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.
Respir Res ; 16: 22, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25763778

ABSTRACT

BACKGROUND: The transcription factor NK2 homeobox 1 (Nkx2-1) plays essential roles in epithelial cell proliferation and differentiation in mouse and human lung development and tumorigenesis. A better understanding of genes and pathways downstream of Nkx2-1 will clarify the multiple roles of this critical lung factor. Nkx2-1 regulates directly or indirectly numerous protein-coding genes; however, there is a paucity of information about Nkx2-1-regulated microRNAs (miRNAs). METHODS AND RESULTS: By miRNA array analyses of mouse epithelial cell lines in which endogenous Nkx2-1 was knocked-down, we revealed that 29 miRNAs were negatively regulated including miR-200c, and 39 miRNAs were positively regulated by Nkx2-1 including miR-1195. Mouse lungs lacking functional phosphorylated Nkx2-1 showed increased expression of miR-200c and alterations in the expression of other top regulated miRNAs. Moreover, chromatin immunoprecipitation assays showed binding of NKX2-1 protein to regulatory regions of these miRNAs. Promoter reporter assays indicated that 1kb of the miR-200c 5' flanking region was transcriptionally active but did not mediate Nkx2-1- repression of miR-200c expression. 3'UTR reporter assays support a direct regulation of the predicted targets Nfib and Myb by miR-200c. CONCLUSIONS: These studies suggest that Nkx2-1 controls the expression of specific miRNAs in lung epithelial cells. In particular, we identified a regulatory link between Nkx2-1, the known tumor suppressor miR-200c, and the developmental and oncogenic transcription factors Nfib and Myb, adding new players to the regulatory mechanisms driven by Nkx2-1 in lung epithelial cells that may have implications in lung development and tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Lung/metabolism , MicroRNAs/metabolism , NFI Transcription Factors/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins v-myb/metabolism , Transcription Factors/metabolism , 3' Untranslated Regions , 5' Flanking Region , Animals , Binding Sites , Cell Line , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genes, Reporter , Mice , MicroRNAs/genetics , NFI Transcription Factors/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Oncogene Proteins v-myb/genetics , Phosphorylation , Promoter Regions, Genetic , Thyroid Nuclear Factor 1 , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic , Transfection
2.
J Biol Chem ; 288(17): 12215-21, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23530051

ABSTRACT

Podoplanin (PDPN) is a transmembrane receptor that affects the activities of Rho, ezrin, and other proteins to promote tumor cell motility, invasion, and metastasis. PDPN is found in many types of cancer and may serve as a tumor biomarker and chemotherapeutic target. The intracellular region of PDPN contains only two serines, and these are conserved in mammals including mice and humans. We generated cells from the embryos of homozygous null Pdpn knock-out mice to investigate the relevance of these serines to cell growth and migration on a clear (PDPN-free) background. We report here that one or both of these serines can be phosphorylated by PKA (protein kinase A). We also report that conversion of these serines to nonphosphorylatable alanine residues enhances cell migration, whereas their conversion to phosphomimetic aspartate residues decreases cell migration. These results indicate that PKA can phosphorylate PDPN to decrease cell migration. In addition, we report that PDPN expression in fibroblasts causes them to facilitate the motility and viability of neighboring melanoma cells in coculture. These findings shed new light on how PDPN promotes cell motility, its role in tumorigenesis, and its utility as a functionally relevant biomarker and chemotherapeutic target.


Subject(s)
Cell Movement , Cyclic AMP-Dependent Protein Kinases/metabolism , Fibroblasts/metabolism , Melanoma/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Animals , Cell Line, Tumor , Coculture Techniques , Cyclic AMP-Dependent Protein Kinases/genetics , Fibroblasts/pathology , Melanoma/genetics , Melanoma/pathology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Phosphorylation/genetics , Serine/genetics , Serine/metabolism
3.
J Biol Chem ; 287(44): 37282-95, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22955271

ABSTRACT

The Grainyhead family of transcription factors controls morphogenesis and differentiation of epithelial cell layers in multicellular organisms by regulating cell junction- and proliferation-related genes. Grainyhead-like 2 (Grhl2) is expressed in developing mouse lung epithelium and is required for normal lung organogenesis. The specific epithelial cells expressing Grhl2 and the genes regulated by Grhl2 in normal lungs are mostly unknown. In these studies we identified the NK2-homeobox 1 transcription factor (Nkx2-1) as a direct transcriptional target of Grhl2. By binding and transcriptional assays and by confocal microscopy we showed that these two transcription factors form a positive feedback loop in vivo and in cell lines and are co-expressed in lung bronchiolar and alveolar type II cells. The morphological changes observed in flattening lung alveolar type II cells in culture are associated with down-regulation of Grhl2 and Nkx2-1. Reduction of Grhl2 in lung epithelial cell lines results in lower expression levels of Nkx2-1 and of known Grhl2 target genes. By microarray analysis we identified that in addition to Cadherin1 and Claudin4, Grhl2 regulates other cell interaction genes such as semaphorins and their receptors, which also play a functional role in developing lung epithelium. Impaired collective cell migration observed in Grhl2 knockdown cell monolayers is associated with reduced expression of these genes and may contribute to the altered epithelial phenotype reported in Grhl2 mutant mice. Thus, Grhl2 functions at the nexus of a novel regulatory network, connecting lung epithelial cell identity, migration, and cell-cell interactions.


Subject(s)
Alveolar Epithelial Cells/physiology , Cell Differentiation , Morphogenesis , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Alveolar Epithelial Cells/metabolism , Animals , Cell Line , Cell Movement , Cell Proliferation , Cell Shape , Chromatin Immunoprecipitation , Gene Expression , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Lung/cytology , Lung/embryology , Mice , Nuclear Proteins/genetics , Phalloidine/metabolism , Phenotype , Promoter Regions, Genetic , Protein Binding , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Transcriptional Activation , Transcriptome
4.
PLoS One ; 7(1): e29907, 2012.
Article in English | MEDLINE | ID: mdl-22242187

ABSTRACT

The homeodomain transcription factor Nkx2-1 is essential for normal lung development and homeostasis. In lung tumors, it is considered a lineage survival oncogene and prognostic factor depending on its expression levels. The target genes directly bound by Nkx2-1, that could be the primary effectors of its functions in the different cellular contexts where it is expressed, are mostly unknown. In embryonic day 11.5 (E11.5) mouse lung, epithelial cells expressing Nkx2-1 are predominantly expanding, and in E19.5 prenatal lungs, Nkx2-1-expressing cells are predominantly differentiating in preparation for birth. To evaluate Nkx2-1 regulated networks in these two cell contexts, we analyzed genome-wide binding of Nkx2-1 to DNA regulatory regions by chromatin immunoprecipitation followed by tiling array analysis, and intersected these data to expression data sets. We further determined expression patterns of Nkx2-1 developmental target genes in human lung tumors and correlated their expression levels to that of endogenous NKX2-1. In these studies we uncovered differential Nkx2-1 regulated networks in early and late lung development, and a direct function of Nkx2-1 in regulation of the cell cycle by controlling the expression of proliferation-related genes. New targets, validated in Nkx2-1 shRNA transduced cell lines, include E2f3, Cyclin B1, Cyclin B2, and c-Met. Expression levels of Nkx2-1 direct target genes identified in mouse development significantly correlate or anti-correlate to the levels of endogenous NKX2-1 in a dosage-dependent manner in multiple human lung tumor expression data sets, supporting alternative roles for Nkx2-1 as a transcriptional activator or repressor, and direct regulator of cell cycle progression in development and tumors.


Subject(s)
Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study , Lung Neoplasms/genetics , Lung/embryology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Cycle/genetics , Cell Proliferation , Chromatin Immunoprecipitation , Conserved Sequence , Down-Regulation/genetics , Humans , Lung/metabolism , Lung Neoplasms/pathology , Mice , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Protein Binding/genetics , Reproducibility of Results , Signal Transduction/genetics , Thyroid Nuclear Factor 1 , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...