Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Toxicol Sci ; 148(2): 409-20, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26354774

ABSTRACT

TGFß2 (transforming growth factor-ß2) is a key growth factor regulating epithelial to mesenchymal transition (EMT). TGFß2 triggers cardiac progenitor cells to differentiate into mesenchymal cells and give rise to the cellular components of coronary vessels as well as cells of aortic and pulmonary valves. TGFß signaling is dependent on a dynamic on and off switch in Smad activity. Arsenite exposure of 1.34 µM for 24-48 h has been reported to disrupt Smad phosphorylation leading to deficits in TGFß2-mediated cardiac precursor differentiation and transformation. In this study, the molecular mechanism of acute arsenite toxicity on TGFß2-induced Smad2/3 nuclear shuttling and TGFß2-mediated cardiac EMT was investigated. A 4-h exposure to 5 µM arsenite blocks nuclear accumulation of Smad2/3 in response to TGFß2 without disrupting Smad phosphorylation or nuclear importation. The depletion of nuclear Smad is restored by knocking-down Smad-specific exportins, suggesting that arsenite augments Smad2/3 nuclear exportation. The blockage in TGFß2-Smad signaling is likely due to the loss of Zn(2+) cofactor in Smad proteins, as Zn(2+) supplementation reverses the disruption in Smad2/3 nuclear translocation and transcriptional activity by arsenite. This coincides with Zn(2+) supplementation rescuing arsenite-mediated deficits in cardiac EMT. Thus, zinc partially protects cardiac EMT from developmental toxicity by arsenite.


Subject(s)
Arsenites/toxicity , Cell Differentiation/drug effects , Myocytes, Cardiac/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Stem Cells/drug effects , Transforming Growth Factor beta2/pharmacology , Zinc/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Epithelial-Mesenchymal Transition/drug effects , HEK293 Cells , Humans , Karyopherins/genetics , Karyopherins/metabolism , Mice, Transgenic , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation , RNA Interference , Signal Transduction/drug effects , Stem Cells/metabolism , Stem Cells/pathology , Time Factors , Transcription, Genetic/drug effects , Transfection
2.
Mol Cancer Res ; 10(12): 1544-54, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23193156

ABSTRACT

The transmembrane mucin MUC1 is overexpressed in most ductal carcinomas, and its overexpression is frequently associated with metastatic progression. MUC1 can drive tumor initiation and progression via interactions with many oncogenic partners, including ß-catenin, the EGF receptor (EGFR) and Src. The decoy peptide protein transduction domain MUC1 inhibitory peptide (PMIP) has been shown to inhibit the tumor promoting activities of MUC1 in breast and lung cancer, including cell growth and invasion, and its usage suppresses metastatic progression in mouse models of breast cancer. To further characterize the reduced metastasis observed upon PMIP treatment, we conducted motility assays and observed that PMIP inhibits cell motility of breast cancer cells. To determine the mechanism by which PMIP inhibits motility, we evaluated changes in global gene transcription upon PMIP treatment, and identified a number of genes with altered expression in response to PMIP. Among these genes is the metastatic mediator, c-Met, a transmembrane tyrosine kinase that can promote cell scattering, migration, and invasion. To further investigate the role of c-Met in MUC1-dependent metastatic events, we evaluated the effects of MUC1 expression and EGFR activation on breast cancer cell scattering, branching, and migration. We found that MUC1 strongly promoted all of these events and this effect was further amplified by EGF treatment. Importantly, the effect of MUC1 and EGF on these phenotypes was dependent upon c-Met activity. Overall, these results indicate that PMIP can block the expression of a key metastatic mediator, further advancing its potential use as a clinical therapeutic.


Subject(s)
Breast Neoplasms/pathology , Cell Movement/genetics , Mucin-1/genetics , Mucin-1/metabolism , Peptides/pharmacology , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Disease Progression , Down-Regulation/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Transcription, Genetic/drug effects
3.
Toxicol Sci ; 116(1): 273-85, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20308225

ABSTRACT

Arsenic is a naturally occurring metalloid and environmental contaminant. Arsenic exposure in drinking water is reported to cause cancer of the liver, kidneys, lung, bladder, and skin as well as birth defects, including neural tube, facial, and vasculogenic defects. The early embryonic period most sensitive to arsenic includes a variety of cellular processes. One key cellular process is epithelial-mesenchymal transition (EMT) where epithelial sheets develop into three-dimensional structures. An embryonic prototype of EMT is found in the atrioventricular (AV) canal of the developing heart, where endothelia differentiate to form heart valves. Effects of arsenic on this cellular process were examined by collagen gel invasion assay (EMT assay) using explanted AV canals from chicken embryo hearts. AV canals treated with 12.5-500 ppb arsenic showed a loss of mesenchyme at 12.5 ppb, and mesenchyme formation was completely inhibited at 500 ppb. Altered gene expression in arsenic-treated explants was investigated by microarray analysis. Genes whose expression was altered consistently at exposure levels of 10, 25, and 100 ppb were identified, and results showed that 25 ppb in vitro was particularly effective. Three hundred and eighty two genes were significantly altered at this exposure level. Cytoscape analysis of the microarray data using the chicken interactome identified four clusters of altered genes based on published relationships and pathways. This analysis identified cytoskeleton and cell adhesion-related genes whose disruption is consistent with an altered ability to undergo EMT. These studies show that EMT is sensitive to arsenic and that an interactome-based approach can be useful in identifying targets.


Subject(s)
Arsenic/toxicity , Collagen , Epithelial Cells/drug effects , Gene Expression/drug effects , Mesoderm/drug effects , Animals , Base Sequence , Chick Embryo , DNA Primers , In Situ Nick-End Labeling , Mesoderm/cytology
4.
Circ Res ; 103(12): 1430-40, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-19008476

ABSTRACT

Congenital heart defects occur at a rate of 5% and are the most prevalent birth defects. A better understanding of the complex signaling networks regulating heart development is necessary to improve repair strategies for congenital heart defects. The mitogen-activated protein 3 kinase (MEKK3) is important to early embryogenesis, but developmental processes affected by MEKK3 during heart morphogenesis have not been fully examined. We identify MEKK3 as a critical signaling molecule during endocardial cushion development. We report the detection of MEKK3 transcripts to embryonic hearts before, during, and after cardiac cushion cells have executed epithelial-to-mesenchymal transition (EMT). MEKK3 is observed to endocardial cells of the cardiac cushions with a diminishing gradient of expression into the cushions. These observations suggest that MEKK3 may function during production of cushion mesenchyme as required for valvular development and septation of the heart. We used a kinase inactive form of MEKK3 (MEKK3(KI)) in an in vitro assay that recapitulates in vivo EMT and show that MEKK3(KI) attenuates mesenchyme formation. Conversely, constitutively active MEKK3 (ca-MEKK3) triggers mesenchyme production in ventricular endocardium, a tissue that does not normally undergo EMT. MEKK3-driven mesenchyme production is further substantiated by increased expression of EMT-relevant genes, including TGFbeta(2), Has2, and periostin. Furthermore, we show that MEKK3 stimulates EMT via a TGFbeta(2)-dependent mechanism. Thus, the activity of MEKK3 is sufficient for developmental EMT in the heart. This knowledge provides a basis to understand how MEKK3 integrates signaling cascades activating endocardial cushion EMT.


Subject(s)
Endocardial Cushions/embryology , Endocardial Cushions/enzymology , Epithelial Cells/cytology , Epithelial Cells/enzymology , MAP Kinase Kinase Kinase 3/physiology , Mesoderm/embryology , Morphogenesis/physiology , Transforming Growth Factor beta2/physiology , Animals , Cell Differentiation/physiology , Endocardial Cushions/cytology , Endocardial Cushions/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Enzymologic/physiology , MAP Kinase Kinase Kinase 3/deficiency , MAP Kinase Kinase Kinase 3/genetics , MAP Kinase Kinase Kinase 3/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...