Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
Dev Dyn ; 250(10): 1432-1449, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33811421

RESUMO

BACKGROUND: Mitral valve prolapse (MVP) is a common and progressive cardiovascular disease with developmental origins. How developmental errors contribute to disease pathogenesis are not well understood. RESULTS: A multimeric complex was identified that consists of the MVP gene Dzip1, Cby1, and ß-catenin. Co-expression during valve development revealed overlap at the basal body of the primary cilia. Biochemical studies revealed a DZIP1 peptide required for stabilization of the complex and suppression of ß-catenin activities. Decoy peptides generated against this interaction motif altered nuclear vs cytosolic levels of ß-catenin with effects on transcriptional activity. A mutation within this domain was identified in a family with inherited non-syndromic MVP. This novel mutation and our previously identified DZIP1S24R variant resulted in reduced DZIP1 and CBY1 stability and increased ß-catenin activities. The ß-catenin target gene, MMP2 was up-regulated in the Dzip1S14R/+ valves and correlated with loss of collagenous ECM matrix and myxomatous phenotype. CONCLUSION: Dzip1 functions to restrain ß-catenin signaling through a CBY1 linker during cardiac development. Loss of these interactions results in increased nuclear ß-catenin/Lef1 and excess MMP2 production, which correlates with developmental and postnatal changes in ECM and generation of a myxomatous phenotype.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Transporte/metabolismo , Valvas Cardíacas/embriologia , Prolapso da Valva Mitral/metabolismo , Organogênese/fisiologia , beta Catenina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células HEK293 , Valvas Cardíacas/metabolismo , Humanos , Camundongos , Camundongos Knockout , Prolapso da Valva Mitral/genética , Fenótipo , Transdução de Sinais/fisiologia
2.
Sci Rep ; 10(1): 20094, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33208886

RESUMO

Protein kinase R-like endoplasmic reticulum kinase (PERK) is one of the endoplasmic reticulum (ER) stress sensors. PERK loss-of-function mutations are known to cause Wolcott-Rallison syndrome. This disease is characterized by early-onset diabetes mellitus, skeletal dysplasia, and cardiac valve malformation. To understand the role of PERK in valve formation in vivo, we used an endothelial-specific PERK conditional knockout mice as well as in vitro PERK inhibition assays. We used ProteoStat dyes to visualize the accumulation of misfolded proteins in the endocardial cushion and valve mesenchymal cells (VMCs). Then, VMCs were isolated from E12.5 fetal mice, by fluorescence assisted cell sorting. Proteomic analysis of PERK-deleted VMCs identified the suppression of proteins related to fatty acid oxidation (FAO), especially carnitine palmitoyltransferase II (CPT2). CPT2 is a critical regulator of endocardial-mesenchymal transformation (EndoMT); however how TGF-ß downstream signaling controls CPT2 expression remains unclear. Here, we showed that PERK inhibition suppressed, not only EndoMT but also CPT2 protein expression in human umbilical vein endothelial cells (HUVECs) under TGF-ß1 stimulation. As a result, PERK inhibition suppressed mitochondrial metabolic activity. Taken together, these results demonstrate that PERK signaling is required for cardiac valve formation via FAO and EndoMT.


Assuntos
Endocárdio/embriologia , Ácidos Graxos/química , Valvas Cardíacas/embriologia , Valvas Cardíacas/metabolismo , Mesoderma/embriologia , Organogênese , eIF-2 Quinase/fisiologia , Animais , Endocárdio/metabolismo , Ácidos Graxos/metabolismo , Feminino , Masculino , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução
3.
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
4.
Artigo em Inglês | MEDLINE | ID: mdl-31988139

RESUMO

Endocardial cells are specialized endothelial cells that, during embryogenesis, form a lining on the inside of the developing heart, which is maintained throughout life. Endocardial cells are an essential source for several lineages of the cardiovascular system including coronary endothelium, endocardial cushion mesenchyme, cardiomyocytes, mural cells, fibroblasts, liver vasculature, adipocytes, and hematopoietic cells. Alterations in the differentiation programs that give rise to these lineages has detrimental effects, including premature lethality or significant structural malformations present at birth. Here, we will review the literature pertaining to the contribution of endocardial cells to valvular, and nonvalvular lineages and highlight critical pathways required for these processes. The lineage differentiation potential of embryonic, and possibly adult, endocardial cells has therapeutic potential in the regeneration of damaged cardiac tissue or treatment of cardiovascular diseases.


Assuntos
Endocárdio/embriologia , Valvas Cardíacas/embriologia , Miocárdio/citologia , Animais , Desenvolvimento Embrionário , Coxins Endocárdicos/embriologia , Valvas Cardíacas/metabolismo , Humanos , Transdução de Sinais
5.
Mol Cell Proteomics ; 18(9): 1782-1795, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31249105

RESUMO

The endocardium is a specialized endothelium that lines the inner surface of the heart. Functional studies in mice and zebrafish have established that the endocardium is a source of instructive signals for the development of cardiac structures, including the heart valves and chambers. Here, we characterized the NOTCH-dependent endocardial secretome by manipulating NOTCH activity in mouse embryonic endocardial cells (MEEC) followed by mass spectrometry-based proteomics. We profiled different sets of soluble factors whose secretion not only responds to NOTCH activation but also shows differential ligand specificity, suggesting that ligand-specific inputs may regulate the expression of secreted proteins involved in different cardiac development processes. NOTCH signaling activation correlates with a transforming growth factor-ß2 (TGFß2)-rich secretome and the delivery of paracrine signals involved in focal adhesion and extracellular matrix (ECM) deposition and remodeling. In contrast, NOTCH inhibition is accompanied by the up-regulation of specific semaphorins that may modulate cell migration. The secretome protein expression data showed a good correlation with gene profiling of RNA expression in embryonic endocardial cells. Additional characterization by in situ hybridization in mouse embryos revealed expression of various NOTCH candidate effector genes (Tgfß2, Loxl2, Ptx3, Timp3, Fbln2, and Dcn) in heart valve endocardium and/or mesenchyme. Validating these results, mice with conditional Dll4 or Jag1 loss-of-function mutations showed gene expression alterations similar to those observed at the protein level in vitro These results provide the first description of the NOTCH-dependent endocardial secretome and validate MEEC as a tool for assaying the endocardial secretome response to a variety of stimuli and the potential use of this system for drug screening.


Assuntos
Endocárdio/embriologia , Endocárdio/metabolismo , Valvas Cardíacas/embriologia , Receptores Notch/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Benzazepinas/farmacologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Endocárdio/citologia , Endocárdio/efeitos dos fármacos , Matriz Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Valvas Cardíacas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Camundongos Mutantes , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores Notch/genética , Reprodutibilidade dos Testes
6.
Cell Death Differ ; 26(11): 2430-2446, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30824836

RESUMO

TAMM41, located within the congenital heart diseases (CHD) sensitive region of 3p25 deletion syndrome, is a mitochondrial membrane maintenance protein critical for yeast survival, but its function in higher vertebrates remains unknown. Via in vivo zebrafish model, we found that tamm41 is highly expressed in the developing heart and deficiency of which led to heart valve abnormalities. Molecular mechanistic studies revealed that TAMM41 interacts and modulates the PINK1-PARK2 dependent mitophagy pathway, thereby implicating TAMM41 in heart valve development during zebrafish embryonic cardiogenesis. Furthermore, through screening of the congenital heart diseases (CHD) sensitive region of 3p25 deletion syndrome among 118 sporadic atrioventricular septal defect (AVSD) patients, we identified three cases carrying heterozygous pathogenic intronic variants of TAMM41. All three cases lacked normal full-length TAMM41 transcripts, most likely due to specific expression of the mutant allele. Collectively, our studies highlight essential roles for TAMM41-dependent mitophagy in development of the heart and provide novel insights into the etiology of AVSD.


