scholarly journals Response gene to complement 32 interacts with Smad3 to promote epithelial‐mesenchymal transition of renal tubule cells

2010 ◽  
Vol 24 (S1) ◽  
Author(s):  
Xia Guo ◽  
Shi‐You Chen
2021 ◽  
Vol 2021 ◽  
pp. 1-17
Author(s):  
Shoujun Bai ◽  
Xiaoyan Xiong ◽  
Bo Tang ◽  
Tingting Ji ◽  
Xiaoying Li ◽  
...  

Diabetic nephropathy (DN) is the leading cause of end-stage renal disease. The association between epithelial-mesenchymal transition (EMT) and fibrosis is quite ascertained, but its link to eventual tubule dysfunction is missing. Here, we show that human microRNA- (hsa-miR-) 199b-3p protects renal tubules from diabetic-induced injury by repressing KDM6A, a histone lysine demethylase regulating E-cadherin expression. Lower E-cadherin expression is related to a higher level of KDM6A, while E-cadherin is promoted upon treatment with the KDM6A inhibitor GSK-J4 in both high glucose- (HG-) induced HK2 cells and the kidneys from streptozotocin- (STZ-) induced type 1 diabetic mice. However, overexpression or RNA silencing of E-cadherin fails to alter KDM6A expression. We also show that the upregulation of KDM6A is associated with the increased methylation level of the E-cadherin promoter. Then, the target prediction results and a dual-luciferase assay show that hsa-miR-199b-3p is a new miRNA that targets KDM6A. Overexpression of hsa-miR-199b-3p increases E-cadherin expression and prevents EMT through repressing KDM6A expression in HG-induced HK2 cells. In contrast, inhibitor-induced hsa-miR-199b-3p knockdown has opposite effects, as it decreases E-cadherin level and worsens EMT, accompanied by increased levels of KDM6A. Besides, Mir199b-knockout mice without mmu-miR-119b-3p expression exhibit more renal tubule dysfunction and more serious kidney tissue damage upon treatment with STZ. These results demonstrate that hsa-miR-199b-3p improves E-cadherin expression and prevents the progression of DN through targeting KDM6A. miR-199b-3p could be a future biomarker or target for the diagnosis or treatment of DN.


2021 ◽  
Vol 22 (19) ◽  
pp. 10252
Author(s):  
Rohan Reddy Nagavally ◽  
Siddharth Sunilkumar ◽  
Mumtaz Akhtar ◽  
Louis D. Trombetta ◽  
Sue M. Ford

Cyclosporine A (CsA) is a nephrotoxicant that causes fibrosis via induction of epithelial–mesenchymal transition (EMT). The flavonoid chrysin has been reported to have anti-fibrotic activity and inhibit signaling pathways that are activated during EMT. This study investigated the nephroprotective role of chrysin in the prevention of CsA-induced renal fibrosis and elucidated a mechanism of inhibition against CsA-induced EMT in proximal tubule cells. Treatment with chrysin prevented CsA-induced renal dysfunction in Sprague Dawley rats measured by blood urea nitrogen (BUN), serum creatinine and creatinine clearance. Chrysin inhibited CsA-induced tubulointerstitial fibrosis, characterized by reduced tubular damage and collagen deposition. In vitro, chrysin significantly inhibited EMT in LLC-PK1 cells, evidenced by inhibition of cell migration, decreased collagen expression, reduced presence of mesenchymal markers and elevated epithelial junction proteins. Furthermore, chrysin co-treatment diminished CsA-induced TGF-β1 signaling pathways, decreasing Smad 3 phosphorylation which lead to a subsequent reduction in Snail expression. Chrysin also inhibited activation of the Akt/ GSK-3β pathway. Inhibition of both pathways diminished the cytosolic accumulation of β-catenin, a known trigger for EMT. In conclusion, flavonoids such as chrysin offer protection against CsA-induced renal dysfunction and interstitial fibrosis. Chrysin was shown to inhibit CsA-induced TGF-β1-dependent EMT in proximal tubule cells by modulation of Smad-dependent and independent signaling pathways.


2018 ◽  
Vol 33 (suppl_1) ◽  
pp. i58-i59
Author(s):  
Kyeong Min Kim ◽  
Ji Hyun Jeon ◽  
So Mi Kim ◽  
Eun Kyoung Lee ◽  
Jong Tae Cho ◽  
...  

Life ◽  
2020 ◽  
Vol 10 (12) ◽  
pp. 354
Author(s):  
Eun-Yeung Gong ◽  
Hyung Ah Jo ◽  
Sang Hyun Park ◽  
Dae Ryong Cha ◽  
Dae Young Hur ◽  
...  

High glucose-mediated tubular injury contributes to the development and progression of diabetic nephropathy through renal tubulointerstitial fibrosis. V-set immunoglobulin-domain-containing 4 (VSIG4), a B7 family-related protein, is a complement receptor. Although the role of epithelial–mesenchymal transition (EMT) has been reported in several diseases, little is known about its relationship with VSIG4 under diabetic conditions. This study aimed to investigate the role of VSIG4 in human tubule cells stimulated by high glucose (HG, 55 mM). HG upregulated both mRNA and protein levels of VSIG4 in proximal tubule cells (HK-2 cells) and Madin Darby Canine Kidney cells. These upregulations were accompanied by increased expression of mesenchymal markers such as fibronectin, N-cadherin, matrix metalloproteinase 9, and vimentin, and by decreased expression of the epithelial marker, E-cadherin. The siRNA-mediated inhibition of VSIG4 in HK-2 cells restored the dysregulation of EMT in cells. Interestingly, VSIG4 inhibition did not affect the expression of transforming growth factor (TGF)-β, whereas inhibition of TGF-β reduced VSIG4 expression, subsequently suppressing fibrosis markers. These findings suggest that VSIG4 plays an important role in mediating renal tubular EMT through the downstream action of HG-induced TGF-β activation.


2020 ◽  
Author(s):  
Bo Tang ◽  
Weiliang Li ◽  
Tingting Ji ◽  
Xiaoying Li ◽  
Xiaolei Qu ◽  
...  

Abstract Background Diabetic nephropathy (DN) is the leading cause of end-stage renal disease. Although dysfunction of renal tubule, also exhibited as epithelial-mesenchymal transition (EMT) and fibrosis, is closely associated with DN, the mechanism underlying renal tubule dysfunction still remains obscure. Methods Here, we identify that miR-199b-3p protect renal tubule from diabetic- induced injury by repressing KDM6A, a histone lysine demethylase reinforcing diabetic renal tubule dysfunction through regulating E-cadherin expression. We investigated the relationship between KDM6A, E-cadherin and miR-199b-3p with a series of gain- and loss- function assay in different cell models and animal models. The expression of KDM6A, E-cadherin and miR-199b-3p was tested by qPCR, western bolt or IHC. The EMT was measured by wound healing assay. The dysfunction of renal tubule were observed through HE and PAS stain and the kidney functions were monitored through several pathological signs detection assay, such as albumin to creatinine ratio (ACR), blood urea nitrogen (BUN), creatinine (Cr), proteinuria. Results Lower expression of E-cadherin was related to a higher level of KDM6A, while E-cadherin was increased with KDM6A-inhibitor GSK-J4 treatment both in HG-induced HK-2 cells and STZ-induced kidney. However, the variable expression of E-cadherin caused by overexpression or silence RNA was failed to alter the KDM6A expression. The target prediction and dual-luciferase assay results showed miR-199b-3p is a new miRNA targeting KDM6A. Overexpression of miR-199b-3p increased E-cadherin expression and prevented EMT through repressing KDM6A expression in HG-induced HK-2 cells, whereas miR-199b-3p knockdown by inhibitor displayed opposite results with lower E-cadherin and worse EMT accompanying with higher level of KDM6A. In addition, the miR-199b-3p knockout mice exhibited more dysfunctional renal tubule and more serious damage in kidney tissue treated with STZ. Conclusions These results demonstrate that miR-199b-3p improve E-cadherin expression and prevent the progression of DN through targeting KDM6A. MiR-199b-3p could be a potential biomarker or target for the diagnosis and treatment of diabetic nephropathy in the future.


2011 ◽  
Vol 300 (6) ◽  
pp. C1415-C1421 ◽  
Author(s):  
Xia Guo ◽  
Pedro A. Jose ◽  
Shi-You Chen

Previous studies demonstrate that response gene to complement 32 (RGC-32) mediates transforming growth factor-β1-induced epithelial-mesenchymal transition (EMT) of human renal proximal tubular cells. However, the mechanisms underlying RGC-32 function remain largely unknown. In the present study, we found that RGC-32 function in EMT is associated with Smad3. Coexpression of RGC-32 and Smad3, but not Smad2, induces a higher mesenchymal marker α-smooth muscle actin (α-SMA) protein expression as compared with RGC-32 or Smad3 alone, while knockdown of Smad3 using short hairpin interfering RNA blocks RGC-32-induced α-SMA expression. These data suggest that RGC-32 interacts with Smad3, but not Smad2, in the regulation of EMT. In addition to α-SMA, RGC-32 and Smad3 also synergistically activate the expression of extracellular matrix protein fibronectin and downregulate the epithelial marker E-cadherin. RGC-32 colocalizes with Smad3 in the nuclei of renal proximal tubular cells. Coimmunoprecipitation assays showed that Smad3, but not Smad2, physically interacts with RGC-32 in renal proximal tubular cells. Mechanistically, RGC-32 and Smad3 coordinate the induction of EMT by regulating the EMT regulators Slug and Snail. Taken together, our data demonstrate for the first time that RGC-32 interacts with Smad3 to mediate the EMT of human renal proximal tubular cells.


Sign in / Sign up

Export Citation Format

Share Document