Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 280
Filtrar
1.
J Am Heart Assoc ; 12(18): e029683, 2023 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-37702066

RESUMEN

Background Endocardial cells are a major progenitor population that gives rise to heart valves through endocardial cushion formation by endocardial to mesenchymal transformation and the subsequent endocardial cushion remodeling. Genetic variants that affect these developmental processes can lead to congenital heart valve defects. Crk and Crkl are ubiquitously expressed genes encoding cytoplasmic adaptors essential for cell signaling. This study aims to explore the specific role of Crk and Crkl in the endocardial lineage during heart valve development. Methods and Results We deleted Crk and Crkl specifically in the endocardial lineage. The resultant heart valve morphology was evaluated by histological analysis, and the underlying cellular and molecular mechanisms were investigated by immunostaining and quantitative reverse transcription polymerase chain reaction. We found that the targeted deletion of Crk and Crkl impeded the remodeling of endocardial cushions at the atrioventricular canal into the atrioventricular valves. We showed that apoptosis was temporally increased in the remodeling atrioventricular endocardial cushions, and this developmentally upregulated apoptosis was repressed by deletion of Crk and Crkl. Loss of Crk and Crkl also resulted in altered extracellular matrix production and organization in the remodeling atrioventricular endocardial cushions. These morphogenic defects were associated with altered expression of genes in BMP (bone morphogenetic protein), connective tissue growth factor, and WNT signaling pathways, and reduced extracellular signal-regulated kinase signaling activities. Conclusions Our findings support that Crk and Crkl have shared functions in the endocardial lineage that critically regulate atrioventricular valve development; together, they likely coordinate the morphogenic signals involved in the remodeling of the atrioventricular endocardial cushions.


Asunto(s)
Endocardio , Válvulas Cardíacas , Apoptosis , Catéteres , Citosol , Endocardio/embriología , Transducción de Señal , Animales , Ratones , Válvulas Cardíacas/embriología
2.
Circ Res ; 130(3): 352-365, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-34995101

RESUMEN

BACKGROUND: Unraveling how new coronary arteries develop may provide critical information for establishing novel therapeutic approaches to treating ischemic cardiac diseases. There are 2 distinct coronary vascular populations derived from different origins in the developing heart. Understanding the formation of coronary arteries may provide insights into new ways of promoting coronary artery formation after myocardial infarction. METHODS: To understand how intramyocardial coronary arteries are generated to connect these 2 coronary vascular populations, we combined genetic lineage tracing, light sheet microscopy, fluorescence micro-optical sectioning tomography, and tissue-specific gene knockout approaches to understand their cellular and molecular mechanisms. RESULTS: We show that a subset of intramyocardial coronary arteries form by angiogenic extension of endocardium-derived vascular tunnels in the neonatal heart. Three-dimensional whole-mount fluorescence imaging showed that these endocardium-derived vascular tunnels or tubes adopt an arterial fate in neonates. Mechanistically, we implicate Mettl3 (methyltransferase-like protein 3) and Notch signaling in regulating endocardium-derived intramyocardial coronary artery formation. Functionally, these intramyocardial arteries persist into adulthood and play a protective role after myocardial infarction. CONCLUSIONS: A subset of intramyocardial coronary arteries form by extension of endocardium-derived vascular tunnels in the neonatal heart.


Asunto(s)
Vasos Coronarios/embriología , Endocardio/embriología , Animales , Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/metabolismo , Endocardio/crecimiento & desarrollo , Endocardio/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Organogénesis
3.
Cell Death Dis ; 12(4): 393, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33846290

RESUMEN

Cardiac septum malformations account for the largest proportion in congenital heart defects. The transcription factor Sox7 has critical functions in the vascular development and angiogenesis. It is unclear whether Sox7 also contributes to cardiac septation development. We identified a de novo 8p23.1 deletion with Sox7 haploinsufficiency in an atrioventricular septal defect (AVSD) patient using whole exome sequencing in 100 AVSD patients. Then, multiple Sox7 conditional loss-of-function mice models were generated to explore the role of Sox7 in atrioventricular cushion development. Sox7 deficiency mice embryos exhibited partial AVSD and impaired endothelial to mesenchymal transition (EndMT). Transcriptome analysis revealed BMP signaling pathway was significantly downregulated in Sox7 deficiency atrioventricular cushions. Mechanistically, Sox7 deficiency reduced the expressions of Bmp2 in atrioventricular canal myocardium and Wnt4 in endocardium, and Sox7 binds to Wnt4 and Bmp2 directly. Furthermore, WNT4 or BMP2 protein could partially rescue the impaired EndMT process caused by Sox7 deficiency, and inhibition of BMP2 by Noggin could attenuate the effect of WNT4 protein. In summary, our findings identify Sox7 as a novel AVSD pathogenic candidate gene, and it can regulate the EndMT involved in atrioventricular cushion morphogenesis through Wnt4-Bmp2 signaling. This study contributes new strategies to the diagnosis and treatment of congenital heart defects.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Defectos de los Tabiques Cardíacos/metabolismo , Factores de Transcripción SOXF/metabolismo , Proteína Wnt4/metabolismo , Animales , Estudios de Casos y Controles , Preescolar , Endocardio/embriología , Endocardio/crecimiento & desarrollo , Endocardio/metabolismo , Femenino , Defectos de los Tabiques Cardíacos/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Factores de Transcripción SOXF/deficiencia , Factores de Transcripción SOXF/genética , Transducción de Señal
4.
Clin Sci (Lond) ; 135(6): 829-846, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33720353

RESUMEN

The endothelial-to-mesenchymal transition (EndMT) is a critical process that occurs during the development of the outflow tract (OFT). Malformations of the OFT can lead to the occurrence of conotruncal defect (CTD). SOX7 duplication has been reported in patients with congenital CTD, but its specific role in OFT development remains poorly understood. To decipher this, histological analysis showed that SRY-related HMG-box 7 (SOX7) was regionally expressed in the endocardial endothelial cells and in the mesenchymal cells of the OFT, where EndMT occurs. Experiments, using in vitro collagen gel culture system, revealed that SOX7 was a negative regulator of EndMT that inhibited endocardial cell (EC) migration and resulted in decreased number of mesenchymal cells. Forced expression of SOX7 in endothelial cells blocked further migration and improved the expression of the adhesion protein vascular endothelial (VE)-cadherin (VE-cadherin). Moreover, a VE-cadherin knockdown could partly reverse the SOX7-mediated repression of cell migration. Luciferase and electrophoretic mobility shift assay (EMSA) demonstrated that SOX7 up-regulated VE-cadherin by directly binding to the gene's promoter in endothelial cells. The coding exons and splicing regions of the SOX7 gene were also scanned in the 536 sporadic CTD patients and in 300 unaffected controls, which revealed four heterozygous SOX7 mutations. Luciferase assays revealed that two SOX7 variants weakened the transactivation of the VE-cadherin promoter. In conclusion, SOX7 inhibited EndMT during OFT development by directly up-regulating the endothelial-specific adhesion molecule VE-cadherin. SOX7 mutations can lead to impaired EndMT by regulating VE-cadherin, which may give rise to the molecular mechanisms associated with SOX7 in CTD pathogenesis.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Endocardio/embriología , Cardiopatías Congénitas/embriología , Factores de Transcripción SOXF/metabolismo , Animales , Antígenos CD/genética , Cadherinas/genética , Movimiento Celular , Embrión de Mamíferos , Endocardio/citología , Endotelio/crecimiento & desarrollo , Transición Epitelial-Mesenquimal/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Ratas , Factores de Transcripción SOXF/genética
5.
Gene Expr Patterns ; 39: 119165, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33373688

