Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Cell Death Dis ; 13(12): 1061, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36539406

ABSTRACT

Mitochondria take part in a network of intracellular processes that regulate homeostasis. Defects in mitochondrial function are key pathophysiological changes during AKI. Although Wnt/ß-catenin signaling mediates mitochondrial dysfunction in chronic kidney fibrosis, little is known of the influence of ß-catenin on mitochondrial function in AKI. To decipher this interaction, we generated an inducible mouse model of tubule-specific ß-catenin overexpression (TubCat), and a model of tubule-specific ß-catenin depletion (TubcatKO), and induced septic AKI in these mice with lipopolysaccharide (LPS) and aseptic AKI with bilateral ischemia-reperfusion. In both AKI models, tubular ß-catenin stabilization in TubCat animals significantly reduced BUN/serum creatinine, tubular damage (NGAL-positive tubules), apoptosis (TUNEL-positive cells) and necroptosis (phosphorylation of MLKL and RIP3) through activating AKT phosphorylation and p53 suppression; enhanced mitochondrial biogenesis (increased PGC-1α and NRF1) and restored mitochondrial mass (increased TIM23) to re-establish mitochondrial homeostasis (increased fusion markers OPA1, MFN2, and decreased fission protein DRP1) through the FOXO3/PGC-1α signaling cascade. Conversely, kidney function loss and histological damage, tubular cell death, and mitochondrial dysfunction were all aggravated in TubCatKO mice. Mechanistically, ß-catenin transfection maintained mitochondrial mass and activated PGC-1α via FOXO3 in LPS-exposed HK-2 cells. Collectively, these findings provide evidence that tubular ß-catenin mitigates cell death and restores mitochondrial homeostasis in AKI through the common mechanisms associated with activation of AKT/p53 and FOXO3/PGC-1α signaling pathways.


Subject(s)
Acute Kidney Injury , Reperfusion Injury , Mice , Animals , beta Catenin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Acute Kidney Injury/metabolism , Kidney/pathology , Cell Death , Mitochondria/metabolism , Wnt Signaling Pathway , Reperfusion Injury/metabolism
2.
Mol Ther Nucleic Acids ; 26: 1280-1290, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34853727

ABSTRACT

Lipotoxicity has been implicated in the pathogenesis of obesity-related kidney damage and propagates chronic kidney injury like diabetic kidney disease; however, the underlying mechanisms have not yet been fully elucidated. To date, reduction of lipid acquisition and enhancement of lipid metabolism are the major, albeit non-specific, approaches to improve lipotoxic kidney damage. In the kidneys of high-fat diet (HFD)-fed mice and tubule cells cultured with palmitic acid (PA), we observed a dramatic upregulation of the long intergenic non-coding RNA-p21 (LincRNA-p21) through a p53-dependent mechanism. Kidney tubule cell-specific deletion of LincRNA-p21 attenuated oxidative stress, inflammation, apoptosis, and endoplasmic reticulum stress, leading to reduction of histological and functional kidney injury despite persistent obesity and hyperlipidemia. Mechanistically, HFD- or PA-initiated lipotoxicity suppressed the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR)/murine double minute 2 homolog (MDM2) signaling cascade to activate p53 and enhance the transcriptional activity of LincRNA-p21. Collectively, our findings suggest that the p53/LincRNA-p21 axis is the downstream effector in lipotoxic kidney injury and that targeting this axis particularly in the kidney tubule could be a novel therapeutic strategy.

3.
Front Physiol ; 12: 650888, 2021.
Article in English | MEDLINE | ID: mdl-33790807

ABSTRACT

Spleen tyrosine kinase (Syk) is a non-receptor tyrosine kinase involved in signal transduction in a variety of immune responses. It has been demonstrated that Syk plays a pathogenic role in orchestrating inflammatory responses and cell proliferation in human mesangial cells (HMC) in IgA nephropathy (IgAN). However, whether Syk is involved in tubular damage in IgAN remains unknown. Using human kidney biopsy specimens, we found that Syk was activated in renal tubules of biopsy-proven IgAN patients with an increase in total and phosphorylated levels compared to that from healthy control subjects. In vitro, cultured proximal tubular epithelial cells (PTECs) were stimulated with conditioned medium prepared from human mesangial cells incubated with polymeric IgA (IgA-HMC) from patients with IgAN or healthy control. Induction of IL-6, IL-8, and ICAM-1 synthesis from cultured PTECs incubated with IgA-HMC conditioned medium was significantly suppressed by treatment with the Syk inhibitor R406 compared to that from healthy control. Furthermore, R406 downregulated expression of phosphorylated p65 NF-κB and p-42/p-44 MAPK, and attenuated TNF-α-induced cytokine production in PTECs. Taken together, our findings suggest that Syk mediates IgA-HMC conditioned medium-induced inflammation in tubular cells via activation of NF-κB and p-42/p-44 MAPK signaling. Inhibition of Syk may be a potential therapeutic approach for tubulointerstitial injury in IgAN.

4.
Clin Sci (Lond) ; 135(3): 429-446, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33458750

ABSTRACT

Kallistatin is a multiple functional serine protease inhibitor that protects against vascular injury, organ damage and tumor progression. Kallistatin treatment reduces inflammation and fibrosis in the progression of chronic kidney disease (CKD), but the molecular mechanisms underlying this protective process and whether kallistatin plays an endogenous role are incompletely understood. In the present study, we observed that renal kallistatin levels were significantly lower in patients with CKD. It was also positively correlated with estimated glomerular filtration rate (eGFR) and negatively correlated with serum creatinine level. Unilateral ureteral obstruction (UUO) in animals also led to down-regulation of kallistatin protein in the kidney, and depletion of endogenous kallistatin by antibody injection resulted in aggravated renal fibrosis, which was accompanied by enhanced Wnt/ß-catenin activation. Conversely, overexpression of kallistatin attenuated renal inflammation, interstitial fibroblast activation and tubular injury in UUO mice. The protective effect of kallistatin was due to the suppression of TGF-ß and ß-catenin signaling pathways and subsequent inhibition of epithelial-to-mesenchymal transition (EMT) in cultured tubular cells. In addition, kallistatin could inhibit TGF-ß-mediated fibroblast activation via modulation of Wnt4/ß-catenin signaling pathway. Therefore, endogenous kallistatin protects against renal fibrosis by modulating Wnt/ß-catenin-mediated EMT and fibroblast activation. Down-regulation of kallistatin in the progression of renal fibrosis underlies its potential as a valuable clinical biomarker and therapeutic target in CKD.


Subject(s)
Renal Insufficiency, Chronic/pathology , Serpins/metabolism , Ureteral Obstruction/pathology , Wnt Signaling Pathway , Adult , Aged , Animals , Disease Models, Animal , Female , Fibrosis/pathology , Humans , Kidney/pathology , Male , Mice, Inbred BALB C , Middle Aged , Rats, Sprague-Dawley , Transforming Growth Factor beta/metabolism , beta Catenin/metabolism
5.
Clin Sci (Lond) ; 134(21): 2873-2891, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33078834

ABSTRACT

Protease-activated receptor (PAR)-1 has emerged as a key profibrotic player in various organs including kidney. PAR-1 activation leads to deposition of extracellular matrix (ECM) proteins in the tubulointerstitium and induction of epithelial-mesenchymal transition (EMT) during renal fibrosis. We tested the anti-fibrotic potential of vorapaxar, a clinically approved PAR-1 antagonist for cardiovascular protection, in an experimental kidney fibrosis model of unilateral ureteral obstruction (UUO) and an AKI-to-chronic kidney disease (CKD) transition model of unilateral ischemia-reperfusion injury (UIRI), and dissected the underlying renoprotective mechanisms using rat tubular epithelial cells. PAR-1 is activated mostly in the renal tubules in both the UUO and UIRI models of renal fibrosis. Vorapaxar significantly reduced kidney injury and ameliorated morphologic changes in both models. Amelioration of kidney fibrosis was evident from down-regulation of fibronectin (Fn), collagen and α-smooth muscle actin (αSMA) in the injured kidney. Mechanistically, inhibition of PAR-1 inhibited MAPK ERK1/2 and transforming growth factor-ß (TGF-ß)-mediated Smad signaling, and suppressed oxidative stress, overexpression of pro-inflammatory cytokines and macrophage infiltration into the kidney. These beneficial effects were recapitulated in cultured tubular epithelial cells in which vorapaxar ameliorated thrombin- and hypoxia-induced TGF-ß expression and ECM accumulation. In addition, vorapaxar mitigated capillary loss and the expression of adhesion molecules on the vascular endothelium during AKI-to-CKD transition. The PAR-1 antagonist vorapaxar protects against kidney fibrosis during UUO and UIRI. Its efficacy in human CKD in addition to CV protection warrants further investigation.


Subject(s)
Kidney/injuries , Lactones/pharmacology , Pyridines/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Animals , Biomarkers/metabolism , Cell Hypoxia/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Extracellular Matrix Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Inflammation/pathology , Kidney/drug effects , Kidney/pathology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Macrophages/drug effects , Macrophages/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Receptor, PAR-1/metabolism , Reperfusion Injury/complications , Smad3 Protein/metabolism , Thrombin/pharmacology , Transforming Growth Factor beta/metabolism , Up-Regulation/drug effects , Ureteral Obstruction/complications , Ureteral Obstruction/pathology
6.
Prog Lipid Res ; 77: 101020, 2020 01.
Article in English | MEDLINE | ID: mdl-31870728

ABSTRACT

Obesity is featured by chronic systemic low-grade inflammation that eventually contributes to the development of insulin resistance. Toll-like receptor 4 (TLR4) is an important mediator that triggers the innate immune response by activating inflammatory signaling cascades. Human, animal and cell culture studies identified saturated fatty acids (SFAs), the dominant non-esterified fatty acid (NEFA) in the circulation of obese subjects, as non-microbial agonists that trigger the inflammatory response via activating TLR4 signaling, which acts as an important causative link between fatty acid overload, chronic low-grade inflammation and the related metabolic aberrations. The interaction between SFAs and TLR4 may be modulated through the myeloid differentiation primary response gene 88-dependent and independent signaling pathway. Greater understanding of the crosstalk between dietary SFAs and TLR4 signaling in the pathogenesis of metabolic imbalance may facilitate the design of a more efficient pharmacological strategy to alleviate the risk of developing chronic diseases elicited in part by fatty acid overload. The current review discusses recent advances in the impact of crosstalk between SFAs and TLR4 on inflammation and insulin resistance in multiple cell types, tissues and organs in the context of metabolic dysregulation.


Subject(s)
Fatty Acids/genetics , Inflammation/genetics , Insulin Resistance/genetics , Toll-Like Receptor 4/genetics , Animals , Humans , Immunity, Innate/genetics , Inflammation/metabolism , Inflammation/pathology , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Signal Transduction/genetics
7.
Stem Cells Transl Med ; 8(9): 898-910, 2019 09.
Article in English | MEDLINE | ID: mdl-31054183

ABSTRACT

Recent advances in the understanding of lipid metabolism suggest a critical role of endoplasmic reticulum (ER) stress in obesity-induced kidney injury. Hepatocyte growth factor (HGF) is a pleiotropic cytokine frequently featured in stem cell therapy with distinct renotropic benefits. This study aims to define the potential link between human induced pluripotent stem cell-derived mesenchymal stem cells (iPS-MSCs)/bone marrow-derived MSCs (BM-MSCs) and ER stress in lipotoxic kidney injury induced by palmitic acid (PA) in renal tubular cells and by high-fat diet (HFD) in mice. iPS-MSCs or BM-MSCs alleviated ER stress (by preventing induction of Bip, chop, and unfolded protein response), inflammation (Il6, Cxcl1, and Cxcl2), and apoptosis (Bax/Bcl2 and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling-positive cells) in renal cortex of animals exposed to HFD thus mitigating histologic damage and albuminuria, via activating HGF/c-Met paracrine signaling that resulted in enhanced HGF secretion in the glomerular compartment and c-Met expression in the tubules. Coculture experiments identified glomerular endothelial cells (GECs) to be the exclusive source of glomerular HGF when incubated with either iPS-MSCs or BM-MSCs in the presence of PA. Furthermore, both GEC-derived HGF and exogenous recombinant HGF attenuated PA-induced ER stress in cultured tubular cells, and this effect was abrogated by a neutralizing anti-HGF antibody. Taken together, this study is the first to demonstrate that MSCs ameliorate lipotoxic kidney injury via a novel microenvironment-dependent paracrine HGF/c-Met signaling mechanism to suppress ER stress and its downstream pro-inflammatory and pro-apoptotic consequences. Stem Cells Translational Medicine 2019;8:898&910.


Subject(s)
Endoplasmic Reticulum Stress , Hepatocyte Growth Factor/metabolism , Obesity/pathology , Proto-Oncogene Proteins c-met/metabolism , Animals , Apoptosis/drug effects , Coculture Techniques , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Diet, High-Fat , Endoplasmic Reticulum Stress/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hepatocyte Growth Factor/genetics , Kidney/metabolism , Kidney/pathology , Kidney Tubules/cytology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Obesity/metabolism , Palmitic Acid/toxicity , Paracrine Communication/drug effects , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Signal Transduction/drug effects
8.
Semin Nephrol ; 38(5): 485-495, 2018 09.
Article in English | MEDLINE | ID: mdl-30177020

ABSTRACT

IgA nephropathy (IgAN), a common primary glomerulonephritis worldwide, is associated with a substantial risk of progression to end-stage renal failure. The disease runs a highly variable clinical course with frequent involvement of tubulointerstitial damage. A subgroup of IgAN with proximal tubular epithelial cells (PTECs) and tubulointerstitial damage often is associated with rapid progression to end-stage renal failure. Human mesangial cell-derived mediators lead to podocyte and tubulointerstitial injury via mesangial-podocytic-tubular cross-talk. Although mesangial-podocytic communication plays a pathogenic role in podocytic injury, the implication of a podocyte-PTEC cross-talk pathway in the progression of tubulointerstitial injury in IgAN should not be underscored. We review the role of mesangial-podocytic-tubular cross-talk in the progression of IgAN. We discuss how podocytopathy in IgAN promotes subsequent PTEC dysfunction and whether tubulointerstitial injury affects the propagation of podocytic injury in IgAN. A thorough understanding of the cross-talk mechanisms among mesangial cells, podocytes, and PTECs may lead to better design of potential therapeutic options for IgAN.


Subject(s)
Epithelial Cells/metabolism , Glomerulonephritis, IGA/metabolism , Kidney Tubules/metabolism , Mesangial Cells/metabolism , Podocytes/metabolism , Epithelial Cells/cytology , Humans , Kidney Tubules/cytology , Mesangial Cells/cytology , Podocytes/cytology
9.
Kidney Int ; 93(6): 1367-1383, 2018 06.
Article in English | MEDLINE | ID: mdl-29605095

ABSTRACT

Imbalance of Wnt/ß-catenin signaling in renal cells is associated with renal dysfunction, yet the precise mechanism is poorly understood. Previously we observed activated Wnt/ß-catenin signaling in renal tubules during proteinuric nephropathy with an unknown net effect. Therefore, to identify the definitive role of tubular Wnt/ß-catenin, we generated a novel transgenic "Tubcat" mouse conditionally expressing stabilized ß-catenin specifically in renal tubules following tamoxifen administration. Four weeks after tamoxifen injection, uninephrectomized Tubcat mice displayed proteinuria and elevated blood urea nitrogen levels compared to non-transgenic mice, implying a detrimental effect of the activated signaling. This was associated with infiltration of the tubulointerstitium predominantly by M1 macrophages and overexpression of the inflammatory chemocytokines CCL-2 and RANTES. Induction of overload proteinuria by intraperitoneal injection of low-endotoxin bovine serum albumin following uninephrectomy for four weeks aggravated proteinuria and increased blood urea nitrogen levels to a significantly greater extent in Tubcat mice. Renal dysfunction correlated with the degree of M1 macrophage infiltration in the tubulointerstitium and renal cortical up-regulation of CCL-2, IL-17A, IL-1ß, CXCL1, and ICAM-1. There was overexpression of cortical TLR-4 and NLRP-3 in Tubcat mice, independent of bovine serum albumin injection. Finally, there was no fibrosis, activation of epithelial-mesenchymal transition or non-canonical Wnt pathways observed in the kidneys of Tubcat mice. Thus, conditional activation of renal tubular Wnt/ß-catenin signaling in a novel transgenic mouse model demonstrates that this pathway enhances intrarenal inflammation via the TLR-4/NLRP-3 inflammasome axis in overload proteinuria.


Subject(s)
Inflammation Mediators/metabolism , Kidney Tubules/metabolism , Macrophages/metabolism , Nephritis/metabolism , Proteinuria/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Chemokine CCL2/metabolism , Chemokine CCL5/metabolism , Disease Models, Animal , Inflammasomes/metabolism , Kidney Tubules/pathology , Kidney Tubules/physiopathology , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephrectomy , Nephritis/genetics , Nephritis/pathology , Nephritis/physiopathology , Proteinuria/genetics , Proteinuria/pathology , Proteinuria/physiopathology , Serum Albumin, Bovine , Toll-Like Receptor 4/metabolism , Up-Regulation , Wnt Signaling Pathway/genetics , beta Catenin/genetics
10.
Nephrol Dial Transplant ; 33(8): 1323-1332, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29294056

ABSTRACT

Background: Complement C5 mediates pro-inflammatory responses in many immune-related renal diseases. Given that the C5a level is elevated in diabetes, we investigated whether activation of C5a/C5aR signalling plays a pathogenic role in diabetic nephropathy (DN) and the therapeutic potential of C5a inhibition for renal fibrosis. Methods: Human renal biopsies from patients with DN and control subjects were used for immunohistochemical staining of complement C5 components. Renal function and tubulointerstitial injury were compared between db/m mice, vehicle-treated mice and C5a inhibitor-treated db/db mice. A cell culture model of tubule epithelial cells (HK-2) was used to demonstrate the effect of C5a on the renal fibrotic pathway. Results: Increased levels of C5a, but not of its receptor C5aR, were detected in renal tubules from patients with DN. The intensity of C5a staining was positively correlated with the progression of the disease. In db/db mice, administration of a novel C5a inhibitor, NOX-D21, reduced the serum triglyceride level and attenuated the upregulation of diacylglycerolacyltransferase-1 and sterol-regulatory element binding protein-1 expression and lipid accumulation in diabetic kidney. NOX-D21-treated diabetic mice also had reduced serum blood urea nitrogen and creatinine levels with less glomerular and tubulointerstitial damage. Renal transforming growth factor beta 1 (TGF-ß1), fibronectin and collagen type I expressions were reduced by NOX-D21. In HK-2 cells, C5a stimulated TGF-ß production through the activation of the PI3K/Akt signalling pathway. Conclusions: Blockade of C5a signalling by NOX-D21 moderates altered lipid metabolism in diabetes and improved tubulointerstitial fibrosis by reduction of lipid accumulation and TGF-ß-driven fibrosis in diabetic kidney.


Subject(s)
Aptamers, Nucleotide/pharmacology , Complement C5a/antagonists & inhibitors , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/complications , Fibrosis/prevention & control , Kidney Diseases/prevention & control , Lipid Metabolism/drug effects , Animals , Fibrosis/etiology , Fibrosis/metabolism , Humans , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Serine Endopeptidases/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism
11.
Oncotarget ; 8(61): 103640-103656, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29262590

ABSTRACT

Human induced pluripotent stem cell-derived mesenchymal stem cells (iPS-MSCs) are emerging as attractive options for use in cell replacement therapy, but their effect in kidney diseases remains unknown. Here, we showed that intravenous injection of iPS-MSCs protect against renal function loss in both short-term and long-term models of adriamycin nephropathy (AN). In the short-term AN model, iPS-MSCs conferred a substantial anti-apoptotic effect on tubular cells, associated with a downregulation of Bax and Bax/Bcl2 ratio and an upregulation of survivin expression. In vitro, conditioned medium from iPS-MSCs (iPSMSC-CM) significantly limited albumin-induced tubular apoptosis and enhanced tubular proliferation, accompanied by a reduced expression of tubular Bax and an elevated expression of Bcl2 and survivin. Oxidative stress was markedly attenuated by iPS-MSCs both in AN mice and in protein-overloaded tubular cells. In the long-term AN model, repeated injections of iPS-MSCs significantly inhibited tubulointerstitial fibrosis and reduced intrarenal deposition of collagen I, collagen IV and αSMA. Modulation of the hedgehog signaling pathway contributed to the anti-fibrotic effect of iPS-MSCs in chronic AN. Finally, we detected that most of the infused iPS-MSCs were entrapped in the lungs. In conclusion, our data support a beneficial role of iPS-MSCs in both acute and chronic AN.

12.
F1000Res ; 52016.
Article in English | MEDLINE | ID: mdl-26918170

ABSTRACT

Since its first description in 1968, IgA nephropathy has remained the most common form of primary glomerulonephritis leading to chronic kidney disease in developed countries. The clinical progression varies, and consequent end-stage renal disease occurs in 30% to 40% of patients 20 to 30 years after the first clinical presentation. Current data implicate overproduction of aberrantly glycosylated IgA1 as being pivotal in the induction of renal injury. Effective and specific treatment is still lacking, and new therapeutic approaches will be developed after better understanding the disease pathogenesis.

13.
Kidney Int ; 89(2): 386-98, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26536000

ABSTRACT

Kallistatin is a serine protease inhibitor with anti-inflammatory, anti-angiogenic, and anti-oxidative properties. Since oxidative stress plays a critical role in the pathogenesis of diabetic nephropathy, we studied the effect and mechanisms of action of kallistatin superinduction. Using ultrasound-microbubble-mediated gene transfer, kallistatin overexpression was induced in kidney tubules. In db/db mice, kallistatin overexpression reduced serum creatinine and BUN levels, ameliorated glomerulosclerosis and tubulointerstitial injury, and attenuated renal fibrosis by inhibiting TGF-ß signaling. Additionally, downstream PAI-1 and collagens I and IV expression were reduced and kallistatin partially suppressed renal inflammation by inhibiting NF-κB signaling and decreasing tissue kallikrein activity. Kallistatin lowered blood pressure and attenuated oxidative stress as evidenced by suppressed levels of NADPH oxidase 4, and oxidative markers (nitrotyrosine, 8-hydroxydeoxyguanosine, and malondialdehyde) in diabetic renal tissue. Kallistatin also inhibited RAGE expression in the diabetic kidney and AGE-stimulated cultured proximal tubular cells. Reduced AGE-induced reactive oxygen species generation reflected an anti-oxidative mechanism via the AGE-RAGE-reactive oxygen species axis. These results indicate a renoprotective role of kallistatin against diabetic nephropathy by multiple mechanisms including suppression of oxidative stress, anti-fibrotic and anti-inflammatory actions, and blood pressure lowering.


Subject(s)
Diabetic Nephropathies/prevention & control , Genetic Therapy , Receptor for Advanced Glycation End Products/metabolism , Serpins/physiology , Animals , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Fibrosis , Gene Transfer Techniques , Kallikreins/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Function Tests , Male , Mice, Inbred C57BL , Mice, Transgenic , NADPH Oxidase 4 , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Neovascularization, Pathologic , Oxidative Stress , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vascular Endothelial Growth Factor A/metabolism
14.
Kidney Int ; 89(3): 683-92, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26581012

ABSTRACT

Immunoglobulin A nephropathy (IgAN) is a worldwide disease characterized by the presence of galactose-deficient IgA1 deposits in the glomerular mesangium. A kidney biopsy for diagnosis is required. Here, we measured two miRNAs (let-7b and miR-148b), previously identified as regulators of the O-glycosylation process of IgA1, in serum samples from patients with IgAN and healthy blood donors (controls) recruited in an international multicenter study. Two predictive models, based on these miRNAs, were developed and the diagnostic accuracy of the combined biomarkers was assessed by the area under the receiver operating characteristic (ROC) curve (AUC) carried out in three steps. In a training study, the combined miRNAs were able to discriminate between 100 patients with IgAN and 119 controls (AUC, 0.82). A validation study confirmed the model in an independent cohort of 145 patients with IgAN and 64 controls (AUC, 0.78). Finally, in a test study, the combined biomarkers were able to discriminate patients with IgAN from 105 patients affected by other forms of primary glomerulonephritis, supporting the specificity (AUC, 0.76). Using the same study design, we also performed two subgroup analyses (one for Caucasians and one for East Asians) and found that race-specific models were the best fit to distinguish IgAN patients from controls. Thus, serum levels of the combined miRNA biomarker, let-7b and miR-148b, appears to be a novel, reliable, and noninvasive test to predict the probability of having IgAN.


Subject(s)
Glomerulonephritis, IGA/blood , MicroRNAs/blood , Adult , Area Under Curve , Asian People/genetics , Female , Genetic Markers , Glomerulonephritis, IGA/diagnosis , Glomerulonephritis, IGA/ethnology , Glomerulonephritis, IGA/genetics , Greece/epidemiology , Hong Kong/epidemiology , Humans , Italy/epidemiology , Japan/epidemiology , Male , MicroRNAs/genetics , Middle Aged , Predictive Value of Tests , Prognosis , ROC Curve , Reproducibility of Results , Retrospective Studies , White People/genetics
15.
Gene ; 571(1): 43-51, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26095808

ABSTRACT

BACKGROUND: Previously, a large proportion of the genetic components predisposing individuals to IgA nephropathy (IgAN) have been unidentified. Familial IgAN is enriched with genetic variations predisposing individuals to the disease. Whole exome sequencing is an effective way to explore disease-causing genes and gene variants. METHODS: We performed exome sequencing on the probands from each of ten IgAN families, and on one of the unaffected member from 7 of the families. Sanger sequencing, bioinformatics and co-segregation analysis were performed for all available family members to detect deleterious genetic variation. The relatedness of the families was tested by haplotype analyses. RESULTS: Six deleterious variants in 4 genes were observed to be associated with IgA nephropathy by co-segregating with the disease phenotypes in study families. MYCT1 p.Asp22Glufs*34 was associated with IgAN by co-segregating with its phenotypes in families 2, 7, and 9; DEFA4 p.Ala8Pro, p.Ala8Val, c.172+1G>T co-segregated in families 1, 2, and 3; ZNF543 p.Pro226Ala co-segregated in families 3, 5, and 6 and CARD8 p.Val98Lysfs*26 co-segregated in families 7 and 8. Among these genes, MYCT1, CARD8 and ZNF543 are novel. Our haplotype analyses showed that families in which the same variation(s) were co-segregating with IgAN were unrelated, except for DEFA4. Of the families carrying DEFA4, families 2 and 3 were possibly related, but not family 1, indicating that common genes/variations in these families were not due to the same founder. Interfamilial sharing of different co-segregating genes was also observed, demonstrating the polygenic nature of this disease. CONCLUSIONS: We discovered 6 deleterious variants in 4 genes associated with familial IgAN. These genes are good candidate genes that appear to be causally related to IgAN and warrant further study.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease/genetics , Glomerulonephritis, IGA/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Adult , Aged , Exome/genetics , Family Health , Glomerulonephritis, IGA/pathology , Haplotypes , Humans , Middle Aged , Pedigree , Phenotype , Sequence Analysis, DNA/methods , alpha-Defensins/genetics
16.
Apoptosis ; 20(7): 907-20, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25808596

ABSTRACT

Glomerulo-podocytic communication plays an important role in the podocytic injury in IgA nephropathy (IgAN). In this study, we examine the role of podocytic angiotensin II receptor subtype 1 (AT1R) and prorenin receptor (PRR) in podocytic apoptosis in IgAN. Polymeric IgA (pIgA) was isolated from patients with IgAN and healthy controls. Conditioned media were prepared from growth arrested human mesangial cells (HMC) incubated with pIgA from patients with IgAN (IgA-HMC media) or healthy controls (Ctl-HMC media). A human podocyte cell line was used as a model to examine the regulation of the expression of AT1R, PRR, TNF-α and CTGF by IgA-HMC media. Podocytic nephrin expression, annexin V binding and caspase 3 activity were used as the functional readout of podocytic apoptosis. IgA-HMC media had no effect on AngII release by podocytes. IgA-HMC media significantly up-regulated the expression of AT1R and PRR, down-regulated nephrin expression and induced apoptosis in podocytes. Mono-blockade of AT1R, PRR, TNF-α or CTGF partially reduced podocytic apoptosis. IgA-HMC media activated NFκB, notch1 and HEY1 expression by podocytes and dual blockade of AT1R with PRR, or anti-TNF-α with anti-CTGF, effectively rescued the podocytic apoptosis induced by IgA-HMC media. Our data suggests that pIgA-activated HMC up-regulates the expression of AT1R and PRR expression by podocytes and the associated activation of NFκB and notch signalling pathways play an essential role in the podocytic apoptosis induced by glomerulo-podocytic communication in IgAN. Simultaneously targeting the AT1R and PRR could be a potential therapeutic option to reduce the podocytic injury in IgAN.


Subject(s)
Apoptosis , Mesangial Cells/cytology , Podocytes/metabolism , Receptors, Angiotensin/metabolism , Receptors, Cell Surface/metabolism , Asian People , Female , Humans , Immunoglobulin A/metabolism , Male , Mesangial Cells/metabolism , Receptors, Notch/metabolism , Renin-Angiotensin System , Signal Transduction , Prorenin Receptor
17.
Clin Sci (Lond) ; 128(4): 269-80, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25200314

ABSTRACT

Bone morphogenetic protein 7 (BMP7) has been reported to confer renoprotective effects in acute and chronic kidney disease models, but its potential role in Type 2 diabetic nephropathy remains unknown. In cultured human proximal tubular epithelial cells (PTECs), exposure to advanced glycation end-products (AGEs) induced overexpression of intercellular adhesion molecule 1 (ICAM1), monocyte chemoattractant protein 1 (MCP1), interleukin 8 (IL-8) and interleukin 6 (IL-6), involving activation of p44/42 and p38 mitogen-activated protein kinase (MAPK) signalling. BMP7 dose-dependently attenuated AGE-induced up-regulation of ICAM1, MCP1, IL-8 and IL-6 at both mRNA and protein levels. Moreover, BMP7 suppressed AGE-induced p38 and p44/42 MAPK phosphorylation and reactive oxygen species production in PTECs. Compared with vehicle control, uninephrectomized db/db mice treated with BMP7 for 8 weeks had significantly lower urinary albumin-to-creatinine ratio (3549±816.2 µg/mg compared with 8612±2037 µg/mg, P=0.036), blood urea nitrogen (33.26±1.09 mg/dl compared with 37.49±0.89 mg/dl, P=0.006), and renal cortical expression of ICAM1 and MCP1 at both gene and protein levels. In addition, BMP7-treated animals had significantly less severe tubular damage, interstitial inflammatory cell infiltration, renal cortical p38 and p44/42 phosphorylation and lipid peroxidation. Our results demonstrate that BMP7 attenuates tubular pro-inflammatory responses in diabetic kidney disease by suppressing oxidative stress and multiple inflammatory signalling pathways including p38 and p44/42 MAPK. Its potential application as a therapeutic molecule in diabetic nephropathy warrants further investigation.


Subject(s)
Bone Morphogenetic Protein 7/pharmacology , Bone Morphogenetic Protein 7/therapeutic use , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Inflammation/pathology , Oxidative Stress/drug effects , Animals , Diabetic Nephropathies/complications , Diabetic Nephropathies/physiopathology , Glycation End Products, Advanced/metabolism , Humans , Inflammation/complications , Inflammation/drug therapy , Kidney Function Tests , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiopathology , Lipid Peroxidation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Kidney Dis (Basel) ; 1(1): 19-26, 2015 May.
Article in English | MEDLINE | ID: mdl-27536661

ABSTRACT

BACKGROUND: IgA nephropathy (IgAN) is a very common glomerulonephritis worldwide. Nevertheless, treatment options for primary IgAN are still largely based on opinion or weak evidence. There is a lack of large randomized controlled trials (RCT) that provide a definitive immunosuppressive protocol for IgAN. The recent KDIGO Clinical Practice Guidelines for Glomerulonephritis have assigned low levels of evidence for almost all recommendations and suggestions related to this nephropathy. SUMMARY: In this article, we review different treatment options and emphasize that the key to therapeutic decision-making is the assessment of an individual's prognosis. The risk of disease progression is closely related to clinical parameters such as proteinuria, hypertension, and impaired glomerular filtration rate. For patients with minor urinary abnormalities, the mainstay of treatment is long-term regular follow-up to detect renal progression and hypertension. Optimized supportive care aiming to maintain proteinuria <1 g/day is preferred in the typical patient presenting with microhematuria, significant but nonnephrotic proteinuria, hypertension, and variable degrees of renal failure. The atypical patient with overt nephritic syndrome or rapidly progressive kidney injury that represents a vasculitic form of IgAN should be treated with immunosuppression. Finally, the variant of overlapping syndrome of IgAN and lipoid nephrosis that runs a good prognosis should be treated as lipoid nephrosis. KEY MESSAGE: The treatment of IgAN should be structured according to the clinical scenario.

19.
Inflamm Res ; 63(10): 831-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25063374

ABSTRACT

OBJECTIVE: Kidney injury molecule-1 (KIM-1) serves as a useful marker for monitoring tubular injury, and sustained KIM-1 expression may be implicated in chronic kidney fibrosis. In this study, we examine the kinetics and mechanisms of KIM-1 release in human proximal tubular epithelial cells (PTEC) under the activation by major pathologic players in diabetic nephropathy, including human serum albumin (HSA), glycated albumin (AGE-BSA) and high glucose. MATERIALS AND METHODS: The kinetics of KIM-1 release by PTEC under activation with HSA, AGE-BSA and high glucose, were determined by RT-PCR and ELISA. The activation profiles of major signaling pathways in PTEC were identified by PCR array. Based on the array data, blockade experiments were designed to assess their regulatory roles in KIM-1 release. RESULTS: Prompt shedding of KIM-1 was observed in PTEC cultured for 4 h with HSA and AGE-BSA, but not with high glucose. Culturing PTEC for 3 days with AGE-BSA exhibited sustained up-regulation of KIM-1 release, but not with HSA. In all culture experiments, high glucose did not induce KIM-1 release in PTEC. HSA and AGE-BSA activated multiple signaling pathways in PTEC including NFκB, ERK1/2 and the oxidative stress pathways. Long-term culturing PTEC with AGE-BSA but not HSA activated the Jak-Stat pathway. While incubation of PTEC with diphenylene iodonium blocked the short-term release of KIM-1 mediated by HSA or AGE-BSA, Jak-Stat inhibitors diminished the long-term KIM-1 release by PTEC induced by AGE-BSA. CONCLUSION: KIM-1 release in PTEC was differentially up-regulated by HSA and AGE-BSA. The short-term KIM-1 shedding was mediated by the reactive oxygen species, whereas Jak-Stat pathway regulates the long-term KIM-1 release.


Subject(s)
Epithelial Cells/drug effects , Glucose/pharmacology , Glycation End Products, Advanced/pharmacology , Membrane Glycoproteins/metabolism , Receptors, Virus/metabolism , Serum Albumin, Bovine/pharmacology , Serum Albumin/pharmacology , Cells, Cultured , Epithelial Cells/metabolism , Hepatitis A Virus Cellular Receptor 1 , Humans , Janus Kinases/metabolism , Kidney Tubules, Proximal/cytology , Kinetics , Matrix Metalloproteinase 3/metabolism , Membrane Glycoproteins/genetics , Reactive Oxygen Species/metabolism , Receptors, Virus/genetics , STAT Transcription Factors/metabolism , Signal Transduction
20.
PLoS One ; 9(3): e90883, 2014.
Article in English | MEDLINE | ID: mdl-24646687

ABSTRACT

Bone marrow-derived mesenchymal stem cells (BM-MSCs) have recently shown promise as a therapeutic tool in various types of chronic kidney disease (CKD) models. However, the mechanism of action is incompletely understood. As renal prognosis in CKD is largely determined by the degree of renal tubular injury that correlates with residual proteinuria, we hypothesized that BM-MSCs may exert modulatory effects on renal tubular inflammation and epithelial-to-mesenchymal transition (EMT) under a protein-overloaded milieu. Using a co-culture model of human proximal tubular epithelial cells (PTECs) and BM-MSCs, we showed that concomitant stimulation of BM-MSCs by albumin excess was a prerequisite for them to attenuate albumin-induced IL-6, IL-8, TNF-α, CCL-2, CCL-5 overexpression in PTECs, which was partly mediated via deactivation of tubular NF-κB signaling. In addition, albumin induced tubular EMT, as shown by E-cadherin loss and α-SMA, FN and collagen IV overexpression, was also prevented by BM-MSC co-culture. Albumin-overloaded BM-MSCs per se retained their tri-lineage differentiation capacity and overexpressed hepatocyte growth factor (HGF) and TNFα-stimulating gene (TSG)-6 via P38 and NF-κB signaling. Albumin-induced tubular CCL-2, CCL-5 and TNF-α overexpression were suppressed by recombinant HGF treatment, while the upregulation of α-SMA, FN and collagen IV was attenuated by recombinant TSG-6. Neutralizing HGF and TSG-6 abolished the anti-inflammatory and anti-EMT effects of BM-MSC co-culture in albumin-induced PTECs, respectively. In vivo, albumin-overloaded mice treated with mouse BM-MSCs had markedly reduced BUN, tubular CCL-2 and CCL-5 expression, α-SMA and collagen IV accumulation independent of changes in proteinuria. These data suggest anti-inflammatory and anti-fibrotic roles of BM-MSCs on renal tubular cells under a protein overloaded condition, probably mediated via the paracrine action of HGF and TSG-6.


Subject(s)
Albumins/pharmacology , Bone Marrow Cells/cytology , Epithelial Cells/pathology , Kidney Tubules, Proximal/pathology , Mesenchymal Stem Cells/cytology , Actins/genetics , Actins/metabolism , Bone Marrow Cells/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Coculture Techniques , Collagen Type IV/genetics , Collagen Type IV/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Fibrosis/chemically induced , Fibrosis/metabolism , Fibrosis/pathology , Fibrosis/prevention & control , Gene Expression Regulation , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Mesenchymal Stem Cells/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Primary Cell Culture , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL