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.
J Clin Invest ; 132(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35166232

ABSTRACT

Kidney function decreases with age and may soon limit millions of lives as the proportion of the population over 70 years of age increases. Glycogen synthase kinase 3ß (GSK3ß) is involved with metabolism and may have a role in kidney senescence, positioning it as a target for complications from chronic kidney disease. However, different studies suggest GSK3 has contrasting effects. In this issue of the JCI, Fang et al. explored the function of GSK3ß and the interplay with lithium using human tissue and mouse models. Notably, GSK3ß was overexpressed and activated in aging mice, and depleting GSK3ß reduced senescence and glomerular aging. In this Commentary, we explore the similarities and differences between Fang et al. and previous findings by Hurcombe et al. These findings should prompt further study of lithium and other GSK3ß inhibitors as a means of extending glomerular function in individuals with chronic kidney disease.


Subject(s)
Glycogen Synthase Kinase 3 , Kidney , Aging , Animals , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Kidney Glomerulus/metabolism , Mice
2.
Circulation ; 134(1): 61-72, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27358438

ABSTRACT

BACKGROUND: In nephrotic syndrome, damage to the podocytes of the kidney produces severe hypercholesterolemia for which novel treatments are urgently needed. PCSK9 (proprotein convertase subtilisin/kexin type 9) has emerged as an important regulator of plasma cholesterol levels and therapeutic target. Here, we tested the role of PCSK9 in mediating the hypercholesterolemia of nephrotic syndrome. METHODS: PCSK9 and plasma lipids were studied in nephrotic syndrome patients before and after remission of disease, mice with genetic ablation of the podocyte (Podocyte Apoptosis Through Targeted Activation of Caspase-8, Pod-ATTAC mice) and mice treated with nephrotoxic serum (NTS), which triggers immune-mediated podocyte damage. In addition, mice with hepatic deletion of Pcsk9 were treated with NTS to determine the contribution of PCSK9 to the dyslipidemia of nephrotic syndrome. RESULTS: Patients with nephrotic syndrome showed a decrease in plasma cholesterol and plasma PCSK9 on remission of their disease (P<0.05, n=47-50). Conversely, Pod-ATTAC mice and NTS-treated mice showed hypercholesterolemia and a 7- to 24-fold induction in plasma PCSK9. The induction of plasma PCSK9 appeared to be attributable to increased secretion of PCSK9 from the hepatocyte coupled with decreased clearance. Interestingly, knockout of Pcsk9ameliorated the effects of NTS on plasma lipids. Thus, in the presence of NTS, mice lacking hepatic Pcsk9 showed a 40% to 50% decrease in plasma cholesterol and triglycerides. Moreover, the ability of NTS treatment to increase the percentage of low-density lipoprotein-associated cholesterol (from 9% in vehicle-treated Flox mice to 47% after NTS treatment), was lost in mice with hepatic deletion of Pcsk9 (5% in both the presence and absence of NTS). CONCLUSIONS: Podocyte damage triggers marked inductions in plasma PCSK9, and knockout of Pcsk9 ameliorates dyslipidemia in a mouse model of nephrotic syndrome. These data suggest that PCSK9 inhibitors may be beneficial in patients with nephrotic syndrome-associated hypercholesterolemia.


Subject(s)
Hypercholesterolemia/etiology , Nephrotic Syndrome/complications , Proprotein Convertase 9/physiology , Animals , Humans , Hypercholesterolemia/enzymology , Lipids/blood , Liver/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephrotic Syndrome/blood , Nephrotic Syndrome/enzymology , Podocytes/pathology , Proprotein Convertase 9/deficiency , Proprotein Convertase 9/genetics , Proprotein Convertase 9/therapeutic use , Recombinant Proteins/therapeutic use
3.
Development ; 142(7): 1254-66, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25804736

ABSTRACT

Development of the metanephric kidney depends on tightly regulated interplay between self-renewal and differentiation of a nephron progenitor cell (NPC) pool. Several key factors required for the survival of NPCs have been identified, including fibroblast growth factor (FGF) signaling and the transcription factor Wilms' tumor suppressor 1 (WT1). Here, we present evidence that WT1 modulates FGF signaling by activating the expression of growth arrest-specific 1 (Gas1), a novel WT1 target gene and novel modulator of FGF signaling. We show that WT1 directly binds to a conserved DNA binding motif within the Gas1 promoter and activates Gas1 mRNA transcription in NPCs. We confirm that WT1 is required for Gas1 expression in kidneys in vivo. Loss of function of GAS1 in vivo results in hypoplastic kidneys with reduced nephron mass due to premature depletion of NPCs. Although kidney development in Gas1 knockout mice progresses normally until E15.5, NPCs show decreased rates of proliferation at this stage and are depleted as of E17.5. Lastly, we show that Gas1 is selectively required for FGF-stimulated AKT signaling in vitro. In summary, our data suggest a model in which WT1 modulates receptor tyrosine kinase signaling in NPCs by directing the expression of Gas1.


Subject(s)
Cell Cycle Proteins/metabolism , Fibroblast Growth Factors/metabolism , Nephrons/metabolism , Signal Transduction , Stem Cells/metabolism , WT1 Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Proliferation , DNA/genetics , Enzyme Activation/drug effects , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice, Knockout , Models, Animal , Nephrons/abnormalities , Nephrons/embryology , Nephrons/pathology , Organ Culture Techniques , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-ret/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
J Biol Chem ; 288(25): 18407-20, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23645677

ABSTRACT

Gain-of-function mutations in the canonical transient receptor potential 6 (TRPC6) gene are a cause of autosomal dominant focal segmental glomerulosclerosis (FSGS). The mechanisms whereby abnormal TRPC6 activity results in proteinuria remain unknown. The ERK1/2 MAPKs are activated in glomeruli and podocytes in several proteinuric disease models. We therefore examined whether FSGS-associated mutations in TRPC6 result in activation of these kinases. In 293T cells and cultured podocytes, overexpression of gain-of-function TRPC6 mutants resulted in increased ERK1/2 phosphorylation, an effect dependent upon channel function. Pharmacologic inhibitor studies implicated several signaling mediators, including calmodulin and calcineurin, supporting the importance of TRPC6-mediated calcium influx in this process. Through medium transfer experiments, we uncovered two distinct mechanisms for ERK activation by mutant TRPC6, a cell-autonomous, EGF receptor-independent mechanism and a non-cell-autonomous mechanism involving metalloprotease-mediated release of a presumed EGF receptor ligand. The inhibitors KN-92 and H89 were able to block both pathways in mutant TRPC6 expressing cells as well as the prolonged elevation of intracellular calcium levels upon carbachol stimulation seen in these cells. However, these effects appear to be independent of their effects on calcium/calmodulin-dependent protein kinase II and PKA, respectively. Phosphorylation of Thr-70, Ser-282, and Tyr-31/285 were not necessary for ERK activation by mutant TRPC6, although a phosphomimetic TRPC6 S282E mutant was capable of ERK activation. Taken together, these results identify two pathways downstream of mutant TRPC6 leading to ERK activation that may play a role in the development of FSGS.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , TRPC Cation Channels/physiology , Animals , Benzylamines/pharmacology , Calcineurin/metabolism , Calmodulin/metabolism , Cell Line , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , HEK293 Cells , Humans , Immunoblotting , Isoquinolines/pharmacology , Phosphorylation/drug effects , Podocytes/cytology , Podocytes/drug effects , Podocytes/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfonamides/pharmacology , TRPC Cation Channels/genetics , TRPC6 Cation Channel
5.
Cancer Cell ; 20(6): 768-80, 2011 Dec 13.
Article in English | MEDLINE | ID: mdl-22172722

ABSTRACT

Angiogenesis is regulated by the balance of proangiogenic VEGF(165) and antiangiogenic VEGF(165)b splice isoforms. Mutations in WT1, the Wilms' tumor suppressor gene, suppress VEGF(165)b and cause abnormal gonadogenesis, renal failure, and Wilms' tumors. In WT1 mutant cells, reduced VEGF(165)b was due to lack of WT1-mediated transcriptional repression of the splicing-factor kinase SRPK1. WT1 bound to the SRPK1 promoter, and repressed expression through a specific WT1 binding site. In WT1 mutant cells SRPK1-mediated hyperphosphorylation of the oncogenic RNA binding protein SRSF1 regulated splicing of VEGF and rendered WT1 mutant cells proangiogenic. Altered VEGF splicing was reversed by wild-type WT1, knockdown of SRSF1, or SRPK1 and inhibition of SRPK1, which prevented in vitro and in vivo angiogenesis and associated tumor growth.


Subject(s)
Neovascularization, Pathologic/genetics , Protein Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor B/genetics , WT1 Proteins/genetics , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Denys-Drash Syndrome/genetics , Denys-Drash Syndrome/metabolism , Denys-Drash Syndrome/pathology , Gene Expression , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Neoplasms/blood supply , Nuclear Proteins/metabolism , Podocytes/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Transport , RNA Interference , RNA Splicing/genetics , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
J Am Soc Nephrol ; 22(7): 1286-96, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21719793

ABSTRACT

Paracrine signaling between podocytes and glomerular endothelial cells through vascular endothelial growth factor A (VEGFA) maintains a functional glomerular filtration barrier. Heparan sulfate proteoglycans (HSPGs), located on the cell surface or in the extracellular matrix, bind signaling molecules such as VEGFA and affect their local concentrations, but whether modulation of these moieties promotes normal crosstalk between podocytes and endothelial cells is unknown. Here, we found that the transcription factor Wilms' Tumor 1 (WT1) modulates VEGFA and FGF2 signaling by increasing the expression of the 6-O-endosulfatases Sulf1 and Sulf2, which remodel the heparan sulfate 6-O-sulfation pattern in the extracellular matrix. Mice deficient in both Sulf1 and Sulf2 developed age-dependent proteinuria as a result of ultrastructural abnormalities in podocytes and endothelial cells, a phenotype similar to that observed in children with WT1 mutations and in Wt1(+/-) mice. These kidney defects associated with a decreased distribution of VEGFA in the glomerular basement membrane and on endothelial cells. Collectively, these data suggest that WT1-dependent sulfatase expression plays a critical role in maintaining the glomerular filtration barrier by modulating the bioavailability of growth factors, thereby promoting normal crosstalk between podocytes and endothelial cells.


Subject(s)
Kidney Glomerulus/enzymology , Sulfatases/metabolism , Sulfotransferases/metabolism , WT1 Proteins/metabolism , Animals , Cell Communication , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation , Heterozygote , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mutation , Permeability , Promoter Regions, Genetic , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...