Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Molecules ; 26(23)2021 Dec 02.
Article in English | MEDLINE | ID: mdl-34885910

ABSTRACT

Novel indolocarbazole derivatives named LCS were synthesized by our research group. Two of them were selected as the most active anticancer agents in vivo. We studied the mechanisms of anticancer activity in accordance with the previously described effects of indolocarbazoles. Cytotoxicity was estimated by MTT assay. We analyzed LCS-DNA interactions by circular dichroism in cholesteric liquid crystals and fluorescent indicator displacement assay. The effect on the activity of topoisomerases I and II was studied by DNA relaxation assay. Expression of interferon signaling target genes was estimated by RT-PCR. Chromatin remodeling was analyzed-the effect on histone H1 localization and reactivation of epigenetically silenced genes. LCS-induced change in the expression of a wide gene set was counted by means of PCR array. Our study revealed the cytotoxic activity of the compounds against 11 cancer cell lines and it was higher than in immortalized cells. Both compounds bind DNA; binding constants were estimated-LCS-1208 demonstrated higher affinity than LCS-1269; it was shown that LCS-1208 intercalates into DNA that is typical for rebeccamycin derivatives. LCS-1208 also inhibits topoisomerases I and IIα. Being a strong intercalator and topoisomerase inhibitor, LCS-1208 upregulates the expression of interferon-induced genes. In view of LCSs binding to DNA we analyzed their influence on chromatin stability and revealed that LCS-1269 displaces histone H1. Our analysis of chromatin remodeling also included a wide set of epigenetic experiments in which LCS-1269 demonstrated complex epigenetic activity. Finally, we revealed that the antitumor effect of the compounds is based not only on binding to DNA and chromatin remodeling but also on alternative mechanisms. Both compounds induce expression changes in genes involved in neoplastic transformation and target genes of the signaling pathways in cancer cells. Despite of being structurally similar, each compound has unique biological activities. The effects of LCS-1208 are associated with intercalation. The mechanisms of LCS-1269 include influence on higher levels such as chromatin remodeling and epigenetic effects.


Subject(s)
Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Glycosides/pharmacology , Antineoplastic Agents/chemistry , Carbazoles/chemistry , Cell Line, Tumor , Epigenesis, Genetic/drug effects , Glycosides/chemistry , Humans , Indoles/chemistry , Indoles/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics
2.
J Exp Med ; 216(4): 936-949, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30886059

ABSTRACT

Diabetic nephropathy is a leading cause of end-stage kidney failure. Reduced angiopoietin-TIE2 receptor tyrosine kinase signaling in the vasculature leads to increased vascular permeability, inflammation, and endothelial cell loss and is associated with the development of diabetic complications. Here, we identified a mechanism to explain how TIE2 signaling is attenuated in diabetic animals. Expression of vascular endothelial protein tyrosine phosphatase VE-PTP (also known as PTPRB), which dephosphorylates TIE2, is robustly up-regulated in the renal microvasculature of diabetic rodents, thereby reducing TIE2 activity. Increased VE-PTP expression was dependent on hypoxia-inducible factor transcriptional activity in vivo. Genetic deletion of VE-PTP restored TIE2 activity independent of ligand availability and protected kidney structure and function in a mouse model of severe diabetic nephropathy. Mechanistically, inhibition of VE-PTP activated endothelial nitric oxide synthase and led to nuclear exclusion of the FOXO1 transcription factor, reducing expression of pro-inflammatory and pro-fibrotic gene targets. In sum, we identify inhibition of VE-PTP as a promising therapeutic target to protect the kidney from diabetic injury.


Subject(s)
Diabetic Nephropathies/metabolism , Receptor, TIE-2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Animals , Cell Line , Disease Models, Animal , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Forkhead Box Protein O1/metabolism , Gene Knockdown Techniques , Humans , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase/metabolism , RNA, Small Interfering/genetics
3.
Proc Natl Acad Sci U S A ; 115(6): 1298-1303, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29358379

ABSTRACT

The angiopoietin (ANGPT)-TIE2/TEK signaling pathway is essential for blood and lymphatic vascular homeostasis. ANGPT1 is a potent TIE2 activator, whereas ANGPT2 functions as a context-dependent agonist/antagonist. In disease, ANGPT2-mediated inhibition of TIE2 in blood vessels is linked to vascular leak, inflammation, and metastasis. Using conditional knockout studies in mice, we show TIE2 is predominantly activated by ANGPT1 in the cardiovascular system and by ANGPT2 in the lymphatic vasculature. Mechanisms underlying opposing actions of ANGPT2 in blood vs. lymphatic endothelium are poorly understood. Here we show the endothelial-specific phosphatase VEPTP (vascular endothelial protein tyrosine phosphatase) determines TIE2 response to ANGPT2. VEPTP is absent from lymphatic endothelium in mouse in vivo, permitting ANGPT2/TIE2-mediated lymphangiogenesis. Inhibition of VEPTP converts ANGPT2 into a potent TIE2 activator in blood endothelium. Our data support a model whereby VEPTP functions as a rheostat to modulate ANGPT2 ligand effect on TIE2.


Subject(s)
Angiopoietin-2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Angiopoietin-2/genetics , Animals , Endothelium, Lymphatic/embryology , Endothelium, Lymphatic/metabolism , Endothelium, Vascular/metabolism , HEK293 Cells , Humans , Mice, Knockout , Mice, Transgenic , Receptor, TIE-2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Signal Transduction
4.
J Am Soc Nephrol ; 29(1): 92-103, 2018 01.
Article in English | MEDLINE | ID: mdl-29018139

ABSTRACT

Nephrin is a key structural component of the podocyte slit diaphragm, and proper expression of nephrin on the cell surface is critical to ensure integrity of the blood filtration barrier. Maintenance of nephrin within this unique cell junction has been proposed to require dynamic phosphorylation events and endocytic recycling, although the molecular mechanisms that control this interplay are poorly understood. Here, we investigated the possibility that the phosphotyrosine adaptor protein ShcA regulates nephrin turnover. Western blotting and immunostaining analysis confirmed that ShcA is expressed in podocytes. In immunoprecipitation and pulldown assays, ShcA, via its SH2 domain, was associated with several phosphorylated tyrosine residues on nephrin. Overexpression of ShcA promoted nephrin tyrosine phosphorylation and reduced nephrin signaling and cell surface expression in vitro In a rat model of reversible podocyte injury and proteinuria, phosphorylated nephrin temporally colocalized with endocytic structures coincident with upregulation of ShcA expression. In vivo biotinylation assays confirmed that nephrin expression decreased at the cell surface and correspondingly increased in the cytosol during the injury time course. Finally, immunostaining in kidney biopsy specimens demonstrated overexpression of ShcA in several human proteinuric kidney diseases compared with normal conditions. Our results suggest that increases in ShcA perturb nephrin phosphosignaling dynamics, leading to aberrant nephrin turnover and slit diaphragm disassembly.


Subject(s)
Endocytosis , Kidney Diseases/metabolism , Membrane Proteins/metabolism , Podocytes/metabolism , Proteinuria/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Animals , Biotinylation , Cell Membrane/metabolism , Cytosol/metabolism , HEK293 Cells , Humans , Kidney Diseases/pathology , Male , Nephrosis/chemically induced , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Tyrosine/metabolism , Up-Regulation
5.
Sci Rep ; 7(1): 11351, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900259

ABSTRACT

Hypoxia-inducible factor (HIF) is a key transcriptional factor in the response to hypoxia. Although the effect of HIF activation in chronic kidney disease (CKD) has been widely evaluated, the results have been inconsistent until now. This study aimed to investigate the effects of HIF-2α activation on renal fibrosis according to the activation timing in inducible tubule-specific transgenic mice with non-diabetic CKD. HIF-2α activation in renal tubular cells upregulated mRNA and protein expressions of fibronectin and type 1 collagen associated with the activation of p38 mitogen-activated protein kinase. In CKD mice, activation of HIF-2α at the beginning of CKD significantly aggravated renal fibrosis, whereas it did not lead to renal dysfunction. However, activation at a late-stage of CKD abrogated both renal dysfunction and fibrosis, which was associated with restoration of renal vasculature and amelioration of hypoxia through increased renal tubular expression of VEGF and its isoforms. As with tubular cells with HIF-2α activation, those under hypoxia also upregulated VEGF, fibronectin, and type 1 collagen expressions associated with HIF-1α activation. In conclusion, late-stage renal tubular HIF-2α activation has protective effects on renal fibrosis and the resultant renal dysfunction, thus it could represent a therapeutic target in late stage of CKD.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Tubules/metabolism , Transcriptional Activation , Animals , Atrophy , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers , Disease Models, Animal , Disease Progression , Epithelial Cells/metabolism , Fibrosis , Glomerulonephritis, IGA/etiology , Glomerulonephritis, IGA/metabolism , Glomerulonephritis, IGA/pathology , Humans , Hypoxia/genetics , Hypoxia/metabolism , Kidney Diseases/pathology , Kidney Function Tests , Kidney Tubules/pathology , Male , Mice , Mice, Transgenic , Middle Aged
6.
J Am Soc Nephrol ; 27(8): 2422-35, 2016 08.
Article in English | MEDLINE | ID: mdl-26802179

ABSTRACT

Podocytes are specialized epithelial cells of the kidney blood filtration barrier that contribute to permselectivity via a series of interdigitating actin-rich foot processes. Positioned between adjacent projections is a unique cell junction known as the slit diaphragm, which is physically connected to the actin cytoskeleton via the transmembrane protein nephrin. Evidence indicates that tyrosine phosphorylation of the intracellular tail of nephrin initiates signaling events, including recruitment of cytoplasmic adaptor proteins Nck1 and Nck2 that regulate actin cytoskeletal dynamics. Nephrin tyrosine phosphorylation is altered in human and experimental renal diseases characterized by pathologic foot process remodeling, prompting the hypothesis that phosphonephrin signaling directly influences podocyte morphology. To explore this possibility, we generated and analyzed knockin mice with mutations that disrupt nephrin tyrosine phosphorylation and Nck1/2 binding (nephrin(Y3F/Y3F) mice). Homozygous nephrin(Y3F/Y3F) mice developed progressive proteinuria accompanied by structural changes in the filtration barrier, including podocyte foot process effacement, irregular thickening of the glomerular basement membrane, and dilated capillary loops, with a similar but later onset phenotype in heterozygous animals. Furthermore, compared with wild-type mice, nephrin(Y3F/Y3F) mice displayed delayed recovery in podocyte injury models. Profiling of nephrin tyrosine phosphorylation dynamics in wild-type mice subjected to podocyte injury indicated site-specific differences in phosphorylation at baseline, injury, and recovery, which correlated with loss of nephrin-Nck1/2 association during foot process effacement. Our results define an essential requirement for nephrin tyrosine phosphorylation in stabilizing podocyte morphology and suggest a model in which dynamic changes in phosphotyrosine-based signaling confer plasticity to the podocyte actin cytoskeleton.


Subject(s)
Podocytes/physiology , Podocytes/ultrastructure , Tyrosine/metabolism , Animals , Female , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Phosphorylation , Signal Transduction
7.
J Am Soc Nephrol ; 25(11): 2459-70, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24904088

ABSTRACT

Podocytes are terminally differentiated cells with an elaborate cytoskeleton and are critical components of the glomerular barrier. We identified a bHLH transcription factor, Tcf21, that is highly expressed in developing and mature podocytes. Because conventional Tcf21 knockout mice die in the perinatal period with major cardiopulmonary defects, we generated a conditional Tcf21 knockout mouse to explore the role of this transcription factor in podocytes in vivo. Tcf21 was deleted from podocytes and podocyte progenitors using podocin-cre (podTcf21) and wnt4-cre (wnt4creTcf21) driver strains, respectively. Loss of Tcf21 from capillary-loop stage podocytes (podTcf21) results in simplified glomeruli with a decreased number of endothelial and mesangial cells. By 5 weeks of age, 40% of podTcf21 mice develop massive proteinuria and lesions similar to FSGS. Notably, the remaining 60% of mice do not develop proteinuria even when aged to 8 months. By contrast, earlier deletion of Tcf21 from podocyte precursors (wnt4creTcf21) results in a profound developmental arrest of podocyte differentiation and renal failure in 100% of mice during the perinatal period. Taken together, our results demonstrate a critical role for Tcf21 in the differentiation and maintenance of podocytes. Identification of direct targets of this transcription factor may provide new therapeutic avenues for proteinuric renal disease, including FSGS.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Diabetes Mellitus, Experimental/physiopathology , Glomerulosclerosis, Focal Segmental/physiopathology , Podocytes/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Cell Line , Cellular Senescence/physiology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Kidney Glomerulus/embryology , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Lac Operon , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Phenotype , Podocytes/pathology , Proteinuria/genetics , Proteinuria/pathology , Proteinuria/physiopathology
8.
Diabetes ; 61(11): 2958-66, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23093658

ABSTRACT

Vascular endothelial growth factor A (VEGFA) expression is increased in glomeruli in the context of diabetes. Here, we tested the hypothesis that this upregulation of VEGFA protects the glomerular microvasculature in diabetes and that therefore inhibition of VEGFA will accelerate nephropathy. To determine the role of glomerular Vegfa in the development and progression of diabetic nephropathy, we used an inducible Cre-loxP gene-targeting system that enabled genetic deletion of Vegfa selectively from glomerular podocytes of wild-type or diabetic mice. Type 1 diabetes was induced in mice using streptozotocin (STZ). We then assessed the extent of glomerular dysfunction by measuring proteinuria, glomerular pathology, and glomerular cell apoptosis. Vegfa expression increased in podocytes in the STZ model of diabetes. After 7 weeks of diabetes, diabetic mice lacking Vegfa in podocytes exhibited significantly greater proteinuria with profound glomerular scarring and increased apoptosis compared with control mice with diabetes or Vegfa deletion without diabetes. Reduced local production of glomerular Vegfa in a mouse model of type 1 diabetes promotes endothelial injury accelerating the progression of glomerular injury. These results suggest that upregulation of VEGFA in diabetic kidneys protects the microvasculature from injury and that reduction of VEGFA in diabetes may be harmful.


Subject(s)
Diabetic Nephropathies/metabolism , Kidney Glomerulus/blood supply , Kidney Glomerulus/metabolism , Microvessels/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis , Cells, Cultured , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Diabetic Nephropathies/urine , Disease Progression , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Hyperglycemia/etiology , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Microvessels/pathology , Podocytes/metabolism , Podocytes/pathology , Proteinuria/etiology , RNA, Messenger/metabolism , Random Allocation , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics
9.
Semin Nephrol ; 30(6): 582-90, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21146123

ABSTRACT

Anti-vascular endothelial growth factor (VEGF) agents are an important component in the treatment of many solid tumors. As the indications for these targeted therapies grow, the expected number of patients to receive these drugs will increase exponentially. Despite the great promise, serious toxicities may arise. Here, we discuss the incidence, pathogenesis, and management of proteinuria and renal insufficiency associated with this class of drugs.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/pharmacology , Proteinuria/chemically induced , Renal Insufficiency/chemically induced , Thrombotic Microangiopathies/chemically induced , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Humans , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Mice , Models, Animal , Proteinuria/epidemiology , Proteinuria/pathology , Proteinuria/therapy , Renal Insufficiency/epidemiology , Renal Insufficiency/pathology , Renal Insufficiency/therapy , Signal Transduction/genetics , Thrombotic Microangiopathies/pathology , Thrombotic Microangiopathies/physiopathology , Vascular Endothelial Growth Factor A/drug effects
10.
Cell Metab ; 12(4): 329-340, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20889126

ABSTRACT

Diabetic nephropathy (DN) is the leading cause of renal failure in the world. It is characterized by albuminuria and abnormal glomerular function and is considered a hyperglycemic "microvascular" complication of diabetes, implying a primary defect in the endothelium. However, we have previously shown that human podocytes have robust responses to insulin. To determine whether insulin signaling in podocytes affects glomerular function in vivo, we generated mice with specific deletion of the insulin receptor from their podocytes. These animals develop significant albuminuria together with histological features that recapitulate DN, but in a normoglycemic environment. Examination of "normal" insulin-responsive podocytes in vivo and in vitro demonstrates that insulin signals through the MAPK and PI3K pathways via the insulin receptor and directly remodels the actin cytoskeleton of this cell. Collectively, this work reveals the critical importance of podocyte insulin sensitivity for kidney function.


Subject(s)
Insulin/physiology , Kidney/physiology , Podocytes/physiology , Animals , Diabetic Nephropathies , Kidney Glomerulus/cytology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction/physiology
11.
J Am Soc Nephrol ; 21(10): 1691-701, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20688931

ABSTRACT

VEGF is a potent vascular growth factor produced by podocytes in the developing and mature glomerulus. Specific deletion of VEGF from podocytes causes glomerular abnormalities including profound endothelial cell injury, suggesting that paracrine signaling is critical for maintaining the glomerular filtration barrier (GFB). However, it is not clear whether normal GFB function also requires autocrine VEGF signaling in podocytes. In this study, we sought to determine whether an autocrine VEGF-VEGFR-2 loop in podocytes contributes to the maintenance of the GFB in vivo. We found that induced, whole-body deletion of VEGFR-2 caused marked abnormalities in the kidney and also other tissues, including the heart and liver. By contrast, podocyte-specific deletion of the VEGFR-2 receptor had no effect on glomerular development or function even up to 6 months old. Unlike cell culture models, enhanced expression of VEGF by podocytes in vivo caused foot process fusion and alterations in slit diaphragm-associated proteins; however, inhibition of VEGFR-2 could not rescue this defect. Although VEGFR-2 was dispensable in the podocyte, glomerular endothelial cells depended on VEGFR-2 expression: postnatal deletion of the receptor resulted in global defects in the glomerular microvasculature. Taken together, our results provide strong evidence for dominant actions of a paracrine VEGF-VEGFR-2 signaling loop both in the developing and in the filtering glomerulus. VEGF produced by the podocyte regulates the structure and function of the adjacent endothelial cell.


Subject(s)
Autocrine Communication , Paracrine Communication , Podocytes/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Endothelial Cells/physiology , Kidney/pathology , Mice , Mice, Transgenic , Phenotype , Proteinuria/pathology , Up-Regulation , Vascular Endothelial Growth Factor Receptor-2/genetics
12.
J Am Soc Nephrol ; 20(7): 1533-43, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19443634

ABSTRACT

Within the glomerulus, the scaffolding protein nephrin bridges the actin-rich foot processes that extend from adjacent podocytes to form the slit diaphragm. Mutations affecting a number of slit diaphragm proteins, including nephrin, cause glomerular disease through rearrangement of the actin cytoskeleton and disruption of the filtration barrier. We recently established that the Nck family of Src homology 2 (SH2)/SH3 cytoskeletal adaptor proteins can mediate nephrin-dependent actin reorganization. Formation of foot processes requires expression of Nck in developing podocytes, but it is unknown whether Nck maintains podocyte structure and function throughout life. Here, we used an inducible transgenic strategy to delete Nck expression in adult mouse podocytes and found that loss of Nck expression rapidly led to proteinuria, glomerulosclerosis, and altered morphology of foot processes. We also found that podocyte injury reduced phosphorylation of nephrin in adult kidneys. These data suggest that Nck is required to maintain adult podocytes and that phosphotyrosine-based interactions with nephrin may occur in foot processes of resting, mature podocytes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Glomerular Filtration Rate/physiology , Kidney Glomerulus/metabolism , Oncogene Proteins/metabolism , Podocytes/metabolism , Tight Junctions/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Anti-Bacterial Agents/pharmacology , Cell Line , Disease Models, Animal , Doxycycline/pharmacology , Glomerulonephritis/chemically induced , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Kidney Glomerulus/pathology , Male , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Oncogene Proteins/genetics , Phosphorylation , Podocytes/drug effects , Podocytes/ultrastructure , Proteinuria/metabolism , Proteinuria/pathology , Puromycin Aminonucleoside/adverse effects , Rats , Rats, Sprague-Dawley , Tight Junctions/ultrastructure
13.
Nat Genet ; 40(8): 1010-5, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18604206

ABSTRACT

Tissue organization in Drosophila is regulated by the core planar cell polarity (PCP) proteins Frizzled, Dishevelled, Prickle, Van Gogh and Flamingo. Core PCP proteins are conserved in mammals and function in mammalian tissue organization. Recent studies have identified another group of Drosophila PCP proteins, consisting of the protocadherins Fat and Dachsous (Ds) and the transmembrane protein Four-jointed (Fj). In Drosophila, Fat represses fj transcription, and Ds represses Fat activity in PCP. Here we show that Fat4 is an essential gene that has a key role in vertebrate PCP. Loss of Fat4 disrupts oriented cell divisions and tubule elongation during kidney development, leading to cystic kidney disease. Fat4 genetically interacts with the PCP genes Vangl2 and Fjx1 in cyst formation. In addition, Fat4 represses Fjx1 expression, indicating that Fat signaling is conserved. Together, these data suggest that Fat4 regulates vertebrate PCP and that loss of PCP signaling may underlie some cystic diseases in humans.


Subject(s)
Cadherins/metabolism , Cell Division , Cell Polarity , Kidney Diseases, Cystic/metabolism , Kidney/embryology , Animals , Cadherins/genetics , Drosophila melanogaster , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Mice
14.
Dev Biol ; 316(2): 288-301, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18328474

ABSTRACT

Integrins are transmembrane heteromeric receptors that mediate interactions between cells and extracellular matrix (ECM). beta1, the most abundantly expressed integrin subunit, binds at least 12 alpha subunits. beta1 containing integrins are highly expressed in the glomerulus of the kidney; however their role in glomerular morphogenesis and maintenance of glomerular filtration barrier integrity is poorly understood. To study these questions we selectively deleted beta1 integrin in the podocyte by crossing beta1(flox/flox) mice with podocyte specific podocin-cre mice (pod-Cre), which express cre at the time of glomerular capillary formation. We demonstrate that podocyte abnormalities are visualized during glomerulogenesis of the pod-Cre;beta1(flox/flox) mice and proteinuria is present at birth, despite a grossly normal glomerular basement membrane. Following the advent of glomerular filtration there is progressive podocyte loss and the mice develop capillary loop and mesangium degeneration with little evidence of glomerulosclerosis. By 3 weeks of age the mice develop severe end stage renal failure characterized by both tubulointerstitial and glomerular pathology. Thus, expression of beta1 containing integrins by the podocyte is critical for maintaining the structural integrity of the glomerulus.


Subject(s)
Gene Expression Regulation , Integrin beta1/genetics , Kidney Glomerulus/growth & development , Podocytes/physiology , Aging , Animals , Animals, Newborn , DNA Primers , In Situ Hybridization , Kidney Glomerulus/anatomy & histology , Kidney Glomerulus/physiology , Mice , Mice, Knockout , Mice, Transgenic , Polymerase Chain Reaction , Proteinuria/genetics
15.
N Engl J Med ; 358(11): 1129-36, 2008 Mar 13.
Article in English | MEDLINE | ID: mdl-18337603

ABSTRACT

The glomerular microvasculature is particularly susceptible to injury in thrombotic microangiopathy, but the mechanisms by which this occurs are unclear. We report the cases of six patients who were treated with bevacizumab, a humanized monoclonal antibody against vascular endothelial growth factor (VEGF), in whom glomerular disease characteristic of thrombotic microangiopathy developed. To show that local reduction of VEGF within the kidney is sufficient to trigger the pathogenesis of thrombotic microangiopathy, we used conditional gene targeting to delete VEGF from renal podocytes in adult mice; this resulted in a profound thrombotic glomerular injury. These observations provide evidence that glomerular injury in patients who are treated with bevacizumab is probably due to direct targeting of VEGF by antiangiogenic therapy.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Antibodies, Monoclonal/adverse effects , Kidney Glomerulus/drug effects , Podocytes/metabolism , Thrombosis/chemically induced , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Aged , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Bevacizumab , Female , Gene Targeting , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/pathology , Male , Mice , Mice, Knockout , Microcirculation/drug effects , Middle Aged , Neoplasms/drug therapy , Proteinuria/chemically induced , RNA, Messenger/metabolism , Renal Circulation , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
16.
J Am Soc Nephrol ; 19(3): 495-502, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18199804

ABSTRACT

The cellular composition of crescents in glomerular disease is controversial. The role of podocytes in crescent formation has been especially difficult to study because podocytes typically lose their characteristic terminally differentiated phenotype under disease conditions, making them difficult to identify. We reasoned that the intermediate filament protein nestin, a marker of progenitor cells that has recently been identified in podocytes, may allow the investigation of podocyte involvement in glomerular crescents. In a series of 35 biopsies with crescentic glomerular disease, all showed nestin-positive cells in the crescents, ranging in number from occasional to approximately 50% of crescent cells. Other podocyte markers, such as podocin and WT1, failed to identify cells in crescents, and no contribution by endothelial or myogenic cells was noted. CD68-positive cells were observed in 80% of cases but were never as numerous as the nestin-positive cells. Nestin and CD68 were not coexpressed by the same cells, providing no evidence of trans-differentiation of podocytes into a macrophage phenotype. Keratin-positive cells were found in crescents in 51% of cases, but only as occasional cells. Up to one third of crescent cells were cycling in 48% of biopsies, and double immunostaining identified these cells as a mixture of nestin-positive cells and "null" cells (negative for nestin, CD68, and keratin). In addition to our observations in human disease, we also identified nestin-positive proliferating podocytes in the crescents of 2 mouse models of crescentic glomerulonephritis. We conclude that podocytes play a role in the formation of glomerular crescents.


Subject(s)
Anti-Glomerular Basement Membrane Disease/pathology , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Podocytes/physiology , Animals , Anti-Glomerular Basement Membrane Disease/metabolism , Cell Cycle/physiology , Humans , Immunophenotyping , Ki-67 Antigen/metabolism , Mice , Nestin , Podocytes/metabolism
17.
Nephron Physiol ; 106(2): p32-7, 2007.
Article in English | MEDLINE | ID: mdl-17570946

ABSTRACT

BACKGROUND/AIMS: Vascular endothelial growth factor is a major regulator of angiogenesis and vascular permeability [Carmeliet et al.: Nature 1996;380:435-439]. The podocyte, the outermost layer of the glomerular filtration barrier, produces large amounts of VEGF-A. The observation that levels of VEGF-A are altered in glomerular diseases, the identification of a link between pre-eclampsia and elevated levels of a circulating soluble VEGF receptor, and the entry of anti-VEGF therapies into the clinical arena have generated intense interest in the functional role of VEGF-A in the glomerulus. METHODS: A variety of studies have been performed to address the role of VEGF-A signaling in the glomerulus. These include descriptions of expression patterns in human renal biopsies, cell culture studies to dissect paracrine versus autocrine signaling roles, and manipulation of VEGF-A expression in animal models using pharmacologic, biologic or genetic approaches. RESULTS: Exquisite dosage sensitivity to VEGF-A exists in the developing glomerulus as small reductions in the expression of VEGF-A lead to profound changes in glomerular structure and function in mice. The use of VEGF inhibitors is associated with damage to the glomerular endothelium in animal models and proteinuria in patients, suggesting that local VEGF-A production is also required for maintenance of this specialized vascular bed. CONCLUSIONS: Tight regulation of VEGF-A signaling is required for development and maintenance of the glomerular filtration barrier (GFB) and emphasizes the role of podocyte-endothelial crosstalk in the glomerulus. The relative contributions of various VEGF-A isoforms, the role of autocrine signaling in vivo and identification of factors and mechanisms that regulate constitutive expression, storage and delivery of VEGF-A in the glomerulus are still under investigation.


Subject(s)
Autocrine Communication/physiology , Glomerular Filtration Rate/physiology , Kidney Glomerulus/physiology , Podocytes/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Humans
18.
Nature ; 440(7085): 818-23, 2006 Apr 06.
Article in English | MEDLINE | ID: mdl-16525419

ABSTRACT

The glomerular filtration barrier in the kidney is formed in part by a specialized intercellular junction known as the slit diaphragm, which connects adjacent actin-based foot processes of kidney epithelial cells (podocytes). Mutations affecting a number of slit diaphragm proteins, including nephrin (encoded by NPHS1), lead to renal disease owing to disruption of the filtration barrier and rearrangement of the actin cytoskeleton, although the molecular basis for this is unclear. Here we show that nephrin selectively binds the Src homology 2 (SH2)/SH3 domain-containing Nck adaptor proteins, which in turn control the podocyte cytoskeleton in vivo. The cytoplasmic tail of nephrin has multiple YDxV sites that form preferred binding motifs for the Nck SH2 domain once phosphorylated by Src-family kinases. We show that this Nck-nephrin interaction is required for nephrin-dependent actin reorganization. Selective deletion of Nck from podocytes of transgenic mice results in defects in the formation of foot processes and in congenital nephrotic syndrome. Together, these findings identify a physiological signalling pathway in which nephrin is linked through phosphotyrosine-based interactions to Nck adaptors, and thus to the underlying actin cytoskeleton in podocytes. Simple and widely expressed SH2/SH3 adaptor proteins can therefore direct the formation of a specialized cellular morphology in vivo.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Kidney/cytology , Kidney/metabolism , Membrane Proteins/metabolism , Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Animals , Cell Line , Cytoskeleton/chemistry , Humans , Kidney/pathology , Membrane Proteins/genetics , Mice , Mutation/genetics , Nephrotic Syndrome/congenital , Nephrotic Syndrome/genetics , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Oncogene Proteins/chemistry , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Phosphorylation , Phosphotyrosine/metabolism , src Homology Domains
19.
J Am Soc Nephrol ; 17(3): 724-35, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16436493

ABSTRACT

The glomerular filtration barrier separates the blood from the urinary space and consists of two major cell types: podocytes and fenestrated endothelial cells. Mesangial cells sit between the capillary loops and provide structural support. Proliferation and loss of mesangial cells both are central findings in a number of renal diseases, including diabetic nephropathy and mesangiolysis, respectively. Using cell-specific gene targeting, it was shown previously that vascular endothelial growth factor A (VEGF-A) production by podocytes is required for glomerular endothelial cell migration, differentiation, and survival. For further investigation of the effect of gene dose and VEGF-A knockdown within the glomerulus, mice that carry one hypomorphic VEGF-A allele and one podocyte-specific null VEGF-A allele (VEGFhypo/loxP,Neph-Cre+/-) were generated; in these mice, the "allelic dose" of VEGF-A is intermediate between glomerular-specific heterozygous and null states. VEGFhypo/loxP,Neph-Cre+/- mice die at 3 wk of age from renal failure. Although endothelial cell defects are observed, striking loss of mesangial cells occurs postnatally. In addition, differentiated mesangial cells cannot be found in glomeruli of podocyte-specific null VEGF-A mice (VEGFloxP/loxP,Cre+/-). Together, these results demonstrate a key role for VEGF-A production in the podocyte for mesangial cell survival and differentiation.


Subject(s)
Cell Death/physiology , Cell Movement/physiology , Glomerular Mesangium/cytology , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Death/genetics , Cell Movement/genetics , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Endothelial Cells/physiology , Gene Expression Regulation, Developmental , Genotype , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Transgenic , Phenotype , Podocytes/metabolism , Probability , Sensitivity and Specificity , Signal Transduction
20.
Curr Opin Nephrol Hypertens ; 13(1): 9-15, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15090854

ABSTRACT

PURPOSE OF REVIEW: Vascular endothelial growth factor is a major regulator of blood vessel biology and is highly expressed in presumptive and mature podocytes within the glomerulus. It has long been recognized that dysregulation of this factor occurs in a number of glomerular diseases; however, definitive proof that it plays a pathogenic or developmental role in glomerular biology has remained elusive. This review will summarize some of the recent advances in our understanding of the role(s) of VEGF in these processes. RECENT FINDINGS: Gene targeting in the mouse has shown that tight regulation of vascular endothelial growth factor is required for development and maintenance of the glomerular filtration barrier. Podocyte-specific deletion of both alleles leads to congenital nephropathy and perinatal lethality. The glomeruli of mice that lack the 164 and 184 isoforms but express the 120 isoform, are smaller and have fewer capillary loops, whereas mice with podocyte-specific haploinsufficiency for all isoforms develop glomerular endotheliosis, the renal lesion seen in preeclampsia. Elevated levels of the soluble vascular endothelial growth factor receptor 1, which binds and inhibits circulating forms of VEGF were identified in patients with preeclampsia; rats injected with this soluble receptor develop hypertension, endotheliosis and proteinuria, similar to the lesion seen in podocyte-specific haploinsufficient VEGF mice. Conversely, podocyte-specific overexpression of the 164 isoform leads to collapsing glomerulopathy, the classic lesion seen in HIV-associated nephropathy. SUMMARY: These results demonstrate that vascular endothelial growth factor plays a critical role in glomerular development and function, and provides the foundation to develop novel diagnostic or therapeutic tools for patients with glomerular disease.


Subject(s)
Kidney Glomerulus/growth & development , Kidney Glomerulus/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Autocrine Communication/physiology , Cell Membrane Permeability/physiology , Endothelial Cells/pathology , Endothelial Cells/physiology , Female , Glomerular Filtration Rate , Humans , Pre-Eclampsia/complications , Pre-Eclampsia/pathology , Pregnancy , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...