Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 12(4): 393, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846290

ABSTRACT

Cardiac septum malformations account for the largest proportion in congenital heart defects. The transcription factor Sox7 has critical functions in the vascular development and angiogenesis. It is unclear whether Sox7 also contributes to cardiac septation development. We identified a de novo 8p23.1 deletion with Sox7 haploinsufficiency in an atrioventricular septal defect (AVSD) patient using whole exome sequencing in 100 AVSD patients. Then, multiple Sox7 conditional loss-of-function mice models were generated to explore the role of Sox7 in atrioventricular cushion development. Sox7 deficiency mice embryos exhibited partial AVSD and impaired endothelial to mesenchymal transition (EndMT). Transcriptome analysis revealed BMP signaling pathway was significantly downregulated in Sox7 deficiency atrioventricular cushions. Mechanistically, Sox7 deficiency reduced the expressions of Bmp2 in atrioventricular canal myocardium and Wnt4 in endocardium, and Sox7 binds to Wnt4 and Bmp2 directly. Furthermore, WNT4 or BMP2 protein could partially rescue the impaired EndMT process caused by Sox7 deficiency, and inhibition of BMP2 by Noggin could attenuate the effect of WNT4 protein. In summary, our findings identify Sox7 as a novel AVSD pathogenic candidate gene, and it can regulate the EndMT involved in atrioventricular cushion morphogenesis through Wnt4-Bmp2 signaling. This study contributes new strategies to the diagnosis and treatment of congenital heart defects.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Heart Septal Defects/metabolism , SOXF Transcription Factors/metabolism , Wnt4 Protein/metabolism , Animals , Case-Control Studies , Child, Preschool , Endocardium/embryology , Endocardium/growth & development , Endocardium/metabolism , Female , Heart Septal Defects/genetics , Human Umbilical Vein Endothelial Cells , Humans , Mice , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/genetics , Signal Transduction
2.
Adv Wound Care (New Rochelle) ; 9(11): 591-601, 2020 11.
Article in English | MEDLINE | ID: mdl-33095124

ABSTRACT

Objective: This study aimed to elucidate the role of the proangiogenic transcription factors Sox7 and Sox17 in the wound healing process and investigate the therapeutic potential of Dll4 blockade, which is an upstream regulator of Sox17, for the treatment of nonhealing wounds. Approach: After generating a full-thickness skin defect wound model of endothelial Sox7- and/or Sox17-deficient mice, we measured the wound healing rates and performed histological analysis. The effects of an anti-Dll4 antibody on wound angiogenesis in Sox7-deficient mice and db/db diabetic mice were assessed. Results: Sox7 and/or Sox17 deletion delayed wound healing. Moreover, the loss of Sox7 and Sox17 inhibited wound angiogenesis, without affecting the expression of the other. Of interest, after anti-Dll4 antibody treatment, Sox17 levels were increased and the suppression of angiogenesis was alleviated in Sox7-deficient mice and db/db diabetic mice. Consequently, Dll4 blockade effectively recovered the observed delay in wound healing. Innovation: The proangiogenic role of Sox7 and Sox17 in wound angiogenesis was addressed and effective treatment of nonhealing wounds by Dll4 blockade was suggested. Conclusion: This study revealed the proangiogenic role of the transcription factors Sox7 and Sox17 in wound angiogenesis. Furthermore, we suggest a novel method for treating nonhealing wounds by particularly targeting the Dll4-Sox17 axis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/physiology , SOXF Transcription Factors/metabolism , Wound Healing/drug effects , Adaptor Proteins, Signal Transducing , Angiogenesis Inhibitors/pharmacology , Animals , Calcium-Binding Proteins , HMGB Proteins/deficiency , Male , Mice , Mice, Inbred NOD , Morphogenesis/genetics , SOXF Transcription Factors/deficiency , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism
3.
Biol Reprod ; 99(3): 578-589, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29635272

ABSTRACT

In mouse conceptus, two yolk-sac membranes, the parietal endoderm (PE) and visceral endoderm (VE), are involved in protecting and nourishing early-somite-stage embryos prior to the establishment of placental circulation. Both PE and VE membranes are tightly anchored to the marginal edge of the developing placental disk, in which the extraembryonic endoderm (marginal zone endoderm: ME) shows the typical flat epithelial morphology intermediate between those of PE and VE in vivo. However, the molecular characteristics and functions of the ME in mouse placentation remain unclear. Here, we show that SOX17, not SOX7, is continuously expressed in the ME cells, whereas both SOX17 and SOX7 are coexpressed in PE cells, by at least 10.5 days postconception. The Sox17-null conceptus, but not the Sox7-null one, showed the ectopic appearance of squamous VE-like epithelial cells in the presumptive ME region, together with reduced cell density and aberrant morphology of PE cells. Such aberrant ME formation in the Sox17-null extraembryonic endoderm was not rescued by the chimeric embryo replaced with the wild-type gut endoderm by the injection of wild-type ES cells into the Sox17-null blastocyst, suggesting the cell autonomous defects in the extraembryonic endoderm of Sox17-null concepti. These findings provide direct evidence of the crucial roles of SOX17 in proper formation and maintenance of the ME region, highlighting a novel entry point to understand the in vivo VE-to-PE transition in the marginal edge of developing placenta.


Subject(s)
Embryonic Development/physiology , Endoderm/physiology , HMGB Proteins/physiology , Placentation/physiology , SOXF Transcription Factors/physiology , Yolk Sac/physiology , Animals , Cell Proliferation , Female , Gene Expression , Genotype , HMGB Proteins/deficiency , HMGB Proteins/genetics , Male , Mice , Mice, Knockout , Pregnancy , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/genetics
4.
Development ; 144(14): 2629-2639, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28619820

ABSTRACT

Arterial specification and differentiation are influenced by a number of regulatory pathways. While it is known that the Vegfa-Notch cascade plays a central role, the transcriptional hierarchy controlling arterial specification has not been fully delineated. To elucidate the direct transcriptional regulators of Notch receptor expression in arterial endothelial cells, we used histone signatures, DNaseI hypersensitivity and ChIP-seq data to identify enhancers for the human NOTCH1 and zebrafish notch1b genes. These enhancers were able to direct arterial endothelial cell-restricted expression in transgenic models. Genetic disruption of SoxF binding sites established a clear requirement for members of this group of transcription factors (SOX7, SOX17 and SOX18) to drive the activity of these enhancers in vivo Endogenous deletion of the notch1b enhancer led to a significant loss of arterial connections to the dorsal aorta in Notch pathway-deficient zebrafish. Loss of SoxF function revealed that these factors are necessary for NOTCH1 and notch1b enhancer activity and for correct endogenous transcription of these genes. These findings position SoxF transcription factors directly upstream of Notch receptor expression during the acquisition of arterial identity in vertebrates.


Subject(s)
Arteries/embryology , Arteries/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Arteriovenous Malformations/embryology , Arteriovenous Malformations/genetics , Arteriovenous Malformations/metabolism , Enhancer Elements, Genetic , Female , Gene Expression Regulation, Developmental , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pregnancy , Receptor, Notch1/deficiency , SOXF Transcription Factors/deficiency , Sequence Homology, Amino Acid , Signal Transduction , Zebrafish , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Dev Biol ; 414(2): 219-27, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27102016

ABSTRACT

The importance of canonical Wnt signaling to murine uterine development is well established. Mouse models in which uterine-specific Wnt ligands, ß-catenin, or Lef1 are disrupted result in failure of postnatal endometrial gland development. Sox17 is a transcription factor characterized in numerous tissues as an antagonist of Wnt signaling. Thus, we hypothesized that conditional ablation of Sox17 would lead to hyperproliferation of endometrial glands in mice. Contrary to our prediction, disruption of Sox17 in epithelial and stromal compartments led to inhibition of endometrial adenogenesis and a loss of reproductive capacity. Epithelium-specific Sox17 disruption resulted in normal adenogenesis although reproductive capacity remained impaired. These findings suggest that non-epithelial, Sox17-positive cells are necessary for adenogenesis and that glands require Sox17 to properly function. To our knowledge, these findings are the first to implicate Sox17 in endometrial gland formation and reproductive success. The data presented herein underscore the importance of studying Sox17 in uterine homeostasis and function.


Subject(s)
Endometrium/growth & development , Epithelial Cells/physiology , Exocrine Glands/growth & development , HMGB Proteins/physiology , SOXF Transcription Factors/physiology , Animals , Endometrium/metabolism , Endometrium/pathology , Exocrine Glands/metabolism , Female , HMGB Proteins/deficiency , HMGB Proteins/genetics , Hepatocyte Nuclear Factor 3-beta/biosynthesis , Hepatocyte Nuclear Factor 3-beta/genetics , Homeostasis , Hyperplasia , Infertility, Female/genetics , Infertility, Female/pathology , Luminescent Proteins/analysis , Lymphoid Enhancer-Binding Factor 1/biosynthesis , Lymphoid Enhancer-Binding Factor 1/genetics , Mice , Mice, Knockout , Pregnancy , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/genetics , Specific Pathogen-Free Organisms , Stromal Cells/physiology , Wnt Proteins/biosynthesis , Wnt Proteins/genetics , Wnt Signaling Pathway/physiology
6.
PLoS One ; 10(12): e0143650, 2015.
Article in English | MEDLINE | ID: mdl-26630461

ABSTRACT

Vascular development and maintenance are controlled by a complex transcriptional program, which integrates both extracellular and intracellular signals in endothelial cells. Here we study the roles of three closely related SoxF family transcription factors-Sox7, Sox17, and Sox18 -in the developing and mature mouse vasculature using targeted gene deletion on a mixed C57/129/CD1 genetic background. In the retinal vasculature, each SoxF gene exhibits a distinctive pattern of expression in different classes of blood vessels. On a mixed genetic background, vascular endothelial-specific deletion of individual SoxF genes has little or no effect on vascular architecture or differentiation, a result that can be explained by overlapping function and by reciprocal regulation of gene expression between Sox7 and Sox17. By contrast, combined deletion of Sox7, Sox17, and Sox18 at the onset of retinal angiogenesis leads to a dense capillary plexus with a nearly complete loss of radial arteries and veins, whereas the presence of a single Sox17 allele largely restores arterial identity, as determined by vascular smooth muscle cell coverage. In the developing retina, expression of all three SoxF genes is reduced in the absence of Norrin/Frizzled4-mediated canonical Wnt signaling, but SoxF gene expression is unaffected by reduced VEGF signaling in response to deletion of Neuropilin1 (Npn1). In adulthood, Sox7, Sox17, and Sox18 act in a largely redundant manner to maintain blood vessel function, as adult onset vascular endothelial-specific deletion of all three SoxF genes leads to massive edema despite nearly normal vascular architecture. These data reveal critical and partially redundant roles for Sox7, Sox17 and Sox18 in vascular growth, differentiation, and maintenance.


Subject(s)
HMGB Proteins/genetics , Retinal Vessels/growth & development , Retinal Vessels/metabolism , SOXF Transcription Factors/genetics , Animals , Cell Differentiation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Gene Expression Regulation, Developmental , HMGB Proteins/deficiency , HMGB Proteins/metabolism , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Multigene Family , Neovascularization, Physiologic/genetics , Papilledema/etiology , Papilledema/genetics , Papilledema/pathology , Receptors, Notch/metabolism , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/metabolism , Wnt Signaling Pathway
7.
J Immunol ; 194(5): 2424-38, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25653427

ABSTRACT

Human and murine studies showed that GM-CSF exerts beneficial effects in intestinal inflammation. To explore whether GM-CSF mediates its effects via monocytes, we analyzed effects of GM-CSF on monocytes in vitro and assessed the immunomodulatory potential of GM-CSF-activated monocytes (GMaMs) in vivo. We used microarray technology and functional assays to characterize GMaMs in vitro and used a mouse model of colitis to study GMaM functions in vivo. GM-CSF activates monocytes to increase adherence, migration, chemotaxis, and oxidative burst in vitro, and primes monocyte response to secondary microbial stimuli. In addition, GMaMs accelerate epithelial healing in vitro. Most important, in a mouse model of experimental T cell-induced colitis, GMaMs show therapeutic activity and protect mice from colitis. This is accompanied by increased production of IL-4, IL-10, and IL-13, and decreased production of IFN-γ in lamina propria mononuclear cells in vivo. Confirming this finding, GMaMs attract T cells and shape their differentiation toward Th2 by upregulating IL-4, IL-10, and IL-13 in T cells in vitro. Beneficial effects of GM-CSF in Crohn's disease may possibly be mediated through reprogramming of monocytes to simultaneously improved bacterial clearance and induction of wound healing, as well as regulation of adaptive immunity to limit excessive inflammation.


Subject(s)
Adaptive Immunity/drug effects , Colitis/drug therapy , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Intestine, Large/drug effects , Monocytes/drug effects , Adoptive Transfer , Animals , Cell Adhesion/drug effects , Chemotaxis/drug effects , Colitis/immunology , Colitis/pathology , Gene Expression Regulation , Humans , Interferon-gamma/pharmacology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/pharmacology , Intestine, Large/immunology , Intestine, Large/pathology , Mice , Mice, Knockout , Monocytes/cytology , Monocytes/immunology , Primary Cell Culture , Respiratory Burst/drug effects , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/genetics , SOXF Transcription Factors/immunology , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/transplantation
8.
Circulation ; 131(11): 995-1005, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25596186

ABSTRACT

BACKGROUND: Intracranial aneurysm (IA) is a common vascular disorder that frequently leads to fatal vascular rupture. Although various acquired risk factors associated with IA have been identified, the hereditary basis of IA remains poorly understood. As a result, genetically modified animals accurately modeling IA and related pathogenesis have been lacking, and subsequent drug development has been delayed. METHODS AND RESULTS: The transcription factor Sox17 is robustly expressed in endothelial cells of normal intracerebral arteries. The combination of Sox17 deficiency and angiotensin II infusion in mice induces vascular abnormalities closely resembling the cardinal features of IA such as luminal dilation, wall thinning, tortuosity, and subarachnoid hemorrhages. This combination impairs junctional assembly, cell-matrix adhesion, regeneration capacity, and paracrine secretion in endothelial cells of intracerebral arteries, highlighting key endothelial dysfunctions that lead to IA pathogenesis. Moreover, human IA samples showed reduced Sox17 expression and impaired endothelial integrity, further strengthening the applicability of this animal model to clinical settings. CONCLUSIONS: Our findings demonstrate that Sox17 deficiency in mouse can induce IA under hypertensive conditions, suggesting Sox17 deficiency as a potential genetic factor for IA formation. The Sox17-deficient mouse model provides a novel platform to develop therapeutics for incurable IA.


Subject(s)
Endothelium, Vascular/pathology , HMGB Proteins/deficiency , Intracranial Aneurysm/genetics , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/physiology , Adult , Aged , Angiotensin II/toxicity , Animals , Aorta/pathology , Cells, Cultured , Cerebral Arteries/chemistry , Cerebral Arteries/pathology , Cyclin-Dependent Kinase Inhibitor Proteins/biosynthesis , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Dilatation, Pathologic/genetics , Dilatation, Pathologic/pathology , Disease Models, Animal , Endothelium, Vascular/metabolism , Female , HMGB Proteins/genetics , HMGB Proteins/physiology , Humans , Hypertension/complications , Intracranial Aneurysm/etiology , Intracranial Aneurysm/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocytes, Smooth Muscle/chemistry , Paracrine Communication , RNA Interference , SOXF Transcription Factors/analysis , SOXF Transcription Factors/genetics , Specific Pathogen-Free Organisms , Subarachnoid Hemorrhage/etiology , Transcription, Genetic , Up-Regulation , Veins/chemistry
9.
Development ; 141(6): 1228-38, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24523456

ABSTRACT

In mammals, the homeodomain transcription factor Prox1 acts as the central regulator of lymphatic cell fate. Its restricted expression in a subset of cardinal vein cells leads to a switch towards lymphatic specification and hence represents a prerequisite for the initiation of lymphangiogenesis. Murine Prox1-null embryos lack lymphatic structures, and sustained expression of Prox1 is indispensable for the maintenance of lymphatic cell fate even at adult stages, highlighting the unique importance of this gene for the lymphatic lineage. Whether this pre-eminent role of Prox1 within the lymphatic vasculature is conserved in other vertebrate classes has remained unresolved, mainly owing to the lack of availability of loss-of-function mutants. Here, we re-examine the role of Prox1a in zebrafish lymphangiogenesis. First, using a transgenic reporter line, we show that prox1a is initially expressed in different endothelial compartments, becoming restricted to lymphatic endothelial cells only at later stages. Second, using targeted mutagenesis, we show that Prox1a is dispensable for lymphatic specification and subsequent lymphangiogenesis in zebrafish. In line with this result, we found that the functionally related transcription factors Coup-TFII and Sox18 are also dispensable for lymphangiogenesis. Together, these findings suggest that lymphatic commitment in zebrafish and mice is controlled in fundamentally different ways.


Subject(s)
Homeodomain Proteins/physiology , Lymphangiogenesis/physiology , Tumor Suppressor Proteins/physiology , Zebrafish Proteins/physiology , Zebrafish/growth & development , Animals , Animals, Genetically Modified , COUP Transcription Factor II/deficiency , COUP Transcription Factor II/genetics , COUP Transcription Factor II/metabolism , Cell Differentiation , Cell Lineage , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Lymphangiogenesis/genetics , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Mice , Mice, Knockout , Mutation , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Species Specificity , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Zebrafish/genetics , Zebrafish/physiology , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
10.
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
11.
Hum Mol Genet ; 21(18): 4115-25, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22723016

ABSTRACT

Recurrent microdeletions of 8p23.1 that include GATA4 and SOX7 confer a high risk of both congenital diaphragmatic hernia (CDH) and cardiac defects. Although GATA4-deficient mice have both CDH and cardiac defects, no humans with cardiac defects attributed to GATA4 mutations have been reported to have CDH. We were also unable to identify deleterious GATA4 sequence changes in a CDH cohort. This suggested that haploinsufficiency of another 8p23.1 gene may contribute, along with GATA4, to the development of CDH. To determine if haploinsufficiency of SOX7-another transcription factor encoding gene-contributes to the development of CDH, we generated mice with a deletion of the second exon of Sox7. A portion of these Sox7(Δex2/+) mice developed retrosternal diaphragmatic hernias located in the anterior muscular portion of the diaphragm. Anterior CDH is also seen in Gata4(+/-) mice and has been described in association with 8p23.1 deletions in humans. Immunohistochemistry revealed that SOX7 is expressed in the vascular endothelial cells of the developing diaphragm and may be weakly expressed in some diaphragmatic muscle cells. Sox7(Δex2/Δex2) embryos die prior to diaphragm development with dilated pericardial sacs and failure of yolk sac remodeling suggestive of cardiovascular failure. Similar to our experience screening GATA4, no clearly deleterious SOX7 sequence changes were identified in our CDH cohort. We conclude that haploinsufficiency of Sox7 or Gata4 is sufficient to produce anterior CDH in mice and that haploinsufficiency of SOX7 and GATA4 may each contribute to the development of CDH in individuals with 8p23.1 deletions.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 8/genetics , Hernias, Diaphragmatic, Congenital , SOXF Transcription Factors/genetics , Animals , Base Sequence , DNA Mutational Analysis , Diaphragm/metabolism , Diaphragm/pathology , Disease Models, Animal , Female , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Genes, Lethal , Genetic Association Studies , Haploinsufficiency , Hernia, Diaphragmatic/genetics , Hernia, Diaphragmatic/pathology , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , SOXF Transcription Factors/deficiency
12.
Int J Dev Biol ; 55(1): 45-58, 2011.
Article in English | MEDLINE | ID: mdl-21305474

ABSTRACT

Sox17 is a transcription factor that is required for maintenance of the definitive endoderm in mouse embryos. By expression profiling of wild-type and mutant embryos and Sox17-overexpressing hepatoma cells, we identified genes with Sox17-dependent expression. Among the genes that were up-regulated in Sox17-null embryos and down-regulated by Sox17 expressing HepG2 cells is a set of genes that are expressed in the developing liver, suggesting that one function of Sox17 is the repression of liver gene expression, which is compatible with a role for Sox17 in maintaining the definitive endoderm in a progenitor state. Consistent with these findings, Sox17(-/-) cells display a diminished capacity to contribute to the definitive endoderm when transplanted into wild-type hosts. Analysis of gene ontology further revealed that many genes related to heart development were downregulated in Sox17-null embryos. This is associated with the defective development of the heart in the mutant embryos, which is accompanied by localised loss of Myocd-expressing cardiogenic progenitors and the malformation of the anterior intestinal portal.


Subject(s)
Embryo, Mammalian/metabolism , Gastrointestinal Tract/metabolism , Gene Expression Regulation, Developmental , HMGB Proteins/genetics , Myocardium/metabolism , SOXF Transcription Factors/genetics , Animals , Cell Transplantation/methods , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Endoderm/embryology , Endoderm/metabolism , Female , Gastrointestinal Tract/embryology , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HMGB Proteins/deficiency , Heart/embryology , Hep G2 Cells , Humans , In Situ Hybridization , Male , Mice , Mice, 129 Strain , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , SOXF Transcription Factors/deficiency , Somites/embryology , Somites/metabolism
13.
Blood ; 114(23): 4813-22, 2009 Nov 26.
Article in English | MEDLINE | ID: mdl-19801444

ABSTRACT

The molecular mechanisms that regulate the balance between proliferation and differentiation of precursors at the onset of hematopoiesis specification are poorly understood. By using a global gene expression profiling approach during the course of embryonic stem cell differentiation, we identified Sox7 as a potential candidate gene involved in the regulation of blood lineage formation from the mesoderm germ layer. In the present study, we show that Sox7 is transiently expressed in mesodermal precursors as they undergo specification to the hematopoietic program. Sox7 knockdown in vitro significantly decreases the formation of both primitive erythroid and definitive hematopoietic progenitors as well as endothelial progenitors. In contrast, Sox7-sustained expression in the earliest committed hematopoietic precursors promotes the maintenance of their multipotent and self-renewing status. Removal of this differentiation block driven by Sox7-enforced expression leads to the efficient differentiation of hematopoietic progenitors to all erythroid and myeloid lineages. This study identifies Sox7 as a novel and important player in the molecular regulation of the first committed blood precursors. Furthermore, our data demonstrate that the mere sustained expression of Sox7 is sufficient to completely alter the balance between proliferation and differentiation at the onset of hematopoiesis.


Subject(s)
Gene Expression Regulation, Developmental , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , SOXF Transcription Factors/physiology , Animals , Cell Division , Cell Lineage , Cells, Cultured/cytology , Cells, Cultured/metabolism , Erythroid Cells/cytology , Gastrula/cytology , Gastrula/metabolism , Gene Knockdown Techniques , Genetic Vectors/pharmacology , Hemangioblasts/cytology , Hemangioblasts/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Mesoderm/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/cytology , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/physiology , SOXF Transcription Factors/biosynthesis , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
Development ; 136(14): 2385-91, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19515696

ABSTRACT

Developmental defects caused by targeted gene inactivation in mice are commonly subject to strain-specific modifiers that modulate the severity of the phenotype. Although several genetic modifier loci have been mapped in mice, the gene(s) residing at these loci are mostly unidentified, and the molecular mechanisms of modifier action remain poorly understood. Mutations in Sox18 cause a variable phenotype in the human congenital syndrome hypotrichosis-lymphedema-telangiectasia, and the phenotype of Sox18-null mice varies from essentially normal to completely devoid of lymphatic vasculature and lethal, depending on the strain of the mice, suggesting a crucial role for strain-specific modifiers in this system. Here we show that two closely related Group F Sox factors, SOX7 and SOX17, are able to functionally substitute for SOX18 in vitro and in vivo. SOX7 and SOX17 are not normally expressed during lymphatic development, excluding a conventional redundancy mechanism. Instead, these genes are activated specifically in the absence of SOX18 function, and only in certain strains. Our studies identify Sox7 and Sox17 as modifiers of the Sox18 mutant phenotype, and reveal their mechanism of action as a novel mode of strain-specific compensatory upregulation.


Subject(s)
HMGB Proteins/physiology , Lymphangiogenesis/physiology , SOXF Transcription Factors/physiology , Animals , Base Sequence , DNA Primers/genetics , Gene Expression Regulation, Developmental , HMGB Proteins/genetics , Homeodomain Proteins/genetics , Humans , Hypotrichosis/genetics , Lymphangiogenesis/genetics , Lymphedema/genetics , Mice , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/genetics , Species Specificity , Syndrome , Telangiectasis/genetics , Tumor Suppressor Proteins/genetics , Up-Regulation
15.
Nature ; 456(7222): 643-7, 2008 Dec 04.
Article in English | MEDLINE | ID: mdl-18931657

ABSTRACT

The lymphatic system plays a key role in tissue fluid regulation and tumour metastasis, and lymphatic defects underlie many pathological states including lymphoedema, lymphangiectasia, lymphangioma and lymphatic dysplasia. However, the origins of the lymphatic system in the embryo, and the mechanisms that direct growth of the network of lymphatic vessels, remain unclear. Lymphatic vessels are thought to arise from endothelial precursor cells budding from the cardinal vein under the influence of the lymphatic hallmark gene Prox1 (prospero homeobox 1; ref. 4). Defects in the transcription factor gene SOX18 (SRY (sex determining region Y) box 18) cause lymphatic dysfunction in the human syndrome hypotrichosis-lymphoedema-telangiectasia, suggesting that Sox18 may also play a role in lymphatic development or function. Here we use molecular, cellular and genetic assays in mice to show that Sox18 acts as a molecular switch to induce differentiation of lymphatic endothelial cells. Sox18 is expressed in a subset of cardinal vein cells that later co-express Prox1 and migrate to form lymphatic vessels. Sox18 directly activates Prox1 transcription by binding to its proximal promoter. Overexpression of Sox18 in blood vascular endothelial cells induces them to express Prox1 and other lymphatic endothelial markers, while Sox18-null embryos show a complete blockade of lymphatic endothelial cell differentiation from the cardinal vein. Our findings demonstrate a critical role for Sox18 in developmental lymphangiogenesis, and suggest new avenues to investigate for therapeutic management of human lymphangiopathies.


Subject(s)
Cell Differentiation , Lymphatic Vessels/cytology , Lymphatic Vessels/embryology , SOXF Transcription Factors/metabolism , Animals , Biomarkers/analysis , Cell Movement , Cells, Cultured , Edema/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Ephrin-B2/genetics , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Hypotrichosis/genetics , Lymphangiogenesis , Lymphatic Vessels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Promoter Regions, Genetic/genetics , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/genetics , Telangiectasis/genetics , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Veins/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...