RESUMEN

Endoglin (ENG) is essential for cardiovascular development and is expressed in the heart from its earliest developmental stages. ENG expression has been reported in the cardiac crescent, endocardium, valve mesenchyme and coronary vascular endothelial cells. However, its expression in these cell types is non-uniform and the dynamic changes in ENG expression during heart development have not been systematically studied. Using immunofluorescent staining we tracked ENG protein expression in mouse embryonic hearts aged from 11.5 to 17.5 days, and in postnatal and adult hearts. ENG is expressed in the endocardium and in venous endothelial cells throughout these developmental stages. ENG protein is down-regulated by approximately two-fold as a subset of early coronary veins reprogram to form arteries within the developing myocardium from E13.5. This two-fold higher ratio of ENG protein in veins versus arteries is maintained throughout cardiac development and in the adult heart. ENG is also down-regulated two-fold following mesenchymal transition of endocardial cells to form cardiac valve mesenchyme, whilst expression of the pan-endothelial marker CD31 is completely lost. A subset of epicardial cells (which do not express ENG protein) delaminate and undergo a similar mesenchymal transition to form epicardially derived cells (EPDCs). This transient intra-myocardial mesenchymal cell population expresses low levels of ENG protein, similar to valve mesenchyme. In conclusion, ENG shows dynamic changes of expression in vascular endothelial cells, endocardial cells and mesenchymal cells in the developing heart that vary according to cardiovascular cell type.


Asunto(s)
Endoglina/genética , Corazón/embriología , Miocitos Cardíacos/metabolismo , Animales , Vasos Coronarios/embriología , Vasos Coronarios/metabolismo , Endocardio/citología , Endocardio/embriología , Endocardio/metabolismo , Endoglina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL
6.
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
7.
Sci Rep ; 10(1): 20094, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33208886

RESUMEN

Protein kinase R-like endoplasmic reticulum kinase (PERK) is one of the endoplasmic reticulum (ER) stress sensors. PERK loss-of-function mutations are known to cause Wolcott-Rallison syndrome. This disease is characterized by early-onset diabetes mellitus, skeletal dysplasia, and cardiac valve malformation. To understand the role of PERK in valve formation in vivo, we used an endothelial-specific PERK conditional knockout mice as well as in vitro PERK inhibition assays. We used ProteoStat dyes to visualize the accumulation of misfolded proteins in the endocardial cushion and valve mesenchymal cells (VMCs). Then, VMCs were isolated from E12.5 fetal mice, by fluorescence assisted cell sorting. Proteomic analysis of PERK-deleted VMCs identified the suppression of proteins related to fatty acid oxidation (FAO), especially carnitine palmitoyltransferase II (CPT2). CPT2 is a critical regulator of endocardial-mesenchymal transformation (EndoMT); however how TGF-ß downstream signaling controls CPT2 expression remains unclear. Here, we showed that PERK inhibition suppressed, not only EndoMT but also CPT2 protein expression in human umbilical vein endothelial cells (HUVECs) under TGF-ß1 stimulation. As a result, PERK inhibition suppressed mitochondrial metabolic activity. Taken together, these results demonstrate that PERK signaling is required for cardiac valve formation via FAO and EndoMT.


Asunto(s)
Endocardio/embriología , Ácidos Grasos/química , Válvulas Cardíacas/embriología , Válvulas Cardíacas/metabolismo , Mesodermo/embriología , Organogénesis , eIF-2 Quinasa/fisiología , Animales , Endocardio/metabolismo , Ácidos Grasos/metabolismo , Femenino , Masculino , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción
8.
Dev Biol ; 465(1): 46-57, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32628938

RESUMEN

Endocardium is critically important for proper function of the cardiovascular system. Not only does endocardium connect the heart to blood vasculature, it also plays an important role in heart morphogenesis, valve formation, and ventricular trabeculation. The extracellular protein Fibronectin (Fn1) promotes endocardial differentiation, but the signaling pathways downstream of Fn1 that regulate endocardial development are not understood. Here, we analyzed the role of the Fibronectin receptors Integrin alpha5 (Itga5) and Integrin alpha4 (Itga4) in zebrafish heart development. We show that itga5 mRNA is expressed in both endocardium and myocardium during early stages of heart development. Through analysis of both itga5 single mutants and itga4;itga5 double mutants, we show that loss of both itga5 and itga4 results in enhanced defects in endocardial differentiation and morphogenesis compared to loss of itga5 alone. Loss of both itga5 and itga4 results in cardia bifida and severe myocardial morphology defects. Finally, we find that loss of itga5 and itga4 results in abnormally narrow anterior endodermal sheet morphology. Together, our results support a model in which Itga5 and Itga4 cooperate to promote endocardial differentiation, medial migration of endocardial and myocardial cells, and morphogenesis of anterior endoderm.


Asunto(s)
Diferenciación Celular , Endocardio/embriología , Integrina alfa4/metabolismo , Integrina alfa5/metabolismo , Modelos Biológicos , Organogénesis , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Integrina alfa4/genética , Integrina alfa5/genética , Mutación , Pez Cebra/genética , Proteínas de Pez Cebra/genética
9.
Anat Histol Embryol ; 49(5): 643-655, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32319704

RESUMEN

The initial intraembryonic vasculogenesis occurs in the cardiogenic mesoderm. Here, a cell population of proendocardial cells detaches from the mesoderm that subsequently generates the single endocardial tube by forming vascular plexuses. In the course of embryogenesis, the endocardium retains vasculogenic, angiogenic and haematopoietic potential. The coronary blood vessels that sustain the rapidly expanding myocardium develop in the course of the formation of the cardiac loop by vasculogenesis and angiogenesis from progenitor cells of the proepicardial serosa at the venous pole of the heart as well as from the endocardium and endothelial cells of the sinus venosus. Prospective coronary endothelial cells and progenitor cells of the coronary blood vessel walls (smooth muscle cells, perivascular cells) originate from different cell populations that are in close spatial as well as regulatory connection with each other. Vasculo- and angiogenesis of the coronary blood vessels are for a large part regulated by the epicardium and epicardium-derived cells. Vasculogenic and angiogenic signalling pathways include the vascular endothelial growth factors, the angiopoietins and the fibroblast growth factors and their receptors.


Asunto(s)
Vasos Sanguíneos/embriología , Vasos Sanguíneos/crecimiento & desarrollo , Corazón/embriología , Corazón/crecimiento & desarrollo , Animales , Vasos Coronarios/embriología , Vasos Coronarios/crecimiento & desarrollo , Endocardio/citología , Endocardio/embriología , Endocardio/crecimiento & desarrollo , Endotelio/citología , Humanos
10.
Artículo en Inglés | MEDLINE | ID: mdl-31988139

RESUMEN

Endocardial cells are specialized endothelial cells that, during embryogenesis, form a lining on the inside of the developing heart, which is maintained throughout life. Endocardial cells are an essential source for several lineages of the cardiovascular system including coronary endothelium, endocardial cushion mesenchyme, cardiomyocytes, mural cells, fibroblasts, liver vasculature, adipocytes, and hematopoietic cells. Alterations in the differentiation programs that give rise to these lineages has detrimental effects, including premature lethality or significant structural malformations present at birth. Here, we will review the literature pertaining to the contribution of endocardial cells to valvular, and nonvalvular lineages and highlight critical pathways required for these processes. The lineage differentiation potential of embryonic, and possibly adult, endocardial cells has therapeutic potential in the regeneration of damaged cardiac tissue or treatment of cardiovascular diseases.


Asunto(s)
Endocardio/embriología , Válvulas Cardíacas/embriología , Miocardio/citología , Animales , Desarrollo Embrionario , Cojinetes Endocárdicos/embriología , Válvulas Cardíacas/metabolismo , Humanos , Transducción de Señal
11.
Sci Rep ; 9(1): 11953, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31420575

RESUMEN

The endocardium is the endothelial component of the vertebrate heart and plays a key role in heart development. Where, when, and how the endocardium segregates during embryogenesis have remained largely unknown, however. We now show that Nkx2-5+ cardiac progenitor cells (CPCs) that express the Sry-type HMG box gene Sox17 from embryonic day (E) 7.5 to E8.5 specifically differentiate into the endocardium in mouse embryos. Although Sox17 is not essential or sufficient for endocardium fate, it can bias the fate of CPCs toward the endocardium. On the other hand, Sox17 expression in the endocardium is required for heart development. Deletion of Sox17 specifically in the mesoderm markedly impaired endocardium development with regard to cell proliferation and behavior. The proliferation of cardiomyocytes, ventricular trabeculation, and myocardium thickening were also impaired in a non-cell-autonomous manner in the Sox17 mutant, likely as a consequence of down-regulation of NOTCH signaling. An unknown signal, regulated by Sox17 and required for nurturing of the myocardium, is responsible for the reduction in NOTCH-related genes in the mutant embryos. Our results thus provide insight into differentiation of the endocardium and its role in heart development.


Asunto(s)
Diferenciación Celular , Embrión de Mamíferos/embriología , Endocardio/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/biosíntesis , Factores de Transcripción SOXF/biosíntesis , Transducción de Señal , Células Madre/metabolismo , Animales , Embrión de Mamíferos/citología , Endocardio/citología , Proteínas HMGB/genética , Mesodermo/citología , Mesodermo/embriología , Ratones , Ratones Transgénicos , Receptores Notch/genética , Receptores Notch/metabolismo , Factores de Transcripción SOXF/genética , Células Madre/citología
12.
Mol Cell Proteomics ; 18(9): 1782-1795, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31249105

RESUMEN

The endocardium is a specialized endothelium that lines the inner surface of the heart. Functional studies in mice and zebrafish have established that the endocardium is a source of instructive signals for the development of cardiac structures, including the heart valves and chambers. Here, we characterized the NOTCH-dependent endocardial secretome by manipulating NOTCH activity in mouse embryonic endocardial cells (MEEC) followed by mass spectrometry-based proteomics. We profiled different sets of soluble factors whose secretion not only responds to NOTCH activation but also shows differential ligand specificity, suggesting that ligand-specific inputs may regulate the expression of secreted proteins involved in different cardiac development processes. NOTCH signaling activation correlates with a transforming growth factor-ß2 (TGFß2)-rich secretome and the delivery of paracrine signals involved in focal adhesion and extracellular matrix (ECM) deposition and remodeling. In contrast, NOTCH inhibition is accompanied by the up-regulation of specific semaphorins that may modulate cell migration. The secretome protein expression data showed a good correlation with gene profiling of RNA expression in embryonic endocardial cells. Additional characterization by in situ hybridization in mouse embryos revealed expression of various NOTCH candidate effector genes (Tgfß2, Loxl2, Ptx3, Timp3, Fbln2, and Dcn) in heart valve endocardium and/or mesenchyme. Validating these results, mice with conditional Dll4 or Jag1 loss-of-function mutations showed gene expression alterations similar to those observed at the protein level in vitro These results provide the first description of the NOTCH-dependent endocardial secretome and validate MEEC as a tool for assaying the endocardial secretome response to a variety of stimuli and the potential use of this system for drug screening.


Asunto(s)
Endocardio/embriología , Endocardio/metabolismo , Válvulas Cardíacas/embriología , Receptores Notch/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Benzazepinas/farmacología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Células Cultivadas , Endocardio/citología , Endocardio/efectos de los fármacos , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Válvulas Cardíacas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Ratones Mutantes , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores Notch/genética , Reproducibilidad de los Resultados
13.
JCI Insight ; 52019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31112136

RESUMEN

The ang1-Tie2 pathway is required for normal vascular development, but its molecular effectors are not well-defined during cardiac ontogeny. Here we show that endocardial specific attenuation of Tie2 results in mid-gestation lethality due to heart defects associated with a hyperplastic but simplified trabecular meshwork (fewer but thicker trabeculae). Reduced proliferation and production of endocardial cells (ECs) following endocardial loss of Tie2 results in decreased endocardial sprouting required for trabecular assembly and extension. The hyperplastic trabeculae result from enhanced proliferation of trabecular cardiomyocyte (CMs), which is associated with upregulation of Bmp10, increased retinoic acid (RA) signaling, and Erk1/2 hyperphosphorylation in the myocardium. Intriguingly, myocardial phenotypes in Tie2-cko hearts could be partially rescued by inhibiting in utero RA signaling with pan-retinoic acid receptor antagonist BMS493. These findings reveal two complimentary functions of endocardial Tie2 during ventricular chamber formation: ensuring normal trabeculation by supporting EC proliferation and sprouting, and preventing hypertrabeculation via suppression of RA signaling in trabecular CMs.


Asunto(s)
Desarrollo Embrionario/fisiología , Cardiopatías Congénitas/metabolismo , Corazón/embriología , Corazón/crecimiento & desarrollo , Receptor TIE-2/metabolismo , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Desarrollo Embrionario/genética , Endocardio/embriología , Endocardio/crecimiento & desarrollo , Endocardio/metabolismo , Endocardio/patología , Femenino , Regulación del Desarrollo de la Expresión Génica , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Masculino , Ratones , Receptor TIE-2/genética , Transducción de Señal
14.
Curr Top Dev Biol ; 132: 395-416, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30797515

RESUMEN

Heart formation involves a complex series of tissue rearrangements, during which regions of the developing organ expand, bend, converge, and protrude in order to create the specific shapes of important cardiac components. Much of this morphogenesis takes place while cardiac function is underway, with blood flowing through the rapidly contracting chambers. Fluid forces are therefore likely to influence the regulation of cardiac morphogenesis, but it is not yet clear how these biomechanical cues direct specific cellular behaviors. In recent years, the optical accessibility and genetic amenability of zebrafish embryos have facilitated unique opportunities to integrate the analysis of flow parameters with the molecular and cellular dynamics underlying cardiogenesis. Consequently, we are making progress toward a comprehensive view of the biomechanical regulation of cardiac chamber emergence, atrioventricular canal differentiation, and ventricular trabeculation. In this review, we highlight a series of studies in zebrafish that have provided new insight into how cardiac function can shape cardiac morphology, with a particular focus on how hemodynamics can impact cardiac cell behavior. Over the long-term, this knowledge will undoubtedly guide our consideration of the potential causes of congenital heart disease.


Asunto(s)
Líquidos Corporales/fisiología , Corazón/embriología , Corazón/fisiología , Morfogénesis , Pez Cebra/embriología , Animales , Fenómenos Biomecánicos , Diferenciación Celular/genética , Cojinetes Endocárdicos/citología , Cojinetes Endocárdicos/embriología , Cojinetes Endocárdicos/metabolismo , Endocardio/citología , Endocardio/embriología , Endocardio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/anatomía & histología , Pez Cebra/genética
15.
J Cell Biol ; 218(3): 1039-1054, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30635353

RESUMEN

Elucidating the morphogenetic events that shape vertebrate heart valves, complex structures that prevent retrograde blood flow, is critical to understanding valvular development and aberrations. Here, we used the zebrafish atrioventricular (AV) valve to investigate these events in real time and at single-cell resolution. We report the initial events of collective migration of AV endocardial cells (ECs) into the extracellular matrix (ECM), and their subsequent rearrangements to form the leaflets. We functionally characterize integrin-based focal adhesions (FAs), critical mediators of cell-ECM interactions, during valve morphogenesis. Using transgenes to block FA signaling specifically in AV ECs as well as loss-of-function approaches, we show that FA signaling mediated by Integrin α5ß1 and Talin1 promotes AV EC migration and overall shaping of the valve leaflets. Altogether, our investigation reveals the critical processes driving cardiac valve morphogenesis in vivo and establishes the zebrafish AV valve as a vertebrate model to study FA-regulated tissue morphogenesis.


Asunto(s)
Endocardio/embriología , Adhesiones Focales/metabolismo , Válvulas Cardíacas/embriología , Organogénesis , Transducción de Señal , Pez Cebra/embriología , Animales , Movimiento Celular , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Adhesiones Focales/genética , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Talina/genética , Talina/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
16.
Sci Rep ; 8(1): 15369, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30337622

RESUMEN

SWI/SNF or BAF chromatin-remodeling complexes are polymorphic assemblies of homologous subunit families that remodel nucleosomes and facilitate tissue-specific gene regulation during development. BAF57/SMARCE1 is a BAF complex subunit encoded in animals by a single gene and is a component of all mammalian BAF complexes. In vivo, the loss of SMARCE1 would lead to the formation of deficient combinations of the complex which might present limited remodeling activities. To address the specific contribution of SMARCE1 to the function of the BAF complex, we generated CRISPR/Cas9 mutations of smarce1 in zebrafish. Smarce1 mutants showed visible defects at 72 hpf, including smaller eyes, abnormal body curvature and heart abnormalities. Gene expression analysis revealed that the mutant embryos displayed defects in endocardial development since early stages, which led to the formation of a misshapen heart tube. The severe morphological and functional cardiac problems observed at 4 dpf were correlated with the substantially increased expression of different cardiac transcription factors. Additionally, we showed that Smarce1 binds to cis-regulatory regions of the gata5 gene and is necessary for the recruitment of the BAF complex to these regions.


Asunto(s)
Proteínas Cromosómicas no Histona , Endocardio/embriología , Regulación del Desarrollo de la Expresión Génica , Mutación , Factores de Transcripción , Proteínas de Pez Cebra , Pez Cebra , Animales , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Embrión no Mamífero/embriología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
17.
Dis Model Mech ; 11(9)2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-30061196

RESUMEN

Deletions of chromosome 1p36 are associated with a high incidence of congenital heart defects (CHDs). The arginine-glutamic acid dipeptide repeats gene (RERE) is located in a critical region for CHD on chromosome 1p36 and encodes a cardiac-expressed nuclear receptor co-regulator. Mutations affecting RERE cause atrial and ventricular septal defects (VSDs) in humans, and RERE-deficient mice also develop VSDs. During cardiac development, mesenchymal cells destined to form part of the atrioventricular (AV) septum are generated when endocardial cells in the AV canal undergo epithelial-to-mesenchymal transition (EMT) and migrate into the space between the endocardium and the myocardium. These newly generated mesenchymal cells then proliferate to fill the developing AV endocardial cushions. Here, we demonstrate that RERE-deficient mouse embryos have reduced numbers of mesenchymal cells in their AV endocardial cushions owing to decreased levels of EMT and mesenchymal cell proliferation. In the endocardium, RERE colocalizes with GATA4, a transcription factor required for normal levels of EMT and mesenchymal cell proliferation. Using a combination of in vivo and in vitro studies, we show that Rere and Gata4 interact genetically in the development of CHDs, RERE positively regulates transcription from the Gata4 promoter and GATA4 levels are reduced in the AV canals of RERE-deficient embryos. Tissue-specific ablation of Rere in the endocardium leads to hypocellularity of the AV endocardial cushions, defective EMT and VSDs, but does not result in decreased GATA4 expression. We conclude that RERE functions in the AV canal to positively regulate the expression of GATA4, and that deficiency of RERE leads to the development of VSDs through its effects on EMT and mesenchymal cell proliferation. However, the cell-autonomous role of RERE in promoting EMT in the endocardium must be mediated by its effects on the expression of proteins other than GATA4.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Proteínas Portadoras/metabolismo , Factor de Transcripción GATA4/genética , Regulación del Desarrollo de la Expresión Génica , Defectos del Tabique Interventricular/embriología , Defectos del Tabique Interventricular/genética , Proteínas del Tejido Nervioso/deficiencia , Proteínas Represoras/deficiencia , Alelos , Animales , Proliferación Celular , Embrión de Mamíferos/metabolismo , Cojinetes Endocárdicos/embriología , Cojinetes Endocárdicos/metabolismo , Cojinetes Endocárdicos/patología , Endocardio/embriología , Endocardio/metabolismo , Endocardio/patología , Transición Epitelial-Mesenquimal/genética , Factor de Transcripción GATA4/metabolismo , Mesodermo/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , Proteínas Represoras/genética
18.
Development ; 145(13)2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29853617

RESUMEN

Signaling interactions between the myocardium and endocardium pattern embryonic cardiac regions, instructing their development to fulfill specific functions in the mature heart. We show that ectopic Bmp2 expression in the mouse chamber myocardium changes the transcriptional signature of adjacent chamber endocardial cells into valve tissue, and enables them to undergo epithelial-mesenchyme transition. This induction is independent of valve myocardium specification and requires high levels of Notch1 activity. Biochemical experiments suggest that Bmp2-mediated Notch1 induction is achieved through transcriptional activation of the Notch ligand Jag1, and physical interaction of Smad1/5 with the intracellular domain of the Notch1 receptor. Thus, widespread myocardial Bmp2 and endocardial Notch signaling drive presumptive ventricular endocardium to differentiate into valve endocardium. Understanding the molecular basis of valve development is instrumental to designing therapeutic strategies for congenital heart valve defects.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Embrión de Mamíferos/embriología , Endocardio/embriología , Válvulas Cardíacas/embriología , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Proteína Morfogenética Ósea 2/genética , Embrión de Mamíferos/citología , Endocardio/citología , Válvulas Cardíacas/citología , Ratones , Ratones Transgénicos , Miocardio/citología , Miocardio/metabolismo , Receptores Notch/genética , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo
19.
Cell Death Differ ; 25(6): 1118-1130, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29358667

RESUMEN

Activation of p53-dependent apoptosis is critical for tumor suppression but aberrant activation of p53 also leads to developmental defects and heart failure. Here, we found that Rbm24 RNA-binding protein, a target of p53, regulates p53 mRNA translation. Mechanistically, we found that through binding to p53 mRNA and interaction with translation initiation factor eIF4E, Rbm24 prevents eIF4E from binding to p53 mRNA and inhibits the assembly of translation initiation complex. Importantly, we showed that mice deficient in Rbm24 die in utero due to the endocardial cushion defect in the heart at least in part due to aberrant activation of p53-dependent apoptosis. We also showed that the heart developmental defect in Rbm24-null mice can be partially rescued by p53 deficiency through decreased apoptosis in the heart. Together, we postulate that the p53-Rbm24 loop is critical for the heart development and may be explored for mitigating congenital heart diseases and heart failure.


Asunto(s)
Endocardio/embriología , Regulación del Desarrollo de la Expresión Génica , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/genética , Humanos , Ratones , Ratones Noqueados , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Proteína p53 Supresora de Tumor/genética
20.
Elife ; 62017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28271994

RESUMEN

Novel regenerative therapies may stem from deeper understanding of the mechanisms governing cardiovascular lineage diversification. Using enhancer mapping and live imaging in avian embryos, and genetic lineage tracing in mice, we investigated the spatio-temporal dynamics of cardiovascular progenitor populations. We show that expression of the cardiac transcription factor Nkx2.5 marks a mesodermal population outside of the cardiac crescent in the extraembryonic and lateral plate mesoderm, with characteristics of hemogenic angioblasts. Extra-cardiac Nkx2.5 lineage progenitors migrate into the embryo and contribute to clusters of CD41+/CD45+ and RUNX1+ cells in the endocardium, the aorta-gonad-mesonephros region of the dorsal aorta and liver. We also demonstrated that ectopic expression of Nkx2.5 in chick embryos activates the hemoangiogenic gene expression program. Taken together, we identified a hemogenic angioblast cell lineage characterized by transient Nkx2.5 expression that contributes to hemogenic endothelium and endocardium, suggesting a novel role for Nkx2.5 in hemoangiogenic lineage specification and diversification.


Asunto(s)
Aorta/embriología , Endocardio/embriología , Hemangioblastos/fisiología , Proteína Homeótica Nkx-2.5/metabolismo , Animales , Embrión de Pollo , Ratones , Análisis Espacio-Temporal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...