Assuntos
Valvas Cardíacas/embriologia , Mitocôndrias/metabolismo , Mitofagia/fisiologia , Nucleotidiltransferases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Adolescente , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Linhagem Celular , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Proteínas Mitocondriais/metabolismo , Miócitos Cardíacos/patologia , Nucleotidiltransferases/genética , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Adulto Jovem , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética
7.
Basic Res Cardiol ; 113(1): 1, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29101484

RESUMO

Tissue development and homeostasis are dependent upon the concerted synthesis, maintenance, and degradation of extracellular matrix (ECM) molecules. Cardiac fibrosis is now recognized as a primary contributor to incidence of heart failure, particularly heart failure with preserved ejection fraction, wherein cardiac filling in diastole is compromised. Periostin is a cell-associated protein involved in cell fate determination, proliferation, tumorigenesis, and inflammatory responses. As a non-structural component of the ECM, secreted 90 kDa periostin is emerging as an important matricellular factor in cardiac mesenchymal tissue development. In addition, periostin's role as a mediator in cell-matrix crosstalk has also garnered attention for its association with fibroproliferative diseases in the myocardium, and for its association with TGF-ß/BMP signaling. This review summarizes the phylogenetic history of periostin, its role in cardiac development, and the major signaling pathways influencing its expression in cardiovascular pathology. Further, we provide a synthesis of the current literature to distinguish the multiple roles of periostin in cardiac health, development and disease. As periostin may be targeted for therapeutic treatment of cardiac fibrosis, these insights may shed light on the putative timing for application of periostin-specific therapies.


Assuntos
Doenças Cardiovasculares/metabolismo , Moléculas de Adesão Celular/metabolismo , Valvas Cardíacas/embriologia , Animais , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Matriz Extracelular/metabolismo , Coração/fisiologia , Humanos , Mesoderma/metabolismo , Família Multigênica , Domínios Proteicos , Regeneração
8.
Am J Physiol Heart Circ Physiol ; 313(6): H1143-H1154, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28842437

RESUMO

During postnatal heart valve development, glycosaminoglycan (GAG)-rich valve primordia transform into stratified valve leaflets composed of GAGs, fibrillar collagen, and elastin layers accompanied by decreased cell proliferation as well as thinning and elongation. The neonatal period is characterized by the transition from a uterine environment to atmospheric O2, but the role of changing O2 levels in valve extracellular matrix (ECM) composition or morphogenesis is not well characterized. Here, we show that tissue hypoxia decreases in mouse aortic valves in the days after birth, concomitant with ECM remodeling and cell cycle arrest of valve interstitial cells. The effects of hypoxia on late embryonic valve ECM composition, Sox9 expression, and cell proliferation were examined in chicken embryo aortic valve organ cultures. Maintenance of late embryonic chicken aortic valve organ cultures in a hypoxic environment promotes GAG expression, Sox9 nuclear localization, and indicators of hyaluronan remodeling but does not affect fibrillar collagen content or cell proliferation. Chronic hypoxia also promotes GAG accumulation in murine adult heart valves in vivo. Together, these results support a role for hypoxia in maintaining a primitive GAG-rich matrix in developing heart valves before birth and also in the induction of hyaluronan remodeling in adults.NEW & NOTEWORTHY Tissue hypoxia decreases in mouse aortic valves after birth, and exposure to hypoxia promotes glycosaminoglycan accumulation in cultured chicken embryo valves and adult murine heart valves. Thus, hypoxia maintains a primitive extracellular matrix during heart valve development and promotes extracellular matrix remodeling in adult mice, as occurs in myxomatous disease.


Assuntos
Microambiente Celular , Matriz Extracelular/metabolismo , Valvas Cardíacas/metabolismo , Ácido Hialurônico/metabolismo , Oxigênio/metabolismo , Animais , Animais Recém-Nascidos , Hipóxia Celular , Proliferação de Células , Embrião de Galinha , Colágenos Fibrilares/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Valvas Cardíacas/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Organogênese , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Fatores de Tempo
9.
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
10.
Am J Physiol Heart Circ Physiol ; 311(5): H1150-H1159, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27542407

RESUMO

Cardiac neural crest cell (CNCC) ablation creates congenital heart defects (CHDs) that resemble those observed in many syndromes with craniofacial and cardiac consequences. The loss of CNCCs causes a variety of great vessel defects, including persistent truncus arteriosus and double-outlet right ventricle. However, because of the lack of quantitative volumetric measurements, less severe defects, such as great vessel size changes and valve defects, have not been assessed. Also poorly understood is the role of abnormal cardiac function in the progression of CNCC-related CHDs. CNCC ablation was previously reported to cause abnormal cardiac function in early cardiogenesis, before the CNCCs arrive in the outflow region of the heart. However, the affected functional parameters and how they correlate with the structural abnormalities were not fully characterized. In this study, using a CNCC-ablated quail model, we contribute quantitative phenotyping of CNCC ablation-related CHDs and investigate abnormal early cardiac function, which potentially contributes to late-stage CHDs. Optical coherence tomography was used to assay early- and late-stage embryos and hearts. In CNCC-ablated embryos at four-chambered heart stages, great vessel diameter and left atrioventricular valve leaflet volumes are reduced. Earlier, at cardiac looping stages, CNCC-ablated embryos exhibit abnormally twisted bodies, abnormal blood flow waveforms, increased retrograde flow percentage, and abnormal cardiac cushions. The phenotypes observed in this CNCC-ablation model were also strikingly similar to those found in an established avian fetal alcohol syndrome model, supporting the contribution of CNCC dysfunction to the development of alcohol-induced CHDs.


Assuntos
Comunicação Atrioventricular/embriologia , Coração/embriologia , Crista Neural/cirurgia , Animais , Aorta/anormalidades , Aorta/diagnóstico por imagem , Aorta/embriologia , Embrião não Mamífero , Comunicação Atrioventricular/diagnóstico por imagem , Transtornos do Espectro Alcoólico Fetal , Coração/diagnóstico por imagem , Cardiopatias Congênitas/diagnóstico por imagem , Cardiopatias Congênitas/embriologia , Valvas Cardíacas/anormalidades , Valvas Cardíacas/diagnóstico por imagem , Valvas Cardíacas/embriologia , Terapia a Laser , Crista Neural/embriologia , Tamanho do Órgão , Fenótipo , Artéria Pulmonar/anormalidades , Artéria Pulmonar/diagnóstico por imagem , Artéria Pulmonar/embriologia , Codorniz , Tomografia de Coerência Óptica
11.
Biochem Biophys Res Commun ; 477(4): 581-588, 2016 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-27343557

RESUMO

In search for novel key regulators of cardiac valve formation, we isolated the zebrafish cardiac valve mutant ping pong (png). We find that an insertional promoter mutation within the zebrafish mediator complex subunit 10 (med10) gene is leading to impaired heart valve formation. Expression of the T-box transcription factor 2b (Tbx2b), known to be essential in cardiac valve development, is severely reduced in png mutant hearts. We demonstrate here that transient reconstitution of Tbx2b expression rescues AV canal development in png mutant zebrafish. By contrast, overexpression of Forkhead box N4 (Foxn4), a known upstream regulator of Tbx2b, is not capable to reconstitute tbx2b expression and heart valve formation in Med10-deficient png mutant hearts. Interestingly, hyaluronan synthase 2 (has2), a known downstream target of Tbx2 and producer of hyaluronan (HA) - a major ECM component of the cardiac jelly and critical for proper heart valve development - is completely absent in ping pong mutant hearts. We propose here a rather unique role of Med10 in orchestrating cardiac valve formation by mediating Foxn4 dependent tbx2b transcription, expression of Has2 and subsequently proper development of the cardiac jelly.


Assuntos
Glucuronosiltransferase/metabolismo , Valvas Cardíacas/embriologia , Complexo Mediador/fisiologia , Proteínas com Domínio T/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Animais , Fatores de Transcrição Forkhead/metabolismo , Valvas Cardíacas/metabolismo , Hialuronan Sintases , Mutação , Transdução de Sinais , Proteínas com Domínio T/metabolismo , Peixe-Zebra/genética
12.
Development ; 143(3): 473-82, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26674310

RESUMO

The elucidation of mechanisms in semilunar valve development might enable the development of new therapies for congenital heart disorders. Here, we found differences in proliferation-associated genes and genes repressed by VEGF between human semilunar valve leaflets from first and second trimester hearts. The proliferation of valve interstitial cells and ventricular valve endothelial cells (VECs) and cellular density declined from the first to the second trimester. Cytoplasmic expression of NFATC1 was detected in VECs (4 weeks) and, later, cells in the leaflet/annulus junction mesenchyme expressing inactive NFATC1 (5.5-9 weeks) were detected, indicative of endocardial-to-mesenchymal transformation (EndMT) in valvulogenesis. At this leaflet/annulus junction, CD44(+) cells clustered during elongation (11 weeks), extending toward the tip along the fibrosal layer in second trimester leaflets. Differing patterns of maturation in the fibrosa and ventricularis were detected via increased fibrosal periostin content, which tracked the presence of the CD44(+) cells in the second trimester. We revealed that spatiotemporal NFATC1 expression actively regulates EndMT during human valvulogenesis, as early as 4 weeks. Additionally, CD44(+) cells play a role in leaflet maturation toward the trilaminar structure, possibly via migration of VECs undergoing EndMT, which subsequently ascend from the leaflet/annulus junction.


Assuntos
Endocárdio/embriologia , Valvas Cardíacas/citologia , Valvas Cardíacas/embriologia , Mesoderma/citologia , Mesoderma/embriologia , Moléculas de Adesão Celular/metabolismo , Contagem de Células , Diferenciação Celular , Proliferação de Células , Células Endoteliais/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Receptores de Hialuronatos/metabolismo , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Gravidez , Segundo Trimestre da Gravidez , Análise Espaço-Temporal , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
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
14.
J Appl Physiol (1985) ; 118(1): 124-31, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25359717

RESUMO

Tightly regulated cellular signaling is critical for correct heart valve development, but how and why signaling is dysregulated in congenital heart disease is not very well known. We focused on protein tyrosine phosphatase Shp2, because mutations in this signaling modulator frequently cause valve malformations associated with Noonan syndrome or Noonan syndrome with multiple lentigines (NSML). To model NSML-associated valve disease, we targeted overexpression of Q510E-Shp2 to mouse endocardial cushions (ECs) using a Tie2-Cre-based approach. At midgestation, Q510E-Shp2 expression increased the size of atrioventricular ECs by 80%. To dissect the underlying cellular mechanisms, we explanted ECs from chick embryonic hearts and induced Q510E-Shp2 expression using adenoviral vectors. Valve cell outgrowth from cultured EC explants into surrounding matrix was significantly increased by Q510E-Shp2 expression. Because focal adhesion kinase (FAK) is a critical regulator of cell migration, we tested whether FAK inhibition counteracts the Q510E-Shp2-induced effects in explanted ECs. The FAK/src inhibitor PP2 normalized valve cell outgrowth from Q510E-Shp2-expressing ECs. Next, chick ECs were further dissociated to assess cell proliferation and migration. Valve cell proliferation was not increased by Q510E-Shp2 as determined by label incorporation. In contrast, valve cell migration as reflected in a wound-healing assay was increased by Q510E-Shp2 expression, indicating that increased migration is the predominant effect of Q510E-Shp2 expression in ECs. In conclusion, PP2-sensitive signaling mediates the pathogenic effects of Q510E-Shp2 on cell migration in EC explant cultures. This suggests a central role for FAK and provides new mechanistic insight into the molecular basis of valve defects in NSML.


Assuntos
Movimento Celular/genética , Valvas Cardíacas/embriologia , Mutação , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Animais , Proliferação de Células/genética , Embrião de Galinha , Proteína-Tirosina Quinases de Adesão Focal/genética , Camundongos , Camundongos Transgênicos , Transdução de Sinais/genética
15.
J Biol Chem ; 289(12): 8545-61, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24469446

RESUMO

Periostin (PN), a novel fasciclin-related matricellular protein, has been implicated in cardiac development and postnatal remodeling, but the mechanism remains unknown. We examined the role of PN in mediating intracellular kinase activation for atrioventricular valve morphogenesis using well defined explant cultures, gene transfection systems, and Western blotting. The results show that valve progenitor (cushion) cells secrete PN into the extracellular matrix, where it can bind to INTEGRINs and activate INTEGRIN/focal adhesion kinase signaling pathways and downstream kinases, PI3K/AKT and ERK. Functional assays with prevalvular progenitor cells showed that activating these signaling pathways promoted adhesion, migration, and anti-apoptosis. Through activation of PI3K/ERK, PN directly enhanced collagen expression. Comparing PN-null to WT mice also revealed that expression of hyaluronan (HA) and activation of hyaluronan synthase-2 (Has2) are also enhanced upon PN/INTEGRIN/focal adhesion kinase-mediated activation of PI3K and/or ERK, an effect confirmed by the reduction of HA synthase-2 in PN-null mice. We also identified in valve progenitor cells a potential autocrine signaling feedback loop between PN and HA through PI3K and/or ERK. Finally, in a three-dimensional assay to simulate normal valve maturation in vitro, PN promoted collagen compaction in a kinase-dependent fashion. In summary, this study provides the first direct evidence that PN can act to stimulate a valvulogenic signaling pathway.


Assuntos
Moléculas de Adesão Celular/metabolismo , Valvas Cardíacas/embriologia , Ácido Hialurônico/metabolismo , Transdução de Sinais , Animais , Adesão Celular , Moléculas de Adesão Celular/genética , Movimento Celular , Proliferação de Células , Células Cultivadas , Embrião de Galinha , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Deleção de Genes , Valvas Cardíacas/citologia , Valvas Cardíacas/metabolismo , Integrinas/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ovinos
16.
Cells Tissues Organs ; 198(4): 300-10, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24356423

RESUMO

Proper fibroblast cell migration and differentiation are critical for valve formation and homeostasis, but uncontrolled myofibroblastic activation may precede osteogenic differentiation and calcification. Cadherin-11 (cad-11) is a cell-cell adhesion protein classically expressed at mesenchymal-osteoblast interfaces that participates in mesenchymal differentiation to osteochondral lineages. This suggests cad-11 may have an important role in heart valve development and pathogenesis, but its expression patterns in valves are largely unknown. In this study, we profiled the spatial and temporal expression patterns of cad-11 in embryonic chick and mouse heart development. We determined that cad-11 is expressed in both endocardial and mesenchymal cells of the atrioventricular and outflow tract cushions (pre-HH30/E14), but becomes restricted to the valve endocardial/endothelial cells during late fetal remodeling and throughout postnatal life. We then investigated changes in cad-11 expression in a murine aortic valve disease model (the ApoE(-/-)). Unlike wild-type mice, cad-11 becomes dramatically re-expressed in the interstitium. Similarly, in calcified human aortic valve leaflets, cad-11 loses endothelial confinement and becomes significantly re-expressed in the valve interstitium. Double labeling identified that 91% of myofibroblastic and 96% of osteoblastic cells in calcified aortic valves were also cad-11 positive. Collectively, our results suggest that cad-11 is important for proper embryonic cushion formation and remodeling, but may also participate in aortic valve pathogenesis if re-expressed in adulthood.


Assuntos
Estenose da Valva Aórtica/embriologia , Estenose da Valva Aórtica/metabolismo , Valva Aórtica/patologia , Caderinas/biossíntese , Calcinose/embriologia , Calcinose/metabolismo , Valvas Cardíacas/embriologia , Valvas Cardíacas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Animais , Valva Aórtica/embriologia , Valva Aórtica/metabolismo , Caderinas/genética , Caderinas/metabolismo , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Embrião de Galinha , Expressão Gênica , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transdução de Sinais
17.
PLoS One ; 8(10): e77611, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24204893

RESUMO

Nitric oxide synthase-3 (NOS3) has recently been shown to promote endothelial-to-mesenchymal transition (EndMT) in the developing atrioventricular (AV) canal. The present study was aimed to investigate the role of NOS3 in embryonic development of AV valves. We hypothesized that NOS3 promotes embryonic development of AV valves via EndMT. To test this hypothesis, morphological and functional analysis of AV valves were performed in wild-type (WT) and NOS3(-/-) mice at postnatal day 0. Our data show that the overall size and length of mitral and tricuspid valves were decreased in NOS3(-/-) compared with WT mice. Echocardiographic assessment showed significant regurgitation of mitral and tricuspid valves during systole in NOS3(-/-) mice. These phenotypes were all rescued by cardiac specific NOS3 overexpression. To assess EndMT, immunostaining of Snail1 was performed in the embryonic heart. Both total mesenchymal and Snail1(+) cells in the AV cushion were decreased in NOS3(-/-) compared with WT mice at E10.5 and E12.5, which was completely restored by cardiac specific NOS3 overexpression. In cultured embryonic hearts, NOS3 promoted transforming growth factor (TGFß), bone morphogenetic protein (BMP2) and Snail1expression through cGMP. Furthermore, mesenchymal cell formation and migration from cultured AV cushion explants were decreased in the NOS3(-/-) compared with WT mice. We conclude that NOS3 promotes AV valve formation during embryonic heart development and deficiency in NOS3 results in AV valve insufficiency.


Assuntos
Desenvolvimento Embrionário/fisiologia , Valvas Cardíacas/embriologia , Óxido Nítrico Sintase Tipo III/metabolismo , Animais , Proteína Morfogenética Óssea 2/metabolismo , GMP Cíclico/metabolismo , Valvas Cardíacas/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/embriologia , Camundongos Transgênicos/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo
18.
J Mol Cell Cardiol ; 65: 137-46, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24157418

RESUMO

Mature heart valves are complex structures consisting of three highly organized extracellular matrix layers primarily composed of collagens, proteoglycans and elastin. Collectively, these diverse matrix components provide all the necessary biomechanical properties for valve function throughout life. In contrast to healthy valves, myxomatous valve disease is the most common cause of mitral valve prolapse in the human population and is characterized by an abnormal abundance of proteoglycans within the valve tri-laminar structure. Despite the clinical significance, the etiology of this phenotype is not known. Scleraxis (Scx) is a basic-helix-loop-helix transcription factor that we previously showed to be required for establishing heart valve structure during remodeling stages of valvulogenesis. In this study, we report that remodeling heart valves from Scx null mice express decreased levels of proteoglycans, particularly chondroitin sulfate proteoglycans (CSPGs), while overexpression in embryonic avian valve precursor cells and adult porcine valve interstitial cells increases CSPGs. Using these systems we further identify that Scx is positively regulated by canonical Tgfß2 signaling during this process and this is attenuated by MAPK activity. Finally, we show that Scx is increased in myxomatous valves from human patients and mouse models, and overexpression in human mitral valve interstitial cells modestly increases proteoglycan expression consistent with myxomatous mitral valve phenotypes. Together, these studies identify an important role for Scx in regulating proteoglycans in embryonic and mature valve cells and suggest that imbalanced regulation could influence myxomatous pathogenesis.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Valvas Cardíacas/metabolismo , Sistema de Sinalização das MAP Quinases , Proteoglicanas/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Animais Recém-Nascidos , Galinhas , Modelos Animais de Doenças , Valvas Cardíacas/embriologia , Valvas Cardíacas/patologia , Humanos , Camundongos , Valva Mitral/embriologia , Valva Mitral/metabolismo , Valva Mitral/patologia , Células NIH 3T3 , Sus scrofa
19.
PLoS One ; 8(2): e57032, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23437302

RESUMO

Jun is a highly conserved member of the multimeric activator protein 1 transcription factor complex and plays an important role in human cancer where it is known to be critical for proliferation, cell cycle regulation, differentiation, and cell death. All of these biological functions are also crucial for embryonic development. Although all Jun null mouse embryos die at mid-gestation with persistent truncus arteriosus, a severe cardiac outflow tract defect also seen in human congenital heart disease, the developmental mechanisms are poorly understood. Here we show that murine Jun is expressed in a restricted pattern in several cell populations important for cardiovascular development, including the second heart field, pharyngeal endoderm, outflow tract and atrioventricular endocardial cushions and post-migratory neural crest derivatives. Several genes, including Isl1, molecularly mark the second heart field. Isl1 lineages include myocardium, smooth muscle, neural crest, endocardium, and endothelium. We demonstrate that conditional knockout mouse embryos lacking Jun in Isl1-expressing progenitors display ventricular septal defects, double outlet right ventricle, semilunar valve hyperplasia and aortic arch artery patterning defects. In contrast, we show that conditional deletion of Jun in Tie2-expressing endothelial and endocardial precursors does not result in aortic arch artery patterning defects or embryonic death, but does result in ventricular septal defects and a low incidence of semilunar valve defects, atrioventricular valve defects and double outlet right ventricle. Our results demonstrate that Jun is required in Isl1-expressing progenitors and, to a lesser extent, in endothelial cells and endothelial-derived endocardium for cardiovascular development but is dispensable in both cell types for embryonic survival. These data provide a cellular framework for understanding the role of Jun in the pathogenesis of congenital heart disease.


Assuntos
Sistema Cardiovascular/embriologia , Sistema Cardiovascular/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Animais , Desenvolvimento Embrionário/genética , Endocárdio/embriologia , Endocárdio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Genótipo , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/genética , Valvas Cardíacas/embriologia , Valvas Cardíacas/metabolismo , Proteínas com Homeodomínio LIM/genética , Camundongos , Camundongos Knockout , Mutação , Proteínas Proto-Oncogênicas c-jun/genética , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Fatores de Transcrição/genética , Septo Interventricular/enzimologia , Septo Interventricular/metabolismo
20.
Trends Cardiovasc Med ; 23(5): 135-42, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23295082

RESUMO

During embryonic development, cardiac valves arise at specific regions in the cardiac endothelium that swell up due to enhanced extracellular matrix production (so-called endocardial cushions). An important extracellular matrix component that is produced by the endocardial cells is the glycosaminoglycan hyaluronan. A deficiency in hyaluronan synthesis results in a failure to form endocardial cushions and a loss of their cellularization by a process called endothelial-to-mesenchymal transformation. Expression of the major hyaluronan synthase Has2 is under the influence of both positive and negative regulators. MicroRNA-dependent degradation of Has2 is required to control extracellular hyaluronan levels and thereby the size of the endocardial cushions. In this article, we review the current literature on hyaluronan synthesis during cardiac valve formation and propose that a balanced activity of both positive and negative regulators is required to maintain the critical homeostasis of hyaluronan levels in the extracellular matrix and thereby the size of the endocardial cushions. The activating and inhibitory interactions between microRNA-23, Has2, and hyaluronan are reminiscent of a reaction-diffusion system. Using a mathematical modeling approach we show that the system can produce a confined expression of hyaluronan, but only if the inhibitory signal is transferred to adjacent cells in exosomes.


Assuntos
Transição Epitelial-Mesenquimal , Valvas Cardíacas/metabolismo , Ácido Hialurônico/metabolismo , Transdução de Sinais , Animais , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Doenças das Valvas Cardíacas/metabolismo , Valvas Cardíacas/embriologia , Humanos , Hialuronan Sintases , Ácido Hialurônico/genética , MicroRNAs/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA