Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Cardiovasc Dev Dis ; 11(4)2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38667724

RESUMO

There is increasing evidence that some adult mitral valve pathologies may have developmental origins involving errors in cell signaling and protein deposition during valvulogenesis. While early and late gestational stages are well-documented in zebrafish, chicks, and small mammalian models, longitudinal studies in large mammals with a similar gestational period to humans are lacking. Further, the mechanism of chordae tendineae formation and multiplication remains unclear. The current study presents a comprehensive examination of mitral anterior leaflet and chordae tendineae development in a bovine model (a large mammal with the same gestational period as humans). Remarkably distinct from small mammals, bovine development displayed early branched chordae, with increasing attachments only until birth, while the anterior leaflet grew both during gestation and postnatally. Chordae also exhibited accelerated collagen deposition, maturation, and crimp development during gestation. These findings suggest that the bovine anterior leaflet and chordae tendineae possess unique processes of development despite being a continuous collagenous structure and could provide greater insight into human valve development.

2.
Dev Cell ; 59(3): 339-350.e4, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38198889

RESUMO

Congenital heart malformations include mitral valve defects, which remain largely unexplained. During embryogenesis, a restricted population of endocardial cells within the atrioventricular canal undergoes an endothelial-to-mesenchymal transition to give rise to mitral valvular cells. However, the identity and fate decisions of these progenitors as well as the behavior and distribution of their derivatives in valve leaflets remain unknown. We used single-cell RNA sequencing (scRNA-seq) of genetically labeled endocardial cells and microdissected mouse embryonic and postnatal mitral valves to characterize the developmental road. We defined the metabolic processes underlying the specification of the progenitors and their contributions to subtypes of valvular cells. Using retrospective multicolor clonal analysis, we describe specific modes of growth and behavior of endocardial cell-derived clones, which build up, in a proper manner, functional valve leaflets. Our data identify how both genetic and metabolic mechanisms specifically drive the fate of a subset of endocardial cells toward their distinct clonal contribution to the formation of the valve.


Assuntos
Desenvolvimento Embrionário , Valva Mitral , Animais , Camundongos , Valva Mitral/anormalidades , Valva Mitral/metabolismo , Estudos Retrospectivos , Diferenciação Celular
3.
J Cell Sci ; 135(17)2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35946425

RESUMO

Mitral and tricuspid valves are essential for unidirectional blood flow in the heart. They are derived from similar cell sources, and yet congenital dysplasia affecting both valves is clinically rare, suggesting the presence of differential regulatory mechanisms underlying their development. Here, we specifically inactivated Dicer1 in the endocardium during cardiogenesis and found that Dicer1 deletion caused congenital mitral valve stenosis and regurgitation, whereas it had no impact on other valves. We showed that hyperplastic mitral valves were caused by abnormal condensation and extracellular matrix (ECM) remodeling. Our single-cell RNA sequencing analysis revealed impaired maturation of mesenchymal cells and abnormal expression of ECM genes in mutant mitral valves. Furthermore, expression of a set of miRNAs that target ECM genes was significantly lower in tricuspid valves compared to mitral valves, consistent with the idea that the miRNAs are differentially required for mitral and tricuspid valve development. We thus reveal miRNA-mediated gene regulation as a novel molecular mechanism that differentially regulates mitral and tricuspid valve development, thereby enhancing our understanding of the non-association of inborn mitral and tricuspid dysplasia observed clinically.


Assuntos
MicroRNAs , Valva Tricúspide , Matriz Extracelular/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Valva Mitral , Valva Tricúspide/anormalidades
4.
Dev Dyn ; 251(3): 481-497, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34535945

RESUMO

BACKGROUND: While much is known about the genetic regulation of early valvular morphogenesis, mechanisms governing fetal valvular growth and remodeling remain unclear. Hemodynamic forces strongly influence morphogenesis, but it is unknown whether or how they interact with valvulogenic signaling programs. Side-specific activity of valvulogenic programs motivates the hypothesis that shear stress pattern-specific endocardial signaling controls the elongation of leaflets. RESULTS: We determined that extension of the semilunar valve occurs via fibrosa sided endocardial proliferation. Low OSS was necessary and sufficient to induce canonical Wnt/ß-catenin activation in fetal valve endothelium, which in turn drives BMP receptor/ligand expression, and pSmad1/5 activity essential for endocardial proliferation. In contrast, ventricularis endocardial cells expressed active Notch1 but minimal pSmad1/5. Endocardial monolayers exposed to LSS attenuate Wnt signaling in a Notch1 dependent manner. CONCLUSIONS: Low OSS is transduced by endocardial cells into canonical Wnt signaling programs that regulate BMP signaling and endocardial proliferation. In contrast, high LSS induces Notch signaling in endocardial cells, inhibiting Wnt signaling and thereby restricting growth on the ventricular surface. Our results identify a novel mechanically regulated molecular switch, whereby fluid shear stress drives the growth of valve endothelium, orchestrating the extension of the valve in the direction of blood flow.


Assuntos
Valva Aórtica , Endocárdio , Endocárdio/metabolismo , Feminino , Humanos , Morfogênese , Gravidez , Estresse Mecânico , Via de Sinalização Wnt
5.
Cell Rep ; 37(1): 109782, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34610316

RESUMO

In the zebrafish embryo, the onset of blood flow generates fluid shear stress on endocardial cells, which are specialized endothelial cells that line the interior of the heart. High levels of fluid shear stress activate both Notch and Klf2 signaling, which play crucial roles in atrioventricular valvulogenesis. However, it remains unclear why only individual endocardial cells ingress into the cardiac jelly and initiate valvulogenesis. Here, we show that lateral inhibition between endocardial cells, mediated by Notch, singles out Delta-like-4-positive endocardial cells. These cells ingress into the cardiac jelly, where they form an abluminal cell population. Delta-like-4-positive cells ingress in response to Wnt9a, which is produced in parallel through an Erk5-Klf2-Wnt9a signaling cascade also activated by blood flow. Hence, mechanical stimulation activates parallel mechanosensitive signaling pathways that produce binary effects by driving endocardial cells toward either luminal or abluminal fates. Ultimately, these cell fate decisions sculpt cardiac valve leaflets.


Assuntos
Endocárdio/metabolismo , Mecanotransdução Celular , Transdução de Sinais , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados/metabolismo , Embrião não Mamífero/metabolismo , Embrião não Mamífero/patologia , Desenvolvimento Embrionário , Endocárdio/citologia , Valvas Cardíacas/crescimento & desenvolvimento , Valvas Cardíacas/metabolismo , Valvas Cardíacas/patologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Morfolinos/metabolismo , Receptores de Neurotransmissores/antagonistas & inibidores , Receptores de Neurotransmissores/genética , Receptores de Neurotransmissores/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética
6.
Development ; 147(16)2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32843528

RESUMO

The Hippo-Yap pathway regulates multiple cellular processes in response to mechanical and other stimuli. In Drosophila, the polarity protein Lethal (2) giant larvae [L(2)gl], negatively regulates Hippo-mediated transcriptional output. However, in vertebrates, little is known about its homolog Llgl1. Here, we define a novel role for vertebrate Llgl1 in regulating Yap stability in cardiomyocytes, which impacts heart development. In contrast to the role of Drosophila L(2)gl, Llgl1 depletion in cultured rat cardiomyocytes decreased Yap protein levels and blunted target gene transcription without affecting Yap transcript abundance. Llgl1 depletion in zebrafish resulted in larger and dysmorphic cardiomyocytes, pericardial effusion, impaired blood flow and aberrant valvulogenesis. Cardiomyocyte Yap protein levels were decreased in llgl1 morphants, whereas Notch, which is regulated by hemodynamic forces and participates in valvulogenesis, was more broadly activated. Consistent with the role of Llgl1 in regulating Yap stability, cardiomyocyte-specific overexpression of Yap in Llgl1-depleted embryos ameliorated pericardial effusion and restored blood flow velocity. Altogether, our data reveal that vertebrate Llgl1 is crucial for Yap stability in cardiomyocytes and its absence impairs cardiac development.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Coração/embriologia , Miócitos Cardíacos/metabolismo , Transativadores/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Proteínas de Ciclo Celular/genética , Estabilidade Proteica , Transativadores/genética , Proteínas de Sinalização YAP , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
7.
Cell Rep ; 32(2): 107883, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32668254

RESUMO

The formation of cardiac valves depends on mechanical forces exerted by blood flow. Endocardial cells lining the interior of the heart are sensitive to these stimuli and respond by rearranging into luminal cells subjected to shear stress and abluminal cells not exposed to it. The mechanisms by which endocardial cells sense these dynamic biomechanical stimuli and how they evoke different cellular responses are largely unknown. Here, we show that blood flow activates two parallel mechanosensitive pathways, one mediated by Notch and the other by Klf2a. Both pathways negatively regulate the angiogenesis receptor Vegfr3/Flt4, which becomes restricted to abluminal endocardial cells. Its loss disrupts valve morphogenesis and results in the occurrence of Notch signaling within abluminal endocardial cells. Our work explains how antagonistic activities by Vegfr3/Flt4 on the abluminal side and by Notch on the luminal side shape cardiac valve leaflets by triggering unique differences in the fates of endocardial cells.


Assuntos
Valvas Cardíacas/embriologia , Mecanotransdução Celular , Organogênese , Receptor Notch1/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like , Camundongos Endogâmicos C57BL , Transdução de Sinais
8.
Dev Biol ; 458(1): 88-97, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31669335

RESUMO

Atrioventricular valve development requires endothelial-to-mesenchymal transition (EndMT) that induces cushion endocardial cells to give rise to mesenchymal cells crucial to valve formation. In the adult endothelium, deletion of the docking protein FRS2α induces EndMT by activating TGFß signaling in a miRNA let-7-dependent manner. To study the role of endothelial FRS2α during embryonic development, we generated mice with an inducible endothelial-specific deletion of Frs2α (FRS2αiECKO). Analysis of the FRS2αiECKO embryos uncovered a combination of impaired EndMT in AV cushions and defective maturation of AV valves leading to development of thickened, abnormal valves when Frs2α was deleted early (E7.5) in development. At the same time, no AV valve developmental abnormalities were observed after late (E10.5) deletion. These observations identify FRS2α as a pivotal controller of cell fate transition during both EndMT and post-EndMT valvulogenesis.


Assuntos
Coxins Endocárdicos/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/fisiologia , Animais , Contagem de Células , Linhagem da Célula , Comunicação Atrioventricular/embriologia , Comunicação Atrioventricular/genética , Coxins Endocárdicos/citologia , Coxins Endocárdicos/patologia , Células Endoteliais/citologia , Deleção de Genes , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Mesoderma/citologia , Mesoderma/embriologia , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/fisiologia , Valva Mitral/anormalidades , Valva Mitral/embriologia , Morfogênese/genética , Fenótipo , Valva Tricúspide/anormalidades , Valva Tricúspide/embriologia
9.
Front Cell Dev Biol ; 7: 277, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31799250

RESUMO

A variety of cardiac transcription factors/cofactors, signaling pathways, and downstream structural genes integrate to form the regulatory hierarchies to ensure proper cardiogenesis in vertebrate. Major interaction proteins of the transcription cofactor vestigial like family member 4 (VGLL4) include myocyte enhancer factor 2 (MEF2) and TEA domain transcription factors (TEAD), both of which play important roles in embryonic cardiac development and in adulthood. In this study, we identified that the deficiency of zebrafish vgll4b paralog, a unique family member expressed in developing heart, led to an impaired valve development. Mechanistically, in vgll4b mutant embryos the disruption of Vgll4b-Mef2c complex, rather than that of Vgll4b-Tead complex, resulted in an aberrant expression of krüppel-like factor 2a (klf2a) in endocardium. Such misexpression of klf2a eventually evoked the valvulogenesis defects. Our findings suggest that zebrafish Vgll4b plays an important role in modulating the transcription activity of Mef2c on klf2a during valve development in a blood-flow-independent manner.

10.
J Mol Cell Cardiol ; 133: 233-246, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31175858

RESUMO

Bicuspid aortic valve (BAV) arises during valvulogenesis when 2 leaflets/cusps of the aortic valve (AOV) are fused together. Its clinical manifestations pertain to faulty AOV function, the associated aortopathy, and other complications surveyed in Part 1 of the present bipartite-series. Part 2 examines mainly genetic and epigenetic causes of BAV and BAV-associated aortopathies (BAVAs) and disease syndromes (BAVD). Part 1 explored the heterogeneity among subsets of patients with BAV and BAVA/BAVD, and investigated abnormal fluid dynamic stress and strain patterns sustained by the cusps. Specific BAV morphologies engender systolic outflow asymmetries, associated with abnormal aortic regional wall-shear-stress distributions and the expression/localization of BAVAs. Understanding fluid dynamic factors besides the developmental mechanisms and underlying genetics governing these congenital anomalies is necessary to explain patient predisposition to aortopathy and phenotypic heterogeneity. BAV aortopathy entails complex/multifactorial pathophysiology, involving alterations in genetics, epigenetics, hemodynamics, and in cellular and molecular pathways. There is always an interdependence between organismic developmental signals and genes-no systemic signals, no gene-expression; no active gene, no next step. An apposite signal induces the expression of the next developmental gene, which needs be expressed to trigger the next signal, and so on. Hence, embryonic, then post-partum, AOV and thoracic aortic development comprise cascades of developmental genes and their regulation. Interdependencies between them arise, entailing reciprocal/cyclical mutual interactions and adaptive feedback loops, by which developmental morphogenetic processes self-correct responding to environmental inputs/reactions. This Survey can serve as a reference point and driver for further pluridisciplinary BAV/BAVD studies and their clinical translation.


Assuntos
Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/patologia , Valva Aórtica/anormalidades , Doenças das Valvas Cardíacas/genética , Doenças das Valvas Cardíacas/patologia , Animais , Valva Aórtica/patologia , Doença da Válvula Aórtica Bicúspide , Epigênese Genética , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Padrões de Herança , Camundongos , Fenótipo , Transcriptoma
11.
Dev Cell ; 48(5): 617-630.e3, 2019 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-30799229

RESUMO

During mammalian embryogenesis, de novo hematopoiesis occurs transiently in multiple anatomical sites including the yolk sac, dorsal aorta, and heart tube. A long-unanswered question is whether these local transient hematopoietic mechanisms are essential for embryonic growth. Here, we show that endocardial hematopoiesis is critical for cardiac valve remodeling as a source of tissue macrophages. Colony formation assay from explanted heart tubes and genetic lineage tracing with the endocardial specific Nfatc1-Cre mouse revealed that hemogenic endocardium is a de novo source of tissue macrophages in the endocardial cushion, the primordium of the cardiac valves. Surface marker characterization, gene expression profiling, and ex vivo phagocytosis assay revealed that the endocardially derived cardiac tissue macrophages play a phagocytic and antigen presenting role. Indeed, genetic ablation of endocardially derived macrophages caused severe valve malformation. Together, these data suggest that transient hemogenic activity in the endocardium is indispensable for the valvular tissue remodeling in the heart.


Assuntos
Endocárdio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Valvas Cardíacas/citologia , Macrófagos/metabolismo , Animais , Embrião de Mamíferos/metabolismo , Hematopoese/fisiologia , Mesoderma/metabolismo , Camundongos Transgênicos , Fatores de Transcrição NFATC/metabolismo , Saco Vitelino
12.
Curr Cardiol Rep ; 20(4): 21, 2018 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-29520694

RESUMO

PURPOSE OF REVIEW: This review aims to highlight the past and more current literature related to the multifaceted pathogenic programs that contribute to calcific aortic valve disease (CAVD) with a focus on the contribution of developmental programs. RECENT FINDINGS: Calcification of the aortic valve is an active process characterized by calcific nodule formation on the aortic surface leading to a less supple and more stiffened cusp, thereby limiting movement and causing clinical stenosis. The mechanisms underlying these pathogenic changes are largely unknown, but emerging studies have suggested that signaling pathways common to valvulogenesis and bone development play significant roles and include Transforming Growth Factor-ß (TGF-ß), bone morphogenetic protein (BMP), Wnt, Notch, and Sox9. This comprehensive review of the literature highlights the complex nature of CAVD but concurrently identifies key regulators that can be targeted in the development of mechanistic-based therapies beyond surgical intervention to improve patient outcome.


Assuntos
Estenose da Valva Aórtica/fisiopatologia , Valva Aórtica/patologia , Transdução de Sinais , Calcificação Vascular/fisiopatologia , Valva Aórtica/fisiopatologia , Estenose da Valva Aórtica/etiologia , Biologia do Desenvolvimento , Matriz Extracelular/metabolismo , Humanos , Calcificação Vascular/etiologia
13.
Dev Dyn ; 247(3): 531-541, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28884516

RESUMO

BACKGROUND: The formation of healthy heart valves throughout embryonic development is dependent on both genetic and epigenetic factors. Hemodynamic stimuli are important epigenetic regulators of valvulogenesis, but the resultant molecular pathways that control valve development are poorly understood. Here we describe how the heart and valves recover from the removal of a partial constriction (banding) of the OFT/ventricle junction (OVJ) that temporarily alters blood flow velocity through the embryonic chicken heart (HH stage 16/17). Recovery is described in terms of 24- and 48-hr gene expression, morphology, and OVJ hemodynamics. RESULTS: Collectively, these studies show that after 24 hr of recovery, important epithelial-mesenchymal transformation (EMT) genes TGFßRIII and Cadherin 11 (CDH11) transcript levels normalize return to control levels, in contrast to Periostin and TGFß,3 which remain altered. In addition, after 48 hr of recovery, TGFß3 and CDH11 transcript levels remain normalized, whereas TGFßRIII and Periostin are down-regulated. Analyses of OFT cushion volumes in the hearts show significant changes, as does the ratio of cushion to cell volume at 24 hr post band removal (PBR). Morphologically, the hearts show visible alteration following band removal when compared to their control age-matched counterparts. CONCLUSIONS: Although some aspects of the genetic/cellular profiles affected by altered hemodynamics seem to be reversed, not all gene expression and cardiac growth normalize following 48 hr of band removal. Developmental Dynamics 247:531-541, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Constrição , Valvas Cardíacas/embriologia , Coração/embriologia , Animais , Caderinas/genética , Caderinas/metabolismo , Moléculas de Adesão Celular/metabolismo , Embrião de Galinha , Expressão Gênica , Hemodinâmica , Proteoglicanas/genética , Proteoglicanas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo
14.
J Mol Cell Cardiol ; 108: 114-126, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28576718

RESUMO

Intracardiac haemodynamics is crucial for normal cardiogenesis, with recent evidence showing valvulogenesis is haemodynamically dependent and inextricably linked with shear stress. Although valve anomalies have been associated with genetic mutations, often the cause is unknown. However, altered haemodynamics have been suggested as a pathogenic contributor to bicuspid aortic valve disease. Conversely, how abnormal haemodynamics impacts mitral valve development is still poorly understood. In order to analyse altered blood flow, the outflow tract of the chick heart was constricted using a ligature to increase cardiac pressure overload. Outflow tract-banding was performed at HH21, with harvesting at crucial valve development stages (HH26, HH29 and HH35). Although normal valve morphology was found in HH26 outflow tract banded hearts, smaller and dysmorphic mitral valve primordia were seen upon altered haemodynamics in histological and stereological analysis at HH29 and HH35. A decrease in apoptosis, and aberrant expression of a shear stress responsive gene and extracellular matrix markers in the endocardial cushions were seen in the chick HH29 outflow tract banded hearts. In addition, dysregulation of extracellular matrix (ECM) proteins fibrillin-2, type III collagen and tenascin were further demonstrated in more mature primordial mitral valve leaflets at HH35, with a concomitant decrease of ECM cross-linking enzyme, transglutaminase-2. These data provide compelling evidence that normal haemodynamics are a prerequisite for normal mitral valve morphogenesis, and abnormal blood flow could be a contributing factor in mitral valve defects, with differentiation as a possible underlying mechanism.


Assuntos
Coração/embriologia , Coração/fisiologia , Hemodinâmica , Valva Mitral/embriologia , Animais , Biomarcadores , Embrião de Galinha , Matriz Extracelular , Perfilação da Expressão Gênica , Cardiopatias Congênitas/etiologia , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/patologia , Valva Mitral/anormalidades , Valva Mitral/metabolismo , Organogênese/genética , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
15.
Curr Top Dev Biol ; 124: 1-40, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28335857

RESUMO

Cardiac development is a dynamic process regulated by spatial and temporal cues that are integrated to effect molecular, cellular, and tissue-level events that form the adult heart. Disruption of these highly orchestrated events can be devastating for cardiac form and function. Aberrations in heart development result in congenital heart defects (CHDs), which affect 1 in 100 infants in the United States each year. Zebrafish have proven informative as a model organism to understand both heart development and the mechanisms associated with CHDs due to the similarities in heart morphogenesis among vertebrates, as well as their genetic tractability and amenability to live imaging. In this review, we discuss the mechanisms of zebrafish heart development and the utility of zebrafish for understanding syndromic CHDs, those cardiac abnormalities that occur in the context of multisystem disorders. We conclude with avenues of zebrafish research that will potentially inform future therapeutic approaches for the treatment of CHDs.


Assuntos
Modelos Animais de Doenças , Cardiopatias Congênitas/patologia , Peixe-Zebra/fisiologia , Animais , Coração/embriologia , Humanos , Modelos Biológicos , Síndrome
16.
J Cell Sci ; 130(7): 1321-1332, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28232522

RESUMO

Heparin-binding EGF-like growth factor (HB-EGF) plays an indispensable role in suppression of cell proliferation during mouse valvulogenesis. However, ligands of the EGF receptor (EGFR/ErbB1), including HB-EGF, are generally considered as growth-promoting factors, as shown in cancers. HB-EGF binds to and activates ErbB1 and ErbB4. We investigated the role of ErbB receptors in valvulogenesis in vivo using ErbB1- and ErbB4-deficient mice, and an ex vivo model of endocardial cushion explants. We show that HB-EGF suppresses valve mesenchymal cell proliferation through a heterodimer of ErbB1 and ErbB4, and an ErbB1 ligand (or ligands) promotes cell proliferation through a homodimer of ErbB1. Moreover, a rescue experiment with cleavable or uncleavable isoforms of ErbB4 in ERBB4-null cells indicates that the cleavable JM-A, but not the uncleavable JM-B, splice variant of ErbB4 rescues the defect of the null cells. These data suggest that the cytoplasmic intracellular domain of ErbB4, rather than the membrane-anchored tyrosine kinase, achieves this suppression. Our study demonstrates that opposing signals generated by different ErbB dimer combinations function in the same cardiac cushion mesenchymal cells for proper cardiac valve formation.


Assuntos
Receptores ErbB/metabolismo , Valvas Cardíacas/embriologia , Valvas Cardíacas/metabolismo , Mesoderma/citologia , Organogênese , Receptor ErbB-4/metabolismo , Transdução de Sinais , Alelos , Animais , Proliferação de Células , Embrião de Mamíferos/metabolismo , Genes Dominantes , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Ligantes , Camundongos Knockout , Modelos Biológicos , Mutação/genética , Domínios Proteicos , Isoformas de Proteínas/metabolismo , Receptor ErbB-4/química , Regulação para Cima
17.
Genesis ; 53(5): 337-45, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25950518

RESUMO

Heart valve development begins with the endothelial-to-mesenchymal transition (EMT) of endocardial cells. Although lineage studies have demonstrated contributions from cardiac neural crest and epicardium to semilunar and atrioventricular (AV) valve formation, respectively, most valve mesenchyme derives from the endocardial EMT. Specific Cre mouse lines for fate-mapping analyses of valve endocardial cells are limited. Msx1 displayed expression in AV canal endocardium and cushion mesenchyme between E9.5 and E11.5, when EMT is underway. Additionally, previous studies have demonstrated that deletion of Msx1 and its paralog Msx2 results in hypoplastic AV cushions and impaired endocardial signaling. A knock-in tamoxifen-inducible Cre line was recently generated (Msx1CreERT2) and characterized during embryonic development and after birth, and was shown to recapitulate the endogenous Msx1 expression pattern. Here, we further analyze this knock-in allele and track the Msx1-expressing cells and their descendants during cardiac development with a particular focus on their contribution to the valves and their precursors. Thus, Msx1CreERT2 mice represent a useful model for lineage tracing and conditional gene manipulation of endocardial and mesenchymal cushion cells essential to understand mechanisms of valve development and remodeling.


Assuntos
Alelos , Técnicas de Introdução de Genes , Valvas Cardíacas/embriologia , Valvas Cardíacas/metabolismo , Integrases/genética , Fator de Transcrição MSX1/genética , Receptores de Estrogênio/genética , Animais , Endocárdio/enzimologia , Endocárdio/metabolismo , Transição Epitelial-Mesenquimal/genética , Regulação da Expressão Gênica no Desenvolvimento , Integrases/metabolismo , Fator de Transcrição MSX1/metabolismo , Camundongos , Organogênese/genética , Receptores de Estrogênio/metabolismo
18.
Prog Mol Biol Transl Sci ; 124: 155-88, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24751430

RESUMO

The zinc-finger transcription factor KLF2 transduces the physical forces exerted by blood flow into molecular signals responsible for a wide range of biological responses. Following its initial recognition as a flow-responsive endothelial transcription factor, KLF2 is now known to be expressed in a range of cell types and to participate in a number of processes during development and disease such as endothelial homeostasis, vasoregulation, vascular growth/remodeling, and inflammation. In this review, we summarize the current understanding about KLF2 with a focus on its effects on vascular biology.


Assuntos
Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Doenças Vasculares/metabolismo , Animais , Hemorreologia , Humanos , Mecanotransdução Celular , Modelos Biológicos , Doenças Vasculares/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA