Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Annu Rev Genomics Hum Genet ; 22: 257-284, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34061573

RESUMO

Congenital heart disease is the most frequent birth defect and the leading cause of death for the fetus and in the first year of life. The wide phenotypic diversity of congenital heart defects requires expert diagnosis and sophisticated repair surgery. Although these defects have been described since the seventeenth century, it was only in 2005 that a consensus international nomenclature was adopted, followed by an international classification in 2017 to help provide better management of patients. Advances in genetic engineering, imaging, and omics analyses have uncovered mechanisms of heart formation and malformation in animal models, but approximately 80% of congenital heart defects have an unknown genetic origin. Here, we summarize current knowledge of congenital structural heart defects, intertwining clinical and fundamental research perspectives, with the aim to foster interdisciplinary collaborations at the cutting edge of each field. We also discuss remaining challenges in better understanding congenital heart defects and providing benefits to patients.


Assuntos
Cardiopatias Congênitas , Animais , Cardiopatias Congênitas/genética , Humanos , Modelos Animais
2.
Development ; 145(22)2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-30467108

RESUMO

Extensive studies have shown how bilateral symmetry of the vertebrate embryo is broken during early development, resulting in a molecular left-right bias in the mesoderm. However, how this early asymmetry drives the asymmetric morphogenesis of visceral organs remains poorly understood. The heart provides a striking model of left-right asymmetric morphogenesis, undergoing rightward looping to shape an initially linear heart tube and align cardiac chambers. Importantly, abnormal left-right patterning is associated with severe congenital heart defects, as exemplified in heterotaxy syndrome. Here, we compare the mechanisms underlying the rightward looping of the heart tube in fish, chick and mouse embryos. We propose that heart looping is not only a question of direction, but also one of fine-tuning shape. This is discussed in the context of evolutionary and clinical perspectives.


Assuntos
Padronização Corporal , Cardiopatias/embriologia , Coração/embriologia , Animais , Humanos , Modelos Biológicos , Morfogênese , Vertebrados/embriologia
3.
Development ; 140(2): 395-404, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23250213

RESUMO

Anisotropies that underlie organ morphogenesis have been quantified in 2D, taking advantage of a reference axis. However, morphogenesis is a 3D process and it remains a challenge to analyze cell polarities in 3D. Here, we have designed a novel procedure that integrates multidisciplinary tools, including image segmentation, statistical analyses, axial clustering and correlation analysis. The result is a sensitive and unbiased assessment of the significant alignment of cell orientations in 3D, compared with a random axial distribution. Taking the mouse heart as a model, we validate the procedure at the fetal stage, when cardiomyocytes are known to be aligned. At the embryonic stage, our study reveals that ventricular cells are already coordinated locally. The centrosome-nucleus axes and the cell division axes are biased in a plane parallel to the outer surface of the heart, with a minor transmural component. We show further alignment of these axes locally in the plane of the heart surface. Our method is generally applicable to other sets of vectors or axes in 3D tissues to map the regions where they show significant alignment.


Assuntos
Biologia do Desenvolvimento/métodos , Coração/embriologia , Imageamento Tridimensional/métodos , Animais , Anisotropia , Padronização Corporal , Divisão Celular , Núcleo Celular/metabolismo , Centrossomo/metabolismo , Processamento de Imagem Assistida por Computador , Camundongos , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Fatores de Tempo
4.
Bioinformatics ; 29(6): 772-9, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23337749

RESUMO

MOTIVATION: In developmental biology, quantitative tools to extract features from fluorescence microscopy images are becoming essential to characterize organ morphogenesis at the cellular level. However, automated image analysis in this context is a challenging task, owing to perturbations induced by the acquisition process, especially in organisms where the tissue is dense and opaque. RESULTS: We propose an automated framework for the segmentation of 3D microscopy images of highly cluttered environments such as developing tissues. The approach is based on a partial differential equation framework that jointly takes advantage of the nuclear and cellular membrane information to enable accurate extraction of nuclei and cells in dense tissues. This framework has been used to study the developing mouse heart, allowing the extraction of quantitative information such as the cell cycle duration; the method also provides qualitative information on cell division and cell polarity through the creation of 3D orientation maps that provide novel insight into tissue organization during organogenesis.


Assuntos
Imageamento Tridimensional/métodos , Microscopia de Fluorescência/métodos , Miocárdio/citologia , Animais , Ciclo Celular , Divisão Celular , Núcleo Celular/ultraestrutura , Centrossomo/ultraestrutura , Coração/embriologia , Camundongos , Miocárdio/ultraestrutura
5.
Circ Res ; 111(10): 1313-22, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22855565

RESUMO

RATIONALE: Genetic tracing experiments and cell lineage analyses are complementary approaches that give information about the progenitor cells of a tissue. Approaches based on gene expression have led to conflicting views about the origin of the venous pole of the heart. Whereas the heart forms from 2 sources of progenitor cells, the first and second heart fields, genetic tracing has suggested a distinct origin for caval vein myocardium, from a proposed third heart field. OBJECTIVE: To determine the cell lineage history of the myocardium at the venous pole of the heart. METHODS AND RESULTS: We used retrospective clonal analyses to investigate lineage segregation for myocardium at the venous pole of the mouse heart, independent of gene expression. CONCLUSIONS: Our lineage analysis unequivocally shows that caval vein and atrial myocardium share a common origin and demonstrates a clonal relationship between the pulmonary vein and progenitors of the left venous pole. Clonal characteristics give insight into the development of the veins. Unexpectedly, we found a lineage relationship between the venous pole and part of the arterial pole, which is derived exclusively from the second heart field. Integration of results from genetic tracing into the lineage tree adds a further temporal dimension to this reconstruction of the history of venous myocardium and the arterial pole.


Assuntos
Linhagem da Célula/fisiologia , Células-Tronco Embrionárias/citologia , Coração/embriologia , Miocárdio/citologia , Organogênese/fisiologia , Animais , Células Clonais/citologia , Células Clonais/fisiologia , Células-Tronco Embrionárias/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Átrios do Coração/citologia , Átrios do Coração/embriologia , Masculino , Camundongos , Camundongos Mutantes , Veias Pulmonares/citologia , Veias Pulmonares/embriologia , Veia Cava Inferior/citologia , Veia Cava Inferior/embriologia , Veia Cava Superior/citologia , Veia Cava Superior/embriologia
6.
Circ Res ; 111(10): 1323-35, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22955731

RESUMO

RATIONALE: The second heart field (SHF) contains progenitors of all heart chambers, excluding the left ventricle. The SHF is patterned, and the anterior region is known to be destined to form the outflow tract and right ventricle. OBJECTIVE: The aim of this study was to map the fate of the posterior SHF (pSHF). METHODS AND RESULTS: We examined the contribution of pSHF cells, labeled by lipophilic dye at the 4- to 6-somite stage, to regions of the heart at 20 to 25 somites, using mouse embryo culture. Cells more cranial in the pSHF contribute to the atrioventricular canal (AVC) and atria, whereas those more caudal generate the sinus venosus, but there is intermixing of fate throughout the pSHF. Caudal pSHF contributes symmetrically to the sinus venosus, but the fate of cranial pSHF is left/right asymmetrical. Left pSHF moves to dorsal left atrium and superior AVC, whereas right pSHF contributes to right atrium, ventral left atrium, and inferior AVC. Retrospective clonal analysis shows the relationships between AVC and atria to be clonal and that right and left progenitors diverge before first and second heart lineage separation. Cranial pSHF cells also contribute to the outflow tract: proximal and distal at 4 somites, and distal only at 6 somites. All outflow tract-destined cells are intermingled with those that will contribute to inflow and AVC. CONCLUSIONS: These observations show asymmetric fate of the pSHF, resulting in unexpected left/right contributions to both poles of the heart and can be integrated into a model of the morphogenetic movement of cells during cardiac looping.


Assuntos
Células-Tronco Embrionárias/citologia , Coração/embriologia , Coração/fisiologia , Organogênese/fisiologia , Animais , Animais não Endogâmicos , Seio Coronário/citologia , Seio Coronário/embriologia , Técnicas de Cultura Embrionária , Células-Tronco Embrionárias/fisiologia , Fator 10 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica no Desenvolvimento , Átrios do Coração/citologia , Átrios do Coração/embriologia , Ventrículos do Coração/citologia , Ventrículos do Coração/embriologia , Óperon Lac , Camundongos , Camundongos Transgênicos , Somitos/citologia , Somitos/embriologia
7.
Med Sci (Paris) ; 30(11): 996-1003, 2014 Nov.
Artigo em Francês | MEDLINE | ID: mdl-25388582

RESUMO

After the seminal discovery in 2000 that primary cilia are functional organelles which are essential for embryonic development, several mouse models of ciliopathies have been generated. The heart is frequently affected, with a large spectrum of malformations. The cilia of the node are required early in development in the determination of the left/right laterality of the embryo, which has secondary consequences on the formation of the heart. Thus, abnormal looping of the heart is a recurrent phenotype in models of ciliopathies. However, the function of primary cilia in cardiac cells remains poorly understood. Receptors such as polycystins or hedgehog receptors are usually localized in the primary cilium, raising the possibility that these signalling pathways, which are important for the septation and the growth of the heart, are transduced in primary cilia of cardiac cells. Knowledge of the roles of primary cilia at different steps of heart development and in different cardiac cell types will be essential to better understand the origin of human cardiopathies associated with ciliopathies.


Assuntos
Cílios/fisiologia , Coração/embriologia , Animais , Transtornos da Motilidade Ciliar/genética , Transtornos da Motilidade Ciliar/patologia , Vasos Coronários/embriologia , Desenvolvimento Embrionário , Coração Fetal/crescimento & desenvolvimento , Coração Fetal/ultraestrutura , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Proteínas Hedgehog/genética , Proteínas Hedgehog/fisiologia , Humanos , Camundongos , Morfogênese/fisiologia , Receptores Patched , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia , Transdução de Sinais/fisiologia , Receptor Smoothened , Síndrome , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/fisiologia , Vertebrados/embriologia
8.
Dev Dyn ; 242(6): 665-77, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23526457

RESUMO

BACKGROUND: The ventricular conduction system (VCS) coordinates the heartbeat and is composed of central components (the atrioventricular node, bundle, and right and left bundle branches) and a peripheral Purkinje fiber network. Conductive myocytes develop from common progenitor cells with working myocytes in a bimodal process of lineage restriction followed by limited outgrowth. The lineage relationship between progenitor cells giving rise to different components of the VCS is unclear. RESULTS: Cell lineage contributions to different components of the VCS were analysed by a combination of retrospective clonal analysis, regionalized transgene expression studies, and genetic tracing experiments using Connexin40-GFP mice that precisely delineate the VCS. Analysis of a library of hearts containing rare large clusters of clonally related myocytes identifies two VCS lineages encompassing either the right Purkinje fiber network or left bundle branch. Both lineages contribute to the atrioventricular bundle and right bundle branch that segregate early from working myocytes. Right and left VCS lineages share the transcriptional program of the respective ventricular working myocytes and genetic tracing experiments discount alternate progenitor cell contributions to the VCS. CONCLUSIONS: The mammalian VCS is comprised of cells derived from two lineages, supporting a dual contribution of first and second heart field progenitor cells.


Assuntos
Linhagem da Célula , Conexinas/genética , Sistema de Condução Cardíaco/embriologia , Ventrículos do Coração/embriologia , Células-Tronco/citologia , Alelos , Animais , Fascículo Atrioventricular/metabolismo , Conexinas/fisiologia , Feminino , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Microscopia de Fluorescência , Miocárdio/citologia , Miócitos Cardíacos/citologia , Fatores de Tempo , Transcrição Gênica , Transgenes , Proteína alfa-5 de Junções Comunicantes
9.
Nat Commun ; 15(1): 1982, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38438357

RESUMO

De novo synthesis of the pyrimidine, cytidine triphosphate (CTP), is crucial for DNA/RNA metabolism and depends on the CTP synthetases, CTPS1 and -2. Partial CTPS1 deficiency in humans has previously been shown to lead to immunodeficiency, with impaired expansion of T and B cells. Here, we examine the effects of conditional and inducible inactivation of Ctps1 and/or Ctps2 on mouse embryonic development and immunity. We report that deletion of Ctps1, but not Ctps2, is embryonic-lethal. Tissue and cells with high proliferation and renewal rates, such as intestinal epithelium, erythroid and thymic lineages, activated B and T lymphocytes, and memory T cells strongly rely on CTPS1 for their maintenance and growth. However, both CTPS1 and CTPS2 are required for T cell proliferation following TCR stimulation. Deletion of Ctps1 in T cells or treatment with a CTPS1 inhibitor rescued Foxp3-deficient mice from fatal systemic autoimmunity and reduced the severity of experimental autoimmune encephalomyelitis. These findings support that CTPS1 may represent a target for immune suppression.


Assuntos
Autoimunidade , Desenvolvimento Embrionário , Feminino , Gravidez , Humanos , Animais , Camundongos , Citidina Trifosfato , Autoimunidade/genética , Linfócitos B , Proliferação de Células
10.
Development ; 137(19): 3269-79, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20823066

RESUMO

Head muscle progenitors in pharyngeal mesoderm are present in close proximity to cells of the second heart field and show overlapping patterns of gene expression. However, it is not clear whether a single progenitor cell gives rise to both heart and head muscles. We now show that this is the case, using a retrospective clonal analysis in which an nlaacZ sequence, converted to functional nlacZ after a rare intragenic recombination event, is targeted to the alpha(c)-actin gene, expressed in all developing skeletal and cardiac muscle. We distinguish two branchiomeric head muscle lineages, which segregate early, both of which also contribute to myocardium. The first gives rise to the temporalis and masseter muscles, which derive from the first branchial arch, and also to the extraocular muscles, thus demonstrating a contribution from paraxial as well as prechordal mesoderm to this anterior muscle group. Unexpectedly, this first lineage also contributes to myocardium of the right ventricle. The second lineage gives rise to muscles of facial expression, which derive from mesoderm of the second branchial arch. It also contributes to outflow tract myocardium at the base of the arteries. Further sublineages distinguish myocardium at the base of the aorta or pulmonary trunk, with a clonal relationship to right or left head muscles, respectively. We thus establish a lineage tree, which we correlate with genetic regulation, and demonstrate a clonal relationship linking groups of head muscles to different parts of the heart, reflecting the posterior movement of the arterial pole during pharyngeal morphogenesis.


Assuntos
Linhagem da Célula , Coração/embriologia , Músculo Esquelético/citologia , Músculo Esquelético/embriologia , Miocárdio/citologia , Animais , Padronização Corporal , Camundongos , Camundongos Transgênicos
11.
Dev Cell ; 58(21): 2217-2234.e8, 2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37852253

RESUMO

Despite their burden, most congenital defects remain poorly understood, due to lack of knowledge of embryological mechanisms. Here, we identify Greb1l mutants as a mouse model of crisscross heart. Based on 3D quantifications of shape changes, we demonstrate that torsion of the atrioventricular canal occurs together with supero-inferior ventricles at E10.5, after heart looping. Mutants phenocopy partial deficiency in retinoic acid signaling, which reflect overlapping pathways in cardiac precursors. Spatiotemporal gene mapping and cross-correlated transcriptomic analyses further reveal the role of Greb1l in maintaining a pool of dorsal pericardial wall precursor cells during heart tube elongation, likely by controlling ribosome biogenesis and cell differentiation. Consequently, we observe growth arrest and malposition of the outflow tract, which are predictive of abnormal tube remodeling in mutants. Our work on a rare cardiac malformation opens novel perspectives on the origin of a broader spectrum of congenital defects associated with GREB1L in humans.


Assuntos
Coração Entrecruzado , Humanos , Animais , Camundongos , Morfogênese/genética , Coração , Ventrículos do Coração , Células-Tronco
12.
Circ Res ; 107(1): 153-61, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20466980

RESUMO

RATIONALE: The ventricular conduction system controls the propagation of electric activity through the heart to coordinate cardiac contraction. This system is composed of specialized cardiomyocytes organized in defined structures including central components and a peripheral Purkinje fiber network. How the mammalian ventricular conduction system is established during development remains controversial. OBJECTIVE: To define the lineage relationship between cells of the murine ventricular conduction system and surrounding working myocytes. METHODS AND RESULTS: A retrospective clonal analysis using the alpha-cardiac actin(nlaacZ/+) mouse line was carried out in three week old hearts. Clusters of clonally related myocytes were screened for conductive cells using connexin40-driven enhanced green fluorescent protein expression. Two classes of clusters containing conductive cells were obtained. Mixed clusters, composed of conductive and working myocytes, reveal that both cell types develop from common progenitor cells, whereas smaller unmixed clusters, composed exclusively of conductive cells, show that proliferation continues after lineage restriction to the conduction system lineage. Differences in the working component of mixed clusters between the right and left ventricles reveal distinct progenitor cell histories in these cardiac compartments. These results are supported by genetic fate mapping using Cre recombinase revealing progressive restriction of connexin40-positive myocytes to a conductive fate. CONCLUSIONS: A biphasic mode of development, lineage restriction followed by limited outgrowth, underlies establishment of the mammalian ventricular conduction system.


Assuntos
Sistema de Condução Cardíaco/crescimento & desenvolvimento , Ventrículos do Coração/crescimento & desenvolvimento , Fatores Etários , Animais , Feminino , Técnicas de Introdução de Genes , Masculino , Camundongos , Camundongos Transgênicos , Gravidez , Estudos Retrospectivos
13.
Curr Opin Genet Dev ; 73: 101896, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35026527

RESUMO

As other tubular organs, the embryonic heart develops from an epithelial sheet of cells, referred to as the heart field. The second heart field, which lies in the dorsal pericardial wall, constitutes a transient cell reservoir, integrating patterning and polarity cues. Conditional mutants have shown that impairment of the epithelial architecture of the second heart field is associated with congenital heart defects. Here, taking the mouse as a model, we review the epithelial properties of the second heart field and how they are modulated upon cardiomyocyte differentiation. Compared to other cases of tubulogenesis, the cellular dynamics in the second heart field are only beginning to be revealed. A challenge for the future will be to unravel key physical forces driving heart tube morphogenesis.


Assuntos
Coração , Organogênese , Animais , Diferenciação Celular/genética , Epitélio , Camundongos , Morfogênese , Organogênese/genética
14.
Dev Biol ; 331(2): 210-21, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19422818

RESUMO

In the mouse blastocyst, some cells of the inner cell mass (ICM) develop into primitive endoderm (PE) at the surface, while deeper cells form the epiblast. It remained unclear whether the position of cells determines their fate, such that gene expression is adjusted to cell position, or if cells are pre-specified at random positions and then sort. We have tracked and characterised dynamics of all ICM cells from the early to late blastocyst stage. Time-lapse microscopy in H2B-EGFP embryos shows that a large proportion of ICM cells change position between the surface and deeper compartments. Most of this cell movement depends on actin and is associated with cell protrusions. We also find that while most cells are precursors for only one lineage, some give rise to both, indicating that lineage segregation is not complete in the early ICM. Finally, changing the expression levels of the PE marker Gata6 reveals that it is required in surface cells but not sufficient for the re-positioning of deeper cells. We provide evidence that Wnt9A, known to be expressed in the surface ICM, facilitates re-positioning of Gata6-expressing cells. Combining these experimental results with computer modelling suggests that PE formation involves both cell sorting movements and position-dependent induction.


Assuntos
Massa Celular Interna do Blastocisto/citologia , Linhagem da Célula/fisiologia , Movimento Celular/fisiologia , Animais , Antígenos de Diferenciação/metabolismo , Massa Celular Interna do Blastocisto/fisiologia , Padronização Corporal/fisiologia , Diferenciação Celular/fisiologia , Endoderma/citologia , Endoderma/embriologia , Endoderma/fisiologia , Fator de Transcrição GATA6/metabolismo , Camundongos , Modelos Biológicos , Proteínas Wnt/metabolismo
15.
Philos Trans R Soc Lond B Biol Sci ; 375(1809): 20190556, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-32829679

RESUMO

Retinoic acid (RA), derived from vitamin A, is a major teratogen, clinically recognized in 1983. Identification of its natural presence in the embryo and dissection of its molecular mechanism of action became possible in the animal model with the advent of molecular biology, starting with the cloning of its nuclear receptor. In normal development, the dose of RA is tightly controlled to regulate organ formation. Its production depends on enzymes, which have a dynamic expression profile during embryonic development. As a small molecule, it diffuses rapidly and acts as a morphogen. Here, we review advances in deciphering how endogenously produced RA provides positional information to cells. We compare three mesodermal tissues, the limb, the somites and the heart, and discuss how RA signalling regulates antero-posterior and left-right patterning. A common principle is the establishment of its spatio-temporal dynamics by positive and negative feedback mechanisms and by antagonistic signalling by FGF. However, the response is cell-specific, pointing to the existence of cofactors and effectors, which are as yet incompletely characterized. This article is part of a discussion meeting issue 'Contemporary morphogenesis'.


Assuntos
Padronização Corporal , Embrião de Mamíferos/embriologia , Embrião não Mamífero/embriologia , Mesoderma/embriologia , Transdução de Sinais , Tretinoína/metabolismo , Animais , Camundongos , Peixe-Zebra
16.
Dev Cell ; 55(4): 413-431.e6, 2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-33171097

RESUMO

The secreted factor Nodal, known as a major left determinant, is associated with severe heart defects. Yet, it has been unclear how it regulates asymmetric morphogenesis such as heart looping, which align cardiac chambers to establish the double blood circulation. Here, we report that Nodal is transiently active in precursors of the mouse heart tube poles, before looping. In conditional mutants, we show that Nodal is not required to initiate asymmetric morphogenesis. We provide evidence of a heart-specific random generator of asymmetry that is independent of Nodal. Using 3D quantifications and simulations, we demonstrate that Nodal functions as a bias of this mechanism: it is required to amplify and coordinate opposed left-right asymmetries at the heart tube poles, thus generating a robust helical shape. We identify downstream effectors of Nodal signaling, regulating asymmetries in cell proliferation, differentiation, and extracellular matrix composition. Our study uncovers how Nodal regulates asymmetric organogenesis.


Assuntos
Padronização Corporal , Coração/embriologia , Proteína Nodal/metabolismo , Transdução de Sinais , Animais , Diferenciação Celular , Proliferação de Células , Simulação por Computador , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Matriz Extracelular/metabolismo , Cardiopatias Congênitas/metabolismo , Mesoderma/metabolismo , Camundongos , Miocárdio/metabolismo , Miocárdio/patologia , Transgenes
17.
Cell Rep ; 32(3): 107932, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32698004

RESUMO

Cilia and the intraflagellar transport (IFT) proteins involved in ciliogenesis are associated with congenital heart diseases (CHDs). However, the molecular links between cilia, IFT proteins, and cardiogenesis are yet to be established. Using a combination of biochemistry, genetics, and live-imaging methods, we show that IFT complex B proteins (Ift88, Ift54, and Ift20) modulate the Hippo pathway effector YAP1 in zebrafish and mouse. We demonstrate that this interaction is key to restrict the formation of the proepicardium and the myocardium. In cellulo experiments suggest that IFT88 and IFT20 interact with YAP1 in the cytoplasm and functionally modulate its activity, identifying a molecular link between cilia-related proteins and the Hippo pathway. Taken together, our results highlight a noncanonical role for IFT complex B proteins during cardiogenesis and shed light on a mechanism of action for ciliary proteins in YAP1 regulation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Flagelos/metabolismo , Coração/embriologia , Organogênese , Proteínas Serina-Treonina Quinases/metabolismo , Transativadores/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Transporte Biológico , Proteínas Morfogenéticas Ósseas/metabolismo , Cílios/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos Endogâmicos C57BL , Pericárdio/metabolismo , Ligação Proteica , Transdução de Sinais , Proteínas de Sinalização YAP
18.
Dev Biol ; 313(1): 25-34, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18005956

RESUMO

Outflow tract myocardium in the mouse heart is derived from the anterior heart field, a subdomain of the second heart field. We have recently characterized a transgene (y96-Myf5-nlacZ-16), which is expressed in the inferior wall of the outflow tract and then predominantly in myocardium at the base of the pulmonary trunk. Transgene A17-Myf5-nlacZ-T55 is expressed in the developing heart in a complementary pattern to y96-Myf5-nlacZ-16, in the superior wall of the outflow tract at E10.5 and in myocardium at the base of the aorta at E14.5. At E9.5, the two transgenes are transcribed in different subdomains of the anterior heart field. A clonal analysis of cardiomyocytes in the outflow tract, at E10.5 and E14.5, provides insight into the behaviour of myocardial cells and their progenitors. At E14.5, most clones are located at the base of either the pulmonary trunk or the aorta, indicating that these derive from distinct myocardial domains. At E10.5, clones are observed in subdomains of the outflow tract. The distribution of small clones indicates proliferative differences, whereas regionalization of large clones, that derive from an early myocardial progenitor cell, reflect coherent cell growth in the heart field as well as in the myocardium. Our results suggest that myocardial differences at the base of the great arteries are prefigured in distinct progenitor cell populations in the anterior heart field, with important implications for understanding the etiology of congenital heart defects affecting the arterial pole of the heart.


Assuntos
Coração/embriologia , Miocárdio/citologia , Animais , Aorta/embriologia , Mesoderma/citologia , Camundongos , Camundongos Transgênicos , Fator Regulador Miogênico 5/genética , Células-Tronco/citologia
19.
Dev Cell ; 6(5): 685-98, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15130493

RESUMO

When and how cells form and pattern the myocardium is a central issue for heart morphogenesis. Many genes are differentially expressed and function in subsets of myocardial cells. However, the lineage relationships between these cells remain poorly understood. To examine this, we have adopted a retrospective approach in the mouse embryo, based on the use of the laacZ reporter gene, targeted to the alpha-cardiac actin locus. This clonal analysis demonstrates the existence of two lineages that segregate early from a common precursor. The primitive left ventricle and the presumptive outflow tract are derived exclusively from a single lineage. Unexpectedly, all other regions of the heart, including the primitive atria, are colonized by both lineages. These results are not consistent with the prespecification of the cardiac tube as a segmented structure. They are discussed in the context of different heart fields and of the evolution of the heart.


Assuntos
Diferenciação Celular/genética , Linhagem da Célula/genética , Células Clonais/metabolismo , Coração/embriologia , Miocárdio/citologia , Actinas/metabolismo , Animais , Evolução Biológica , Biomarcadores , Padronização Corporal/genética , Células Clonais/citologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes Reporter , Óperon Lac , Camundongos , Células-Tronco/citologia , Células-Tronco/metabolismo
20.
J Cell Biol ; 164(1): 97-109, 2004 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-14709543

RESUMO

During heart morphogenesis, cardiac chambers arise by differential expansion of regions of the primitive cardiac tube. This process is under the control of specific transcription factors such as Tbx5 and dHAND. To gain insight into the cellular mechanisms that underlie cardiogenesis, we have used a retrospective clonal approach based on the spontaneous recombination of an nlaacZ reporter gene targeted to the murine alpha-cardiac actin locus. We show that clonal growth of myocardial cells is oriented. At embryonic day (E) 10.5, the shape of clones is characteristic of a given cardiac region and reflects its morphology. This is already detectable in the primitive cardiac tube at E8.5, and is maintained after septation at E14.5 with additional modulations. The clonal analysis reveals new subdivisions of the myocardium, including an interventricular boundary region. Our results show that the myocardium, from the time of its formation, is a polarized and regionalized tissue and point to the role of oriented clonal cell growth in cardiac chamber morphogenesis.


Assuntos
Polaridade Celular/genética , Células Clonais/metabolismo , Coração/embriologia , Miocárdio/metabolismo , Organogênese/genética , Actinas/genética , Animais , Diferenciação Celular/genética , Divisão Celular/genética , Tamanho Celular/genética , Células Clonais/citologia , Genes Reporter/genética , Coração/fisiologia , Átrios do Coração/embriologia , Ventrículos do Coração/embriologia , Óperon Lac/genética , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Organogênese/fisiologia , Função Ventricular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA