Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Dev Biol ; 444 Suppl 1: S337-S351, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30292786

RESUMO

Transcription factors that coordinate migration, differentiation or proliferation of enteric nervous system (ENS) precursors are not well defined. To identify novel transcriptional regulators of ENS development, we performed microarray analysis at embryonic day (E) 17.5 and identified many genes that were enriched in the ENS compared to other bowel cells. We decided to investigate the T-box transcription factor Tbx3, which is prominently expressed in developing and mature ENS. Haploinsufficiency for TBX3 causes ulnar-mammary syndrome (UMS) in humans, a multi-organ system disorder. TBX3 also regulates several genes known to be important for ENS development. To test the hypothesis that Tbx3 is important for ENS development or function, we inactivated Tbx3 in all neural crest derivatives, including ENS progenitors using Wnt1-Cre and a floxed Tbx3 allele. Tbx3 fl/fl; Wnt1-Cre conditional mutant mice die shortly after birth with cleft palate and difficulty feeding. The ENS of mutants was well-organized with a normal density of enteric neurons and nerve fiber bundles, but small bowel glial cell density was reduced. Despite this, bowel motility appeared normal. Furthermore, although Tbx3 is expressed in cardiac neural crest, Tbx3 fl/fl; Wnt1-Cre mice had structurally normal hearts. Thus, loss of Tbx3 within neural crest has selective effects on Tbx3-expressing neural crest derivatives.


Assuntos
Sistema Nervoso Entérico/embriologia , Crista Neural/embriologia , Proteínas com Domínio T/fisiologia , Animais , Diferenciação Celular , Movimento Celular , Fissura Palatina/embriologia , Fissura Palatina/genética , Coração/embriologia , Intestinos/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Crista Neural/metabolismo , Crista Neural/fisiologia , Neurogênese , Neuroglia/fisiologia , Neurônios , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/genética , Proteína Wnt1
2.
Development ; 141(22): 4320-31, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25371366

RESUMO

Elongation of the vertebrate heart occurs by progressive addition of second heart field (SHF) cardiac progenitor cells from pharyngeal mesoderm to the poles of the heart tube. The importance of these cells in the etiology of congenital heart defects has led to extensive research into the regulation of SHF deployment by signaling pathways and transcription factors. However, the basic cellular features of these progenitor cells, including epithelial polarity, cell shape and cell dynamics, remain poorly characterized. Here, using immunofluorescence, live imaging and embryo culture, we demonstrate that SHF cells constitute an atypical, apicobasally polarized epithelium in the dorsal pericardial wall, characterized by apical monocilia and dynamic actin-rich basal filopodia. We identify the 22q11.2 deletion syndrome gene Tbx1, required in the SHF for outflow tract development, as a regulator of the epithelial properties of SHF cells. Cell shape changes in mutant embryos include increased circularity, a reduced basolateral membrane domain and impaired filopodial activity, and are associated with elevated aPKCζ levels. Activation of aPKCζ in embryo culture similarly impairs filopodia activity and phenocopies proliferative defects and ectopic differentiation observed in the SHF of Tbx1 null embryos. Our results reveal that epithelial and progenitor cell status are coupled in the SHF, identifying control of cell shape as a regulatory step in heart tube elongation and outflow tract morphogenesis.


Assuntos
Polaridade Celular/fisiologia , Epitélio/embriologia , Coração/embriologia , Morfogênese/fisiologia , Pseudópodes/fisiologia , Proteínas com Domínio T/genética , Animais , Western Blotting , Síndrome de DiGeorge/genética , Imuno-Histoquímica , Camundongos , Proteína Quinase C/metabolismo
3.
Circ Res ; 115(9): 790-9, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25190705

RESUMO

RATIONALE: Cardiac progenitor cells from the second heart field (SHF) contribute to rapid growth of the embryonic heart, giving rise to right ventricular and outflow tract (OFT) myocardium at the arterial pole of the heart, and atrial myocardium at the venous pole. Recent clonal analysis and cell-tracing experiments indicate that a common progenitor pool in the posterior region of the SHF gives rise to both OFT and atrial myocytes. The mechanisms regulating deployment of this progenitor pool remain unknown. OBJECTIVE: To evaluate the role of TBX1, the major gene implicated in congenital heart defects in 22q11.2 deletion syndrome patients, in posterior SHF development. METHODS AND RESULTS: Using transcriptome analysis, genetic tracing, and fluorescent dye-labeling experiments, we show that Tbx1-dependent OFT myocardium originates in Hox-expressing cells in the posterior SHF. In Tbx1 null embryos, OFT progenitor cells fail to segregate from this progenitor cell pool, leading to failure to expand the dorsal pericardial wall and altered positioning of the cardiac poles. Unexpectedly, addition of SHF cells to the venous pole of the heart is also impaired, resulting in abnormal development of the dorsal mesenchymal protrusion, and partially penetrant atrioventricular septal defects, including ostium primum defects. CONCLUSIONS: Tbx1 is required for inflow as well as OFT morphogenesis by regulating the segregation and deployment of progenitor cells in the posterior SHF. Our results provide new insights into the pathogenesis of congenital heart defects and 22q11.2 deletion syndrome phenotypes.


Assuntos
Movimento Celular , Vasos Coronários/metabolismo , Síndrome de DiGeorge/metabolismo , Coração/embriologia , Miocárdio/metabolismo , Células-Tronco/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Linhagem da Célula , Proliferação de Células , Vasos Coronários/embriologia , Vasos Coronários/patologia , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Idade Gestacional , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese , Miocárdio/patologia , Fenótipo , Transdução de Sinais , Células-Tronco/patologia , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética
4.
Biochim Biophys Acta ; 1833(4): 795-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23051926

RESUMO

At the end of the first week of mouse gestation, cardiomyocyte differentiation initiates in the cardiac crescent to give rise to the linear heart tube. The heart tube subsequently elongates by addition of cardiac progenitor cells from adjacent pharyngeal mesoderm to the growing arterial and venous poles. These progenitor cells, termed the second heart field, originate in splanchnic mesoderm medial to cells of the cardiac crescent and are patterned into anterior and posterior domains adjacent to the arterial and venous poles of the heart, respectively. Perturbation of second heart field cell deployment results in a spectrum of congenital heart anomalies including conotruncal and atrial septal defects seen in human patients. Here, we briefly review current knowledge of how the properties of second heart field cells are controlled by a network of transcriptional regulators and intercellular signaling pathways. Focus will be on 1) the regulation of cardiac progenitor cell proliferation in pharyngeal mesoderm, 2) the control of progressive progenitor cell differentiation and 3) the patterning of cardiac progenitor cells in the dorsal pericardial wall. Coordination of these three processes in the early embryo drives progressive heart tube elongation during cardiac morphogenesis. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Cardiac Pathways of Differentiation, Metabolism and Contraction.


Assuntos
Coração/embriologia , Mesoderma/citologia , Miócitos Cardíacos/citologia , Organogênese/fisiologia , Células-Tronco/citologia , Animais , Diferenciação Celular , Proliferação de Células , Embrião de Mamíferos , Regulação da Expressão Gênica , Coração/anatomia & histologia , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Fatores de Regulação Miogênica/genética , Fatores de Regulação Miogênica/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Transcrição Gênica
5.
Hum Mol Genet ; 21(6): 1217-29, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22116936

RESUMO

The 22q11.2 deletion syndrome (22q11.2DS) is the most common microdeletion disorder and is characterized by abnormal development of the pharyngeal apparatus and heart. Cardiovascular malformations (CVMs) affecting the outflow tract (OFT) are frequently observed in 22q11.2DS and are among the most commonly occurring heart defects. The gene encoding T-box transcription factor 1 (Tbx1) has been identified as a major candidate for 22q11.2DS. However, CVMs are generally considered to have a multigenic basis and single-gene mutations underlying these malformations are rare. The T-box family members Tbx2 and Tbx3 are individually required in regulating aspects of OFT and pharyngeal development. Here, using expression and three-dimensional reconstruction analysis, we show that Tbx1 and Tbx2/Tbx3 are largely uniquely expressed but overlap in the caudal pharyngeal mesoderm during OFT development, suggesting potential combinatorial requirements. Cross-regulation between Tbx1 and Tbx2/Tbx3 was analyzed using mouse genetics and revealed that Tbx1 deficiency affects Tbx2 and Tbx3 expression in neural crest-derived cells and pharyngeal mesoderm, whereas Tbx2 and Tbx3 function redundantly upstream of Tbx1 and Hh ligand expression in pharyngeal endoderm and bone morphogenetic protein- and fibroblast growth factor-signaling in cardiac progenitors. Moreover, in vivo, we show that loss of two of the three genes results in severe pharyngeal hypoplasia and heart tube extension defects. These findings reveal an indispensable T-box gene network governing pharyngeal and OFT development and identify TBX2 and TBX3 as potential modifier genes of the cardiopharyngeal phenotypes found in TBX1-haploinsufficient 22q11.2DS patients.


Assuntos
Artérias/anormalidades , Anormalidades Cardiovasculares/genética , Regulação da Expressão Gênica no Desenvolvimento , Morfogênese/fisiologia , Faringe/anormalidades , Proteínas com Domínio T/fisiologia , Animais , Cromossomos Humanos Par 22/genética , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Fatores de Crescimento de Fibroblastos/metabolismo , Genes Modificadores/fisiologia , Humanos , Técnicas Imunoenzimáticas , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Fenótipo , Obstrução do Fluxo Ventricular Externo/etiologia
6.
Circ Res ; 106(4): 686-94, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20110535

RESUMO

RATIONALE: Loss of Tbx1 and decrease of retinoic acid (RA) synthesis result in DiGeorge/velocardiofacial syndrome (DGS/VCFS)-like phenotypes in mouse models, including defects in septation of the outflow tract of the heart and anomalies of pharyngeal arch-derived structures including arteries of the head and neck, laryngeal-tracheal cartilage, and thymus/parathyroid. Wild-type levels of T-box transcription factor (Tbx)1 and RA signaling are required for normal pharyngeal arch artery development. Recent studies have shown that reduction of RA or loss of Tbx1 alters the contribution of second heart field (SHF) progenitor cells to the elongating heart tube. OBJECTIVE: Here we tested whether Tbx1 and the RA signaling pathway interact during the deployment of the SHF and formation of the mature aortic arch. METHODS AND RESULTS: Molecular markers of the SHF, neural crest and smooth muscle cells, were analyzed in Raldh2;Tbx1 compound heterozygous mutants. Our results revealed that the SHF and outflow tract develop normally in Raldh2(+/-);Tbx1(+/-) embryos. However, we found that decreased levels of RA accelerate the recovery from arterial growth delay observed in Tbx1(+/-) mutant embryos. This compensation coincides with the differentiation of smooth muscle cells in the 4th pharyngeal arch arteries, and is associated with severity of neural crest cell migration defects observed in these mutants. CONCLUSIONS: Our data suggest that differences in levels of embryonic RA may contribute to the variability in great artery anomalies observed in DGS/VCFS patients.


Assuntos
Aorta Torácica/metabolismo , Região Branquial/metabolismo , Síndrome de DiGeorge/metabolismo , Músculo Liso Vascular/metabolismo , Transdução de Sinais , Tretinoína/metabolismo , Aldeído Oxirredutases/genética , Aldeído Oxirredutases/metabolismo , Animais , Aorta Torácica/anormalidades , Região Branquial/anormalidades , Diferenciação Celular , Cruzamentos Genéticos , Síndrome de DiGeorge/embriologia , Síndrome de DiGeorge/genética , Modelos Animais de Doenças , Regulação para Baixo , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Idade Gestacional , Coração/embriologia , Heterozigoto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Músculo Liso Vascular/anormalidades , Mutação , Crista Neural/anormalidades , Crista Neural/metabolismo , Fenótipo , Transdução de Sinais/genética , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Técnicas de Cultura de Tecidos
7.
Circ Res ; 104(8): 933-42, 2009 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-19390062

RESUMO

Insight into the mechanisms underlying congenital heart defects and the use of stem cells for cardiac repair are major research goals in cardiovascular biology. In the early embryo, progenitor cells in pharyngeal mesoderm contribute to the rapid growth of the heart tube during looping morphogenesis. These progenitor cells constitute the second heart field (SHF) and were first identified in 2001. Direct or indirect perturbation of SHF addition to the heart results in congenital heart defects, including arterial pole alignment defects. Over the last 3 years, a number of studies have identified key intercellular signaling pathways that control the proliferation and deployment of SHF progenitor cells. Here, we review data concerning Wnt, fibroblast growth factor, bone morphogenetic protein, Hedgehog, and retinoic acid signaling that have begun to identify the ligand sources and responding cell types controlling SHF development. These studies have revealed the importance of signals from pharyngeal mesoderm itself, as well as critical inputs from adjacent pharyngeal epithelia and neural crest cells. Proliferation is emerging as a central checkpoint in the regulation of SHF development. Together, these studies contribute to defining the niche of cardiac progenitor cells in the early embryo, and we discuss the implications of these findings for the regulation of resident stem cell populations in the fetal and postnatal heart. Characterization of signals that maintain, expand, and regulate the differentiation of cardiac progenitor cells is essential for understanding both the etiology of congenital heart defects and the biomedical application of stem cell populations for cardiac repair.


Assuntos
Células-Tronco Embrionárias/metabolismo , Cardiopatias Congênitas/metabolismo , Coração/embriologia , Miocárdio/metabolismo , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais , Animais , Comunicação Autócrina , Proteínas Morfogenéticas Ósseas/metabolismo , Região Branquial , Diferenciação Celular , Movimento Celular , Proliferação de Células , Endoderma/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/cirurgia , Proteínas Hedgehog/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Proteínas com Homeodomínio LIM , Mesoderma/metabolismo , Crista Neural/metabolismo , Transplante de Células-Tronco , Fatores de Transcrição , Tretinoína/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
8.
Birth Defects Res A Clin Mol Teratol ; 91(6): 477-84, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21591244

RESUMO

Conotruncal congenital heart defects, including defects in septation and alignment of the ventricular outlets, account for approximately a third of all congenital heart defects. Failure of the left ventricle to obtain an independent outlet results in incomplete separation of systemic and pulmonary circulation at birth. The embryonic outflow tract, a transient cylinder of myocardium connecting the embryonic ventricles to the aortic sac, plays a critical role in this process during normal development. The outflow tract (OFT) is derived from a population of cardiac progenitor cells called the second heart field that contributes to the arterial pole of the heart tube during cardiac looping. During septation, the OFT is remodeled to form the base of the ascending aorta and pulmonary trunk. Tbx1, the major candidate gene for DiGeorge syndrome, is a critical transcriptional regulator of second heart field development. DiGeorge syndrome patients are haploinsufficient for Tbx1 and present a spectrum of conotruncal anomalies including tetralogy of Fallot, pulmonary atresia, and common arterial trunk. In this review, we focus on the role of Tbx1 in the regulation of second heart field deployment and, in particular, in the development of a specific population of myocardial cells at the base of the pulmonary trunk. Recent data characterizing additional properties and regulators of development of this region of the heart, including the retinoic acid, hedgehog, and semaphorin signaling pathways, are discussed. These findings identify future subpulmonary myocardium as the clinically relevant component of the second heart field and provide new mechanistic insight into a spectrum of common conotruncal congenital heart defects.


Assuntos
Cardiopatias Congênitas/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/genética , Humanos , Pulmão/metabolismo , Transdução de Sinais , Proteínas com Domínio T/genética
9.
Dev Dyn ; 239(12): 3303-11, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20941780

RESUMO

Development of the mammalian heart is mediated by complex interactions between myocardial, endocardial, and neural crest-derived cells. Studies in Drosophila have shown that the Slit-Robo signaling pathway controls cardiac cell shape changes and lumen formation of the heart tube. Here, we demonstrate by in situ hybridization that multiple Slit ligands and Robo receptors are expressed in the developing mouse heart. Slit3 is the predominant ligand transcribed in the early mouse heart and is expressed in the ventral wall of the linear heart tube and subsequently in chamber but not in atrioventricular canal myocardium. Furthermore, we identify that the homeobox gene Nkx2-5 is required for early ventral restriction of Slit3 and that the T-box transcription factor Tbx2 mediates repression of Slit3 in nonchamber myocardium. Our results suggest that patterned Slit-Robo signaling may contribute to the control of oriented cell growth during chamber morphogenesis of the mammalian heart.


Assuntos
Coração/embriologia , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Gravidez , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Roundabout
10.
Circ Res ; 103(7): 743-50, 2008 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-18723448

RESUMO

Conotruncal and ventricular septal congenital heart anomalies result from defects in formation and division of the embryonic outflow tract. Cardiac remodeling during outflow tract and ventricular septation converts the tubular embryonic heart into a parallel circulatory system with an independent left ventricular outlet and right ventricular inlet. Tbx3 encodes a T-box-containing transcription factor expressed in the developing conduction system of the heart. Mutations in TBX3 cause ulnar-mammary syndrome. Here we show that mice lacking Tbx3 develop severe outflow tract defects, including connection of both the aorta and pulmonary trunk with the right ventricle, in addition to aortic arch artery anomalies and abnormal communication between the right atrium and left ventricle. Alignment defects are preceded by a delay in caudal displacement of the arterial pole of the heart during aortic arch artery formation. Embryonic anterior-posterior patterning and cardiac chamber development are unaffected in Tbx3 mutant embryos. However, the contribution of second heart field derived progenitor cells to the arterial pole of the heart is impaired. Tbx3 is expressed in pharyngeal epithelia and neural crest cells in the pharyngeal region, suggesting an indirect role in second heart field deployment. Loss of Tbx3 affects multiple signaling pathways regulating second heart field proliferation and outflow tract morphogenesis, including fibroblast growth factor signaling, leading to a failure of normal heart tube extension and consequent atrioventricular and ventriculoarterial alignment defects.


Assuntos
Aorta/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Coração/embriologia , Organogênese/fisiologia , Transdução de Sinais/fisiologia , Proteínas com Domínio T/metabolismo , Animais , Cardiopatias Congênitas/genética , Camundongos , Camundongos Mutantes , Especificidade de Órgãos/fisiologia , Proteínas com Domínio T/genética
11.
Circ Res ; 103(2): 142-8, 2008 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-18583714

RESUMO

TBX1, encoding a T-box containing transcription factor, is the major candidate gene for del22q11.2 or DiGeorge syndrome, characterized by craniofacial and cardiovascular defects including tetralogy of Fallot and common arterial trunk. Mice lacking Tbx1 have severe defects in the development of pharyngeal derivatives including cardiac progenitor cells of the second heart field that contribute to the arterial pole of the heart. The outflow tract of Tbx1 mutant embryos is short and narrow resulting in common arterial trunk. Here we show by a series of genetic crosses using transgene markers of second heart field derived myocardium and coronary endothelial cells that a subdomain of myocardium normally observed at the base of the pulmonary trunk is reduced and malpositioned in Tbx1 mutant hearts. This defect is associated with anomalous coronary artery patterning. Both right and left coronary ostia form predominantly at the right/ventral sinus in mutant hearts, proximal coronary arteries coursing across the normally coronary free ventral region of the heart. We have identified Semaphorin3c as a Tbx1-dependent gene expressed in subpulmonary myocardium. Our results implicate second heart field development in coronary artery patterning and provide new insights into the association between conotruncal defects and coronary artery anomalies.


Assuntos
Anomalias dos Vasos Coronários/genética , Vasos Coronários/embriologia , Vasos Coronários/fisiopatologia , Proteínas com Domínio T/genética , Animais , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/fisiopatologia , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Coração/embriologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Artéria Pulmonar/embriologia , Artéria Pulmonar/fisiopatologia , Fluxo Sanguíneo Regional/fisiologia , Semaforinas/genética
12.
Cardiovasc Res ; 104(3): 432-42, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25344367

RESUMO

AIMS: Cardiomyocyte proliferation gradually declines during embryogenesis resulting in severely limited regenerative capacities in the adult heart. Understanding the developmental processes controlling cardiomyocyte proliferation may thus identify new therapeutic targets to modulate the cell-cycle activity of cardiomyocytes in the adult heart. This study aims to determine the mechanism by which fibroblast growth factor 10 (FGF10) controls foetal cardiomyocyte proliferation and to test the hypothesis that FGF10 promotes the proliferative capacity of adult cardiomyocytes. METHODS AND RESULTS: Analysis of Fgf10(-/-) hearts and primary cardiomyocyte cultures reveals that altered ventricular morphology is associated with impaired proliferation of right but not left-ventricular myocytes. Decreased FOXO3 phosphorylation associated with up-regulated p27(kip) (1) levels was observed specifically in the right ventricle of Fgf10(-/-) hearts. In addition, cell-type-specific expression analysis revealed that Fgf10 and its receptor, Fgfr2b, are expressed in cardiomyocytes and not cardiac fibroblasts, consistent with a cell-type autonomous role of FGF10 in regulating regional specific myocyte proliferation in the foetal heart. Furthermore, we demonstrate that in vivo overexpression of Fgf10 in adult mice promotes cardiomyocyte but not cardiac fibroblast cell-cycle re-entry. CONCLUSION: FGF10 regulates regional cardiomyocyte proliferation in the foetal heart through a FOXO3/p27(kip1) pathway. In addition, FGF10 triggers cell-cycle re-entry of adult cardiomyocytes and is thus a potential target for cardiac repair.


Assuntos
Fator 10 de Crescimento de Fibroblastos/fisiologia , Coração/embriologia , Miócitos Cardíacos/fisiologia , Animais , Ciclo Celular , Proliferação de Células , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/metabolismo , Camundongos
13.
PLoS One ; 4(7): e6267, 2009 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-19609448

RESUMO

BACKGROUND: Rapid growth of the embryonic heart occurs by addition of progenitor cells of the second heart field to the poles of the elongating heart tube. Failure or perturbation of this process leads to congenital heart defects. In order to provide further insight into second heart field development we characterized the insertion site of a transgene expressed in the second heart field and outflow tract as the result of an integration site position effect. RESULTS: Here we show that the integration site of the A17-Myf5-nlacZ-T55 transgene lies upstream of Hes1, encoding a basic helix-loop-helix containing transcriptional repressor required for the maintenance of diverse progenitor cell populations during embryonic development. Transgene expression in a subset of Hes1 expression sites, including the CNS, pharyngeal epithelia, pericardium, limb bud and lung endoderm suggests that Hes1 is the endogenous target of regulatory elements trapped by the transgene. Hes1 is expressed in pharyngeal endoderm and mesoderm including the second heart field. Analysis of Hes1 mutant hearts at embryonic day 15.5 reveals outflow tract alignment defects including ventricular septal defects and overriding aorta. At earlier developmental stages, Hes1 mutant embryos display defects in second heart field proliferation, a reduction in cardiac neural crest cells and failure to completely extend the outflow tract. CONCLUSIONS: Hes1 is expressed in cardiac progenitor cells in the early embryo and is required for development of the arterial pole of the heart.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Coração/embriologia , Proteínas de Homeodomínio/genética , Miocárdio/metabolismo , Animais , Sequência de Bases , Western Blotting , Proliferação de Células , Primers do DNA , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Morfogênese , Fatores de Transcrição HES-1 , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA