Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Cell ; 151(5): 1083-96, 2012 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-23178125

RESUMO

The origins and developmental mechanisms of coronary arteries are incompletely understood. We show here by fate mapping, clonal analysis, and immunohistochemistry that endocardial cells generate the endothelium of coronary arteries. Dye tracking, live imaging, and tissue transplantation also revealed that ventricular endocardial cells are not terminally differentiated; instead, they are angiogenic and form coronary endothelial networks. Myocardial Vegf-a or endocardial Vegfr-2 deletion inhibited coronary angiogenesis and arterial formation by ventricular endocardial cells. In contrast, lineage and knockout studies showed that endocardial cells make a small contribution to the coronary veins, the formation of which is independent of myocardial-to-endocardial Vegf signaling. Thus, contrary to the current view of a common source for the coronary vessels, our findings indicate that the coronary arteries and veins have distinct origins and are formed by different mechanisms. This information may help develop better cell therapies for coronary artery disease.


Assuntos
Vasos Coronários/embriologia , Células Endoteliais/citologia , Miocárdio/citologia , Neovascularização Fisiológica , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Diferenciação Celular , Vasos Coronários/citologia , Vasos Coronários/metabolismo , Células Endoteliais/metabolismo , Camundongos , Miocárdio/metabolismo , Fatores de Transcrição NFATC/metabolismo
2.
Nature ; 557(7705): 439-445, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29743679

RESUMO

In vertebrate hearts, the ventricular trabecular myocardium develops as a sponge-like network of cardiomyocytes that is critical for contraction and conduction, ventricular septation, papillary muscle formation and wall thickening through the process of compaction 1 . Defective trabeculation leads to embryonic lethality2-4 or non-compaction cardiomyopathy (NCC) 5 . There are divergent views on when and how trabeculation is initiated in different species. In zebrafish, trabecular cardiomyocytes extrude from compact myocardium 6 , whereas in chicks, chamber wall thickening occurs before overt trabeculation 7 . In mice, the onset of trabeculation has not been described, but is proposed to begin at embryonic day 9.0, when cardiomyocytes form radially oriented ribs 2 . Endocardium-myocardium communication is essential for trabeculation, and numerous signalling pathways have been identified, including Notch2,8 and Neuregulin (NRG) 4 . Late disruption of the Notch pathway causes NCC 5 . Whereas it has been shown that mutations in the extracellular matrix (ECM) genes Has2 and Vcan prevent the formation of trabeculae in mice9,10 and the matrix metalloprotease ADAMTS1 promotes trabecular termination 3 , the pathways involved in ECM dynamics and the molecular regulation of trabeculation during its early phases remain unexplored. Here we present a model of trabeculation in mice that integrates dynamic endocardial and myocardial cell behaviours and ECM remodelling, and reveal new epistatic relationships between the involved signalling pathways. NOTCH1 signalling promotes ECM degradation during the formation of endocardial projections that are critical for individualization of trabecular units, whereas NRG1 promotes myocardial ECM synthesis, which is necessary for trabecular rearrangement and growth. These systems interconnect through NRG1 control of Vegfa, but act antagonistically to establish trabecular architecture. These insights enabled the prediction of persistent ECM and cardiomyocyte growth in a mouse NCC model, providing new insights into the pathophysiology of congenital heart disease.


Assuntos
Coração/embriologia , Miocárdio/citologia , Miocárdio/metabolismo , Neuregulina-1/metabolismo , Organogênese , Receptor Notch1/metabolismo , Animais , Modelos Animais de Doenças , Endocárdio/citologia , Endocárdio/metabolismo , Matriz Extracelular/metabolismo , Cardiopatias/congênito , Cardiopatias/metabolismo , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Neuregulina-1/genética , Receptor Notch1/genética , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
PLoS Genet ; 15(5): e1007711, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31120883

RESUMO

Dominant mutations of Gata4, an essential cardiogenic transcription factor (TF), were known to cause outflow tract (OFT) defects in both human and mouse, but the underlying molecular mechanism was not clear. In this study, Gata4 haploinsufficiency in mice was found to result in OFT defects including double outlet right ventricle (DORV) and ventricular septum defects (VSDs). Gata4 was shown to be required for Hedgehog (Hh)-receiving progenitors within the second heart field (SHF) for normal OFT alignment. Restored cell proliferation in the SHF by knocking-down Pten failed to rescue OFT defects, suggesting that additional cell events under Gata4 regulation is important. SHF Hh-receiving cells failed to migrate properly into the proximal OFT cushion, which is associated with abnormal EMT and cell proliferation in Gata4 haploinsufficiency. The genetic interaction of Hh signaling and Gata4 is further demonstrated to be important for OFT development. Gata4 and Smo double heterozygotes displayed more severe OFT abnormalities including persistent truncus arteriosus (PTA). Restoration of Hedgehog signaling renormalized SHF cell proliferation and migration, and rescued OFT defects in Gata4 haploinsufficiency. In addition, there was enhanced Gata6 expression in the SHF of the Gata4 heterozygotes. The Gata4-responsive repressive sites were identified within 1kbp upstream of the transcription start site of Gata6 by both ChIP-qPCR and luciferase reporter assay. These results suggested a SHF regulatory network comprising of Gata4, Gata6 and Hh-signaling for OFT development.


Assuntos
Fator de Transcrição GATA4/genética , Fator de Transcrição GATA6/genética , Proteínas Hedgehog/genética , Receptor Smoothened/genética , Obstrução do Fluxo Ventricular Externo/genética , Septo Interventricular/metabolismo , Animais , Movimento Celular , Proliferação de Células , Embrião de Mamíferos , Fator de Transcrição GATA4/metabolismo , Fator de Transcrição GATA6/metabolismo , Regulação da Expressão Gênica , Haploinsuficiência , Proteínas Hedgehog/metabolismo , Heterozigoto , Humanos , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Transdução de Sinais , Receptor Smoothened/metabolismo , Tronco Arterial/anormalidades , Tronco Arterial/metabolismo , Obstrução do Fluxo Ventricular Externo/metabolismo , Obstrução do Fluxo Ventricular Externo/patologia , Septo Interventricular/patologia
4.
Arterioscler Thromb Vasc Biol ; 40(3): 638-655, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31893948

RESUMO

OBJECTIVE: Although often studied independently, little is known about how aortic valve endothelial cells and valve interstitial cells interact collaborate to maintain tissue homeostasis or drive valve calcific pathogenesis. Inflammatory signaling is a recognized initiator of valve calcification, but the cell-type-specific downstream mechanisms have not been elucidated. In this study, we test how inflammatory signaling via NFκB (nuclear factor κ-light-chain enhancer of activated B cells) activity coordinates unique and shared mechanisms of valve endothelial cells and valve interstitial cells differentiation during calcific progression. Approach and Results: Activated NFκB was present throughout the calcific aortic valve disease (CAVD) process in both endothelial and interstitial cell populations in an established mouse model of hypercholesterolemia-induced CAVD and in human CAVD. NFκB activity induces endothelial to mesenchymal transformation in 3-dimensional cultured aortic valve endothelial cells and subsequent osteogenic calcification of transformed cells. Similarly, 3-dimensional cultured valve interstitial cells calcified via NFκB-mediated osteogenic differentiation. NFκB-mediated endothelial to mesenchymal transformation was directly demonstrated in vivo during CAVD via genetic lineage tracking. Genetic deletion of NFκB in either whole valves or valve endothelium only was sufficient to prevent valve-specific molecular and cellular mechanisms of CAVD in vivo despite the persistence of a CAVD inducing environment. CONCLUSIONS: Our results identify NFκB signaling as an essential molecular regulator for both valve endothelial and interstitial participation in CAVD pathogenesis. Direct demonstration of valve endothelial cell endothelial to mesenchymal transformation transmigration in vivo during CAVD highlights a new cellular population for further investigation in CAVD morbidity. The efficacy of valve-specific NFκB modulation in inhibiting hypercholesterolemic CAVD suggests potential benefits of multicell type integrated investigation for biological therapeutic development and evaluation for CAVD.


Assuntos
Valva Aórtica/metabolismo , Calcinose/metabolismo , Diferenciação Celular , Células Endoteliais/metabolismo , Doenças das Valvas Cardíacas/metabolismo , NF-kappa B/metabolismo , Osteogênese , Animais , Valva Aórtica/patologia , Calcinose/etiologia , Calcinose/patologia , Células Cultivadas , Microambiente Celular , Modelos Animais de Doenças , Células Endoteliais/patologia , Doenças das Valvas Cardíacas/etiologia , Doenças das Valvas Cardíacas/genética , Doenças das Valvas Cardíacas/patologia , Humanos , Hipercolesterolemia/complicações , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/genética , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transdução de Sinais , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
5.
Annu Rev Physiol ; 79: 21-41, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-27959615

RESUMO

Normal aortic valves are composed of valve endothelial cells (VECs) and valve interstitial cells (VICs). VICs are the major cell population and have distinct embryonic origins in the endocardium and cardiac neural crest cells. Cell signaling between the VECs and VICs plays critical roles in aortic valve morphogenesis. Disruption of major cell signaling pathways results in aortic valve malformations, including bicuspid aortic valve (BAV). BAV is a common congenital heart valve disease that may lead to calcific aortic valve disease (CAVD), but there is currently no effective medical treatment for this beyond surgical replacement. Mouse and human studies have identified causative gene mutations for BAV and CAVD via disrupted VEC to VIC signaling. Future studies on the developmental signaling mechanisms underlying aortic valve malformations and the pathogenesis of CAVD using genetically modified mouse models and patient-induced pluripotent stem cells may identify new effective therapeutic targets for the disease.


Assuntos
Valva Aórtica/patologia , Doenças das Valvas Cardíacas/patologia , Animais , Células Endoteliais/patologia , Doenças das Valvas Cardíacas/genética , Humanos , Transdução de Sinais/genética
7.
Nucleic Acids Res ; 45(6): 3102-3115, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-27956497

RESUMO

The dynamic interaction of DNA methylation and transcription factor binding in regulating spatiotemporal gene expression is essential for embryogenesis, but the underlying mechanisms remain understudied. In this study, using mouse models and integration of in vitro and in vivo genetic and epigenetic analyses, we show that the binding of REST (repressor element 1 (RE1) silencing transcription factor; also known as NRSF) to its cognate RE1 sequences is temporally regulated by non-CpG methylation. This process is dependent on DNA methyltransferase 3B (DNMT3B) and leads to suppression of adult cardiac genes in developing hearts. We demonstrate that DNMT3B preferentially mediates non-CpG methylation of REST-targeted genes in the developing heart. Downregulation of DNMT3B results in decreased non-CpG methylation of RE1 sequences, reduced REST occupancy, and consequently release of the transcription suppression during later cardiac development. Together, these findings reveal a critical gene silencing mechanism in developing mammalian hearts that is regulated by the dynamic interaction of DNMT3B-mediated non-CpG methylation and REST binding.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Coração/embriologia , Miocárdio/metabolismo , Proteínas Repressoras/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/fisiologia , Metilação de DNA , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/biossíntese , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Camundongos , Camundongos Endogâmicos C57BL , Canais de Potássio/biossíntese , Canais de Potássio/genética , Ligação Proteica , Proteínas Repressoras/fisiologia , DNA Metiltransferase 3B
8.
J Mol Cell Cardiol ; 125: 98-105, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30347193

RESUMO

NOTCH and WNT signaling pathways play critical roles in cardiac chamber formation. Here we explored the potential interactions between the two pathways in this developmental process by using genetically modified mouse models and whole embryo culture systems. By deletion of Notch1 to inactivate NOTCH1 signaling in the endocardium in vivo and ex vivo rescue experiments, we showed that myocardial WNT5A mediated endocardial NOTCH1 signaling to maintain the gene regulatory network essential for cardiac chamber formation. Furthermore, genetic deletion of ß-catenin in the myocardium and inhibition of the WNT/Ca2+ signaling by FK506 resulted in a similar disruption of the gene regulatory network as inactivation of endocardial NOTCH1 signaling. Together, these findings identify WNT5A as a key myocardial factor that mediates the endocardial NOTCH signaling to maintain the gene regulatory network essential for cardiac chamber formation through WNT/ß-catenin and WNT/Ca2+ signaling pathways.


Assuntos
Receptor Notch1/metabolismo , Receptores Notch/metabolismo , Proteína Wnt-5a/metabolismo , beta Catenina/metabolismo , Animais , Endocárdio/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hibridização In Situ , Masculino , Camundongos , Miocárdio/metabolismo , Receptor Notch1/genética , Receptores Notch/genética , Proteína Wnt-5a/genética , beta Catenina/genética
9.
J Mol Cell Cardiol ; 123: 150-158, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30201295

RESUMO

Abnormal endocardial cushion formation is a major cause of congenital heart valve disease, which is a common birth defect with significant morbidity and mortality. Although ß-catenin and BMP2 are two well-known regulators of endocardial cushion formation, their interaction in this process is largely unknown. Here, we report that deletion of ß-catenin in myocardium results in formation of hypoplastic endocardial cushions accompanying a decrease of mesenchymal cell proliferation. Loss of ß-catenin reduced Bmp2 expression in myocardium and SMAD signaling in cushion mesenchyme. Exogenous BMP2 recombinant proteins fully rescued the proliferation defect of mesenchymal cells in cultured heart explants from myocardial ß-catenin knockout embryos. Using a canonical WNT signaling reporter mouse line, we showed that cushion myocardium exhibited high WNT/ß-catenin activities during endocardial cushion growth. Selective disruption of the signaling function of ß-catenin resulted in a cushion growth defect similar to that caused by the complete loss of ß-catenin. Together, these observations demonstrate that myocardial ß-catenin signaling function promotes mesenchymal cell proliferation and endocardial cushion expansion through inducing BMP signaling.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Coxins Endocárdicos/metabolismo , Miocárdio/metabolismo , Organogênese , Transdução de Sinais , beta Catenina/metabolismo , Animais , Proliferação de Células , Coxins Endocárdicos/embriologia , Endocárdio/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Modelos Biológicos , Comunicação Parácrina , Ratos , Via de Sinalização Wnt
10.
Dev Biol ; 430(1): 113-128, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28790014

RESUMO

Distal outgrowth, maturation and remodeling of the endocardial cushion mesenchyme in the atrioventricular (AV) canal are the essential morphogenetic events during four-chambered heart formation. Mesenchymalized AV endocardial cushions give rise to the AV valves and the membranous ventricular septum (VS). Failure of these processes results in several human congenital heart defects. Despite this clinical relevance, the mechanisms governing how mesenchymalized AV endocardial cushions mature and remodel into the membranous VS and AV valves have only begun to be elucidated. The role of BMP signaling in the myocardial and secondary heart forming lineage has been well studied; however, little is known about the role of BMP2 expression in the endocardial lineage. To fill this knowledge gap, we generated Bmp2 endocardial lineage-specific conditional knockouts (referred to as Bmp2 cKOEndo) by crossing conditionally-targeted Bmp2flox/flox mice with a Cre-driver line, Nfatc1Cre, wherein Cre-mediated recombination was restricted to the endocardial cells and their mesenchymal progeny. Bmp2 cKOEndo mouse embryos did not exhibit failure or delay in the initial AV endocardial cushion formation at embryonic day (ED) 9.5-11.5; however, significant reductions in AV cushion size were detected in Bmp2 cKOEndo mouse embryos when compared to control embryos at ED13.5 and ED16.5. Moreover, deletion of Bmp2 from the endocardial lineage consistently resulted in membranous ventricular septal defects (VSDs), and mitral valve deficiencies, as evidenced by the absence of stratification of mitral valves at birth. Muscular VSDs were not found in Bmp2 cKOEndo mouse hearts. To understand the underlying morphogenetic mechanisms leading to a decrease in cushion size, cell proliferation and cell death were examined for AV endocardial cushions. Phospho-histone H3 analyses for cell proliferation and TUNEL assays for apoptotic cell death did not reveal significant differences between control and Bmp2 cKOEndo in AV endocardial cushions. However, mRNA expression of the extracellular matrix components, versican, Has2, collagen 9a1, and periostin was significantly reduced in Bmp2 cKOEndo AV cushions. Expression of transcription factors implicated in the cardiac valvulogenesis, Snail2, Twist1 and Sox9, was also significantly reduced in Bmp2 cKOEndo AV cushions. These data provide evidence that BMP2 expression in the endocardial lineage is essential for the distal outgrowth, maturation and remodeling of AV endocardial cushions into the normal membranous VS and the stratified AV valves.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Linhagem da Célula , Coxins Endocárdicos/citologia , Coxins Endocárdicos/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Proteína Morfogenética Óssea 2/genética , Moléculas de Adesão Celular/metabolismo , Morte Celular , Proliferação de Células , Colágeno/metabolismo , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Coxins Endocárdicos/metabolismo , Deleção de Genes , Comunicação Interventricular/metabolismo , Comunicação Interventricular/patologia , Imageamento Tridimensional , Imuno-Histoquímica , Mesoderma/citologia , Camundongos Knockout , Valva Mitral/patologia , Fatores de Transcrição NFATC/metabolismo , Proteoglicanas/metabolismo , Fatores de Transcrição/metabolismo , Transformação Genética
11.
Proc Natl Acad Sci U S A ; 112(16): 5141-6, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25848038

RESUMO

We have generated low-expressing and high-expressing endothelin-1 genes (L and H) and have bred mice with four levels of expression: L/L, ∼20%; L/+, ∼65%; +/+ (wild type), 100%; and H/+, ∼350%. The hypomorphic L allele can be spatiotemporally switched to the hypermorphic H allele by Cre-loxP recombination. Young adult L/L and L/+ mice have dilated cardiomyopathy, hypertension, and increased plasma volumes, together with increased ventricular superoxide levels, increased matrix metalloproteinase 9 (Mmp9) expression, and reduced ventricular stiffness. H/+ mice have decreased plasma volumes and significantly heavy stiff hearts. Global or cardiomyocyte-specific switching expression from L to H normalized the abnormalities already present in young adult L/L mice. An epithelial sodium channel antagonist normalized plasma volume and blood pressure, but only partially corrected the cardiomyopathy. A superoxide dismutase mimetic made superoxide levels subnormal, reduced Mmp9 overexpression, and substantially improved cardiac function. Genetic absence of Mmp9 also improved cardiac function, but increased superoxide remained. We conclude that endothelin-1 is critical for maintaining normal contractile function, for controlling superoxide and Mmp9 levels, and for ensuring that the myocardium has sufficient collagen to prevent overstretching. Even a modest (∼35%) decrease in endothelin-1 gene (Edn1) expression is sufficient to cause cardiac dysfunction.


Assuntos
Endotelina-1/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Miocárdio/metabolismo , Superóxidos/metabolismo , Envelhecimento/patologia , Animais , Animais Recém-Nascidos , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Colágeno/metabolismo , Masculino , Camundongos , Miocárdio/enzimologia , Miocárdio/patologia , Especificidade de Órgãos , Fenótipo , Superóxido Dismutase/metabolismo , Análise de Sobrevida
12.
Eur Heart J ; 38(9): 675-686, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26491108

RESUMO

AIMS: Congenital anomalies of arterial valves are common birth defects, leading to valvar stenosis. With no pharmaceutical treatment that can prevent the disease progression, prosthetic replacement is the only choice of treatment, incurring considerable morbidity and mortality. Animal models presenting localized anomalies and stenosis of congenital arterial valves similar to that of humans are critically needed research tools to uncover developmental molecular mechanisms underlying this devastating human condition. METHODS AND RESULTS: We generated and characterized mouse models with conditionally altered Notch signalling in endothelial or interstitial cells of developing valves. Mice with inactivation of Notch1 signalling in valvar endothelial cells (VEC) developed congenital anomalies of arterial valves including bicuspid aortic valves and valvar stenosis. Notch1 signalling in VEC was required for repressing proliferation and activating apoptosis of valvar interstitial cells (VIC) after endocardial-to-mesenchymal transformation (EMT). We showed that Notch signalling regulated Tnfα expression in vivo, and Tnf signalling was necessary for apoptosis of VIC and post-EMT development of arterial valves. Furthermore, activation or inhibition of Notch signalling in cultured pig aortic VEC-promoted or suppressed apoptosis of VIC, respectively. CONCLUSION: We have now met the need of critical animal models and shown that Notch-Tnf signalling balances proliferation and apoptosis for post-EMT development of arterial valves. Our results suggest that mutations in its components may lead to congenital anomaly of aortic valves and valvar stenosis in humans.


Assuntos
Estenose da Valva Aórtica/etiologia , Receptor Notch1/metabolismo , Animais , Valva Aórtica/anormalidades , Estenose da Valva Aórtica/embriologia , Estenose da Valva Aórtica/fisiopatologia , Apoptose/fisiologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Homeostase/fisiologia , Células-Tronco Mesenquimais/fisiologia , Camundongos Knockout , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
13.
J Mol Cell Cardiol ; 97: 278-85, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27266388

RESUMO

Definitively identifying the cell type of newly generated cells in the heart and defining their origins are central questions in cardiac regenerative medicine. Currently, it is challenging to ascertain the myocardial identity and to track myocardial progeny during heart development and disease due to lack of proper genetic tools. This may lead to many misinterpretations of the findings in cardiac regenerative biology. In this study, we developed a set of novel mouse models by inserting double reporter genes nlacZ/H2B-GFP, mGFP/H2B-mCherry into the start codon of Tnnt2 and Myh6. nlacZ (nuclear lacZ) and mGFP (membrane GFP) are flanked by two LoxP sites in these animals. We found that the reporter genes faithfully recapitulated Tnnt2 and Myh6 cardiac expression from embryonic stage and adulthood. The reporter mice provide unprecedented robustness and fidelity for visualizing and tracing cardiomyocytes with nuclear or cell membrane localization signals. These animal models offer superior genetic tools to meet a critical need in studies of heart development, cardiac stem cell biology and cardiac regenerative medicine.


Assuntos
Marcadores Genéticos , Miócitos Cardíacos/metabolismo , Fenótipo , Animais , Linhagem da Célula/genética , Feminino , Imunofluorescência , Expressão Gênica , Genes Reporter , Masculino , Camundongos , Camundongos Transgênicos , Miocárdio , Cadeias Pesadas de Miosina/genética , Especificidade de Órgãos/genética , Proteínas Recombinantes de Fusão , Regeneração , Medicina Regenerativa , Troponina T/genética
14.
Development ; 140(15): 3176-87, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23824573

RESUMO

Cardiac valves are essential to direct forward blood flow through the cardiac chambers efficiently. Congenital valvular defects are prevalent among newborns and can cause an immediate threat to survival as well as long-term morbidity. Valve leaflet formation is a rigorously programmed process consisting of endocardial epithelial-mesenchymal transformation (EMT), mesenchymal cell proliferation, valve elongation and remodeling. Currently, little is known about the coordination of the diverse signals that regulate endocardial cushion development and valve elongation. Here, we report that the T-box transcription factor Tbx20 is expressed in the developing endocardial cushions and valves throughout heart development. Ablation of Tbx20 in endocardial cells causes severe valve elongation defects and impaired cardiac function in mice. Our study reveals that endocardial Tbx20 is crucial for valve endocardial cell proliferation and extracellular matrix development, but is not required for initiation of EMT. Elimination of Tbx20 also causes aberrant Wnt/ß-catenin signaling in the endocardial cushions. In addition, Tbx20 regulates Lef1, a key transcriptional mediator for Wnt/ß-catenin signaling, in this developmental process. Our study suggests a model in which Tbx20 regulates the Wnt pathway to direct endocardial cushion maturation and valve elongation, and provides new insights into the etiology of valve defects in humans.


Assuntos
Coxins Endocárdicos/embriologia , Coxins Endocárdicos/metabolismo , Valvas Cardíacas/embriologia , Valvas Cardíacas/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Proliferação de Células , Transição Epitelial-Mesenquimal , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Fator 1 de Ligação ao Facilitador Linfoide/genética , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Organogênese , Gravidez , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética , Via de Sinalização Wnt , beta Catenina/metabolismo
15.
Circ Res ; 109(2): 183-92, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21597012

RESUMO

RATIONALE: Formation of heart valves requires early endocardial to mesenchymal transformation (EMT) to generate valve mesenchyme and subsequent endocardial cell proliferation to elongate valve leaflets. Nfatc1 (nuclear factor of activated T cells, cytoplasmic 1) is highly expressed in valve endocardial cells and is required for normal valve formation, but its role in the fate of valve endocardial cells during valve development is unknown. OBJECTIVE: Our aim was to investigate the function of Nfatc1 in cell-fate decision making by valve endocardial cells during EMT and early valve elongation. METHODS AND RESULTS: Nfatc1 transcription enhancer was used to generate a novel valve endocardial cell-specific Cre mouse line for fate-mapping analyses of valve endocardial cells. The results demonstrate that a subpopulation of valve endocardial cells marked by the Nfatc1 enhancer do not undergo EMT. Instead, these cells remain within the endocardium as a proliferative population to support valve leaflet extension. In contrast, loss of Nfatc1 function leads to enhanced EMT and decreased proliferation of valve endocardium and mesenchyme. The results of blastocyst complementation assays show that Nfatc1 inhibits EMT in a cell-autonomous manner. We further reveal by gene expression studies that Nfatc1 suppresses transcription of Snail1 and Snail2, the key transcriptional factors for initiation of EMT. CONCLUSIONS: These results show that Nfatc1 regulates the cell-fate decision making of valve endocardial cells during valve development and coordinates EMT and valve elongation by allocating endocardial cells to the 2 morphological events essential for valve development.


Assuntos
Linhagem da Célula , Endocárdio/embriologia , Valvas Cardíacas/embriologia , Fatores de Transcrição NFATC/fisiologia , Animais , Endocárdio/citologia , Transição Epitelial-Mesenquimal , Regulação da Expressão Gênica no Desenvolvimento , Valvas Cardíacas/citologia , Valvas Cardíacas/crescimento & desenvolvimento , Camundongos , Morfogênese , Organogênese , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Transcrição Gênica
16.
STAR Protoc ; 4(4): 102619, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37897735

RESUMO

Developing an ex vivo system that mimics in vivo developmental coronary angiogenesis provides an improved understanding of its underlying molecular and cellular mechanisms. Here, we present a sandwiched embryonic ventricular explant assay to model mouse coronary angiogenesis ex vivo. We describe steps for breeding mice, labeling endocardial cells, isolating murine hearts, dissecting left ventricles, and making sandwiched explants in Matrigel. We then detail procedures for modeling coronary angiogenesis and taking images. For complete details on the use and execution of this protocol, please refer to Lu et al. (2023)1.


Assuntos
Angiogênese , Ventrículos do Coração , Animais , Camundongos , Coração , Bioensaio , Células Endoteliais
17.
Eur J Pharmacol ; 951: 175783, 2023 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-37172927

RESUMO

BACKGROUD: Congenital bicuspid aortic valve (cBAV) develops calcification and stenotic obstruction early compared with degenerative tricuspid aortic valve (dTAV), which requires surgical intervention. Here we report a comparative study of patients with cBAV or dTAV to identify risk factors associated with the rapid development of calcified bicuspid valves. METHODS: A total of 69 aortic valves (24 dTAV and 45 cBAV) were collected at the time of surgical aortic valve replacement for comparative clinical characteristics. Ten samples were randomly selected from each group for histology, pathology, and inflammatory factors expression and comparison analyses. OM-induced calcification in porcine aortic valve interstitial cell cultures were prepared for illustrating the underlying molecular mechanisms about calcification progress of cBAV and dTAV. RESULTS: We found that cBAV patients have increased cases of aortic valve stenosis compared with dTAV patients. Histopathological examinations revealed increased collagens deposition, neovascularization and infiltrations by inflammatory cells, especially T-lymphocytes and macrophages. We identified that tumor necrosis factor α (TNFα) and its regulated inflammatory cytokines are upregulated in cBAV. Further in vitro study indicated that TNFα-NFκB and TNFα-GSK3ß pathway accelerate aortic valve interstitial cells calcification, while inhibition of TNFα significantly delays this process. CONCLUSION: The finding of intensified TNFα-mediated inflammation in the pathological cBAV advocates the inhibition of TNFα as a potential treatment for patients with cBAV by alleviating the progress of inflammation-induced valve damage and calcification.


Assuntos
Estenose da Valva Aórtica , Doença da Válvula Aórtica Bicúspide , Animais , Suínos , Fator de Necrose Tumoral alfa/metabolismo , Doença da Válvula Aórtica Bicúspide/complicações , Doença da Válvula Aórtica Bicúspide/metabolismo , Doença da Válvula Aórtica Bicúspide/patologia , Estenose da Valva Aórtica/etiologia , Estenose da Valva Aórtica/patologia , Estenose da Valva Aórtica/cirurgia , Valva Aórtica/anormalidades , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Inflamação/metabolismo
18.
Life Sci Alliance ; 6(9)2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37385754

RESUMO

Endocardial cushion formation is essential for heart valve development and heart chamber separation. Abnormal endocardial cushion formation often causes congenital heart defects. ß-Catenin is known to be essential for endocardial cushion formation; however, the underlying cellular and molecular mechanisms remain incompletely understood. Here, we show that endothelial-specific deletion of ß-catenin in mice resulted in formation of hypoplastic endocardial cushions due to reduced cell proliferation and impaired cell migration. By using a ß-catenin DM allele in which the transcriptional function of ß-catenin is selectively disrupted, we further reveal that ß-catenin regulated cell proliferation and migration through its transcriptional and non-transcriptional function, respectively. At the molecular level, loss of ß-catenin resulted in increased expression of cell cycle inhibitor p21 in cushion endocardial and mesenchymal cells in vivo. In vitro rescue experiments with HUVECs and pig aortic valve interstitial cells confirmed that ß-catenin promoted cell proliferation by suppressing p21. In addition, one savvy negative observation is that ß-catenin was dispensable for endocardial-to-mesenchymal fate change. Taken together, our findings demonstrate that ß-catenin is essential for cell proliferation and migration but dispensable for endocardial cells to gain mesenchymal fate during endocardial cushion formation. Mechanistically, ß-catenin promotes cell proliferation by suppressing p21. These findings inform the potential role of ß-catenin in the etiology of congenital heart defects.


Assuntos
Coxins Endocárdicos , beta Catenina , Animais , Camundongos , Suínos , beta Catenina/genética , Proliferação de Células/genética , Alelos , Ciclo Celular
19.
Dev Cell ; 58(9): 791-805.e4, 2023 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-37023750

RESUMO

Coronary heart disease damages the trabecular myocardium, and the regeneration of trabecular vessels may alleviate ischemic injury. However, the origins and developmental mechanisms of trabecular vessels remain unknown. Here, we show that murine ventricular endocardial cells generate trabecular vessels through an "angioEMT" mechanism. Time course fate mapping defined a specific wave of trabecular vascularization by ventricular endocardial cells. Single-cell transcriptomics and immunofluorescence identified a subpopulation of ventricular endocardial cells that underwent endocardial-mesenchymal transition (EMT) before these cells generated trabecular vessels. Ex vivo pharmacological activation and in vivo genetic inactivation experiments identified an EMT signal in ventricular endocardial cells involving SNAI2-TGFB2/TGFBR3, which was a prerequisite for later trabecular-vessel formation. Additional loss- and gain-of-function genetic studies showed that VEGFA-NOTCH1 signaling regulated post-EMT trabecular angiogenesis by ventricular endocardial cells. Our finding that trabecular vessels originate from ventricular endocardial cells through a two-step angioEMT mechanism could inform better regeneration medicine for coronary heart disease.


Assuntos
Endocárdio , Coração , Animais , Camundongos , Ventrículos do Coração , Miocárdio , Células Endoteliais
20.
J Am Heart Assoc ; 12(18): e029683, 2023 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-37702066

RESUMO

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.


Assuntos
Endocárdio , Valvas Cardíacas , Apoptose , Catéteres , Citosol , Endocárdio/embriologia , Transdução de Sinais , Animais , Camundongos , Valvas Cardíacas/embriologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa