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.
Sci Rep ; 9(1): 11953, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31420575

ABSTRACT

The endocardium is the endothelial component of the vertebrate heart and plays a key role in heart development. Where, when, and how the endocardium segregates during embryogenesis have remained largely unknown, however. We now show that Nkx2-5+ cardiac progenitor cells (CPCs) that express the Sry-type HMG box gene Sox17 from embryonic day (E) 7.5 to E8.5 specifically differentiate into the endocardium in mouse embryos. Although Sox17 is not essential or sufficient for endocardium fate, it can bias the fate of CPCs toward the endocardium. On the other hand, Sox17 expression in the endocardium is required for heart development. Deletion of Sox17 specifically in the mesoderm markedly impaired endocardium development with regard to cell proliferation and behavior. The proliferation of cardiomyocytes, ventricular trabeculation, and myocardium thickening were also impaired in a non-cell-autonomous manner in the Sox17 mutant, likely as a consequence of down-regulation of NOTCH signaling. An unknown signal, regulated by Sox17 and required for nurturing of the myocardium, is responsible for the reduction in NOTCH-related genes in the mutant embryos. Our results thus provide insight into differentiation of the endocardium and its role in heart development.


Subject(s)
Cell Differentiation , Embryo, Mammalian/embryology , Endocardium/embryology , Gene Expression Regulation, Developmental , HMGB Proteins/biosynthesis , SOXF Transcription Factors/biosynthesis , Signal Transduction , Stem Cells/metabolism , Animals , Embryo, Mammalian/cytology , Endocardium/cytology , HMGB Proteins/genetics , Mesoderm/cytology , Mesoderm/embryology , Mice , Mice, Transgenic , Receptors, Notch/genetics , Receptors, Notch/metabolism , SOXF Transcription Factors/genetics , Stem Cells/cytology
2.
Dev Biol ; 445(2): 256-270, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30472119

ABSTRACT

The enteric nervous system is thought to originate solely from the neural crest. Transgenic lineage tracing revealed a novel population of clonal pancreatic duodenal homeobox-1 (Pdx1)-Cre lineage progenitor cells in the tunica muscularis of the gut that produced pancreatic descendants as well as neurons upon differentiation in vitro. Additionally, an in vivo subpopulation of endoderm lineage enteric neurons, but not glial cells, was seen especially in the proximal gut. Analysis of early transgenic embryos revealed Pdx1-Cre progeny (as well as Sox-17-Cre and Foxa2-Cre progeny) migrating from the developing pancreas and duodenum at E11.5 and contributing to the enteric nervous system. These results show that the mammalian enteric nervous system arises from both the neural crest and the endoderm. Moreover, in adult mice there are separate Wnt1-Cre neural crest stem cells and Pdx1-Cre pancreatic progenitors within the muscle layer of the gut.


Subject(s)
Enteric Nervous System/embryology , Animals , Cell Lineage/genetics , Duodenum/embryology , Duodenum/innervation , Duodenum/metabolism , Endoderm/cytology , Endoderm/embryology , Endoderm/metabolism , Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Gene Expression Regulation, Developmental , HMGB Proteins/genetics , HMGB Proteins/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Pancreas/embryology , Pancreas/innervation , Pancreas/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
3.
Mech Dev ; 139: 51-64, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26643664

ABSTRACT

Pancreas organogenesis is a highly dynamic process where neighboring tissue interactions lead to dynamic changes in gene regulatory networks that orchestrate endocrine, exocrine, and ductal lineage formation. To understand the spatio-temporal regulatory logic we have used the Forkhead transcription factor Foxa2-Venus fusion (FVF) knock-in reporter mouse to separate the FVF(+) pancreatic epithelium from the FVF(−) surrounding tissue (mesenchyme, neurons, blood, and blood vessels) to perform a genome-wide mRNA expression profiling at embryonic days (E) 12.5-15.5. Annotating genes and molecular processes suggest that FVF marks endoderm-derived multipotent epithelial progenitors at several lineage restriction steps, when the bulk of endocrine, exocrine and ductal cells are formed during the secondary transition. In the pancreatic epithelial compartment, we identified most known endocrine and exocrine lineage determining factors and diabetes-associated genes, but also unknown genes with spatio-temporal regulated pancreatic expression. In the non-endoderm-derived compartment, we identified many well-described regulatory genes that are not yet functionally annotated in pancreas development, emphasizing that neighboring tissue interactions are still ill defined. Pancreatic expression of over 635 genes was analyzed with them RNA in situ hybridization Genepaint public database. This validated the quality of the profiling data set and identified hundreds of genes with spatially restricted expression patterns in the pancreas. Some of these genes are also targeted by pancreatic transcription factors and show active chromatin marks in human islets of Langerhans. Thus, with the highest spatio-temporal resolution of a global gene expression profile during the secondary transition, our study enables to shed light on neighboring tissue interactions, developmental timing and diabetes gene regulation.


Subject(s)
Pancreas/embryology , Transcriptome , Animals , Cell Differentiation , Endoderm/embryology , Endoderm/metabolism , Gene Expression Regulation, Developmental , Gene Ontology , Hepatocyte Nuclear Factor 3-beta/physiology , Humans , Mice , Multipotent Stem Cells/physiology , Organogenesis , Pancreas/cytology , Pancreas/metabolism
4.
Genesis ; 51(11): 793-802, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24038996

ABSTRACT

The HMG-box transcription factor Sox17 is essential for endoderm formation, vascular development, and definitive hematopoiesis. To investigate the fate of distinct Sox17-expressing progenitor cells in a spatiotemporal manner, we generated a hormone-inducible CreERT2 knock-in mouse line. By homologous recombination we fused a codon improved, ligand-dependent estrogen receptor Cre recombinase by an intervening viral T2A sequence for co-translational cleavage to the 3' coding region of Sox17. Induction of Cre activity by administration of tamoxifen at defined time points of early mouse development and subsequent genetic lineage tracing confirmed the inducibility and tissue specificity of Cre recombination. Furthermore, Cre activity could be selectively induced in extra-embryonic and embryonic endoderm lineages, the primitive gut tube, and in endothelial cells of the vascular system as well as in the hemogenic endothelium of the dorsal aorta. The Sox17CreERT2 mouse line therefore represents a new tool for genetic lineage tracing in a tissue-specific manner and in addition enables lineage-restricted functional analysis.


Subject(s)
Cell Lineage , Embryonic Stem Cells/metabolism , Gene Knock-In Techniques , Integrases/metabolism , SOXF Transcription Factors/genetics , Animals , Aorta/metabolism , Cell Differentiation , Embryo, Mammalian , Endoderm/metabolism , Endothelial Cells/metabolism , Gastrula/metabolism , Genotype , Hematopoietic Stem Cells/metabolism , Integrases/genetics , Mice , Mice, Transgenic , Models, Animal , Organ Specificity , Tamoxifen/pharmacology
5.
Development ; 140(15): 3128-38, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23824574

ABSTRACT

Several signalling cascades are implicated in the formation and patterning of the three principal germ layers, but their precise temporal-spatial mode of action in progenitor populations remains undefined. We have used conditional gene deletion of mouse ß-catenin in Sox17-positive embryonic and extra-embryonic endoderm as well as vascular endothelial progenitors to address the function of canonical Wnt signalling in cell lineage formation and patterning. Conditional mutants fail to form anterior brain structures and exhibit posterior body axis truncations, whereas initial blood vessel formation appears normal. Tetraploid rescue experiments reveal that lack of ß-catenin in the anterior visceral endoderm results in defects in head organizer formation. Sox17 lineage tracing in the definitive endoderm (DE) shows a cell-autonomous requirement for ß-catenin in midgut and hindgut formation. Surprisingly, wild-type posterior visceral endoderm (PVE) in midgut- and hindgut-deficient tetraploid chimera rescues the posterior body axis truncation, indicating that the PVE is important for tail organizer formation. Upon loss of ß-catenin in the visceral endoderm and DE lineages, but not in the vascular endothelial lineage, Sox17 expression is not maintained, suggesting downstream regulation by canonical Wnt signalling. Strikingly, Tcf4/ß-catenin transactivation complexes accumulated on Sox17 cis-regulatory elements specifically upon endoderm induction in an embryonic stem cell differentiation system. Together, these results indicate that the Wnt/ß-catenin signalling pathway regulates Sox17 expression for visceral endoderm pattering and DE formation and provide the first functional evidence that the PVE is necessary for gastrula organizer gene induction and posterior axis development.


Subject(s)
Endoderm/embryology , Endoderm/metabolism , HMGB Proteins/metabolism , Organizers, Embryonic/embryology , Organizers, Embryonic/metabolism , SOXF Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Body Patterning , Cell Differentiation , Cell Lineage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/cytology , Female , Gene Expression Regulation, Developmental , HMGB Proteins/deficiency , HMGB Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Organizers, Embryonic/cytology , Pregnancy , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/genetics , Tetraploidy , Transcription Factor 4 , beta Catenin/deficiency , beta Catenin/genetics
6.
Genesis ; 47(9): 603-10, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19548312

ABSTRACT

Sox17 encodes an SRY-related high-mobility group (HMG) box transcription factor that is essential for endoderm formation and fetal hematopoietic stem cell maintenance. In the mouse, expression of Sox17 is first observed in the extraembryonic endoderm and is subsequently seen in the definitive endoderm as well as in blood and the endothelial cells of the developing vasculature. To conditionally inactivate genes in these domains, we have targeted the Sox17 locus to generate a bicistronic mRNA linking Sox17 to a codon improved Cre recombinase (iCre) via a viral 2A sequence. Here we report a new Cre knock-in mouse line, Sox17-2A-iCre, with activity in the developing endoderm, the vascular endothelial cells of the cardiovascular system and the hematopoietic system. Our results indicate that the Sox17-2A-iCre is active in an early endoderm progenitor and recombination of the Rosa26 reporter was observed in all previously reported expression domains of Sox17. The Sox17-2A-iCre line will be an excellent tool to conditionally inactivate genes in the definitive endoderm as well as in the vasculature and hematopoietic system.


Subject(s)
Endoderm/metabolism , Endothelial Cells/metabolism , Integrases/metabolism , Models, Animal , SOXF Transcription Factors/metabolism , Animals , Blotting, Southern , DNA Primers/genetics , Embryonic Stem Cells/metabolism , Genetic Vectors/genetics , Genotype , Mice , Mice, Transgenic , Viral Proteins/genetics , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL