Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Am J Physiol Renal Physiol ; 319(2): F335-F344, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32657157

ABSTRACT

Human immunodeficiency virus (HIV) infection of kidney cells can lead to HIV-associated nephropathy (HIVAN) and aggravate the progression of other chronic kidney diseases. Thus, a better understanding of the mechanisms of HIV-induced kidney cell injury is needed for effective therapy against HIV-induced kidney disease progression. We have previously shown that the acetylation and activation of key inflammatory regulators, NF-κB p65 and STAT3, were increased in HIVAN kidneys. Here, we demonstrate the key role of sirtuin 1 (SIRT1) deacetylase in the regulation of NF-κB and STAT3 activity in HIVAN. We found that SIRT1 expression was reduced in the glomeruli of human and mouse HIVAN kidneys and that HIV-1 gene expression was associated with reduced SIRT1 expression and increased acetylation of NF-κB p65 and STAT3 in cultured podocytes. Interestingly, SIRT1 overexpression, in turn, reduced the expression of negative regulatory factor in podocytes stably expressing HIV-1 proviral genes, which was associated with inactivation of NF-κB p65 and a reduction in HIV-1 long terminal repeat promoter activity. In vivo, the administration of the small-molecule SIRT1 agonist BF175 or inducible overexpression of SIRT1 specifically in podocytes markedly attenuated albuminuria, kidney lesions, and expression of inflammatory markers in Tg26 mice. Finally, we showed that the reduction in SIRT1 expression by HIV-1 is in part mediated through miR-34a expression. Together, our data provide a new mechanism of SIRT1 regulation and its downstream effects in HIV-1-infected kidney cells and indicate that SIRT1/miR-34a are potential drug targets to treat HIV-related kidney disease.


Subject(s)
AIDS-Associated Nephropathy/virology , Renal Insufficiency, Chronic/metabolism , Sirtuin 1/metabolism , AIDS-Associated Nephropathy/complications , AIDS-Associated Nephropathy/metabolism , Animals , Humans , Kidney/metabolism , Kidney Glomerulus/metabolism , Kidney Glomerulus/virology , Mice , Podocytes/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/virology , Transcription Factor RelA/metabolism
2.
Physiol Rep ; 7(14): e14172, 2019 07.
Article in English | MEDLINE | ID: mdl-31325249

ABSTRACT

Treatment modalities for kidney disease caused by long-term exposure to heavy metals, such as cadmium (Cd), are limited. Often, chronic, long-term environmental exposure to heavy metal is not recognized in the early stages; therefore, chelation therapy is not an effective option. Extracellular vesicles (EVs) derived from stem cells have been demonstrated to reduce disease pathology in both acute and chronic kidney disease models. To test the ability of EVs derived from human bone marrow mesenchymal stem cells (hBM-MSCs) to treat Cd damage, we generated a Cd-exposed medaka model. This model develops heavy metal-induced cell damage in various organs and tissues, and shows decreased overall survival. Intravenous injection of highly purified EVs from hBM-MSCs repaired the damage to apical and basolateral membranes and mitochondria of kidney proximal tubules, glomerular podocytes, bone deformation, and improved survival. Our system also serves as a model with which to study age- and sex-dependent cell injuries of organs caused by various agents and diseases. The beneficial effects of EVs on the tissue repair process, as shown in our novel Cd-exposed medaka model, may open new broad avenues for interventional strategies.


Subject(s)
Cadmium Poisoning/therapy , Extracellular Vesicles/transplantation , Mesenchymal Stem Cell Transplantation/methods , Animals , Bone Marrow Cells/metabolism , Cadmium Poisoning/metabolism , Cells, Cultured , Extracellular Vesicles/metabolism , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Mesenchymal Stem Cells/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Oryzias , Podocytes/metabolism , Podocytes/pathology
3.
Kidney Int ; 94(6): 1160-1176, 2018 12.
Article in English | MEDLINE | ID: mdl-30366682

ABSTRACT

Mounting evidence suggests that epigenetic modification is important in kidney disease pathogenesis. To determine whether epigenetic regulation is involved in HIV-induced kidney injury, we performed genome-wide methylation profiling and transcriptomic profiling of human primary podocytes infected with HIV-1. Comparison of DNA methylation and RNA sequencing profiles identified several genes that were hypomethylated with corresponding upregulated RNA expression in HIV-infected podocytes. Notably, we found only one hypermethylated gene with corresponding downregulated RNA expression, namely regulator of calcineurin 1 (RCAN1). Further, we found that RCAN1 RNA expression was suppressed in glomeruli in human diabetic nephropathy, IgA nephropathy, and lupus nephritis, and in mouse models of HIV-associated nephropathy and diabetic nephropathy. We confirmed that HIV infection or high glucose conditions suppressed RCAN1 expression in cultured podocytes. This suppression was alleviated upon pretreatment with DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine, suggesting that RCAN1 expression is epigenetically suppressed in the context of HIV infection and diabetic conditions. Mechanistically, increased expression of RCAN1 decreased HIV- or high glucose-induced nuclear factor of activated T cells (NFAT) transcriptional activity. Increased RCAN1 expression also stabilized actin cytoskeleton organization, consistent with the inhibition of the calcineurin pathway. In vivo, knockout of RCAN1 aggravated albuminuria and podocyte injury in mice with Adriamycin-induced nephropathy. Our findings suggest that epigenetic suppression of RCAN1 aggravates podocyte injury in the setting of HIV infection and diabetic nephropathy.


Subject(s)
AIDS-Associated Nephropathy/pathology , Diabetic Nephropathies/pathology , Epigenesis, Genetic , Intracellular Signaling Peptides and Proteins/genetics , Muscle Proteins/genetics , Podocytes/pathology , AIDS-Associated Nephropathy/virology , Animals , Biopsy , Calcium-Binding Proteins , Cells, Cultured , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , DNA-Binding Proteins , Datasets as Topic , Decitabine/pharmacology , Disease Models, Animal , Gene Knockout Techniques , Genome, Human/genetics , Glucose/metabolism , HIV-1 , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Glomerulus/pathology , Mice , Mice, Knockout , Muscle Proteins/metabolism , NFATC Transcription Factors/metabolism , Podocytes/virology , Primary Cell Culture , Up-Regulation
4.
PLoS One ; 9(6): e100660, 2014.
Article in English | MEDLINE | ID: mdl-24949636

ABSTRACT

MYH9 encodes non-muscle myosin heavy chain IIA (NMMHCIIA), the predominant force-generating ATPase in non-muscle cells. Several lines of evidence implicate a role for MYH9 in podocytopathies. However, NMMHCIIA's function in podocytes remains unknown. To better understand this function, we performed immuno-precipitation followed by mass-spectrometry proteomics to identify proteins interacting with the NMMHCIIA-enriched actin-myosin complexes. Computational analyses revealed that these proteins belong to functional networks including regulators of cytoskeletal organization, metabolism and networks regulated by the HIV-1 gene nef. We further characterized the subcellular localization of NMMHCIIA within podocytes in vivo, and found it to be present within the podocyte major foot processes. Finally, we tested the effect of loss of MYH9 expression in podocytes in vitro, and found that it was necessary for cytoskeletal organization. Our results provide the first survey of NMMHCIIA-enriched actin-myosin-interacting proteins within the podocyte, demonstrating the important role of NMMHCIIA in organizing the elaborate cytoskeleton structure of podocytes. Our characterization of NMMHCIIA's functions goes beyond the podocyte, providing important insights into its general molecular role.


Subject(s)
Actins/metabolism , Molecular Motor Proteins/metabolism , Myosin Heavy Chains/metabolism , Proteomics , Actins/biosynthesis , Animals , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/metabolism , Gene Expression Regulation , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Mice , Molecular Motor Proteins/biosynthesis , Multiprotein Complexes/metabolism , Myosin Heavy Chains/biosynthesis , Podocytes/metabolism , Protein Interaction Maps , nef Gene Products, Human Immunodeficiency Virus/metabolism
5.
Am J Physiol Renal Physiol ; 304(8): F1127-36, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23389453

ABSTRACT

The Notch pathway is an evolutionarily conserved signaling cascade that is critical in kidney development and has also been shown to play a pathogenetic role in a variety of kidney diseases. We have previously shown that the Notch signaling pathway is activated in human immunodeficiency virus-associated nephropathy (HIVAN) as well as in a rat model of the disease. In this study, we examined Notch signaling in the well established Tg26 mouse model of HIVAN. Notch signaling components were distinctly upregulated in the kidneys of these mice as well as in immortalized podocytes derived from these mice. Notch1 and Notch4 were upregulated in the Tg26 glomeruli, and Notch4 was also expressed in tubules. Notch ligands Jagged1, Jagged2, Delta-like1, and Delta-like 4 were all upregulated in the tubules of Tg26 mice, but glomeruli showed minimal expression of Notch ligands. To examine a potential pathogenetic role for Notch in HIVAN, Tg26 mice were treated with GSIXX, a gamma secretase inhibitor that blocks Notch signaling. Strikingly, GSIXX treatment resulted in significant improvement in both histological kidney injury scores and renal function. GSIXX-treated Tg26 mice also showed diminished podocyte proliferation and dedifferentiation, cellular hallmarks of the disease. Moreover, GSIXX blocked podocyte proliferation in vitro induced by HIV proteins Nef and Tat. These studies suggest that Notch signaling can promote HIVAN progression and that Notch inhibition may be a viable treatment strategy for HIVAN.


Subject(s)
AIDS-Associated Nephropathy/metabolism , Podocytes/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Notch1/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , AIDS-Associated Nephropathy/drug therapy , AIDS-Associated Nephropathy/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Cell Dedifferentiation/drug effects , Cell Dedifferentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Line, Transformed , Dibenzazepines/pharmacology , Dipeptides/pharmacology , Disease Models, Animal , Disease Progression , Humans , Kidney/drug effects , Kidney/pathology , Kidney/physiology , Ligands , Mice , Mice, Inbred Strains , Mice, Transgenic , Podocytes/cytology , Podocytes/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch4 , Receptors, Notch/antagonists & inhibitors , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology
6.
AIDS ; 26(7): 797-803, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22313957

ABSTRACT

BACKGROUND: The continuing disease burden of HIV-associated nephropathy (HIVAN) warrants better elucidation of its pathogenic mechanisms. Given that loss of MYH9 function causes a Mendelian renal disease, we hypothesized that renal expression of MYH9 is down-regulated by HIV-1 in HIVAN pathogenesis. METHOD AND RESULTS: Using immunofluorescence, we determined that glomerular expression of MYH9 was reduced in the kidneys of HIV-1 transgenic mice. We further determined that Myh9 expression was reduced in HIV-1 transgenic podocytes, statistically significantly at the protein level, and that MYH9 expression was significantly reduced at protein and message level in human podocytes transduced with HIV-1. In analyzing expression in human tissue, we confirmed that MYH9 is abundantly expressed in glomeruli, and podocytes specifically. Finally, we found that MYH9 expression was significantly reduced in human glomeruli in the setting of HIVAN. CONCLUSION: We conclude that the podocyte host response to HIV-1 includes down-regulation of MYH9 expression, and hypothesize that this down-regulation might play a role in the pathogenesis of HIVAN.


Subject(s)
AIDS-Associated Nephropathy/physiopathology , HIV-1/metabolism , Nonmuscle Myosin Type IIA/metabolism , Podocytes/pathology , AIDS-Associated Nephropathy/metabolism , Animals , Disease Models, Animal , HIV Infections/metabolism , HIV Infections/physiopathology , Humans , Mice , Mice, Transgenic , Molecular Motor Proteins/metabolism , Myosin Heavy Chains/metabolism , Podocytes/metabolism , Real-Time Polymerase Chain Reaction
7.
Kidney Int ; 76(1): 54-62, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19367327

ABSTRACT

The regulation of epithelial branching morphogenesis by bone morphogenetic protein-7 depends, in part, on functionally defined cyclic adenosine monophosphate (cAMP)-dependent protein kinases. We previously identified protein kinase-X (PRKX), a cAMP-dependent kinase, as a regulator of epithelial morphogenesis during kidney development and found that it binds to and phosphorylates Polycystin-1. Overexpression of PRKX stimulates renal epithelial cell migration, tubulogenesis, ureteric bud branching, and glomerular induction in embryonic mouse kidney explants in organ cultures. Here we determined the physiological functions of endogenous PRKX. Knockdown by siRNA of PRKX gene expression in a human fetal collecting tubule (HFCT) cell line exceeded 70% and resulted in decreased cell migration and increased adhesion of the cells to a collagen I matrix. In embryonic mouse kidney explants, the same degree of knockdown decreased ureteric bud branching and glomerular induction. Because PRKX BAG-3 PIN-1 and MAGI-1 are all expressed in ureteric bud derivatives, we tested for interactions among them and found that PRKX binds to all three proteins through its WW domain as determined by TransSignal domain arrays, and it coimmunoprecipitated with Pin-1 in HFCT cell lysates. These studies suggest that Polycystin-1 and Pin-1 may mediate the function of PRKX in kidney development.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Kidney/cytology , Kidney/embryology , Peptidylprolyl Isomerase/metabolism , Protein Kinases/metabolism , TRPP Cation Channels/metabolism , Animals , Cell Adhesion/genetics , Cell Movement/genetics , Female , Mice , Morphogenesis/genetics , NIMA-Interacting Peptidylprolyl Isomerase , Organ Culture Techniques , Pregnancy , Protein Serine-Threonine Kinases
8.
Biochim Biophys Acta ; 1782(1): 1-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17980165

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is a common, genetically determined developmental disorder of the kidney that is characterized by cystic expansion of renal tubules and is caused by truncating mutations and haplo-insufficiency of the PKD1 gene. Several defects in cAMP-mediated proliferation and ion secretion have been detected in ADPKD cyst-lining epithelia. Unlike the ubiquitous PKA, the cAMP-dependent CREB-kinase, Protein Kinase X (PRKX) is developmentally regulated, tissue restricted and induces renal epithelial cell migration, and tubulogenesis in vitro as well as branching morphogenesis of ureteric bud in developing kidneys. The possibility of functional interactions between PKD1-encoded polycystin-1 and PRKX was suggested by the renal co-distribution of PRKX and polycystin-1 and the binding and phosphorylation of the C-terminal of polycystin-1 by PRKX at S4166 in vitro. Early consequences of PKD1 mutation include increased tubule epithelial cell-matrix adhesion, decreased migration, reduced ureteric bud branching and aberrant renal tubule dilation. To determine whether PRKX might counteract the adverse effects of PKD1 mutation, human ADPKD epithelial cell lines were transfected with constitutively active PRKX and shown to rescue characteristic adhesion and migration defects. In addition, the co-injection of constitutively active PRKX with inhibitory pMyr-EGFP-PKD1 into the ureteric buds of mouse embryonic kidneys in organ culture resulted in restoration of normal branching morphogenesis without cystic tubular dilations. These results suggest that PRKX can restore normal function to PKD1-deficient kidneys and have implications for the development of preventative therapy for ADPKD.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , TRPP Cation Channels/metabolism , Animals , Cell Adhesion , Cell Line , Cell Movement , Cell Shape , Humans , Kidney/metabolism , Mice , Organ Culture Techniques , Phosphorylation , Polycystic Kidney, Autosomal Dominant/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics
9.
Biochim Biophys Acta ; 1762(7): 647-55, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16797938

ABSTRACT

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a very common lethal monogenetic disease with significant morbidities and a high likelihood of progression to renal failure for which there is no proven disease-specific therapy currently available for clinical use. Human ADPKD cystic epithelia have proliferative abnormalities mediated by EGFR over-expression and mispolarization leading autocrine response to EGF family ligands. We now show that apical localization of EGFR complexes in normal fetal and ADPKD epithelia is associated with heterodimerization of EGFR(HER-1) with HER-2(neu/ErbB2), while basal membrane localization in normal adult renal epithelia is associated with EGFR(HER-1) homodimers. Since ADPKD epithelial cells have reduced migratory function, this was used as a bioassay to evaluate the ability of compounds to rescue the aberrant human ADPKD phenotype. General tyrosine kinase inhibition by herbimycin and specific inhibition of HER-2(neu/ErbB2) by AG825 or pretreatment with ErbB2 siRNA reversed the migration defect of ADPKD epithelia. Selective inhibition of EGFR(HER-1) showed partial rescue. Increased ADPKD cell migration after inhibition of p38MAP kinase but not of PI3-kinase implicated p38MAPK downstream of HER-2(neu/ErbB2) stimulation. Daily administration of AG825 to PKD1 null heterozygous mice significantly inhibited the development of renal cysts. These studies implicate HER2(neu/ErbB2) as an effector of apical EGFR complex mispolarization and that its inhibition should be considered a candidate for clinical therapy of ADPKD.


Subject(s)
Polycystic Kidney Diseases/genetics , Receptor, ErbB-2/genetics , Cell Movement , Cloning, Molecular , Epithelial Cells/metabolism , Epithelial Cells/pathology , ErbB Receptors/metabolism , Humans , Immunoblotting , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , Polycystic Kidney Diseases/therapy , RNA, Small Interfering/genetics , Receptor, ErbB-2/antagonists & inhibitors , Reference Values , Reverse Transcriptase Polymerase Chain Reaction
10.
Nephron Exp Nephrol ; 103(2): e50-4, 2006.
Article in English | MEDLINE | ID: mdl-16543764

ABSTRACT

Advances in microscopy now enable researchers to easily acquire multi-channel three-dimensional (3D) images and 3D time series (4D). However, processing, analyzing, and displaying this data can often be difficult and time- consuming. We discuss some of the software tools and techniques that are available to accomplish these tasks.


Subject(s)
Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Microscopy , Software , Animals , Image Processing, Computer-Assisted/instrumentation , Image Processing, Computer-Assisted/methods
11.
J Am Soc Nephrol ; 16(12): 3543-52, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16236808

ABSTRACT

The human protein kinase X (PRKX) gene was identified previously as a cAMP-dependent serine/threonine kinase that is aberrantly expressed in autosomal dominant polycystic disease kidneys and normally expressed in fetal kidneys. The PRKX kinase belongs to a serine/threonine kinase family that is phylogenetically and functionally distinct from classical protein kinase A kinases. Expression of PRKX activates cAMP-dependent renal epithelial cell migration and tubular morphogenesis in cell culture, suggesting that it might regulate branching growth of the collecting duct system in the fetal kidney. With the use of a mouse embryonic kidney organ culture system that recapitulates early kidney development in vitro, it is demonstrated that lentiviral vector-driven expression of a constitutively active, cAMP-independent PRKX in the ureteric bud epithelium stimulates branching morphogenesis and results in a 2.5-fold increase in glomerular number. These results suggest that PRKX stimulates epithelial branching morphogenesis by activating cell migration and support a role for this kinase in the regulation of nephrogenesis and of collecting system development in the fetal kidney.


Subject(s)
Kidney/embryology , Organogenesis/physiology , Protein Kinases/genetics , Ureter/embryology , Animals , Cells, Cultured , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , In Vitro Techniques , Kidney/pathology , Mice , Organogenesis/genetics , RNA, Messenger/analysis , Sensitivity and Specificity , Ureter/pathology
12.
Dev Biol ; 286(1): 16-30, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16122726

ABSTRACT

The polycystic kidney disease (PKD1) gene-encoded protein, polycystin-1, is developmentally regulated, with highest expression levels seen in normal developing kidneys, where it is distributed in a punctate pattern at the basal surface of ureteric bud epithelia. Overexpression in ureteric epithelial cell membranes of an inhibitory pMyr-GFP-PKD1 fusion protein via a retroviral (VVC) delivery system and microinjection into the ureteric bud lumen of embryonic day 11 mouse metanephric kidneys resulted in disrupted branching morphogenesis. Using confocal quantitative analysis, significant reductions were measured in the numbers of ureteric bud branch points and tips, as well as in the total ureteric bud length, volume and area, while significant increases were seen as dilations of the terminal branches, where significant increases in outer diameter and volumes were measured. Microinjection of an activating 5TM-GFP-PKD1 fusion protein had an opposite effect and showed significant increases in ureteric bud length and area. These are the first studies to experimentally manipulate polycystin-1 expression by transduction in the embryonic mouse kidney and suggest that polycystin-1 plays a critical role in the regulation of epithelial morphogenesis during renal development.


Subject(s)
Kidney/embryology , Proteins/antagonists & inhibitors , Ureter/embryology , Animals , Cell Line , Gene Expression Regulation, Developmental , Genetic Vectors , Humans , Mice , Morphogenesis , Polycystic Kidney, Autosomal Dominant/embryology , Polycystic Kidney, Autosomal Dominant/genetics , Proteins/genetics , Proteins/physiology , Recombinant Fusion Proteins/genetics , TRPP Cation Channels
13.
Organogenesis ; 1(1): 14-21, 2004 Jul.
Article in English | MEDLINE | ID: mdl-19521555

ABSTRACT

During kidney development, the growth and development of the stromal and nephrogenic mesenchyme cell populations and the ureteric bud epithelium is tightly coupled through intricate reciprocal signaling mechanisms between these three tissue compartments. Midkine, a target gene activated by retinoid signaling in the metanephros, encodes a secreted polypeptide with mitogenic and anti-apoptotic activities in a wide variety of cell types. Using immmunohistochemical methods we demonstrated that Midkine is found in the uninduced mesenchyme at the earliest stages of metanephric kidney development and only subsequently concentrated in the ureteric bud epithelium and basement membrane. The biological effects of purified recombinant Midkine were analyzed in metanephric organ culture experiments carried out in serum-free defined media. These studies revealed that Midkine selectively promoted the overgrowth of the Pax-2 and N-CAM positive nephrogenic mesenchymal cells, failed to stimulate expansion of the stromal compartment and suppressed branching morphogenesis of the ureteric bud. Midkine suppressed apoptosis and stimulated cellular proliferation of the nephrogenic mesenchymal cells, and was capable of maintaining the viability of isolated mesenchymes cultured in the absence of the ureteric bud. These results suggest that Midkine may regulate the balance of epithelial and stromal progenitor cell populations of the metanephric mesenchyme during renal organogenesis.

14.
J Biol Chem ; 278(42): 41420-30, 2003 Oct 17.
Article in English | MEDLINE | ID: mdl-12882970

ABSTRACT

WT1 encodes a transcription factor involved in kidney development and tumorigenesis. Using representational difference analysis, we identified a new set of WT1 targets, including a homologue of the Drosophila receptor tyrosine kinase regulator, sprouty. Sprouty1 was up-regulated in cell lines expressing wild-type but not mutant WT1. WT1 bound to the endogenous sprouty1 promoter in vivo and directly regulated sprouty1 through an early growth response gene-1 binding site. Expression of Sprouty1 and WT1 overlapped in the developing metanephric mesenchyme, and Sprouty1, like WT1, plays a key role in the early steps of glomerulus formation. Disruption of Sprouty1 expression in embryonic kidney explants by antisense oligonucleotides reduced condensation of the metanephric mesenchyme, leading to a decreased number of glomeruli. In addition, sprouty1 was expressed in the ureteric tree and antisense-treated ureteric trees had cystic lumens. Therefore, sprouty1 represents a physiologically relevant target gene of WT1 during kidney development.


Subject(s)
Gene Expression Regulation, Developmental , Kidney/embryology , Membrane Proteins/metabolism , Membrane Proteins/physiology , Phosphoproteins/metabolism , Phosphoproteins/physiology , WT1 Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Binding Sites , Blotting, Northern , Chromatin/metabolism , Cloning, Molecular , Drosophila , Genes, Reporter , Immunohistochemistry , Kidney/metabolism , Kidney Glomerulus/embryology , Mice , Models, Genetic , NIH 3T3 Cells , Oligonucleotides/chemistry , Oligonucleotides, Antisense/pharmacology , Precipitin Tests , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Transcription, Genetic , Transfection , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...