Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
PLoS One ; 16(10): e0259426, 2021.
Article in English | MEDLINE | ID: mdl-34714866

ABSTRACT

ERBB family members and their ligands play an essential role in embryonic heart development and adult heart physiology. Among them, ERBB3 is a binding partner of ERBB2; the ERBB2/3 complex mediates downstream signaling for cell proliferation. ERBB3 has seven consensus binding sites to the p85 regulatory subunit of PI3K, which activates the downstream AKT pathway, leading to the proliferation of various cells. This study generated a human ERBB3 knock-in mouse expressing a mutant ERBB3 whose seven YXXM p85 binding sites were replaced with YXXA. Erbb3 knock-in embryos exhibited lethality between E12.5 to E13.5, and showed a decrease in mesenchymal cell numbers and density in AV cushions. We determined that the proliferation of mesenchymal cells in the atrioventricular (AV) cushion in Erbb3 knock-in mutant embryos was temporarily reduced due to the decrease of AKT and ERK1/2 phosphorylation. Overall, our results suggest that AKT/ERK activation by the ERBB3-dependent PI3K signaling is crucial for AV cushion morphogenesis during embryonic heart development.


Subject(s)
Endocardial Cushion Defects/genetics , Endocardial Cushions/metabolism , Receptor, ErbB-3/metabolism , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Endocardial Cushion Defects/metabolism , Endocardial Cushions/embryology , Humans , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-3/chemistry , Receptor, ErbB-3/genetics , Signal Transduction
2.
Am J Physiol Heart Circ Physiol ; 321(2): H294-H305, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34142884

ABSTRACT

The etiology of ethanol-related congenital heart defects has been the focus of much study, but most research has concentrated on cellular and molecular mechanisms. We have shown with optical coherence tomography (OCT) that ethanol exposure led to increased retrograde flow and smaller atrioventricular (AV) cushions compared with controls. Since AV cushions play a role in patterning the conduction delay at the atrioventricular junction (AVJ), this study aims to investigate whether ethanol exposure alters the AVJ conduction in early looping hearts and whether this alteration is related to the decreased cushion size. Quail embryos were exposed to a single dose of ethanol at gastrulation, and Hamburger-Hamilton stage 19-20 hearts were dissected for imaging. Cardiac conduction was measured using an optical mapping microscope and we imaged the endocardial cushions using OCT. Our results showed that, compared with controls, ethanol-exposed embryos exhibited abnormally fast AVJ conduction and reduced cushion size. However, this increased conduction velocity (CV) did not strictly correlate with decreased cushion volume and thickness. By matching the CV map to the cushion-size map along the inflow heart tube, we found that the slowest conduction location was consistently at the atrial side of the AVJ, which had the thinner cushions, not at the thickest cushion location at the ventricular side as expected. Our findings reveal regional differences in the AVJ myocardium even at this early stage in heart development. These findings reveal the early steps leading to the heterogeneity and complexity of conduction at the mature AVJ, a site where arrhythmias can be initiated.NEW & NOTEWORTHY To the best of our knowledge, this is the first study investigating the impact of ethanol exposure on the early cardiac conduction system. Our results showed that ethanol-exposed embryos exhibited abnormally fast atrioventricular conduction. In addition, our findings, in CV measurements and endocardial cushion thickness, reveal regional differences in the AVJ myocardium even at this early stage in heart development, suggesting that the differentiation and maturation at this site are complex and warrant further studies.


Subject(s)
Central Nervous System Depressants/pharmacology , Endocardial Cushions/drug effects , Ethanol/pharmacology , Heart Conduction System/drug effects , Animals , Embryo, Nonmammalian , Endocardial Cushions/diagnostic imaging , Endocardial Cushions/embryology , Gastrulation , Heart/diagnostic imaging , Heart/drug effects , Heart/embryology , Heart Conduction System/diagnostic imaging , Heart Conduction System/embryology , Quail , Tomography, Optical Coherence , Voltage-Sensitive Dye Imaging
3.
Open Biol ; 11(6): 210020, 2021 06.
Article in English | MEDLINE | ID: mdl-34062094

ABSTRACT

Smoothened is a key receptor of the hedgehog pathway, but the roles of Smoothened in cardiac development remain incompletely understood. In this study, we found that the conditional knockout of Smoothened from the mesoderm impaired the development of the venous pole of the heart and resulted in hypoplasia of the atrium/inflow tract (IFT) and a low heart rate. The blockage of Smoothened led to reduced expression of genes critical for sinoatrial node (SAN) development in the IFT. In a cardiac cell culture model, we identified a Gli2-Tbx5-Hcn4 pathway that controls SAN development. In the mutant embryos, the endocardial-to-mesenchymal transition (EndMT) in the atrioventricular cushion failed, and Bmp signalling was downregulated. The addition of Bmp2 rescued the EndMT in mutant explant cultures. Furthermore, we analysed Gli2+ scRNAseq and Tbx5-/- RNAseq data and explored the potential genes downstream of hedgehog signalling in posterior second heart field derivatives. In conclusion, our study reveals that Smoothened-mediated hedgehog signalling controls posterior cardiac progenitor commitment, which suggests that the mutation of Smoothened might be involved in the aetiology of congenital heart diseases related to the cardiac conduction system and heart valves.


Subject(s)
Endocardial Cushions/embryology , Endocardial Cushions/metabolism , Hedgehog Proteins/metabolism , Organogenesis , Signal Transduction , Sinoatrial Node/embryology , Sinoatrial Node/metabolism , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Computational Biology/methods , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Ontology , Immunohistochemistry , Mice , Mice, Knockout , Mice, Transgenic , Smoothened Receptor/genetics , Smoothened Receptor/metabolism
4.
Anat Rec (Hoboken) ; 304(8): 1732-1744, 2021 08.
Article in English | MEDLINE | ID: mdl-33191650

ABSTRACT

Cushion tissues, the primordia of valves and septa of the adult heart, are formed in the atrioventricular (AV) and outflow tract (OFT) regions of the embryonic heart. The cushion tissues are generated by the endothelial-mesenchymal transition (EMT), involving many soluble factors, extracellular matrix, and transcription factors. Moreover, neural crest-derived mesenchymal cells also migrate into the OFT cushion. The transcription factor Msx1 is known to be expressed in the endothelial and mesenchymal cells during cushion tissue formation. However, its exact role in EMT during cushion tissue formation is still unknown. In this study, we investigated the expression patterns of Msx1 mRNA and protein during chick heart development. Msx1 mRNA was localized in endothelial cells of the AV region at Stage 14, and its protein was first detected at Stage 15. Thereafter, Msx1 mRNA and protein were observed in the endothelial and mesenchymal cells of the OFT and AV regions. in vitro assays showed that ectopic Msx1 expression in endothelial cells induced p27, a cell-cycle inhibitor, expression and inhibited fibroblast growth factor 4 (FGF4)-induced cell proliferation. Although the FGF signal reduced the EMT-inducing activities of transforming growth factor ß (TGFß), ectopic Msx1 expression in endothelial cells enhanced TGFß signaling-induced αSMA, an EMT marker, expression. These results suggest that Msx1 may support the transformation of endothelial cells due to a TGFß signal in EMT during cushion tissue formation.


Subject(s)
Cell Proliferation/physiology , Endocardial Cushions/embryology , Gene Expression Regulation, Developmental , Heart/embryology , MSX1 Transcription Factor/metabolism , Myocardium/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Animals , Chick Embryo , Endocardial Cushions/metabolism , MSX1 Transcription Factor/genetics , Proliferating Cell Nuclear Antigen/genetics , Signal Transduction/physiology
5.
Oxid Med Cell Longev ; 2020: 1679045, 2020.
Article in English | MEDLINE | ID: mdl-32655758

ABSTRACT

NADPH oxidases (NOX) are a major source of reactive oxygen species (ROS) production in the heart. ROS signaling regulates gene expression, cell proliferation, apoptosis, and migration. However, the role of NOX2 in embryonic heart development remains elusive. We hypothesized that deficiency of Nox2 disrupts endocardial to mesenchymal transition (EndMT) and results in congenital septal and valvular defects. Our data show that 34% of Nox2-/- neonatal mice had various congenital heart defects (CHDs) including atrial septal defects (ASD), ventricular septal defects (VSD), atrioventricular canal defects (AVCD), and malformation of atrioventricular and aortic valves. Notably, Nox2-/- embryonic hearts show abnormal development of the endocardial cushion as evidenced by decreased cell proliferation and an increased rate of apoptosis. Additionally, Nox2 deficiency disrupted EndMT of atrioventricular cushion explants ex vivo. Furthermore, treatment with N-acetylcysteine (NAC) to reduce ROS levels in the wild-type endocardial cushion explants decreased the number of cells undergoing EndMT. Importantly, deficiency of Nox2 was associated with reduced expression of Gata4, Tgfß2, Bmp2, Bmp4, and Snail1, which are critical to endocardial cushion and valvoseptal development. We conclude that NOX2 is critical to EndMT, endocardial cushion cell proliferation, and normal embryonic heart development.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Heart Defects, Congenital/pathology , Heart/embryology , NADPH Oxidase 2/metabolism , Animals , Apoptosis , Cell Proliferation , Endocardial Cushions/embryology , Endocardial Cushions/metabolism , Endocardial Cushions/pathology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Developmental , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Mice , NADPH Oxidase 2/deficiency , NADPH Oxidase 2/genetics , Reactive Oxygen Species/metabolism , Signal Transduction
6.
Article in English | MEDLINE | ID: mdl-31988139

ABSTRACT

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.


Subject(s)
Endocardium/embryology , Heart Valves/embryology , Myocardium/cytology , Animals , Embryonic Development , Endocardial Cushions/embryology , Heart Valves/metabolism , Humans , Signal Transduction
7.
Dev Biol ; 458(1): 88-97, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31669335

ABSTRACT

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.


Subject(s)
Endocardial Cushions/embryology , Gene Expression Regulation, Developmental , Membrane Proteins/physiology , Animals , Cell Count , Cell Lineage , Endocardial Cushion Defects/embryology , Endocardial Cushion Defects/genetics , Endocardial Cushions/cytology , Endocardial Cushions/pathology , Endothelial Cells/cytology , Gene Deletion , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mesoderm/cytology , Mesoderm/embryology , Mice , Mice, Inbred C57BL , MicroRNAs/physiology , Mitral Valve/abnormalities , Mitral Valve/embryology , Morphogenesis/genetics , Phenotype , Tricuspid Valve/abnormalities , Tricuspid Valve/embryology
8.
Curr Top Dev Biol ; 132: 395-416, 2019.
Article in English | MEDLINE | ID: mdl-30797515

ABSTRACT

Heart formation involves a complex series of tissue rearrangements, during which regions of the developing organ expand, bend, converge, and protrude in order to create the specific shapes of important cardiac components. Much of this morphogenesis takes place while cardiac function is underway, with blood flowing through the rapidly contracting chambers. Fluid forces are therefore likely to influence the regulation of cardiac morphogenesis, but it is not yet clear how these biomechanical cues direct specific cellular behaviors. In recent years, the optical accessibility and genetic amenability of zebrafish embryos have facilitated unique opportunities to integrate the analysis of flow parameters with the molecular and cellular dynamics underlying cardiogenesis. Consequently, we are making progress toward a comprehensive view of the biomechanical regulation of cardiac chamber emergence, atrioventricular canal differentiation, and ventricular trabeculation. In this review, we highlight a series of studies in zebrafish that have provided new insight into how cardiac function can shape cardiac morphology, with a particular focus on how hemodynamics can impact cardiac cell behavior. Over the long-term, this knowledge will undoubtedly guide our consideration of the potential causes of congenital heart disease.


Subject(s)
Body Fluids/physiology , Heart/embryology , Heart/physiology , Morphogenesis , Zebrafish/embryology , Animals , Biomechanical Phenomena , Cell Differentiation/genetics , Endocardial Cushions/cytology , Endocardial Cushions/embryology , Endocardial Cushions/metabolism , Endocardium/cytology , Endocardium/embryology , Endocardium/metabolism , Gene Expression Regulation, Developmental , Heart/anatomy & histology , Zebrafish/genetics
9.
J Mol Cell Cardiol ; 123: 150-158, 2018 10.
Article in English | MEDLINE | ID: mdl-30201295

ABSTRACT

Abnormal endocardial cushion formation is a major cause of congenital heart valve disease, which is a common birth defect with significant morbidity and mortality. Although ß-catenin and BMP2 are two well-known regulators of endocardial cushion formation, their interaction in this process is largely unknown. Here, we report that deletion of ß-catenin in myocardium results in formation of hypoplastic endocardial cushions accompanying a decrease of mesenchymal cell proliferation. Loss of ß-catenin reduced Bmp2 expression in myocardium and SMAD signaling in cushion mesenchyme. Exogenous BMP2 recombinant proteins fully rescued the proliferation defect of mesenchymal cells in cultured heart explants from myocardial ß-catenin knockout embryos. Using a canonical WNT signaling reporter mouse line, we showed that cushion myocardium exhibited high WNT/ß-catenin activities during endocardial cushion growth. Selective disruption of the signaling function of ß-catenin resulted in a cushion growth defect similar to that caused by the complete loss of ß-catenin. Together, these observations demonstrate that myocardial ß-catenin signaling function promotes mesenchymal cell proliferation and endocardial cushion expansion through inducing BMP signaling.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Endocardial Cushions/metabolism , Myocardium/metabolism , Organogenesis , Signal Transduction , beta Catenin/metabolism , Animals , Cell Proliferation , Endocardial Cushions/embryology , Endocardium/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Models, Biological , Paracrine Communication , Rats , Wnt Signaling Pathway
10.
Dis Model Mech ; 11(9)2018 08 28.
Article in English | MEDLINE | ID: mdl-30061196

ABSTRACT

Deletions of chromosome 1p36 are associated with a high incidence of congenital heart defects (CHDs). The arginine-glutamic acid dipeptide repeats gene (RERE) is located in a critical region for CHD on chromosome 1p36 and encodes a cardiac-expressed nuclear receptor co-regulator. Mutations affecting RERE cause atrial and ventricular septal defects (VSDs) in humans, and RERE-deficient mice also develop VSDs. During cardiac development, mesenchymal cells destined to form part of the atrioventricular (AV) septum are generated when endocardial cells in the AV canal undergo epithelial-to-mesenchymal transition (EMT) and migrate into the space between the endocardium and the myocardium. These newly generated mesenchymal cells then proliferate to fill the developing AV endocardial cushions. Here, we demonstrate that RERE-deficient mouse embryos have reduced numbers of mesenchymal cells in their AV endocardial cushions owing to decreased levels of EMT and mesenchymal cell proliferation. In the endocardium, RERE colocalizes with GATA4, a transcription factor required for normal levels of EMT and mesenchymal cell proliferation. Using a combination of in vivo and in vitro studies, we show that Rere and Gata4 interact genetically in the development of CHDs, RERE positively regulates transcription from the Gata4 promoter and GATA4 levels are reduced in the AV canals of RERE-deficient embryos. Tissue-specific ablation of Rere in the endocardium leads to hypocellularity of the AV endocardial cushions, defective EMT and VSDs, but does not result in decreased GATA4 expression. We conclude that RERE functions in the AV canal to positively regulate the expression of GATA4, and that deficiency of RERE leads to the development of VSDs through its effects on EMT and mesenchymal cell proliferation. However, the cell-autonomous role of RERE in promoting EMT in the endocardium must be mediated by its effects on the expression of proteins other than GATA4.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Carrier Proteins/metabolism , GATA4 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Heart Septal Defects, Ventricular/embryology , Heart Septal Defects, Ventricular/genetics , Nerve Tissue Proteins/deficiency , Repressor Proteins/deficiency , Alleles , Animals , Cell Proliferation , Embryo, Mammalian/metabolism , Endocardial Cushions/embryology , Endocardial Cushions/metabolism , Endocardial Cushions/pathology , Endocardium/embryology , Endocardium/metabolism , Endocardium/pathology , Epithelial-Mesenchymal Transition/genetics , GATA4 Transcription Factor/metabolism , Mesoderm/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Nerve Tissue Proteins/genetics , Repressor Proteins/genetics
11.
J Cell Physiol ; 233(3): 1887-1894, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28513838

ABSTRACT

Nephronectin (NPNT), a highly conserved extracellular matrix protein, plays an important role in regulating cell adhesion, differentiation, spreading, and survival. NPNT protein belongs to the epidermal growth factor (EGF)-like superfamily and exhibits several common structural determinants; including EGF-like repeat domains, MAM domain (Meprin, A5 Protein, and Receptor Protein-Tyrosine Phosphatase µ), RGD motif (Arg-Gly-Asp) and a coiled-coil domain. It regulates integrins-mediated signaling pathways via the interaction of its RGD motif with integrin α8ß1. Recent studies revealed that NPNT is involved in kidney development, renal injury repair, atrioventricular canal differentiation, pulmonary function, and muscle cell niche maintenance. Moreover, NPNT regulates osteoblast differentiation and mineralization, as well as osteogenic angiogenesis. Altered expression of NPNT has been linked with the progression of certain types of cancers, such as spontaneous breast tumor metastasis and malignant melanoma. Interestingly, NPNT gene expression can be regulated by a range of external factors such as tumor necrosis factor alpha (TNF-α), transforming growth factor beta (TGF-ß), oncostatin M (OSM), bone morphogenic protein 2 (BMP2), Wnt3a, Vitamin D3 , and microRNA-378 (miR378). Further understanding the cellular and molecular mechanisms by which NPNT regulates tissue homeostasis in an organ-specific manner is critical in exploring NPNT as a therapeutic target for tissue regeneration and tissue engineering.


Subject(s)
Bone and Bones/blood supply , Extracellular Matrix Proteins/metabolism , Kidney/embryology , Neoplasms/pathology , Neovascularization, Physiologic/physiology , Osteogenesis/physiology , Animals , Cell Adhesion/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Endocardial Cushions/cytology , Endocardial Cushions/embryology , Homeostasis/physiology , Humans , Kidney/cytology , Mice , Signal Transduction/physiology
12.
Development ; 144(23): 4322-4327, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29183943

ABSTRACT

Myocardial contractility and blood flow provide essential mechanical cues for the morphogenesis of the heart. In general, endothelial cells change their migratory behavior in response to shear stress patterns, according to flow directionality. Here, we assessed the impact of shear stress patterns and flow directionality on the behavior of endocardial cells, the specialized endothelial cells of the heart. At the early stages of zebrafish heart valve formation, we show that endocardial cells are converging to the valve-forming area and that this behavior depends upon mechanical forces. Quantitative live imaging and mathematical modeling allow us to correlate this tissue convergence with the underlying flow forces. We predict that tissue convergence is associated with the direction of the mean wall shear stress and of the gradient of harmonic phase-averaged shear stresses, which surprisingly do not match the overall direction of the flow. This contrasts with the usual role of flow directionality in vascular development and suggests that the full spatial and temporal complexity of the wall shear stress should be taken into account when studying endothelial cell responses to flow in vivo.


Subject(s)
Heart/embryology , Models, Cardiovascular , Zebrafish/embryology , Animals , Anisotropy , Biomechanical Phenomena , Endocardial Cushions/cytology , Endocardial Cushions/embryology , Endothelial Cells/cytology , Endothelial Cells/physiology , Erythrocytes/physiology , Hemodynamics , Hydrodynamics , Imaging, Three-Dimensional , Organogenesis/physiology , Shear Strength , Stress, Mechanical
13.
Cell Rep ; 19(8): 1602-1613, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28538179

ABSTRACT

The HAND2 transcriptional regulator controls cardiac development, and we uncover additional essential functions in the endothelial to mesenchymal transition (EMT) underlying cardiac cushion development in the atrioventricular canal (AVC). In Hand2-deficient mouse embryos, the EMT underlying AVC cardiac cushion formation is disrupted, and we combined ChIP-seq of embryonic hearts with transcriptome analysis of wild-type and mutants AVCs to identify the functionally relevant HAND2 target genes. The HAND2 target gene regulatory network (GRN) includes most genes with known functions in EMT processes and AVC cardiac cushion formation. One of these is Snai1, an EMT master regulator whose expression is lost from Hand2-deficient AVCs. Re-expression of Snai1 in mutant AVC explants partially restores this EMT and mesenchymal cell migration. Furthermore, the HAND2-interacting enhancers in the Snai1 genomic landscape are active in embryonic hearts and other Snai1-expressing tissues. These results show that HAND2 directly regulates the molecular cascades initiating AVC cardiac valve development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Endocardial Cushions/embryology , Endocardial Cushions/metabolism , Gene Regulatory Networks , Heart Valves/embryology , Heart Valves/metabolism , Animals , Base Sequence , Cell Movement/genetics , Chromatin/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Developmental , Genome , Mesoderm/cytology , Mesoderm/metabolism , Mice , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Transcription, Genetic
14.
Zhonghua Xin Xue Guan Bing Za Zhi ; 43(5): 437-41, 2015 May.
Article in Chinese | MEDLINE | ID: mdl-26419991

ABSTRACT

OBJECTIVE: To investigate the role of GATA4 gene in the endocardial cushions development. METHODS: Target gene eukaryote expression vectors were constructed by pcDNA3.1(-) vector plasmid, and were identified by DNA sequence analysis. Recombinant plasmids were transfected into Hela cells with lipofectamine 2000, meanwhile Hela cells transfected with empty vector or those without transfection served as transfection control group and blank control group, respectively. Real-time PCR and Western blot were performed to detect the relative expression of mRNA and protein of transcription factors GATA4, Sox9, Scleraxis and ECM proteins Aggrecan, Tenascin in each group. RESULTS: The relative mRNA expression of GATA4 in experimental group was significantly higher than in transfection control group and blank control group. GATA4 mRNA expression in Hela(GATA4), Hela(H436Y), Hela(Null) and Hela group was 310.83 ± 2.39, 146.35 ± 1.74, 0.94 ± 0.32, 1.00 ± 0.28, respectively (F = 72.508, P < 0.05). Western blot results were consistent with the results obtained by qRT-PCR. The relative mRNA and protein expressions of Sox9, Scleraxis, Aggrecan and Tenascin in both experimental groups were significantly higher than that in transfection control group and blank control group (P < 0.05), and above gene expressions were significantly downregulated in GATA4(H436Y) group, while they were similar between transfection control group and blank control group (all P > 0.05). CONCLUSIONS: GATA4 H436Y mutation reduces it's transcriptional activation, which might serve as a theoretical framework to demonstrate the roles of GATA4 gene in endocardial cushion development.


Subject(s)
Endocardial Cushions/embryology , GATA4 Transcription Factor/metabolism , Aggrecans/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Down-Regulation , GATA4 Transcription Factor/genetics , Gene Expression , Genetic Vectors , HeLa Cells , Humans , RNA, Messenger , SOX9 Transcription Factor/metabolism , Tenascin/metabolism , Transfection
15.
BMC Dev Biol ; 15: 36, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26472242

ABSTRACT

BACKGROUND: Development of the valves and septa of the heart depends on the formation and remodeling of the endocardial cushions in the atrioventricular canal and outflow tract. These cushions are populated by mesenchyme produced from the endocardium by epithelial-mesenchymal transition (EMT). The endocardial cushions are remodeled into the valves at post-EMT stages via differentiation of the mesenchyme and changes in the extracellular matrix (ECM). Transforming growth factor ß (TGFß) signaling has been implicated in both the induction of EMT in the endocardial cushions and the remodeling of the valves at post-EMT stages. We previously identified the RNA binding protein muscleblind-like 1 (MBNL1) as a negative regulator of TGFß signaling and EMT in chicken endocardial cushions ex vivo. Here, we investigate the role of MBNL1 in endocardial cushion development and valvulogenesis in Mbnl1(∆E3/∆E3) mice, which are null for MBNL1 protein. METHODS: Collagen gel invasion assays, histology, immunohistochemistry, real-time RT-PCR, optical coherence tomography, and echocardiography were used to evaluate EMT and TGFß signaling in the endocardial cushions, and morphogenesis, ECM composition, and function of the heart valves. RESULTS: As in chicken, the loss of MBNL1 promotes precocious TGFß signaling and EMT in the endocardial cushions. Surprisingly, this does not lead to the production of excess mesenchyme, but later valve morphogenesis is aberrant. Adult Mbnl1(∆E3/∆E3) mice exhibit valve dysmorphia with elevated TGFß signaling, changes in ECM composition, and increased pigmentation. This is accompanied by a high incidence of regurgitation across both inflow and outflow valves. Mbnl1(∆E3/∆E3) mice also have a high incidence of ostium secundum septal defects accompanied by atrial communication, but do not develop overt cardiomyopathy. CONCLUSIONS: Together, these data indicate that MBNL1 plays a conserved role in negatively regulating TGFß signaling, and is required for normal valve morphogenesis and homeostasis in vivo.


Subject(s)
DNA-Binding Proteins/metabolism , Endocardial Cushions/embryology , Heart Valves/embryology , Organogenesis , RNA-Binding Proteins/metabolism , Signal Transduction , Animals , DNA-Binding Proteins/genetics , Endocardial Cushions/metabolism , Epithelial-Mesenchymal Transition , Heart/embryology , Heart Valves/cytology , Heart Valves/metabolism , Mice , RNA-Binding Proteins/genetics , Transforming Growth Factor beta/metabolism
16.
Dev Biol ; 407(1): 145-57, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26188246

ABSTRACT

Proper remodeling of the endocardial cushions into thin fibrous valves is essential for gestational progression and long-term function. This process involves dynamic interactions between resident cells and their local environment, much of which is not understood. In this study, we show that deficiency of the cell-cell adhesion protein cadherin-11 (Cad-11) results in significant embryonic and perinatal lethality primarily due to valve related cardiac dysfunction. While endocardial to mesenchymal transformation is not abrogated, mesenchymal cells do not homogeneously cellularize the cushions. These cushions remain thickened with disorganized ECM, resulting in pronounced aortic valve insufficiency. Mice that survive to adulthood maintain thickened and stenotic semilunar valves, but interestingly do not develop calcification. Cad-11 (-/-) aortic valve leaflets contained reduced Sox9 activity, ß1 integrin expression, and RhoA-GTP activity, suggesting that remodeling defects are due to improper migration and/or cellular contraction. Cad-11 deletion or siRNA knockdown reduced migration, eliminated collective migration, and impaired 3D matrix compaction by aortic valve interstitial cells (VIC). Cad-11 depleted cells in culture contained few filopodia, stress fibers, or contact inhibited locomotion. Transfection of Cad-11 depleted cells with constitutively active RhoA restored cell phenotypes. Together, these results identify cadherin-11 mediated adhesive signaling for proper remodeling of the embryonic semilunar valves.


Subject(s)
Aortic Valve/embryology , Cadherins/physiology , Cell Movement , Extracellular Matrix/metabolism , Animals , Aortic Valve/cytology , Cell Polarity , Chickens , Endocardial Cushions/embryology , Mice , Mice, Inbred C57BL , Morphogenesis , Swine , rhoA GTP-Binding Protein/physiology
17.
Int J Dev Biol ; 59(4-6): 247-54, 2015.
Article in English | MEDLINE | ID: mdl-25896281

ABSTRACT

The Pitx2 gene is involved in the establishment of vertebrate left-right axis with an important role in subsequent heart organogenesis. Mutations in the Pitx2 gene have been associated with Axenfeld-Rieger syndrome, which is characterized by ocular, craniofacial, and umbilical anomalies, as well as cardiac defects. In addition, recent data have unravelled a molecular link between PITX2 loss of function and atrial fibrillation (AF), supporting an important role of Pitx2 not only in development but also in heart homeostasis. Three PITX2 isoforms have been described in mice: PITX2A, PITX2B, and PITX2C. During heart organogenesis, PITX2C seems to play a determinant role in left-right signalling from early somitogenesis onwards. However the participation of the PITX2A and/or PITX2B isoforms during cardiogenesis is controversial. Here we report for the first time that the Pitx2a and Pitx2b isoforms are jointly expressed with the Pitx2c isoform during heart development. Interestingly, in terms of relative quantification of mRNA, the Pitx2b and Pitx2c isoforms display similar expression profiles during cardiogenesis, decreasing with further development but maintaining their expression until adult stages. Moreover, a detailed analysis of PITX2B protein during cardiac development shows that PITX2B is dynamically expressed in the developing ventricular septum and asymmetrically expressed in the tricuspid valve primordia, suggesting a putative role of the PITX2B isoform during ventricular septation as well as in the maturation of the right portion of the atrioventricular canal.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Myocardium/metabolism , Transcription Factors/genetics , Animals , Body Patterning/genetics , Endocardial Cushions/embryology , Endocardial Cushions/growth & development , Endocardial Cushions/metabolism , Female , Heart/embryology , Heart/growth & development , Homeodomain Proteins/metabolism , Immunohistochemistry , Mice, Inbred BALB C , Organogenesis/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Homeobox Protein PITX2
18.
Birth Defects Res B Dev Reprod Toxicol ; 101(5): 364-70, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25231192

ABSTRACT

BACKGROUND: Diabetes mellitus in pregnancy causes defects in infant heart, including the outflow tracts (OFTs). Development of the aorta and pulmonary artery, which are derived from the common OFT in the embryo, is regulated by the transforming growth factor ß (TGFß) and Wnt families, and can be perturbed by hyperglycemia-generated intracellular stress conditions. However, the underlying cellular and molecular mechanisms remain to be delineated. METHODS: Female mice were induced diabetic with streptozotocin. Embryonic and fetal OFTs were examined morphologically and histologically. Cell proliferation was assessed using 5'-bromo-2'-deoxyuridine incorporation assay. Oxidative and endoplasmic reticulum (ER) stress markers and TGFß factors were detected using immunohistochemistry. The expression of genes in the Wnt-signaling system was assessed using real-time reverse transcription polymerase chain reaction array. The role of activin-A in cell proliferation was addressed by treating embryos cultured in high glucose with activin-A. RESULTS: Maternal diabetes caused complex abnormalities in the OFTs, including aortic and pulmonary stenosis and persistent truncus arteriosus. The development of the endocardial cushions was suppressed, manifested with insufficient cellularization of the tissues. Cell proliferation was significantly decreased under oxidative and ER stress conditions. The expression of genes in the Wnt signaling was significantly altered. Activin-A and Smad3 were found to be expressed in the OFT. Treatment with activin-A rescued cell proliferation in the endocardial cushions. CONCLUSIONS: Maternal diabetes generates oxidative and ER stress conditions, suppresses TGFß and Wnt signaling, inhibits cell proliferation and cellularization of the endocardial cushions, leading to OFT septal defects. Activin-A plays a role in hyperglycemia-suppressed proliferation of the endocardial cells.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes, Gestational/pathology , Heart Defects, Congenital/pathology , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism , Activins/biosynthesis , Activins/pharmacology , Animals , Aorta/embryology , Aortic Valve Stenosis/pathology , Cardiac Output/physiology , Cell Proliferation , Diabetes Mellitus, Experimental/chemically induced , Embryo Culture Techniques , Embryo, Mammalian/metabolism , Endocardial Cushions/embryology , Endoplasmic Reticulum Stress , Female , Gene Expression Regulation, Developmental , Glucose/pharmacology , Hyperglycemia/metabolism , Mice , Mice, Inbred C57BL , Neural Crest/embryology , Oxidative Stress , Pregnancy , Pulmonary Artery/embryology , Pulmonary Valve Stenosis/pathology , Smad3 Protein/biosynthesis , Streptozocin , Truncus Arteriosus, Persistent/pathology , Wnt Signaling Pathway/genetics
19.
Cardiovasc Res ; 104(1): 49-60, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25100766

ABSTRACT

AIMS: Valvular heart disease is responsible for considerable morbidity and mortality. Cardiac valves develop as the heart contracts, and they function throughout the lifetime of the organism to prevent retrograde blood flow. Their precise morphogenesis is crucial for cardiac function. Zebrafish is an ideal model to investigate cardiac valve development as it allows these studies to be carried out in vivo through non-invasive imaging. Accumulating evidence suggests a role for contractility and intracardiac flow dynamics in cardiac valve development. However, these two factors have proved difficult to uncouple, especially since altering myocardial function affects the intracardiac flow pattern. METHODS AND RESULTS: Here, we describe novel zebrafish models of developmental valve defects. We identified two mutant alleles of myosin heavy chain 6 that can be raised to adulthood despite having only one functional chamber-the ventricle. The adult mutant ventricle undergoes remodelling, and the atrioventricular (AV) valves fail to form four cuspids. In parallel, we characterized a novel mutant allele of southpaw, a nodal-related gene involved in the establishment of left-right asymmetry, which exhibits randomized heart and endoderm positioning. We first observed that in southpaw mutants the relative position of the two cardiac chambers is altered, affecting the geometry of the heart, while myocardial function appears unaffected. Mutant hearts that loop properly or exhibit situs inversus develop normally, whereas midline, unlooped hearts exhibit defects in their transvalvular flow pattern during AV valve development as well as defects in valve morphogenesis. CONCLUSION: Our data indicate that intracardiac flow dynamics regulate valve morphogenesis independently of myocardial contractility.


Subject(s)
Coronary Circulation , Endocardial Cushion Defects/embryology , Endocardial Cushions/embryology , Heart Valves/abnormalities , Hemodynamics , Mechanotransduction, Cellular , Animals , Animals, Genetically Modified , Atrial Function , Endocardial Cushion Defects/genetics , Endocardial Cushion Defects/metabolism , Endocardial Cushion Defects/physiopathology , Endocardial Cushions/metabolism , Endocardial Cushions/physiopathology , Genotype , Heart Valves/metabolism , Heart Valves/physiopathology , Morphogenesis , Mutation , Myocardial Contraction , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Phenotype , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
20.
Mol Cells ; 37(5): 406-11, 2014 May.
Article in English | MEDLINE | ID: mdl-24823359

ABSTRACT

The initial step of atrioventricular (AV) valve development involves the deposition of extracellular matrix (ECM) components of the endocardial cushion and the endocardial-mesenchymal transition. While the appropriately regulated expression of the major ECM components, Versican and Hyaluronan, that form the endocardial cushion is important for heart valve development, the underlying mechanism that regulates ECM gene expression remains unclear. We found that zebrafish crip2 expression is restricted to a subset of cells in the AV canal (AVC) endocardium at 55 hours post-fertilization (hpf). Knockdown of crip2 induced a heart-looping defect in zebrafish embryos, although the development of cardiac chambers appeared to be normal. In the AVC of Crip2-deficient embryos, the expression of both versican a and hyaluronan synthase 2 (has2) was highly upregulated, but the expression of bone morphogenetic protein 4 (bmp4) and T-box 2b (tbx2b) in the myocardium and of notch1b in the endocardium in the AVC did not change. Taken together, these results indicate that crip2 plays an important role in AV valve development by downregulating the expression of ECM components in the endocardial cushion.


Subject(s)
Endocardial Cushions/metabolism , Endocardium/embryology , Extracellular Matrix Proteins/genetics , Heart Valves/embryology , LIM Domain Proteins/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Animals , Down-Regulation , Endocardial Cushions/embryology , Endocardium/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Developmental
SELECTION OF CITATIONS
SEARCH DETAIL
...