Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
J Am Soc Nephrol ; 32(3): 597-613, 2021 03.
Article in English | MEDLINE | ID: mdl-33510039

ABSTRACT

BACKGROUND: The ubiquitin-proteasome system (UPS) and the autophagy-lysosomal system (APLS) are major intracellular degradation procedures. The importance of the APLS in podocytes is established, but the role of the UPS is not well understood. METHODS: To investigate the role of the UPS in podocytes, mice were generated that had deletion of Rpt3 (Rpt3pdKO), which encodes an essential regulatory subunit required for construction of the 26S proteasome and its deubiquitinating function. RESULTS: Rpt3pdKO mice showed albuminuria and glomerulosclerosis, leading to CKD. Impairment of proteasome function caused accumulation of ubiquitinated proteins and of oxidative modified proteins, and it induced podocyte apoptosis. Although impairment of proteasome function normally induces autophagic activity, the number of autophagosomes was lower in podocytes of Rpt3pdKO mice than in control mice, suggesting the autophagic activity was suppressed in podocytes with impairment of proteasome function. In an in vitro study, antioxidant apocynin and autophagy activator rapamycin suppressed podocyte apoptosis induced by proteasome inhibition. Moreover, rapamycin ameliorated the glomerular injury in the Rpt3pdKO mice. The accumulation of ubiquitinated proteins and of oxidative modified proteins, which were detected in the podocytes of Rpt3pdKO mice, is a characteristic feature of aging. An aging marker was increased in the podocytes of Rpt3pdKO mice, suggesting that impairment of proteasome function promoted signs of aging in podocytes. CONCLUSIONS: Impairment of proteasome function in podocytes led to CKD, and antioxidants and autophagy activators can be therapeutic agents for age-dependent CKD.


Subject(s)
Podocytes/enzymology , Proteasome Endopeptidase Complex/deficiency , Renal Insufficiency, Chronic/enzymology , Renal Insufficiency, Chronic/etiology , Aging/metabolism , Aging/pathology , Animals , Apoptosis/drug effects , Autophagy , Bortezomib/pharmacology , Cells, Cultured , Glomerulosclerosis, Focal Segmental/enzymology , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/pathology , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Podocytes/drug effects , Podocytes/pathology , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors/pharmacology , Protein Aggregates , Renal Insufficiency, Chronic/pathology , Sirolimus/pharmacology , Ubiquitination
2.
Am J Physiol Renal Physiol ; 318(6): F1377-F1390, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32308020

ABSTRACT

Ste20-like kinase SLK is critical for embryonic development and may play an important role in wound healing, muscle homeostasis, cell migration, and tumor growth. Mice with podocyte-specific deletion of SLK show albuminuria and damage to podocytes as they age. The present study addressed the role of SLK in glomerular injury. We induced adriamycin nephrosis in 3- to 4-mo-old control and podocyte SLK knockout (KO) mice. Compared with control, SLK deletion exacerbated albuminuria and loss of podocytes, synaptopodin, and podocalyxin. Glomeruli of adriamycin-treated SLK KO mice showed diffuse increases in the matrix and sclerosis as well as collapse of the actin cytoskeleton. SLK can phosphorylate ezrin. The complex of phospho-ezrin, Na+/H+ exchanger regulatory factor 2, and podocalyxin in the apical domain of the podocyte is a key determinant of normal podocyte architecture. Deletion of SLK reduced glomerular ezrin and ezrin phosphorylation in adriamycin nephrosis. Also, deletion of SLK reduced the colocalization of ezrin and podocalyxin in the glomerulus. Cultured glomerular epithelial cells with KO of SLK showed reduced ezrin phosphorylation and podocalyxin expression as well as reduced F-actin. Thus, SLK deletion leads to podocyte injury as mice age and exacerbates injury in adriamycin nephrosis. The mechanism may at least in part involve ezrin phosphorylation as well as disruption of the cytoskeleton and podocyte apical membrane structure.


Subject(s)
Actin Cytoskeleton/enzymology , Doxorubicin , Glomerulosclerosis, Focal Segmental/enzymology , Nephrosis/enzymology , Podocytes/enzymology , Protein Serine-Threonine Kinases/deficiency , Actin Cytoskeleton/pathology , Actins/metabolism , Albuminuria/chemically induced , Albuminuria/enzymology , Albuminuria/genetics , Animals , Cells, Cultured , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Gene Knockdown Techniques , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Mice, Knockout , Microfilament Proteins/metabolism , Nephrosis/chemically induced , Nephrosis/genetics , Nephrosis/pathology , Phosphoproteins/metabolism , Phosphorylation , Podocytes/pathology , Protein Serine-Threonine Kinases/genetics , Proteins/metabolism , Sodium-Hydrogen Exchangers/metabolism
3.
J Cell Mol Med ; 23(10): 6543-6553, 2019 10.
Article in English | MEDLINE | ID: mdl-31368174

ABSTRACT

The NPHS2 gene, encoding the slit diaphragm protein podocin, accounts for genetic and sporadic forms of nephrotic syndrome (NS). Patients with NS often present symptoms of volume retention, such as oedema formation or hypertension. The primary dysregulation in sodium handling involves an inappropriate activation of the epithelial sodium channel, ENaC. Plasma proteases in a proteinuria-dependent fashion have been made responsible; however, referring to the timeline of symptoms occurring and underlying mechanisms, contradictory results have been published. Characterizing the mouse model of podocyte inactivation of NPHS2 (Nphs2∆pod ) with respect to volume handling and proteinuria revealed that sodium retention, hypertension and gross proteinuria appeared sequentially in a chronological order. Detailed analysis of Nphs2∆pod during early sodium retention, revealed increased expression of full-length ENaC subunits and αENaC cleavage product with concomitant increase in ENaC activity as tested by amiloride application, and augmented collecting duct Na+ /K+ -ATPase expression. Urinary proteolytic activity was increased and several proteases were identified by mass spectrometry including cathepsin B, which was found to process αENaC. Renal expression levels of precursor and active cathepsin B were increased and could be localized to glomeruli and intercalated cells. Inhibition of cathepsin B prevented hypertension. With the appearance of gross proteinuria, plasmin occurs in the urine and additional cleavage of γENaC is encountered. In conclusion, characterizing the volume handling of Nphs2∆pod revealed early sodium retention occurring independent to aberrantly filtered plasma proteases. As an underlying mechanism cathepsin B induced αENaC processing leading to augmented channel activity and hypertension was identified.


Subject(s)
Cathepsin B/metabolism , Epithelial Sodium Channels/metabolism , Hypertension/etiology , Hypertension/metabolism , Nephrotic Syndrome/complications , Nephrotic Syndrome/metabolism , Amiloride/pharmacology , Animals , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Epithelial Sodium Channel Blockers/pharmacology , Glomerulosclerosis, Focal Segmental/enzymology , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/urine , Hypertension/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Tubules/cytology , Kidney Tubules/metabolism , Lysosomes/enzymology , Lysosomes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Nephrotic Syndrome/genetics , Proteinuria/metabolism , Proteolysis , Sodium/metabolism
4.
J Clin Invest ; 129(3): 1295-1313, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30776024

ABSTRACT

We identified 2 genes, histone deacetylase 1 (HDAC1) and HDAC2, contributing to the pathogenesis of proteinuric kidney diseases, the leading cause of end-stage kidney disease. mRNA expression profiling from proteinuric mouse glomeruli was linked to Connectivity Map databases, identifying HDAC1 and HDAC2 with the differentially expressed gene set reversible by HDAC inhibitors. In numerous progressive glomerular disease models, treatment with valproic acid (a class I HDAC inhibitor) or SAHA (a pan-HDAC inhibitor) mitigated the degree of proteinuria and glomerulosclerosis, leading to a striking increase in survival. Podocyte HDAC1 and HDAC2 activities were increased in mice podocytopathy models, and podocyte-associated Hdac1 and Hdac2 genetic ablation improved proteinuria and glomerulosclerosis. Podocyte early growth response 1 (EGR1) was increased in proteinuric patients and mice in an HDAC1- and HDAC2-dependent manner. Loss of EGR1 in mice reduced proteinuria and glomerulosclerosis. Longitudinal analysis of the multicenter Veterans Aging Cohort Study demonstrated a 30% reduction in mean annual loss of estimated glomerular filtration rate, and this effect was more pronounced in proteinuric patients receiving valproic acid. These results strongly suggest that inhibition of HDAC1 and HDAC2 activities may suppress the progression of human proteinuric kidney diseases through the regulation of EGR1.


Subject(s)
Glomerulosclerosis, Focal Segmental , Histone Deacetylase 1 , Histone Deacetylase 2 , Histone Deacetylase Inhibitors/pharmacology , Podocytes , Animals , Cell Line, Tumor , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Female , Glomerulosclerosis, Focal Segmental/enzymology , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Humans , Male , Mice, Knockout , Podocytes/enzymology , Podocytes/pathology , Proteinuria/drug therapy , Proteinuria/enzymology , Proteinuria/genetics , Proteinuria/pathology
5.
Front Immunol ; 9: 2113, 2018.
Article in English | MEDLINE | ID: mdl-30333818

ABSTRACT

Calcium calmodulin kinase IV (CaMK4) regulates multiple processes that significantly contribute to the lupus-related pathology by controlling the production of IL-2 and IL-17 by T cells, the proliferation of mesangial cells, and the function and structure of podocytes. CaMK4 is also upregulated in podocytes from patients with focal segmental glomerulosclerosis (FSGS). In both immune and non-immune podocytopathies, CaMK4 disrupts the structure and function of podocytes. In lupus-prone mice, targeted delivery of a CaMK4 inhibitor to CD4+ T cells suppresses both autoimmunity and the development of nephritis. Targeted delivery though to podocytes averts the deposition of immune complexes without affecting autoimmunity in lupus-prone mice and averts pathology induced by adriamycin in normal mice. Therefore, targeted delivery of a CaMK4 inhibitor to podocytes holds high therapeutic promise for both immune (lupus nephritis) and non-immune (FSGS) podocytopathies.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Glomerulosclerosis, Focal Segmental/enzymology , Lupus Nephritis/enzymology , Mesangial Cells/metabolism , Podocytes/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 4/antagonists & inhibitors , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/immunology , Humans , Interleukin-2/metabolism , Lupus Nephritis/drug therapy , Lupus Nephritis/immunology , Mice , Molecular Targeted Therapy
6.
BMC Nephrol ; 19(1): 273, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30340546

ABSTRACT

BACKGROUND: Lenvatinib is a tyrosine kinase inhibitor with novel binding ability. It is considered the standard of care for metastatic thyroid cancer; moreover, whether it is indicated for other malignant tumors has been examined. Lenvatinib increases the risk of kidney injury in some patients. In comparison with sorafenib, which is a conventional tyrosine kinase inhibitor (TKI), lenvatinib results in more side effects, including hypertension and proteinuria. We describe a case of secondary focal segmental glomerulosclerosis (FSGS) that developed following treatment of metastatic thyroid cancer with lenvatinib and reviewed the mechanisms of renal impairment. CASE PRESENTATION: We describe a patient with metastatic thyroid cancer who developed hypertension, nephrotic syndrome, and acute kidney injury after 3 months of lenvatinib treatment. Renal biopsy results revealed that 7 of 16 glomeruli indicated complete hyalinization, and that the glomeruli with incomplete hyalinization showed FSGS due to a vascular endothelial disorder and podocyte damage, which seemed to have been induced by lenvatinib treatment. These findings were similar to those of renal impairment treated with conventional TKIs. Although lenvatinib treatment was discontinued, up to 15 months were required to achieve remission of proteinuria, thus leading to chronic kidney disease with hyalinized lesions. CONCLUSIONS: To the best of our knowledge, this is the first reported case of secondary FSGS by lenvatinib treatment. Renal impairment treated with TKIs is commonly associated with minimal change nephrotic syndrome/FSGS findings, and it is suggested that renal involvement with TKI is different from that with the vascular endothelial growth factor ligand. Overexpression of c-mip due to TKI causes disorders such as podocyte dysregulation and promotion of apoptosis, which cause FSGS. Lenvatinib may result in FSGS by a similar mechanism with another TKI and could cause irreversible renal impairment; therefore caution must be used. It is essential to monitor blood pressure, urinary findings, and the renal function.


Subject(s)
Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/diagnosis , Phenylurea Compounds/adverse effects , Protein Kinase Inhibitors/adverse effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinolines/adverse effects , Aged , Female , Glomerulosclerosis, Focal Segmental/enzymology , Humans , Protein-Tyrosine Kinases/metabolism
7.
J Clin Invest ; 128(8): 3445-3459, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29985166

ABSTRACT

Podocyte malfunction occurs in autoimmune and nonautoimmune kidney disease. Calcium signaling is essential for podocyte injury, but the role of Ca2+/calmodulin-dependent kinase (CaMK) signaling in podocytes has not been fully explored. We report that podocytes from patients with lupus nephritis and focal segmental glomerulosclerosis and lupus-prone and lipopolysaccharide- or adriamycin-treated mice display increased expression of CaMK IV (CaMK4), but not CaMK2. Mechanistically, CaMK4 modulated podocyte motility by altering the expression of the GTPases Rac1 and RhoA and suppressed the expression of nephrin, synaptopodin, and actin fibers in podocytes. In addition, it phosphorylated the scaffold protein 14-3-3ß, which resulted in the release and degradation of synaptopodin. Targeted delivery of a CaMK4 inhibitor to podocytes preserved their ultrastructure, averted immune complex deposition and crescent formation, and suppressed proteinuria in lupus-prone mice and proteinuria in mice exposed to lipopolysaccharide-induced podocyte injury by preserving nephrin/synaptopodin expression. In animals exposed to adriamycin, podocyte-specific delivery of a CaMK4 inhibitor prevented and reversed podocyte injury and renal disease. We conclude that CaMK4 is pivotal in immune and nonimmune podocyte injury and that its targeted cell-specific inhibition preserves podocyte structure and function and should have therapeutic value in lupus nephritis and podocytopathies, including focal segmental glomerulosclerosis.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Glomerulosclerosis, Focal Segmental/enzymology , Kidney Glomerulus/enzymology , Lupus Nephritis/enzymology , Signal Transduction , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 4/immunology , Cell Line, Transformed , Female , Glomerulosclerosis, Focal Segmental/immunology , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred MRL lpr , Mice, Knockout , Proteinuria/enzymology , Proteinuria/immunology , Proteinuria/pathology
8.
J Biol Chem ; 292(51): 21137-21148, 2017 12 22.
Article in English | MEDLINE | ID: mdl-28982981

ABSTRACT

Kidney podocytes represent a key constituent of the glomerular filtration barrier. Identifying the molecular mechanisms of podocyte injury and survival is important for better understanding and management of kidney diseases. KIBRA (kidney brain protein), an upstream regulator of the Hippo signaling pathway encoded by the Wwc1 gene, shares the pro-injury properties of its putative binding partner dendrin and antagonizes the pro-survival signaling of the downstream Hippo pathway effector YAP (Yes-associated protein) in Drosophila and MCF10A cells. We recently identified YAP as an essential component of the glomerular filtration barrier that promotes podocyte survival by inhibiting dendrin pro-apoptotic function. Despite these recent advances, the signaling pathways that mediate podocyte injury remain poorly understood. Here we tested the hypothesis that, similar to its role in other model systems, KIBRA promotes podocyte injury. We found increased expression of KIBRA and phosphorylated YAP protein in glomeruli of patients with biopsy-proven focal segmental glomerulosclerosis (FSGS). KIBRA/WWc1 overexpression in murine podocytes promoted LATS kinase phosphorylation, leading to subsequent YAP Ser-127 phosphorylation, YAP cytoplasmic sequestration, and reduction in YAP target gene expression. Functionally, KIBRA overexpression induced significant morphological changes in podocytes, including disruption of the actin cytoskeletal architecture and reduction of focal adhesion size and number, all of which were rescued by subsequent YAP overexpression. Conversely, constitutive KIBRA knockout mice displayed reduced phosphorylated YAP and increased YAP expression at baseline. These mice were protected from acute podocyte foot process effacement following protamine sulfate perfusion. KIBRA knockdown podocytes were also protected against protamine-induced injury. These findings suggest an important role for KIBRA in the pathogenesis of podocyte injury and the progression of proteinuric kidney disease.


Subject(s)
Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Glomerulosclerosis, Focal Segmental/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Podocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Biopsy , Female , Gene Expression Regulation , Glomerulosclerosis, Focal Segmental/enzymology , Glomerulosclerosis, Focal Segmental/pathology , HEK293 Cells , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphorylation , Podocytes/pathology , Podocytes/ultrastructure , Protein Processing, Post-Translational , RNA Interference , Serine/metabolism , Transcription Factors , YAP-Signaling Proteins
9.
Kidney Int ; 91(4): 896-913, 2017 04.
Article in English | MEDLINE | ID: mdl-27998643

ABSTRACT

The glycoprotein CD44 is barely detected in normal mouse and human glomeruli, but is increased in glomerular parietal epithelial cells following podocyte injury in focal segmental glomerulosclerosis (FSGS). To determine the biological role and regulation of CD44 in these cells, we employed an in vivo and in vitro approach. Experimental FSGS was induced in CD44 knockout and wild-type mice with a cytotoxic podocyte antibody. Albuminuria, focal and global glomerulosclerosis (periodic acid-Schiff stain), and collagen IV staining were lower in CD44 knockout compared with wild-type mice with FSGS. Parietal epithelial cells had lower migration from Bowman's capsule to the glomerular tuft in CD44 knockout mice with disease compared with wild type mice. In cultured murine parietal epithelial cells, overexpressing CD44 with a retroviral vector encoding CD44 was accompanied by significantly increased collagen IV expression and parietal epithelial cell migration. Because our results showed de novo co-staining for activated ERK1/2 (pERK) in parietal epithelial cells in experimental FSGS, and also in biopsies from patients with FSGS, two in vitro strategies were employed to prove that pERK regulated CD44 levels. First, mouse parietal epithelial cells were infected with a retroviral vector for the upstream kinase MEK-DD to increase pERK, which was accompanied by increased CD44 levels. Second, in CD44-overexpressing parietal epithelial cells, decreasing pERK with U0126 was accompanied by reduced CD44. Finally, parietal epithelial cell migration was higher in cells with increased and reduced in cells with decreased pERK. Thus, pERK is a regulator of CD44 expression, and increased CD44 expression leads to a pro-sclerotic and migratory parietal epithelial cell phenotype.


Subject(s)
Extracellular Matrix/enzymology , Glomerulosclerosis, Focal Segmental/enzymology , Hyaluronan Receptors/metabolism , Kidney Glomerulus/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Podocytes/enzymology , Albuminuria/enzymology , Albuminuria/genetics , Albuminuria/prevention & control , Animals , Cell Movement , Cells, Cultured , Collagen Type IV/metabolism , Disease Models, Animal , Enzyme Activation , Extracellular Matrix/drug effects , Extracellular Matrix/pathology , Genetic Predisposition to Disease , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Humans , Hyaluronan Receptors/genetics , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Male , Mice, Knockout , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Phenotype , Phosphorylation , Podocytes/drug effects , Podocytes/pathology , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Time Factors , Transfection
10.
Sci Rep ; 6: 20171, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26822129

ABSTRACT

Obesity-induced kidney injury contributes to albuminuria, which is characterized by a progressive decline in renal function leading to glomerulosclerosis and renal fibrosis. Matrix metalloproteinases (MMPs) modulate inflammation and fibrosis by degrading a variety of extracellular matrix and regulating the activities of effector proteins. Abnormal regulation of MMP-12 expression has been implicated in abdominal aortic aneurysm, atherosclerosis, and emphysema, but the underlying mechanisms remain unclear. The present study examined the function of MMP-12 in glomerular fibrogenesis and inflammation using apo E(-/-) or apo E(-/-)MMP-12(-/-) mice and maintained on a high-fat-diet (HFD) for 3, 6, or 9 months. MMP-12 deletion reduced glomerular matrix accumulation, and downregulated the expression of NADPH oxidase 4 and the subunit-p67(phox), indicating the inhibition of renal oxidative stress. In addition, the expression of the inflammation-associated molecule MCP-1 and macrophage marker-CD11b was decreased in glomeruli of apo E(-/-)MMP-12(-/-) mice fed HFD. MMP-12 produced by macrophages infiltrating into glomeruli contributed to the degradation of collagen type IV and fibronectin. Crescent formation due to renal oxidative stress in Bowman's space was a major factor in the development of fibrogenesis and inflammation. These results suggest that regulating MMP-12 activity could be a therapeutic strategy for the treatment of crescentic glomerulonephritis and fibrogenesis.


Subject(s)
Dietary Fats/adverse effects , Glomerulosclerosis, Focal Segmental/enzymology , Kidney Glomerulus/enzymology , Macrophages/enzymology , Matrix Metalloproteinase 12/biosynthesis , Obesity/enzymology , Animals , Dietary Fats/pharmacology , Disease Models, Animal , Fibrosis , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Kidney Glomerulus/pathology , Macrophages/pathology , Matrix Metalloproteinase 12/genetics , Mice , Mice, Knockout , Obesity/chemically induced , Obesity/genetics , Obesity/pathology
11.
Exp Gerontol ; 74: 37-42, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26680767

ABSTRACT

OBJECTIVE: To investigate the effects of atorvastatin (AVT) on renal function and renal pathological changes in the aged rat and explore their possible mechanisms. METHODS: Twenty-month-old, normal female Wistar rats were divided into three groups: group A (n=8) was fed high-dose AVT (10mg/kg/d); group B (n=8) was fed low-dose AVT (1mg/kg/d); and group C (controls, n=8) received the same volume of normal saline; 3-month-old, normal female Wistar rats served as young normal controls (n=8). All rats were sacrificed following a 4-month treatment period. Serum creatinine and blood lipid levels were measured. The glomerular sclerosis index and tubulointerstitial lesions were determined using renal periodic acid-Schiff-stained paraffin sections. The mRNA and protein expressions of matrix metalloproteinases (MMP)-9 and -2, tissue inhibitors of metalloproteinase (TIMP)-1 and -2, transforming growth factor-ß1 (TGF-ß1), and peroxisome proliferator-activated receptors (PPARs) were examined using reverse transcription polymerase chain reactions and Western blots, respectively. RESULTS: Serum lipid (including serum cholesterol and serum triglycerides) levels in aged rats were significantly higher than those in young rats (p<0.05). Compared to the aged control group, high-dose AVT was associated with significantly lower serum total cholesterol and low-density lipoprotein cholesterol (LDL-C) levels in aged rats (p<0.05); low-dose AVT was associated only with lower serum LDL-C levels (p<0.05). Renal morphological changes in aged rats included focal glomerulosclerosis, infiltration of inflammatory cells, and arteriole sclerosis. Improved renal pathology was observed in aged, AVT-treated rats, and included a decreased glomerular sclerosis index and tubulointerstitial lesion score, especially in those receiving high-dose AVT. Additionally, renal artery wall thickening, luminal narrowing, and arteriolosclerosis were significantly less severe in aged rats receiving high-dose AVT. Upregulated expression of MMP-9 and TGF-ß1 was observed in the renal tissue of aged rats. AVT treatment was associated with a reversal of these phenomena and upregulated expression of TIMP-1, PPARα, PPARß, and PPARγ in aged rats. CONCLUSION: AVT improved the renal pathology of aged rats. These effects may have been induced by the lowering of blood lipids, maintaining the MMP/TIMP balance, and downregulating the expression of TGF-ß1. AVT may reduce the levels of MMP-9 and TGF-ß in aged rats by upregulating the expression of PPARs.


Subject(s)
Atorvastatin/pharmacology , Glomerulosclerosis, Focal Segmental/drug therapy , Kidney/drug effects , Matrix Metalloproteinase 9/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Transforming Growth Factor beta1/metabolism , Age Factors , Animals , Atrophy , Down-Regulation , Female , Glomerulosclerosis, Focal Segmental/enzymology , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Kidney/enzymology , Kidney/pathology , Lipids/blood , Matrix Metalloproteinase 9/genetics , Peroxisome Proliferator-Activated Receptors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Transforming Growth Factor beta1/genetics , Up-Regulation
12.
Biochim Biophys Acta ; 1842(7): 1028-40, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24662305

ABSTRACT

Renal ubiquitin C-terminal hydrolase L1 (UCHL1) is upregulated in a subset of human glomerulopathies, including focal segmental glomerulosclerosis (FSGS), where it may serve to promote ubiquitin pools for degradation of cytotoxic proteins. In the present study, we tested whether UCHL1 is expressed in podocytes of a mouse model of ACTN4-associated FSGS. Podocyte UCHL1 protein was detected in glomeruli of K256E-ACTN4(pod+)/UCHL1+/+ mice. UCHL1+/- mice were intercrossed with K256E-ACTN4(pod+) mice and monitored for features of glomerular disease. 10-week-old K256E-ACTN4(pod+)/UCHL1-/- mice exhibited significantly ameliorated albuminuria, glomerulosclerosis, tubular pathology and blood pressure. Interestingly, while UCHL1 deletion diminished both tubular and glomerular apoptosis, WT1-positive nuclei were unchanged. Finally, UCHL1 levels correlated positively with poly-ubiquitinated proteins but negatively with K256E-α-actinin-4 levels, implying reduced K256E-α-actinin-4 proteolysis in the absence of UCHL1. Our data suggest that UCHL1 upregulation in ACTN4-associated FSGS fuels the proteasome and that UCHL1 deletion may impair proteolysis and thereby preserve K256E/wt-α-actinin-4 heterodimers, maintaining podocyte cytoskeletal integrity and protecting the glomerular filtration barrier.


Subject(s)
Actinin/genetics , Glomerulosclerosis, Focal Segmental/genetics , Sequence Deletion , Ubiquitin Thiolesterase/genetics , Actinin/metabolism , Animals , Cytoskeleton/genetics , Cytoskeleton/metabolism , Disease Models, Animal , Genetic Predisposition to Disease , Glomerulosclerosis, Focal Segmental/enzymology , Glomerulosclerosis, Focal Segmental/metabolism , Kidney Glomerulus/enzymology , Kidney Glomerulus/metabolism , Mice , Mice, Knockout , Podocytes/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Up-Regulation
13.
Kidney Int ; 85(2): 457-70, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24067439

ABSTRACT

Renal toxicity constitutes a dose-limiting side effect of anticancer therapies targeting vascular endothelial growth factor (VEGF). In order to study this further, we followed up 29 patients receiving this treatment, who experienced proteinuria, hypertension, and/or renal insufficiency. Eight developed minimal change nephropathy/focal segmental glomerulopathy (MCN/FSG)-like lesions and 13 developed thrombotic microangiopathy (TMA). Patients receiving receptor tyrosine kinase inhibitors (RTKIs) mainly developed MCN/FSG-like lesions, whereas TMA complicated anti-VEGF therapy. There were no mutations in factor H, factor I, or membrane cofactor protein of the complement alternative pathway, while plasma ADAMTS13 activity persisted and anti-ADAMTS13 antibodies were undetectable in patients with TMA. Glomerular VEGF expression was undetectable in TMA and decreased in MCN/FSG. Glomeruli from patients with TMA displayed a high abundance of RelA in endothelial cells and in the podocyte nuclei, but c-mip was not detected. Conversely, MCN/FSG-like lesions exhibited a high abundance of c-mip, whereas RelA was scarcely detected. RelA binds in vivo to the c-mip promoter and prevents its transcriptional activation, whereas RelA knockdown releases c-mip activation. The RTKI sorafenib inhibited RelA activity, which then promoted c-mip expression. Thus, our results suggest that c-mip and RelA define two distinct types of renal damage associated with VEGF-targeted therapies.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Carrier Proteins/metabolism , Kidney Diseases/chemically induced , Kidney Glomerulus/drug effects , Niacinamide/analogs & derivatives , Phenylurea Compounds/adverse effects , Protein Kinase Inhibitors/adverse effects , Transcription Factor RelA/metabolism , Vascular Endothelial Growth Factors/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Adult , Aged , Animals , Base Sequence , Binding Sites , Biomarkers/metabolism , Carrier Proteins/genetics , Case-Control Studies , Cell Line , Female , Gene Expression Regulation , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/diagnosis , Glomerulosclerosis, Focal Segmental/enzymology , Humans , Hypertension/chemically induced , Hypertension/diagnosis , Hypertension/enzymology , Kidney Diseases/diagnosis , Kidney Diseases/enzymology , Kidney Glomerulus/enzymology , Kidney Glomerulus/pathology , Male , Mice , Mice, Knockout , Middle Aged , Molecular Sequence Data , Nephrosis, Lipoid/chemically induced , Nephrosis, Lipoid/diagnosis , Nephrosis, Lipoid/enzymology , Niacinamide/adverse effects , Predictive Value of Tests , Promoter Regions, Genetic , Proteinuria/chemically induced , Proteinuria/diagnosis , Proteinuria/enzymology , Renal Insufficiency/chemically induced , Renal Insufficiency/diagnosis , Renal Insufficiency/enzymology , Sorafenib , Thrombotic Microangiopathies/chemically induced , Thrombotic Microangiopathies/diagnosis , Thrombotic Microangiopathies/enzymology , Transcription Factor RelA/deficiency , Transcription Factor RelA/genetics , Transcription, Genetic , Transfection , Vascular Endothelial Growth Factors/metabolism , Young Adult
14.
Kidney Int ; 84(5): 861-3, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24172729

ABSTRACT

The kidneys play an important role in protein metabolism. Renal tubules reabsorb 3 g of albumin under normal conditions, and exhibit a 6-fold increase in the reabsorption of albumin in patients with focal segmental glomerulosclerosis. The capacity of tubular lysosomal proteolysis can be increased up to 8-fold; however, proteinuria over the capacity of tubular handling may cause tubulointerstitial damage in patients with nephrotic syndrome.


Subject(s)
Glomerulosclerosis, Focal Segmental/enzymology , Kidney Tubules, Proximal/enzymology , Lysosomes/enzymology , Peptide Hydrolases/metabolism , Animals
17.
Am J Physiol Renal Physiol ; 305(8): F1228-38, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23926186

ABSTRACT

Focal segmental glomerulosclerosis (FSGS) and collapsing glomerulopathy are common causes of nephrotic syndrome. Variants in >20 genes, including genes critical for mitochondrial function, have been associated with these podocyte diseases. One such gene, PDSS2, is required for synthesis of the decaprenyl tail of coenzyme Q10 (Q10) in humans. The mouse gene Pdss2 is mutated in the kd/kd mouse model of collapsing glomerulopathy. We examined the hypothesis that human PDSS2 polymorphisms are associated with podocyte diseases. We genotyped 377 patients with primary FSGS or collapsing glomerulopathy, together with 900 controls, for 9 single-nucleotide polymorphisms in the PDSS2 gene in a case-control study. Subjects included 247 African American (AA) and 130 European American (EA) patients and 641 AA and 259 EA controls. Among EAs, a pair of proxy SNPs was significantly associated with podocyte disease, and patients homozygous for one PDSS2 haplotype had a strongly increased risk for podocyte disease. By contrast, the distribution of PDSS2 genotypes and haplotypes was similar in AA patients and controls. Thus a PDSS2 haplotype, which has a frequency of 13% in the EA control population and a homozygote frequency of 1.2%, is associated with a significantly increased risk for FSGS and collapsing glomerulopathy in EAs. Lymphoblastoid cell lines from FSGS patients had significantly less Q10 than cell lines from controls; contrary to expectation, this finding was independent of PDSS2 haplotype. These results suggest that FSGS patients have Q10 deficiency and that this deficiency is manifested in patient-derived lymphoblastoid cell lines.


Subject(s)
Alkyl and Aryl Transferases/genetics , Glomerulosclerosis, Focal Segmental/enzymology , Glomerulosclerosis, Focal Segmental/genetics , Ubiquinone/analogs & derivatives , Adolescent , Adult , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/pathology , Case-Control Studies , Glomerulosclerosis, Focal Segmental/ethnology , Haplotypes , Humans , Lymphocyte Activation/genetics , Middle Aged , Polymorphism, Single Nucleotide , Ubiquinone/deficiency , Ubiquinone/metabolism , Young Adult
18.
Mol Med Rep ; 8(4): 1183-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23939749

ABSTRACT

In this study, we report the case of a 12-year-old male with X-linked ichthyosis (XLI) in association with glomerular sclerosis, and our investigation into the deletion pattern of the STS gene and the flanking regions in DNA samples of family members. We observed no features typical of renal osteodystrophy or rickets, with the exception of short stature, in the three afffected male family members. Audiometry, visual acuity and olfactory sensation were normal. By performing PCR analysis of the steroid sulfatase (STS) gene and flanking regions on our patients, we discovered a complete deletion that involved the entire region from DXS1139 to DXF22S1. Further studies are required to determine whether the STS gene or the co-deleted flanking sequences are the cause of renal disease associated with XLI.


Subject(s)
Glomerulosclerosis, Focal Segmental/diagnosis , Ichthyosis, X-Linked/diagnosis , Child , DNA Mutational Analysis , Genetic Association Studies , Glomerulosclerosis, Focal Segmental/enzymology , Glomerulosclerosis, Focal Segmental/genetics , Humans , Ichthyosis, X-Linked/enzymology , Ichthyosis, X-Linked/genetics , Male , Pedigree , Sequence Deletion , Skin/pathology , Steryl-Sulfatase/genetics
19.
Kidney Int ; 84(5): 902-10, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23760285

ABSTRACT

Focal segmental glomerulosclerosis (FSGS) is a prevalent cause of end-stage renal disease, but the mechanisms underlying progression are unresolved. Lysosomal protein accumulation in the proximal tubule, mediated by megalin and cubilin endocytosis of increased amounts of filtered protein, is thought to result in inflammation and fibrosis. Here we determine whether release of inflammatory and fibrotic mediators in response to protein overload in the proximal tubule is caused by lysosomal enzyme deficits and insufficient proteolysis. As a model of FSGS, we used inducible podocyte-specific podocin-knockout mice analyzed at different time points. The content of megalin and cubilin ligands increased in the lysosomes after onset of proteinuria; however, protein and mRNA levels of megalin and cubilin showed only minor changes. To determine if the elevated lysosomal ligand content was caused by deficiency of enzymes, we analyzed protein and mRNA levels of lysosomal enzymes and found increased endogenous synthesis. Injection of dye-quenched fluorescent and iodinated albumin showed that proteolytic turnover in lysosomes of knockout mice adapted to the increased protein load. Inflammatory and fibrotic signals were increased early in disease, although the majority of lysosomes degraded endocytosed proteins effectively. Thus, insufficient lysosomal degradation in FSGS is not the cause of the inflammation and fibrosis during kidney disease.


Subject(s)
Glomerulosclerosis, Focal Segmental/enzymology , Kidney Tubules, Proximal/enzymology , Lysosomes/enzymology , Peptide Hydrolases/metabolism , Animals , Disease Models, Animal , Endocytosis , Fibrosis , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Inflammation Mediators/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Kidney Tubules, Proximal/pathology , Ligands , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Lysosomes/pathology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Podocytes/metabolism , Proteinuria/genetics , Proteinuria/metabolism , Proteolysis , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Time Factors , Up-Regulation
20.
J Pathol ; 229(5): 660-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23436459

ABSTRACT

Focal segmental glomerulosclerosis (FSGS) is associated with glomerular podocyte injury. Podocytes undergo dramatic changes in their actin structure, with little mechanistic insight to date into the human disease. Post-transplantation recurrence of FSGS is the archetypal form of the disease caused by unknown circulating plasma 'factors'. There is increasing indication that plasma protease activity could be central to this disease. Using clinical plasma exchange material, collected from patients in relapse and remission stages of disease, the effects of FSGS plasma on human conditionally immortalized podocytes (ciPods) were studied. We show that vasodilator stimulated phosphoprotein (VASP) is phosphorylated in response to relapse plasma from ten consecutively tested patients, and not in response to paired remission plasma or non-FSGS controls. The phosphorylation signal is absent in human podocytes carrying a pathological podocin mutation. To test for a plasma ligand, inhibition of proteases in relapse plasma leads to the loss of VASP phosphorylation. By the use of siRNA technology, we show that proteases in the plasma signal predominantly via protease activated receptor-1 (PAR1) to VASP. Mechanistically, FSGS plasma increases podocyte motility, which is dependent on VASP phosphorylation. These data suggest a specific biomarker for disease activity, as well as revealing a novel and highly specific receptor-mediated signalling pathway to the actin cytoskeleton.


Subject(s)
Cell Adhesion Molecules/metabolism , Glomerulosclerosis, Focal Segmental/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Nephrosis/enzymology , Peptide Hydrolases/blood , Phosphoproteins/metabolism , Podocytes/enzymology , Receptor, PAR-1/metabolism , Actin Cytoskeleton/enzymology , Adolescent , Adult , Cell Line , Cell Movement , Child , Child, Preschool , Female , Glomerulosclerosis, Focal Segmental/blood , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/therapy , Humans , Infant , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Middle Aged , Mutation , Nephrosis/blood , Nephrosis/genetics , Nephrosis/therapy , Phosphorylation , Plasma Exchange , Podocytes/drug effects , Protease Inhibitors/pharmacology , RNA Interference , Receptor, PAR-1/drug effects , Receptor, PAR-1/genetics , Recurrence , Remission Induction , Signal Transduction , Time Factors , Transfection , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...