite and nitrate in children and young adults presenting with left-to-right shunt. METHODS: Twenty-nine patients with ventricular septal defect (n=18), atrial septal defect (n=6) and atrioventricular canal (n=5) were assigned to group I when the ratio of pulmonary to systemic blood flow (Qp/Qs) was less than 1.5 (n=10) and to group II when Qp/Qs > or = 1.5 (n=19). At cardiac catheterization blood samples were taken from the pulmonary vein or left ventricle. In 33 controls peripheral venous blood was obtained. cGMP levels were determined by radioimmunoassay, L-arginine, nitrite and nitrate by high performance liquid chromatography (HPLC). RESULTS: L-arginine plasma levels were lower in group II than in controls (51.7 [23.3-82.2] versus 60.5 [32.4-85.9] pmol/l; p < 0.05 by KRUSKAL-WALLIS). Age did not influence the L-arginine plasma levels (p = 0.30). cGMP levels depended on age (p<0.01) and mean pulmonary artery pressure (p <0.01) but not on high pulmonary blood flow (p=0.85; ANOVA). Plasma nitrite and nitrate were not different in both groups and when compared with controls (nitrite: 26.0 [23.5-31.0] micromol/l; nitrate: 26.8 [24.0-32.0] micromol/l). CONCLUSIONS: Age and pulmonary artery pressure exert important effects on plasma cGMP. Measurement of nitrite and nitrate in plasma alone may not reflect the endogenous NO production. Future studies should evaluate the role of plasma levels of L-arginine in patients with high pulmonary blood flow undergoing repair of their defect.


Asunto(s)
Arginina/metabolismo , Derivación Arteriovenosa Quirúrgica , Defectos de los Tabiques Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Adolescente , Adulto , Factores de Edad , Arginina/sangre , Cateterismo Cardíaco , Estudios de Casos y Controles , Niño , Preescolar , GMP Cíclico/sangre , Defectos de la Almohadilla Endocárdica/sangre , Defectos de la Almohadilla Endocárdica/metabolismo , Defectos de la Almohadilla Endocárdica/fisiopatología , Femenino , Defectos de los Tabiques Cardíacos/sangre , Defectos de los Tabiques Cardíacos/fisiopatología , Hemodinámica , Humanos , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Lactante , Masculino , Nitratos/sangre , Óxido Nítrico/sangre , Circulación Pulmonar
16.
Toxicol Sci ; 53(1): 109-17, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10653528

RESUMEN

Previous epidemiological studies with humans and laboratory studies with chickens and rats linked trichloroethylene (TCE) exposure to cardiac defects. Although the cardiac defects in humans and laboratory animals produced by TCE are diverse, a majority of them involves valvular and septal structures. Progenitors of the valves and septa are formed by an epithelial-mesenchymal cell transformation of endothelial cells in the atrioventricular (AV) canal and outflow tract areas of the heart. Based on these studies, we hypothesized that TCE might cause cardiac valve and septa defects by specifically perturbing epithelial-mesenchymal cell transformation. We tested this hypothesis using an in vitro chick-AV canal culture model. This study shows that TCE affected several elements of epithelial-mesenchymal cell transformation. In particular, TCE blocked the endothelial cell-cell separation process that is associated with endothelial activation. Moreover, TCE inhibited mesenchymal cell formation throughout the concentration range tested (50-250 ppm). In contrast, TCE had no effect on the cell migration rate of the fully formed mesenchymal cells. Finally, the expression of 3 proteins (selected as molecular markers of epithelial-mesenchymal cell transformation) was analyzed in untreated and TCE-treated cultures. TCE inhibited the expression of the transcription factor Mox-1 and extracellular matrix (ECM) protein fibrillin 2. In contrast, TCE had no effect on the expression of alpha-smooth muscle actin. These data suggest that TCE may cause cardiac valvular and septal malformations by inhibiting endothelial separation and early events of mesenchymal cell formation in the heart.


Asunto(s)
Defectos de la Almohadilla Endocárdica/inducido químicamente , Válvulas Cardíacas/efectos de los fármacos , Tricloroetileno/toxicidad , Actinas/metabolismo , Oxidorreductasas de Alcohol/metabolismo , Animales , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Relación Dosis-Respuesta a Droga , Defectos de la Almohadilla Endocárdica/metabolismo , Endotelio/citología , Endotelio/efectos de los fármacos , Endotelio/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Fibrilina-2 , Fibrilinas , Técnica del Anticuerpo Fluorescente Indirecta , Válvulas Cardíacas/embriología , Válvulas Cardíacas/metabolismo , Mesodermo/citología , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Proteínas de Microfilamentos/metabolismo
17.
Pediatr Cardiol ; 17(1): 21-30, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8778697

RESUMEN

Atrioventricular septal defect occurs with a high prevalence in both human Down syndrome (trisomy 21) and the animal model for this disorder, murine trisomy 16 (Ts-16). The embryologic basis of this defect is the failure of the endocardial cushions to fuse. Quantitatively, Ts-16 hearts, when compared to normal mouse embryos, were not significantly different in either the estimates of whole heart volume or endocardial cushion volume. However, both the raw number of cardiac mesenchyme cells and the cellular density were reduced significantly. Qualitatively, endocardial cushion shape was elongated. Immunohistochemistry revealed an apparent delay in the temporally regulated expression of cytotactin and fibronectin during cushion development. Also, anti-heparan sulfate staining was noted on newly formed cardiac mesenchymal cells. These results suggest that the failure of endocardial cushion fusion in the Ts-16 mouse may be related to an elongated shape of the cushions and an inhibition or delay in the induction, transformation, or seeding of cardiac mesenchymal cells.


Asunto(s)
Modelos Animales de Enfermedad , Síndrome de Down/complicaciones , Defectos de la Almohadilla Endocárdica/embriología , Regulación del Desarrollo de la Expresión Génica , Animales , Síndrome de Down/embriología , Defectos de la Almohadilla Endocárdica/metabolismo , Defectos de la Almohadilla Endocárdica/patología , Fibronectinas/metabolismo , Edad Gestacional , Heparitina Sulfato/metabolismo , Inmunohistoquímica , Mesodermo/patología , Ratones , Ratones Mutantes , Tenascina/metabolismo
18.
Arkh Patol ; 48(6): 3-8, 1986.
Artículo en Ruso | MEDLINE | ID: mdl-3753235

RESUMEN

During surgical correction of Fallot's tetrad the authors discovered the decrease of macroergic phosphate concentration and moderately marked signs of cardiomyocytic ultrastructural changes, such as mitochondrial injuries, decrease of glycogen levels, insignificant intracellular edema, no increase of membrane penetration. Changes of endothelial cells were absent. In children not over 1 year old under myocardial protection with hypothermic perfusion (at low volume rates) some decrease of macroergic phosphates occurred, as well as a moderate decrease of the mitochondrial index, glycogen content decrease, there was no intracellular edema. The discovered structural changes are minimal and reversible. A close correlation was found between the electron microscopic data and the character of myocardial energetic metabolism, the biochemical changes preceding the ultrastructural ones. Comparison of morpho- and biochemical changes in the myocardium of the majority of patients with impaired myocardial function confirmed the existence of a complex relationship between them.


Asunto(s)
Adenosina Trifosfato/análisis , Cardiopatías Congénitas/patología , Miocardio/ultraestructura , Fosfocreatina/análisis , Niño , Preescolar , Defectos de la Almohadilla Endocárdica/metabolismo , Defectos de la Almohadilla Endocárdica/patología , Defectos del Tabique Interventricular/metabolismo , Defectos del Tabique Interventricular/patología , Humanos , Miocardio/análisis , Tetralogía de Fallot/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA