Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 16(10): e0259426, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34714866

RESUMO

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.


Assuntos
Comunicação Atrioventricular/genética , Coxins Endocárdicos/metabolismo , Receptor ErbB-3/metabolismo , Animais , Sítios de Ligação , Células CHO , Cricetinae , Cricetulus , Comunicação Atrioventricular/metabolismo , Coxins Endocárdicos/embriologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-3/química , Receptor ErbB-3/genética , Transdução de Sinais
2.
Cardiovasc Res ; 104(1): 49-60, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25100766

RESUMO

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.


Assuntos
Circulação Coronária , Comunicação Atrioventricular/embriologia , Coxins Endocárdicos/embriologia , Valvas Cardíacas/anormalidades , Hemodinâmica , Mecanotransdução Celular , Animais , Animais Geneticamente Modificados , Função Atrial , Comunicação Atrioventricular/genética , Comunicação Atrioventricular/metabolismo , Comunicação Atrioventricular/fisiopatologia , Coxins Endocárdicos/metabolismo , Coxins Endocárdicos/fisiopatologia , Genótipo , Valvas Cardíacas/metabolismo , Valvas Cardíacas/fisiopatologia , Morfogênese , Mutação , Contração Miocárdica , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Fenótipo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
3.
Cardiovasc Res ; 99(3): 452-60, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23723064

RESUMO

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.


Assuntos
Valva Aórtica/embriologia , Coxins Endocárdicos/citologia , Crista Neural/citologia , Animais , Valva Aórtica/anormalidades , Valva Aórtica/citologia , Valva Aórtica/metabolismo , Doença da Válvula Aórtica Bicúspide , Padronização Corporal , Adesão Celular , Comunicação Celular , Células Cultivadas , Anomalias dos Vasos Coronários/embriologia , Anomalias dos Vasos Coronários/metabolismo , Vasos Coronários/embriologia , Vasos Coronários/metabolismo , Modelos Animais de Doenças , Comunicação Atrioventricular/embriologia , Comunicação Atrioventricular/metabolismo , Coxins Endocárdicos/embriologia , Coxins Endocárdicos/metabolismo , Doenças das Valvas Cardíacas/embriologia , Doenças das Valvas Cardíacas/etiologia , Doenças das Valvas Cardíacas/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Modelos Cardiovasculares , Crista Neural/anormalidades , Crista Neural/metabolismo , Transdução de Sinais , Quinases Associadas a rho/deficiência , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
4.
Development ; 139(2): 385-95, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22186728

RESUMO

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.


Assuntos
Comunicação Atrioventricular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Troca Materno-Fetal/fisiologia , Miocárdio/metabolismo , Transdução de Sinais/fisiologia , Proteínas com Domínio T/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Tretinoína/metabolismo , Animais , Imunoprecipitação da Cromatina , Primers do DNA/genética , Comunicação Atrioventricular/etiologia , Feminino , Galactosídeos , Imuno-Histoquímica , Hibridização In Situ , Indóis , Luciferases , Camundongos , Análise em Microsséries , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tretinoína/fisiologia
5.
Dev Biol ; 358(2): 368-78, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21839733

RESUMO

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.


Assuntos
Coração Fetal/embriologia , Coração Fetal/metabolismo , Fator de Transcrição GATA4/metabolismo , Fator de Transcrição GATA5/metabolismo , Fator de Transcrição GATA6/metabolismo , Animais , Estenose da Valva Aórtica/embriologia , Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/metabolismo , Comunicação Atrioventricular/embriologia , Comunicação Atrioventricular/genética , Comunicação Atrioventricular/metabolismo , Feminino , Fator de Transcrição GATA4/deficiência , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA5/deficiência , Fator de Transcrição GATA5/genética , Fator de Transcrição GATA6/deficiência , Fator de Transcrição GATA6/genética , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Comunicação Interventricular/embriologia , Comunicação Interventricular/genética , Comunicação Interventricular/metabolismo , Heterozigoto , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
6.
Circ Res ; 107(10): 1209-19, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20847311

RESUMO

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.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular , Comunicação Atrioventricular/metabolismo , Coxins Endocárdicos/metabolismo , Valvas Cardíacas/metabolismo , Coração/embriologia , Miócitos de Músculo Liso/metabolismo , Crista Neural/metabolismo , Transdução de Sinais , Animais , Sítios de Ligação , Proteína Morfogenética Óssea 4/genética , Diferenciação Celular/genética , Linhagem da Célula , Células Cultivadas , Comunicação Atrioventricular/genética , Comunicação Atrioventricular/patologia , Coxins Endocárdicos/patologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Genótipo , Valvas Cardíacas/anormalidades , Valvas Cardíacas/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Morfogênese , Miócitos de Músculo Liso/patologia , Crista Neural/anormalidades , Crista Neural/patologia , Técnicas de Cultura de Órgãos , Fenótipo , Regiões Promotoras Genéticas , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/genética , Fator de Transcrição AP-1/metabolismo , Transfecção
7.
Dev Dyn ; 238(12): 3103-10, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19890913

RESUMO

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.


Assuntos
Sulfatos de Condroitina/metabolismo , Coxins Endocárdicos/embriologia , Coração/embriologia , Animais , Animais Geneticamente Modificados , Padronização Corporal/efeitos dos fármacos , Padronização Corporal/genética , Padronização Corporal/fisiologia , Movimento Celular/fisiologia , Embrião não Mamífero , Desenvolvimento Embrionário/fisiologia , Comunicação Atrioventricular/genética , Comunicação Atrioventricular/metabolismo , Coxins Endocárdicos/efeitos dos fármacos , Coxins Endocárdicos/metabolismo , Endocárdio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Miocárdio/metabolismo , N-Acetilgalactosaminiltransferases/antagonistas & inibidores , N-Acetilgalactosaminiltransferases/genética , RNA Interferente Pequeno/farmacologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
8.
Dev Biol ; 302(1): 66-79, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17046737

RESUMO

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.


Assuntos
Cardiopatias Congênitas/embriologia , Coração/embriologia , Organogênese , Receptor EphA3/genética , Receptor EphA3/metabolismo , Animais , Malformações Arteriovenosas/embriologia , Malformações Arteriovenosas/metabolismo , Malformações Arteriovenosas/patologia , Eletrocardiografia , Comunicação Atrioventricular/embriologia , Comunicação Atrioventricular/metabolismo , Comunicação Atrioventricular/patologia , Coração/fisiologia , Cardiopatias Congênitas/patologia , Cardiopatias Congênitas/fisiopatologia , Defeitos dos Septos Cardíacos/embriologia , Defeitos dos Septos Cardíacos/metabolismo , Defeitos dos Septos Cardíacos/patologia , Septos Cardíacos/embriologia , Valvas Cardíacas/embriologia , Camundongos , Camundongos Knockout , Mutação , Miocárdio/metabolismo , Miocárdio/patologia
9.
Development ; 133(22): 4585-93, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17050629

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Comunicação Atrioventricular/embriologia , Coração/embriologia , Mesoderma/fisiologia , Modelos Animais , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Comunicação Atrioventricular/metabolismo , Imunofluorescência , Hibridização In Situ , Mesoderma/metabolismo , Camundongos , Camundongos Mutantes , Microdissecção
10.
Birth Defects Res A Clin Mol Teratol ; 76(7): 517-27, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16933305

RESUMO

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.


Assuntos
Comunicação Atrioventricular/induzido quimicamente , Endocárdio/efeitos dos fármacos , Tricloroetileno/toxicidade , Animais , Embrião de Galinha , Relação Dose-Resposta a Droga , Comunicação Atrioventricular/embriologia , Comunicação Atrioventricular/metabolismo , Endocárdio/embriologia , Endocárdio/metabolismo , Átrios do Coração/efeitos dos fármacos , Átrios do Coração/embriologia , Átrios do Coração/metabolismo , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Mesoderma/metabolismo , Mesoderma/ultraestrutura , Microscopia Confocal , Técnicas de Cultura de Órgãos , Fatores de Tempo
11.
Anat Rec A Discov Mol Cell Evol Biol ; 287(2): 1176-82, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16294330

RESUMO

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.


Assuntos
Comunicação Atrioventricular/embriologia , Coração Fetal/embriologia , Mesoderma/citologia , Miócitos Cardíacos/citologia , Situs Inversus/embriologia , Receptores de Ativinas/genética , Receptores de Ativinas/metabolismo , Receptores de Ativinas Tipo I , Animais , Padronização Corporal/genética , Diferenciação Celular , Linhagem da Célula , Comunicação Atrioventricular/genética , Comunicação Atrioventricular/metabolismo , Coração Fetal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/metabolismo , Miócitos Cardíacos/metabolismo , RNA Mensageiro/análise , Situs Inversus/genética , Situs Inversus/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis
12.
Development ; 131(18): 4477-87, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15342473

RESUMO

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.


Assuntos
Diferenciação Celular , Comunicação Atrioventricular/metabolismo , Coração/embriologia , Coração/fisiologia , Morfogênese , Células Musculares/patologia , Proteínas Repressoras/metabolismo , Animais , Divisão Celular , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Regulação para Baixo , Perda do Embrião/metabolismo , Perda do Embrião/patologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Comunicação Atrioventricular/embriologia , Comunicação Atrioventricular/patologia , Endocárdio/metabolismo , Endocárdio/patologia , Inibidores Enzimáticos/farmacologia , Fatores de Transcrição Forkhead , Regulação da Expressão Gênica no Desenvolvimento , Coração/efeitos dos fármacos , Cardiopatias/congênito , Cardiopatias/metabolismo , Cardiopatias/patologia , Proteínas de Grupo de Alta Mobilidade/genética , Hibridização In Situ , Camundongos , Camundongos Knockout , Células Musculares/efeitos dos fármacos , Células Musculares/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição SOXC , Transativadores/genética
13.
Nat Med ; 10(8): 849-57, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15273746

RESUMO

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.


Assuntos
Dosagem de Genes , Mutação/genética , Síndrome de Noonan/genética , Proteínas Tirosina Fosfatases/genética , Animais , Linhagem da Célula/genética , Anormalidades Craniofaciais/genética , Modelos Animais de Doenças , Comunicação Atrioventricular/genética , Comunicação Atrioventricular/metabolismo , Citometria de Fluxo , Genes Dominantes , Cardiopatias Congênitas/genética , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Mutantes , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transtornos Mieloproliferativos/genética , Testes de Precipitina , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteínas Tirosina Fosfatases/metabolismo
14.
Mol Genet Metab ; 80(1-2): 27-35, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14567955

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Comunicação Atrioventricular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Cardiopatias Congênitas/metabolismo , Valvas Cardíacas/crescimento & desenvolvimento , Miocárdio/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Embrião de Mamíferos , Comunicação Atrioventricular/embriologia , Comunicação Atrioventricular/patologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/patologia , Humanos , Técnicas In Vitro , Recém-Nascido , Camundongos , Morfogênese/genética , Morfogênese/fisiologia , Mutação , Miocárdio/patologia , Transdução de Sinais
15.
Clin Lab ; 47(9-10): 441-7, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11596905

RESUMO

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.


Assuntos
Arginina/metabolismo , Derivação Arteriovenosa Cirúrgica , Defeitos dos Septos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Adolescente , Adulto , Fatores Etários , Arginina/sangue , Cateterismo Cardíaco , Estudos de Casos e Controles , Criança , Pré-Escolar , GMP Cíclico/sangue , Comunicação Atrioventricular/sangue , Comunicação Atrioventricular/metabolismo , Comunicação Atrioventricular/fisiopatologia , Feminino , Defeitos dos Septos Cardíacos/sangue , Defeitos dos Septos Cardíacos/fisiopatologia , Hemodinâmica , Humanos , Hipertensão Pulmonar/sangue , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Lactente , Masculino , Nitratos/sangue , Óxido Nítrico/sangue , Circulação Pulmonar
16.
Toxicol Sci ; 53(1): 109-17, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10653528

RESUMO

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.


Assuntos
Comunicação Atrioventricular/induzido quimicamente , Valvas Cardíacas/efeitos dos fármacos , Tricloroetileno/toxicidade , Actinas/metabolismo , Oxirredutases do Álcool/metabolismo , Animais , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , Relação Dose-Resposta a Droga , Comunicação Atrioventricular/metabolismo , Endotélio/citologia , Endotélio/efeitos dos fármacos , Endotélio/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fibrilina-2 , Fibrilinas , Técnica Indireta de Fluorescência para Anticorpo , Valvas Cardíacas/embriologia , Valvas Cardíacas/metabolismo , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Proteínas dos Microfilamentos/metabolismo
17.
Pediatr Cardiol ; 17(1): 21-30, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8778697

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Síndrome de Down/complicações , Comunicação Atrioventricular/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Animais , Síndrome de Down/embriologia , Comunicação Atrioventricular/metabolismo , Comunicação Atrioventricular/patologia , Fibronectinas/metabolismo , Idade Gestacional , Heparitina Sulfato/metabolismo , Imuno-Histoquímica , Mesoderma/patologia , Camundongos , Camundongos Mutantes , Tenascina/metabolismo
18.
Arkh Patol ; 48(6): 3-8, 1986.
Artigo em Russo | MEDLINE | ID: mdl-3753235

RESUMO

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.


Assuntos
Trifosfato de Adenosina/análise , Cardiopatias Congênitas/patologia , Miocárdio/ultraestrutura , Fosfocreatina/análise , Criança , Pré-Escolar , Comunicação Atrioventricular/metabolismo , Comunicação Atrioventricular/patologia , Comunicação Interventricular/metabolismo , Comunicação Interventricular/patologia , Humanos , Miocárdio/análise , Tetralogia de Fallot/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...