Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
PLoS Genet ; 17(4): e1009275, 2021 04.
Article in English | MEDLINE | ID: mdl-33819267

ABSTRACT

Mammalian Hedgehog (HH) signalling pathway plays an essential role in tissue homeostasis and its deregulation is linked to rheumatological disorders. UBR5 is the mammalian homologue of the E3 ubiquitin-protein ligase Hyd, a negative regulator of the Hh-pathway in Drosophila. To investigate a possible role of UBR5 in regulation of the musculoskeletal system through modulation of mammalian HH signaling, we created a mouse model for specific loss of Ubr5 function in limb bud mesenchyme. Our findings revealed a role for UBR5 in maintaining cartilage homeostasis and suppressing metaplasia. Ubr5 loss of function resulted in progressive and dramatic articular cartilage degradation, enlarged, abnormally shaped sesamoid bones and extensive heterotopic tissue metaplasia linked to calcification of tendons and ossification of synovium. Genetic suppression of smoothened (Smo), a key mediator of HH signalling, dramatically enhanced the Ubr5 mutant phenotype. Analysis of HH signalling in both mouse and cell model systems revealed that loss of Ubr5 stimulated canonical HH-signalling while also increasing PKA activity. In addition, human osteoarthritic samples revealed similar correlations between UBR5 expression, canonical HH signalling and PKA activity markers. Our studies identified a crucial function for the Ubr5 gene in the maintenance of skeletal tissue homeostasis and an unexpected mode of regulation of the HH signalling pathway.


Subject(s)
Arthritis, Rheumatoid/genetics , Drosophila Proteins/genetics , Muscle, Skeletal/metabolism , Smoothened Receptor/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cartilage/growth & development , Cartilage/metabolism , Cartilage/pathology , Chondrocytes/metabolism , Disease Models, Animal , Drosophila melanogaster/genetics , Hedgehog Proteins/genetics , Homeostasis/genetics , Humans , Knee Joint/metabolism , Knee Joint/pathology , Mice , Muscle, Skeletal/pathology , Osteogenesis/genetics , Signal Transduction/genetics , Tendons/metabolism , Tendons/pathology
2.
Hum Mol Genet ; 22(25): 5083-95, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-23900076

ABSTRACT

The embryonic epicardium is an important source of cardiovascular precursor cells and paracrine factors that are required for adequate heart formation. Signaling pathways regulated by WT1 that promote heart development have started to be described; however, there is little information on signaling pathways regulated by WT1 that could act in a negative manner. Transcriptome analysis of Wt1KO epicardial cells reveals an unexpected role for WT1 in repressing the expression of interferon-regulated genes that could be involved in a negative regulation of heart morphogenesis. Here, we showed that WT1 is required to repress the expression of the chemokines Ccl5 and Cxcl10 in epicardial cells. We observed an inverse correlation of Wt1 and the expression of Cxcl10 and Ccl5 during epicardium development. Chemokine receptor analyses of hearts from Wt1(gfp/+) mice demonstrate the differential expression of their chemokine receptors in GFP(+) epicardial enriched cells and GFP(-) cells. Functional assays demonstrate that CXCL10 and CCL5 inhibit epicardial cells migration and the proliferation of cardiomyocytes respectively. WT1 regulates the expression levels of Cxcl10 and Ccl5 in epicardial cells directly and indirectly through increasing the levels of IRF7. As epicardial cell reactivation after a myocardial damage is linked with WT1 expression, the present work has potential implications in adult heart repair.


Subject(s)
Chemokine CCL5/biosynthesis , Chemokine CXCL10/biosynthesis , Heart/growth & development , Pericardium/growth & development , WT1 Proteins/genetics , Animals , Chemokine CCL5/genetics , Chemokine CXCL10/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Interferon Regulatory Factor-7/metabolism , Mice , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Pericardium/cytology , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/metabolism , Signal Transduction , WT1 Proteins/biosynthesis
3.
PLoS Genet ; 7(12): e1002404, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22216009

ABSTRACT

There is much interest in the mechanisms that regulate adult tissue homeostasis and their relationship to processes governing foetal development. Mice deleted for the Wilms' tumour gene, Wt1, lack kidneys, gonads, and spleen and die at mid-gestation due to defective coronary vasculature. Wt1 is vital for maintaining the mesenchymal-epithelial balance in these tissues and is required for the epithelial-to-mesenchyme transition (EMT) that generates coronary vascular progenitors. Although Wt1 is only expressed in rare cell populations in adults including glomerular podocytes, 1% of bone marrow cells, and mesothelium, we hypothesised that this might be important for homeostasis of adult tissues; hence, we deleted the gene ubiquitously in young and adult mice. Within just a few days, the mice suffered glomerulosclerosis, atrophy of the exocrine pancreas and spleen, severe reduction in bone and fat, and failure of erythropoiesis. FACS and culture experiments showed that Wt1 has an intrinsic role in both haematopoietic and mesenchymal stem cell lineages and suggest that defects within these contribute to the phenotypes we observe. We propose that glomerulosclerosis arises in part through down regulation of nephrin, a known Wt1 target gene. Protein profiling in mutant serum showed that there was no systemic inflammatory or nutritional response in the mutant mice. However, there was a dramatic reduction in circulating IGF-1 levels, which is likely to contribute to the bone and fat phenotypes. The reduction of IGF-1 did not result from a decrease in circulating GH, and there is no apparent pathology of the pituitary and adrenal glands. These findings 1) suggest that Wt1 is a major regulator of the homeostasis of some adult tissues, through both local and systemic actions; 2) highlight the differences between foetal and adult tissue regulation; 3) point to the importance of adult mesenchyme in tissue turnover.


Subject(s)
Glomerulonephritis/genetics , Homeostasis/genetics , Multiple Organ Failure/genetics , WT1 Proteins/physiology , Animals , Atrophy/genetics , Atrophy/pathology , Cell Lineage/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Deletion , Gene Expression Regulation , Glomerulonephritis/pathology , Gonads/embryology , Gonads/metabolism , Gonads/pathology , Hematopoiesis/genetics , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Kidney Glomerulus/embryology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Multiple Organ Failure/pathology , Pancreas, Exocrine/embryology , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology , Podocytes/metabolism , Podocytes/pathology , Spleen/embryology , Spleen/metabolism , Spleen/pathology , Tamoxifen/pharmacology , WT1 Proteins/genetics
4.
Sci Rep ; 7: 45255, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28345629

ABSTRACT

WT1 is a transcription factor which regulates the epithelial-mesenchymal balance during embryonic development and, if mutated, can lead to the formation of Wilms' tumour, the most common paediatric kidney cancer. Its expression has also been reported in several adult tumour types, including breast cancer, and usually correlates with poor outcome. However, published data is inconsistent and the role of WT1 in this malignancy remains unclear. Here we provide a complete study of WT1 expression across different breast cancer subtypes as well as isoform specific expression analysis. Using in vitro cell lines, clinical samples and publicly available gene expression datasets, we demonstrate that WT1 plays a role in regulating the epithelial-mesenchymal balance of breast cancer cells and that WT1-expressing tumours are mainly associated with a mesenchymal phenotype. WT1 gene expression also correlates with CYP3A4 levels and is associated with poorer response to taxane treatment. Our work is the first to demonstrate that the known association between WT1 expression in breast cancer and poor prognosis is potentially due to cancer-related epithelial-to-mesenchymal transition (EMT) and poor chemotherapy response.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Cytochrome P-450 CYP3A/metabolism , Taxoids/therapeutic use , WT1 Proteins/genetics , WT1 Proteins/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Docetaxel , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , MCF-7 Cells , Mutation , Prognosis , Taxoids/pharmacology , Up-Regulation/drug effects
5.
Dis Model Mech ; 8(8): 903-17, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26035382

ABSTRACT

Wilms' tumours, paediatric kidney cancers, are the archetypal example of tumours caused through the disruption of normal development. The genetically best-defined subgroup of Wilms' tumours is the group caused by biallelic loss of the WT1 tumour suppressor gene. Here, we describe a developmental series of mouse models with conditional loss of Wt1 in different stages of nephron development before and after the mesenchymal-to-epithelial transition (MET). We demonstrate that Wt1 is essential for normal development at all kidney developmental stages under study. Comparison of genome-wide expression data from the mutant mouse models with human tumour material of mutant or wild-type WT1 datasets identified the stage of origin of human WT1-mutant tumours, and emphasizes fundamental differences between the two human tumour groups due to different developmental stages of origin.


Subject(s)
Nephrons/growth & development , Nephrons/metabolism , WT1 Proteins/metabolism , Wilms Tumor/pathology , Animals , Biomarkers/metabolism , Cell Lineage , Gene Expression Regulation, Neoplastic , Genome , Integrases/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , Neoplasm Staging , Nephrons/pathology , Oligonucleotide Array Sequence Analysis , Phenotype , Time-Lapse Imaging , WT1 Proteins/genetics , Wilms Tumor/genetics
6.
Nat Cell Biol ; 16(4): 367-75, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24609269

ABSTRACT

Fuelled by the obesity epidemic, there is considerable interest in the developmental origins of white adipose tissue (WAT) and the stem and progenitor cells from which it arises. Whereas increased visceral fat mass is associated with metabolic dysfunction, increased subcutaneous WAT is protective. There are six visceral fat depots: perirenal, gonadal, epicardial, retroperitoneal, omental and mesenteric, and it is a subject of much debate whether these have a common developmental origin and whether this differs from that for subcutaneous WAT. Here we show that all six visceral WAT depots receive a significant contribution from cells expressing Wt1 late in gestation. Conversely, no subcutaneous WAT or brown adipose tissue arises from Wt1-expressing cells. Postnatally, a subset of visceral WAT continues to arise from Wt1-expressing cells, consistent with the finding that Wt1 marks a proportion of cell populations enriched in WAT progenitors. We show that all visceral fat depots have a mesothelial layer like the visceral organs with which they are associated, and provide several lines of evidence that Wt1-expressing mesothelium can produce adipocytes. These results reveal a major ontogenetic difference between visceral and subcutaneous WAT, and pinpoint the lateral plate mesoderm as a major source of visceral WAT. They also support the notion that visceral WAT progenitors are heterogeneous, and suggest that mesothelium is a source of adipocytes.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , WT1 Proteins/metabolism , Adipocytes/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/embryology , Adipose Tissue, White/cytology , Adipose Tissue, White/embryology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Lineage/genetics , Gene Knock-In Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Tamoxifen/pharmacology , WT1 Proteins/genetics
7.
PLoS One ; 8(4): e62054, 2013.
Article in English | MEDLINE | ID: mdl-23637962

ABSTRACT

There is an increasing need for more efficient generation of transgenic constructs. Here we present a universal multi-site Gateway vector for use in recombineering reactions. Using transgenic mouse models, we show its use for the generation of BAC transgenics and targeting vectors. The modular nature of the vector allows for rapid modification of constructs to generate different versions of the same construct. As such it will help streamline the generation of series of related transgenic models.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Gene Knock-In Techniques/methods , Genetic Engineering/methods , Genetic Vectors/genetics , Recombination, Genetic , Animals , Female , Mice
SELECTION OF CITATIONS
SEARCH DETAIL