Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Development ; 149(14)2022 07 15.
Article in English | MEDLINE | ID: mdl-35877077

ABSTRACT

The mammalian kidney is composed of thousands of nephrons that are formed through reiterative induction of a mesenchymal-to-epithelial transformation by a population of nephron progenitor cells. The number of nephrons in human kidneys ranges from several hundred thousand to nearly a million, and low nephron number has been implicated as a risk factor for kidney disease as an adult. Bmp7 is among a small number of growth factors required to support the proliferation and self-renewal of nephron progenitor cells, in a process that will largely determine the final nephron number. Once induced, each nephron begins as a simple tubule that undergoes extensive proliferation and segmental differentiation. Bmp7 is expressed both by nephron progenitor cells and the ureteric bud derivative branches that induce new nephrons. Here, we show that, in mice, Bmp7 expressed by progenitor cells has a major role in determining nephron number; nephron number is reduced to one tenth its normal value in its absence. Postnatally, Bmp7 also drives proliferation of the proximal tubule cells, and these ultimately constitute the largest segment of the nephron. Bmp7 appears to act through Smad 1,5,9(8), p38 and JNK MAP kinase. In the absence of Bmp7, nephrons undergo a hypertrophic process that involves p38. Following a global inactivation of Bmp7, we also see evidence for Bmp7-driven growth of the nephron postnatally. Thus, we identify a role for Bmp7 in supporting the progenitor population and driving expansion of nephrons to produce a mature kidney.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Kidney , Nephrons , Animals , Cell Differentiation , Humans , Kidney Tubules, Proximal , Mammals , Mice , Nephrons/metabolism , Stem Cells
2.
Development ; 145(14)2018 07 18.
Article in English | MEDLINE | ID: mdl-29945864

ABSTRACT

Epigenetic regulation of gene expression has a crucial role allowing for the self-renewal and differentiation of stem and progenitor populations during organogenesis. The mammalian kidney maintains a population of self-renewing stem cells that differentiate to give rise to thousands of nephrons, which are the functional units that carry out filtration to maintain physiological homeostasis. The polycomb repressive complex 2 (PRC2) epigenetically represses gene expression during development by placing the H3K27me3 mark on histone H3 at promoter and enhancer sites, resulting in gene silencing. To understand the role of PRC2 in nephron differentiation, we conditionally inactivated the Eed gene, which encodes a nonredundant component of the PRC2 complex, in nephron progenitor cells. Resultant kidneys were smaller and showed premature loss of progenitor cells. The progenitors in Eed mutant mice that were induced to differentiate did not develop into properly formed nephrons. Lhx1, normally expressed in the renal vesicle, was overexpressed in kidneys of Eed mutant mice. Thus, PRC2 has a crucial role in suppressing the expression of genes that maintain the progenitor state, allowing nephron differentiation to proceed.


Subject(s)
Cell Differentiation/physiology , Epigenesis, Genetic/physiology , Gene Expression Regulation, Developmental/physiology , Nephrons/embryology , Polycomb Repressive Complex 2/biosynthesis , Stem Cells/metabolism , Animals , LIM-Homeodomain Proteins/biosynthesis , LIM-Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Mutation , Nephrons/cytology , Polycomb Repressive Complex 2/genetics , Stem Cells/cytology , Transcription Factors/biosynthesis , Transcription Factors/genetics
3.
Am J Pathol ; 188(1): 84-94, 2018 01.
Article in English | MEDLINE | ID: mdl-29107072

ABSTRACT

Ectopic cAMP signaling is pathologic in polycystic kidney disease; however, its spatiotemporal actions are unclear. We characterized the expression of phosphorylated Creb (p-Creb), a target and mediator of cAMP signaling, in developing and cystic kidney models. We also examined tubule-specific effects of cAMP analogs in cystogenesis in embryonic kidney explants. In wild-type mice, p-Creb marked nephron progenitors (NP), early epithelial NP derivatives, ureteric bud, and cortical stroma; p-Creb was present in differentiated thick ascending limb of Henle, collecting duct, and stroma; however, it disappeared in mature NP-derived proximal tubules. In Six2cre;Frs2αFl/Fl mice, a renal cystic model, ectopic p-Creb stained proximal tubule-derived cystic segments that lost the differentiation marker lotus tetragonolobus lectin. Furthermore, lotus tetragonolobus lectin-negative/p-Creb-positive cyst segments (re)-expressed Ncam1, Pax2, and Sox9 markers of immature nephron structures and dedifferentiated proximal tubules after acute kidney injury. These dedifferentiation markers were co-expressed with p-Creb in renal cysts in Itf88 knockout mice subjected to ischemia and Six2cre;Pkd1Fl/Fl mice, other renal cystogenesis models. 8-Br-cAMP addition to wild-type embryonic kidney explants induced proximal tubular cystogenesis and p-Creb expression; these effects were blocked by co-addition of protein kinase A inhibitor. Thus p-Creb/cAMP signaling is appropriate in NP and early nephron derivatives, but disappears in mature proximal tubules. Moreover, ectopic p-Creb expression/cAMP signaling marks dedifferentiated proximal tubular cystic segments. Furthermore, proximal tubules are predisposed to become cystic after cAMP stimulation.


Subject(s)
Cell Dedifferentiation/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Kidney Diseases, Cystic/metabolism , Kidney Tubules, Proximal/metabolism , Animals , Disease Models, Animal , Kidney Diseases, Cystic/pathology , Kidney Tubules, Proximal/pathology , Mice , Mice, Knockout , Phosphorylation
4.
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
5.
J Am Soc Nephrol ; 26(9): 2097-104, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25636411

ABSTRACT

The transcription factor Wilms' tumor suppressor 1 (WT1) is key to podocyte development and viability; however, WT1 transcriptional networks in podocytes remain elusive. We provide a comprehensive analysis of the genome-wide WT1 transcriptional network in podocytes in vivo using chromatin immunoprecipitation followed by sequencing (ChIPseq) and RNA sequencing techniques. Our data show a specific role for WT1 in regulating the podocyte-specific transcriptome through binding to both promoters and enhancers of target genes. Furthermore, we inferred a podocyte transcription factor network consisting of WT1, LMX1B, TCF21, Fox-class and TEAD family transcription factors, and MAFB that uses tissue-specific enhancers to control podocyte gene expression. In addition to previously described WT1-dependent target genes, ChIPseq identified novel WT1-dependent signaling systems. These targets included components of the Hippo signaling system, underscoring the power of genome-wide transcriptional-network analyses. Together, our data elucidate a comprehensive gene regulatory network in podocytes suggesting that WT1 gene regulatory function and podocyte cell-type specification can best be understood in the context of transcription factor-regulatory element network interplay.


Subject(s)
Gene Expression Regulation , Podocytes , Repressor Proteins/genetics , Signal Transduction/genetics , Transcriptome , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Chromatin Immunoprecipitation , Forkhead Transcription Factors/genetics , Genomics , Hippo Signaling Pathway , LIM-Homeodomain Proteins/genetics , MafB Transcription Factor/genetics , Mice , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sequence Analysis, DNA , Sequence Analysis, RNA , Transcription Factors/genetics , WT1 Proteins
6.
J Neurosci ; 33(46): 17995-8007, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24227711

ABSTRACT

During development, Schwann cells extend lamellipodia-like processes to segregate large- and small-caliber axons during the process of radial sorting. Radial sorting is a prerequisite for myelination and is arrested in human neuropathies because of laminin deficiency. Experiments in mice using targeted mutagenesis have confirmed that laminins 211, 411, and receptors containing the ß1 integrin subunit are required for radial sorting; however, which of the 11 α integrins that can pair with ß1 forms the functional receptor is unknown. Here we conditionally deleted all the α subunits that form predominant laminin-binding ß1 integrins in Schwann cells and show that only α6ß1 and α7ß1 integrins are required and that α7ß1 compensates for the absence of α6ß1 during development. The absence of either α7ß1 or α6ß1 integrin impairs the ability of Schwann cells to spread and to bind laminin 211 or 411, potentially explaining the failure to extend cytoplasmic processes around axons to sort them. However, double α6/α7 integrin mutants show only a subset of the abnormalities found in mutants lacking all ß1 integrins, and a milder phenotype. Double-mutant Schwann cells can properly activate all the major signaling pathways associated with radial sorting and show normal Schwann cell proliferation and survival. Thus, α6ß1 and α7ß1 are the laminin-binding integrins required for axonal sorting, but other Schwann cell ß1 integrins, possibly those that do not bind laminins, may also contribute to radial sorting during peripheral nerve development.


Subject(s)
Axons/physiology , Integrin alpha6beta1/physiology , Integrins/physiology , Schwann Cells/physiology , Animals , Animals, Newborn , Axons/ultrastructure , Cell Proliferation , Cells, Cultured , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Schwann Cells/ultrastructure
7.
J Proteome Res ; 13(11): 4901-9, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25300029

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is a common genetic disorder caused by mutations in the Pkd1 or Pkd2 genes, in which large cysts replace normal kidney tissue, leading to end-stage kidney disease. In this study we have utilized a powerful nano-HPLC-mass spectrometric approach to characterize patterns of normal and abnormal N-linked glycosylation of α3 integrin subunit in Pkd1(-/-) cells derived from mouse kidneys. Higher molecular weight glycan structures with a different monosaccharide composition were observed at two sites, namely, Asn-925 and Asn-928 sites in α3 integrin isolated from Pkd1(+/+) cells compared with Pkd1(-/-) cells. In addition, an unusual and unique disialic acid glycan structure was observed solely in Pkd1(-/-) cells. Thus, these studies suggest that abnormal protein glycosylation may have a role on the pathogenesis of cyst formation in ADPKD.


Subject(s)
Integrin alpha3/metabolism , Polycystic Kidney Diseases/metabolism , Polysaccharides/metabolism , Animals , Chromatography, High Pressure Liquid , Immunoprecipitation , Mass Spectrometry , Mice , Mice, Knockout , Polycystic Kidney Diseases/pathology , Polysaccharides/isolation & purification , Sialic Acids/metabolism , TRPP Cation Channels/genetics
8.
Am J Physiol Renal Physiol ; 306(7): F764-72, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24500693

ABSTRACT

MicroRNAs (miRNAs) are small, noncoding regulatory RNAs that act as posttranscriptional repressors by binding to the 3'-untranslated region (3'-UTR) of target genes. They require processing by Dicer, an RNase III enzyme, to become mature regulatory RNAs. Previous work from our laboratory revealed critical roles for miRNAs in nephron progenitors at midgestation (Ho J, Pandey P, Schatton T, Sims-Lucas S, Khalid M, Frank MH, Hartwig S, Kreidberg JA. J Am Soc Nephrol 22: 1053-1063, 2011). To interrogate roles for miRNAs in the early metanephric mesenchyme, which gives rise to nephron progenitors as well as the renal stroma during kidney development, we conditionally ablated Dicer function in this lineage. Despite normal ureteric bud outgrowth and condensation of the metanephric mesenchyme to form nephron progenitors, early loss of miRNAs in the metanephric mesenchyme resulted in severe renal dysgenesis. Nephron progenitors are initially correctly specified in the mutant kidneys, with normal expression of several transcription factors known to be critical in progenitors, including Six2, Pax2, Sall1, and Wt1. However, there is premature loss of the nephron progenitor marker Cited1, marked apoptosis, and increased expression of the proapoptotic protein Bim shortly after the initial inductive events in early kidney development. Subsequently, there is a failure in ureteric bud branching and nephron progenitor differentiation. Taken together, our data demonstrate a previously undetermined requirement for miRNAs during early kidney organogenesis and indicate a crucial role for miRNAs in regulating the survival of this lineage.


Subject(s)
DEAD-box RNA Helicases/metabolism , Embryonic Stem Cells/enzymology , Kidney/enzymology , Mesoderm/enzymology , Ribonuclease III/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gestational Age , Kidney/abnormalities , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesoderm/abnormalities , Mice , Mice, Knockout , MicroRNAs/metabolism , Nephrons/abnormalities , Nephrons/enzymology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organogenesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Ribonuclease III/deficiency , Ribonuclease III/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Ureter/abnormalities , Ureter/enzymology
9.
Development ; 137(7): 1189-203, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20215353

ABSTRACT

The Wilms' tumor suppressor 1 (WT1) gene encodes a DNA- and RNA-binding protein that plays an essential role in nephron progenitor differentiation during renal development. To identify WT1 target genes that might regulate nephron progenitor differentiation in vivo, we performed chromatin immunoprecipitation (ChIP) coupled to mouse promoter microarray (ChIP-chip) using chromatin prepared from embryonic mouse kidney tissue. We identified 1663 genes bound by WT1, 86% of which contain a previously identified, conserved, high-affinity WT1 binding site. To investigate functional interactions between WT1 and candidate target genes in nephron progenitors, we used a novel, modified WT1 morpholino loss-of-function model in embryonic mouse kidney explants to knock down WT1 expression in nephron progenitors ex vivo. Low doses of WT1 morpholino resulted in reduced WT1 target gene expression specifically in nephron progenitors, whereas high doses of WT1 morpholino arrested kidney explant development and were associated with increased nephron progenitor cell apoptosis, reminiscent of the phenotype observed in Wt1(-/-) embryos. Collectively, our results provide a comprehensive description of endogenous WT1 target genes in nephron progenitor cells in vivo, as well as insights into the transcriptional signaling networks controlled by WT1 that might direct nephron progenitor fate during renal development.


Subject(s)
Gene Expression Regulation, Developmental , Kidney/cytology , Kidney/embryology , Nephrons/cytology , Stem Cells/physiology , WT1 Proteins/metabolism , Animals , Apoptosis/physiology , Base Sequence , Chromatin Immunoprecipitation , Databases, Factual , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Female , In Situ Hybridization , Kidney/metabolism , Mice , Microarray Analysis , Nephrons/embryology , Nephrons/metabolism , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Pregnancy , Stem Cells/cytology , Tissue Culture Techniques , WT1 Proteins/genetics
10.
Pediatr Nephrol ; 28(2): 219-25, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22660936

ABSTRACT

The discovery of microRNAs (miRNAs) as novel regulators of gene expression has led to a marked change in how gene regulation is viewed, with important implications for development and disease. MiRNAs are endogenous, small, noncoding RNAs that largely repress their target mRNAs post-transcriptionally. The regulation of gene expression by miRNAs represents an evolutionarily conserved mechanism that is broadly applicable to most biological processes. Recent studies have begun to define the role of miRNAs in different cell lineages during kidney development, and to implicate specific miRNAs in developmental and pathophysiological processes in the kidney. This review will focus on novel insights into the role(s) of miRNAs in kidney development, and discuss the implications for pediatric renal disease.


Subject(s)
Kidney Diseases/genetics , Kidney/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Gene Expression Regulation , Humans , Kidney/embryology
11.
J Am Soc Nephrol ; 23(3): 400-4, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22302196

ABSTRACT

MicroRNAs (miRNAs) are a group of small, noncoding RNAs that act as novel regulators of gene expression through the post-transcriptional repression of their target mRNAs. miRNAs have been implicated in diverse biologic processes, and it is estimated that up to half of all transcripts are regulated by miRNAs. Recent studies also demonstrate a critical role for miRNAs in renal development, physiology, and pathophysiology. Understanding the function of miRNAs in the kidney may lead to innovative approaches to renal disease.


Subject(s)
Gene Expression Regulation/physiology , Kidney/physiology , MicroRNAs/physiology , Animals , Disease Models, Animal , Humans , Kidney/embryology , Kidney/physiopathology , Kidney Diseases/physiopathology , Transcription, Genetic/physiology
12.
J Am Soc Nephrol ; 23(8): 1309-18, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22677559

ABSTRACT

The mechanisms of cystogenesis in autosomal dominant polycystic kidney disease (ADPKD) are not fully understood. Hyperactivation of the tyrosine kinase c-Met contributes to cyst formation, but we do not know the downstream mediators. Here, we found that hyperactivated c-Met led to increased NF-κB signaling, which in turn, drove de novo expression of Wnt7a and overexpression of Wnt7b in Pkd1(-/-) mouse kidneys. Hyperactivated Wnt signaling increased expression of the transcription factor Pax2 in the cells lining cysts. Furthermore, blocking Wnt signaling with DKK1 decreased cyst formation in an organ culture model of ADPKD. In summary, these results suggest that the c-Met/NF-κB/Wnt/Pax2 signaling transduction axis may provide pharmacological targets for the treatment of ADPKD.


Subject(s)
NF-kappa B/metabolism , PAX2 Transcription Factor/metabolism , Polycystic Kidney Diseases/metabolism , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins/metabolism , Wnt Proteins/metabolism , 8-Bromo Cyclic Adenosine Monophosphate , Animals , Female , Male , Mice , Mice, Knockout , Organ Culture Techniques , Polycystic Kidney Diseases/etiology , Pregnancy , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Up-Regulation
13.
J Clin Invest ; 119(1): 213-24, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19104148

ABSTRACT

Pulmonary fibrosis, in particular idiopathic pulmonary fibrosis (IPF), results from aberrant wound healing and scarification. One population of fibroblasts involved in the fibrotic process is thought to originate from lung epithelial cells via epithelial-mesenchymal transition (EMT). Indeed, alveolar epithelial cells (AECs) undergo EMT in vivo during experimental fibrosis and ex vivo in response to TGF-beta1. As the ECM critically regulates AEC responses to TGF-beta1, we explored the role of the prominent epithelial integrin alpha3beta1 in experimental fibrosis by generating mice with lung epithelial cell-specific loss of alpha3 integrin expression. These mice had a normal acute response to bleomycin injury, but they exhibited markedly decreased accumulation of lung myofibroblasts and type I collagen and did not progress to fibrosis. Signaling through beta-catenin has been implicated in EMT; we found that in primary AECs, alpha3 integrin was required for beta-catenin phosphorylation at tyrosine residue 654 (Y654), formation of the pY654-beta-catenin/pSmad2 complex, and initiation of EMT, both in vitro and in vivo during the fibrotic phase following bleomycin injury. Finally, analysis of lung tissue from IPF patients revealed the presence of pY654-beta-catenin/pSmad2 complexes and showed accumulation of pY654-beta-catenin in myofibroblasts. These findings demonstrate epithelial integrin-dependent profibrotic crosstalk between beta-catenin and Smad signaling and support the hypothesis that EMT is an important contributor to pathologic fibrosis.


Subject(s)
Epithelial Cells/physiology , Fibroblasts/metabolism , Integrin alpha3beta1/metabolism , Pulmonary Fibrosis , Signal Transduction/physiology , Smad2 Protein/metabolism , beta Catenin/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Cells, Cultured , Epithelial Cells/cytology , Fibroblasts/cytology , Humans , Integrin alpha3beta1/genetics , Lung/cytology , Lung/drug effects , Lung/metabolism , Lung/pathology , Mesoderm/cytology , Mesoderm/physiology , Mice , Mice, Transgenic , Phenotype , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Smad2 Protein/genetics
14.
Biochem Biophys Res Commun ; 425(2): 244-9, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22828505

ABSTRACT

Agents that target the activity of the mammalian target of rapamycin (mTOR) kinase in humans are associated with proteinuria. However, the mechanisms underlying mTOR activity and signaling within the kidney are poorly understood. In this study, we developed a sensitive immunofluorescence technique for the evaluation of activated pmTOR and its associated signals in situ. While we find that pmTOR is rarely expressed in normal non-renal tissues, we consistently find intense expression in glomeruli within normal mouse and human kidneys. Using double staining, we find that the expression of pmTOR co-localizes with nephrin in podocytes and expression appears minimal within other cell types in the glomerulus. In addition, we found that pmTOR was expressed on occasional renal tubular cells within mouse and human kidney specimens. We also evaluated mTOR signaling in magnetic bead-isolated glomeruli from normal mice and, by Western blot analysis, we confirmed function of the pathway in glomerular cells vs. interstitial cells. Furthermore, we found that the activity of the pathway as well as the expression of VEGF, a target of mTOR-induced signaling, were reduced within glomeruli of mice following treatment with rapamycin. Collectively, these findings demonstrate that the mTOR signaling pathway is constitutively hyperactive within podocytes. We suggest that pmTOR signaling functions to regulate glomerular homeostasis in part via the inducible expression of VEGF.


Subject(s)
Kidney Glomerulus/enzymology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Fluorescent Antibody Technique , Homeostasis , Humans , Kidney Glomerulus/drug effects , Mechanistic Target of Rapamycin Complex 1 , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Podocytes/enzymology , Signal Transduction , Sirolimus/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis
15.
Proc Natl Acad Sci U S A ; 106(18): 7595-600, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19383784

ABSTRACT

Cortical GABAergic interneurons, most of which originate in the ganglionic eminences, take distinct tangential migratory trajectories into the developing cerebral cortex. However, the ligand-receptor systems that modulate the tangential migration of distinct groups of interneurons into the emerging cerebral wall remain unclear. Here, we show that netrin-1, a diffusible guidance cue expressed along the migratory routes traversed by GABAergic interneurons, interacts with alpha3beta1 integrin to promote interneuronal migration. In vivo analysis of interneuron-specific alpha3beta1 integrin, netrin-1-deficient mice (alpha3(lox/-)Dlx5/6-CIE, netrin-1(-/-)) reveals specific deficits in the patterns of interneuronal migration along the top of the developing cortical plate, resulting in aberrant interneuronal positioning throughout the cerebral cortex and hippocampus of conditional alpha3(lox/-)Dlx5/6-CIE, netrin-1(-/-) mice. These results indicate that specific guidance mechanisms, such as netrin-1-alpha3beta1 integrin interactions, modulate distinct routes of interneuronal migration and the consequent positioning of groups of cortical interneurons in the developing cerebral cortex.


Subject(s)
Cell Movement , Cerebral Cortex/growth & development , Integrin alpha3beta1/metabolism , Interneurons/physiology , Nerve Growth Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Movement/genetics , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Interneurons/metabolism , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Netrin-1 , Tumor Suppressor Proteins/genetics , gamma-Aminobutyric Acid/metabolism
16.
J Am Soc Nephrol ; 22(6): 1053-63, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21546576

ABSTRACT

Understanding the mechanisms that regulate nephron progenitors during kidney development should aid development of therapies for renal failure. MicroRNAs, which modulate gene expression through post-transcriptional repression of specific target mRNAs, contribute to the differentiation of stem cells, but their role in nephrogenesis is incompletely understood. Here, we found that the loss of miRNAs in nephron progenitors results in a premature depletion of this population during kidney development. Increased apoptosis and expression of the pro-apoptotic protein Bim accompanied this depletion. Profiling of miRNA expression during nephrogenesis identified several highly expressed miRNAs (miR-10a, miR-106b, miR-17-5p) in nephron progenitors that are either known or predicted to target Bim. We propose that modulation of apoptosis by miRNAs may determine congenital nephron endowment. Furthermore, our data implicate the pro-apoptotic protein Bim as a miRNA target in nephron progenitors.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Kidney/embryology , Kidney/physiology , Membrane Proteins/physiology , MicroRNAs/physiology , Proto-Oncogene Proteins/physiology , Stem Cells/physiology , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Differentiation/physiology , Cell Proliferation , Female , Gene Expression Profiling , Kidney/cytology , Male , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Mice, Transgenic , Models, Animal , Pregnancy , Proto-Oncogene Proteins/genetics , Stem Cells/cytology
17.
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
18.
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
19.
Glycobiology ; 21(2): 152-61, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20855470

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) is a heparan sulfate (HS)-binding factor. GDNF is produced by somatic Sertoli cells, where it signals to maintain spermatogonial stem cells (SSCs) and reproduction. Here, we investigate the roles of extracellular HS 6-O-endosulfatases (Sulfs), Sulf1 and Sulf2, in the matrix transmission of GDNF from Sertoli cells to SSCs. Although Sulfs are not required for testis formation, Sulf deficiency leads to the accelerated depletion of SSCs, a testis phenotype similar to that of GDNF+/- mice. Mechanistically, we show that Sulfs are expressed in GDNF-producing Sertoli cells. In addition, reduced Sulf activity profoundly worsens haplo-deficient GDNF phenotypes in our genetic studies. These findings establish a critical role of Sulfs in promoting GDNF signaling and support a model in which Sulfs regulate the bioavailability of GDNF by enzymatically remodeling HS 6-O-desulfation to release GDNF from matrix sequestration. Further, Sertoli cell-specific transcriptional factor Wilm's tumor 1 (WT1) directly activates the transcription of both Sulf1 and Sulf2 genes. Together, our studies not only identify Sulfs as essential regulators of GDNF signaling in the SSC niche, but also as direct downstream targets of WT1, thus establishing a physiological role of WT1 in Sertoli cells.


Subject(s)
Sertoli Cells/metabolism , Spermatogonia , Sulfatases , Sulfotransferases , Animals , Cell Cycle Proteins , Cell Differentiation/physiology , Gene Expression Regulation , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Heparitin Sulfate/metabolism , Humans , Male , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , RNA Splicing Factors , Rats , Signal Transduction/physiology , Spermatogonia/metabolism , Stem Cell Niche/metabolism , Stem Cells/metabolism , Sulfatases/genetics , Sulfatases/metabolism , Sulfotransferases/genetics , Sulfotransferases/metabolism
20.
Am J Physiol Renal Physiol ; 300(3): F811-20, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21209007

ABSTRACT

Dystroglycan (DG or DAG1) is considered a critical link between the basement membrane and the cytoskeleton in multiple tissues. DG consists of two subunits, an extracellular α-subunit that binds laminin and other basement membrane components, and a transmembrane ß-subunit. DG-null mouse embryos die during early embryogenesis because DG is required for Reichert's membrane formation. DG also forms an integral part of the dystrophin-glycoprotein complex in muscle. Although no human DG mutations have been reported, multiple forms of muscular dystrophy have been linked to DG glycosylation defects, and targeted deletion of muscle DG causes muscular dystrophy in mice. Moreover, DG is widely distributed in endothelial and epithelial cells, including those in the kidney. There has therefore been significant interest in DG's role in the kidney, especially in podocytes. Previous reports suggested that DG's disturbance in podocytes might cause glomerular filtration barrier abnormalities. To fully understand DG's contribution to nephrogenesis and kidney function, we used a conditional DG allele and a variety of Cre mice to systematically delete DG from podocytes, ureteric bud, metanephric mesenchyme, and then from the whole kidney. Surprisingly, none of these conditional deletions resulted in significant morphological or functional abnormalities in the kidney. Furthermore, DG-deficient podocytes did not show increased susceptibility to injury, and DG-deficient kidneys did not show delayed recovery. Integrins are therefore likely the primary extracellular matrix receptors in renal epithelia.


Subject(s)
Acute Kidney Injury/metabolism , Dystroglycans/metabolism , Kidney/embryology , Kidney/physiology , Acute Kidney Injury/physiopathology , Animals , Dystroglycans/genetics , Epithelial Cells/metabolism , Integrases/genetics , Integrases/metabolism , Integrin alpha3/genetics , Integrin alpha3/metabolism , Kidney Glomerulus/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Models, Animal , Podocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL