Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Dev Biol ; 483: 22-33, 2022 03.
Article in English | MEDLINE | ID: mdl-34973175

ABSTRACT

The extra-embryonic yolk sac contains adjacent layers of mesoderm and visceral endoderm. The mesodermal layer serves as the first site of embryonic hematopoiesis, while the visceral endoderm provides a means of exchanging nutrients and waste until the development of the chorioallantoic placenta. While defects in chorioallantoic fusion and yolk sac hematopoiesis have been described in Cdx mutant mouse models, little is known about the gene targets and molecular mechanisms through which Cdx members regulate these processes. To this end, we used RNA-seq to examine Cdx-dependent gene expression changes in the yolk sac. We find that loss of Cdx function impacts the expression of genes involved in yolk sac hematopoiesis, as previously described, as well as novel Cdx2 target genes. In addition, we observed Cdx-dependent changes in PRC2 subunit expression accompanied by altered H3K27me3 deposition at a subset of Cdx target genes as early as E7.5 in the embryo proper. This study identifies additional Cdx target genes and provides further evidence for Cdx-dependent epigenetic regulation of gene expression in the early embryo, and that this regulation is required to maintain gene expression programs in the extra-embryonic yolk sac at later developmental stages.


Subject(s)
CDX2 Transcription Factor/genetics , Embryonic Development/genetics , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Polycomb Repressive Complex 2/genetics , Animals , Endoderm/embryology , Female , Gene Expression , Gene Knockout Techniques/methods , Hematopoiesis/genetics , Male , Mesoderm/embryology , Mice , Mice, Knockout , Pregnancy , RNA-Seq/methods , Transcription, Genetic/genetics , Yolk Sac/embryology
2.
Genes (Basel) ; 12(2)2021 01 28.
Article in English | MEDLINE | ID: mdl-33525395

ABSTRACT

The majority of colorectal cancers harbor loss-of-function mutations in APC, a negative regulator of canonical Wnt signaling, leading to intestinal polyps that are predisposed to malignant progression. Comparable murine APC alleles also evoke intestinal polyps, which are typically confined to the small intestine and proximal colon, but do not progress to carcinoma in the absence of additional mutations. The Cdx transcription factors Cdx1 and Cdx2 are essential for homeostasis of the intestinal epithelium, and loss of Cdx2 has been associated with more aggressive subtypes of colorectal cancer in the human population. Consistent with this, concomitant loss of Cdx1 and Cdx2 in a murine APC mutant background leads to an increase in polyps throughout the intestinal tract. These polyps also exhibit a villous phenotype associated with the loss of EphrinB1. However, the basis for these outcomes is poorly understood. To further explore this, we modeled Cdx2 loss in SW480 colorectal cancer cells. We found that Cdx2 impacted Notch signaling in SW480 cells, and that EphrinB1 is a Notch target gene. As EphrinB1 loss also leads to a villus tumor phenotype, these findings evoke a mechanism by which Cdx2 impacts colorectal cancer via Notch-dependent EphrinB1 signaling.


Subject(s)
CDX2 Transcription Factor/genetics , Ephrin-B1/genetics , Gene Expression Regulation , Intestinal Mucosa/metabolism , Receptors, Notch/metabolism , Signal Transduction , Biomarkers , CDX2 Transcription Factor/metabolism , Cell Line, Tumor , Humans , Intestinal Mucosa/pathology , Models, Biological , Promoter Regions, Genetic
3.
Development ; 146(7)2019 04 01.
Article in English | MEDLINE | ID: mdl-30936115

ABSTRACT

Murine cardiac and hematopoietic progenitors are derived from Mesp1+ mesoderm. Cdx function impacts both yolk sac hematopoiesis and cardiogenesis in zebrafish, suggesting that Cdx family members regulate early mesoderm cell fate decisions. We found that Cdx2 occupies a number of transcription factor loci during embryogenesis, including key regulators of both cardiac and blood development, and that Cdx function is required for normal expression of the cardiogenic transcription factors Nkx2-5 and Tbx5 Furthermore, Cdx and Brg1, an ATPase subunit of the SWI/SNF chromatin remodeling complex, co-occupy a number of loci, suggesting that Cdx family members regulate target gene expression through alterations in chromatin architecture. Consistent with this, we demonstrate loss of Brg1 occupancy and altered chromatin structure at several cardiogenic genes in Cdx-null mutants. Finally, we provide evidence for an onset of Cdx2 expression at E6.5 coinciding with egression of cardiac progenitors from the primitive streak. Together, these findings suggest that Cdx functions in multi-potential mesoderm to direct early cell fate decisions through transcriptional regulation of several novel target genes, and provide further insight into a potential epigenetic mechanism by which Cdx influences target gene expression.


Subject(s)
CDX2 Transcription Factor/metabolism , Animals , CDX2 Transcription Factor/genetics , Chromatin Immunoprecipitation , Female , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization , Male , Mice , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
4.
J Biol Chem ; 292(8): 3389-3399, 2017 02 24.
Article in English | MEDLINE | ID: mdl-28082674

ABSTRACT

The packaging of genomic DNA into nucleosomes creates a barrier to transcription that can be relieved through ATP-dependent chromatin remodeling via complexes such as the switch-sucrose non-fermentable (SWI-SNF) chromatin remodeling complex. The SWI-SNF complex remodels chromatin via conformational or positional changes of nucleosomes, thereby altering the access of transcriptional machinery to target genes. The SWI-SNF complex has limited ability to bind to sequence-specific elements, and, therefore, its recruitment to target loci is believed to require interaction with DNA-associated transcription factors. The Cdx family of homeodomain transcript ion factors (Cdx1, Cdx2, and Cdx4) are essential for a number of developmental programs in the mouse. Cdx1 and Cdx2 also regulate intestinal homeostasis throughout life. Although a number of Cdx target genes have been identified, the basis by which Cdx members impact their transcription is poorly understood. We have found that Cdx members interact with the SWI-SNF complex and make direct contact with Brg1, a catalytic member of SWI-SNF. Both Cdx2 and Brg1 co-occupy a number of Cdx target genes, and both factors are necessary for transcriptional regulation of such targets. Finally, Cdx2 and Brg1 occupancy occurs coincident with chromatin remodeling at some of these loci. Taken together, our findings suggest that Cdx transcription factors regulate target gene expression, in part, through recruitment of Brg1-associated SWI-SNF chromatin remodeling activity.


Subject(s)
CDX2 Transcription Factor/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Protein Interaction Maps
5.
Dev Biol ; 422(2): 115-124, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28065741

ABSTRACT

The Cdx transcription factors play essential roles in primitive hematopoiesis in the zebrafish where they exert their effects, in part, through regulation of hox genes. Defects in hematopoiesis have also been reported in Cdx mutant murine embryonic stem cell models, however, to date no mouse model reflecting the zebrafish Cdx mutant hematopoietic phenotype has been described. This is likely due, in part, to functional redundancy among Cdx members and the early lethality of Cdx2 null mutants. To circumvent these limitations, we used Cre-mediated conditional deletion to assess the impact of concomitant loss of Cdx1 and Cdx2 on murine primitive hematopoiesis. We found that Cdx1/Cdx2 double mutants exhibited defects in primitive hematopoiesis and yolk sac vasculature concomitant with reduced expression of several genes encoding hematopoietic transcription factors including Scl/Tal1. Chromatin immunoprecipitation analysis revealed that Scl was occupied by Cdx2 in vivo, and Cdx mutant hematopoietic yolk sac differentiation defects could be rescued by expression of exogenous Scl. These findings demonstrate critical roles for Cdx members in murine primitive hematopoiesis upstream of Scl.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , CDX2 Transcription Factor/genetics , Hematopoiesis/genetics , Homeodomain Proteins/genetics , Neovascularization, Physiologic/genetics , Proto-Oncogene Proteins/biosynthesis , Yolk Sac/blood supply , Animals , Cell Differentiation/genetics , Chromatin Immunoprecipitation , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout , T-Cell Acute Lymphocytic Leukemia Protein 1 , Yolk Sac/embryology
6.
Mol Biol Cell ; 23(20): 4008-19, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22918940

ABSTRACT

Rho GTPases share a common inhibitor, Rho guanine nucleotide dissociation inhibitor (RhoGDI), which regulates their expression levels, membrane localization, and activation state. The selective dissociation of individual Rho GTPases from RhoGDI ensures appropriate responses to cellular signals, but the underlying mechanisms are unclear. Diacylglycerol kinase ζ (DGKζ), which phosphorylates diacylglycerol to yield phosphatidic acid, selectively dissociates Rac1 by stimulating PAK1-mediated phosphorylation of RhoGDI on Ser-101/174. Similarly, phosphorylation of RhoGDI on Ser-34 by protein kinase Cα (PKCα) selectively releases RhoA. Here we show DGKζ is required for RhoA activation and Ser-34 phosphorylation, which were decreased in DGKζ-deficient fibroblasts and rescued by wild-type DGKζ or a catalytically inactive mutant. DGKζ bound directly to the C-terminus of RhoA and the regulatory arm of RhoGDI and was required for efficient interaction of PKCα and RhoA. DGKζ-null fibroblasts had condensed F-actin bundles and altered focal adhesion distribution, indicative of aberrant RhoA signaling. Two targets of the RhoA effector ROCK showed reduced phosphorylation in DGKζ-null cells. Collectively our findings suggest DGKζ functions as a scaffold to assemble a signaling complex that functions as a RhoA-selective, GDI dissociation factor. As a regulator of Rac1 and RhoA activity, DGKζ is a critical factor linking changes in lipid signaling to actin reorganization.


Subject(s)
Diacylglycerol Kinase/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Biocatalysis , Diacylglycerol Kinase/chemistry , Embryo, Mammalian/cytology , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/enzymology , Focal Adhesions/metabolism , Mice , Models, Biological , Multiprotein Complexes/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Kinase C-alpha/metabolism , Protein Stability , Protein Structure, Tertiary , Signal Transduction , Stress Fibers/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors/chemistry , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , rhoA GTP-Binding Protein/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...