Cargando…

Novel aspect of neprilysin in kidney fibrosis via ACSL4‐mediated ferroptosis of tubular epithelial cells

Although inhibition of neprilysin (NEP) might be a therapeutic strategy with the potential to improve the outcome of chronic kidney disease (CKD), the versatile function of NEP with its mechanism remains obscure in kidney fibrosis. In the study, we found that NEP was abnormally increased in tubular...

Descripción completa

Detalles Bibliográficos
Autores principales: Lai, Weijing, Huang, Rongshuang, Wang, Bo, Shi, Min, Guo, Fan, Li, Lingzhi, Ren, Qian, Tao, Sibei, Fu, Ping, Ma, Liang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10349188/
https://www.ncbi.nlm.nih.gov/pubmed/37457659
http://dx.doi.org/10.1002/mco2.330
_version_ 1785073847373922304
author Lai, Weijing
Huang, Rongshuang
Wang, Bo
Shi, Min
Guo, Fan
Li, Lingzhi
Ren, Qian
Tao, Sibei
Fu, Ping
Ma, Liang
author_facet Lai, Weijing
Huang, Rongshuang
Wang, Bo
Shi, Min
Guo, Fan
Li, Lingzhi
Ren, Qian
Tao, Sibei
Fu, Ping
Ma, Liang
author_sort Lai, Weijing
collection PubMed
description Although inhibition of neprilysin (NEP) might be a therapeutic strategy with the potential to improve the outcome of chronic kidney disease (CKD), the versatile function of NEP with its mechanism remains obscure in kidney fibrosis. In the study, we found that NEP was abnormally increased in tubular epithelial cells of CKD patients, as well as unilateral ureteral obstruction and adenine diet‐induced mice. Treatment with a United States Food and Drug Administration‐approved NEP inhibitor Sacubitrilat (LBQ657) could alleviate ferroptosis, tubular injury, and delay the progression of kidney fibrosis in experimental mice. Similarly, genetic knockdown of NEP also inhibited tubular injury and fibrosis in transforming growth factor (TGF)‐β1 ‐induced tubular cells. Mechanically, NEP overexpression aggravated the ferroptotic and fibrotic phenotype, which was restored by acyl‐CoA synthetase long‐chain family member 4 (ACSL4) knockdown. The NEP silencing attenuated TGF‐β1‐induced tubular cell ferroptosis and was exacerbated by ACSL4 overexpression. Collectively, for the first time, a novel aspect of NEP was explored in kidney fibrosis through ACSL4‐mediated tubular epithelial cell ferroptosis. Our data further confirmed that NEP inhibition exerted a promising therapeutic against fibrotic kidney diseases.
format Online
Article
Text
id pubmed-10349188
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-103491882023-07-16 Novel aspect of neprilysin in kidney fibrosis via ACSL4‐mediated ferroptosis of tubular epithelial cells Lai, Weijing Huang, Rongshuang Wang, Bo Shi, Min Guo, Fan Li, Lingzhi Ren, Qian Tao, Sibei Fu, Ping Ma, Liang MedComm (2020) Original Articles Although inhibition of neprilysin (NEP) might be a therapeutic strategy with the potential to improve the outcome of chronic kidney disease (CKD), the versatile function of NEP with its mechanism remains obscure in kidney fibrosis. In the study, we found that NEP was abnormally increased in tubular epithelial cells of CKD patients, as well as unilateral ureteral obstruction and adenine diet‐induced mice. Treatment with a United States Food and Drug Administration‐approved NEP inhibitor Sacubitrilat (LBQ657) could alleviate ferroptosis, tubular injury, and delay the progression of kidney fibrosis in experimental mice. Similarly, genetic knockdown of NEP also inhibited tubular injury and fibrosis in transforming growth factor (TGF)‐β1 ‐induced tubular cells. Mechanically, NEP overexpression aggravated the ferroptotic and fibrotic phenotype, which was restored by acyl‐CoA synthetase long‐chain family member 4 (ACSL4) knockdown. The NEP silencing attenuated TGF‐β1‐induced tubular cell ferroptosis and was exacerbated by ACSL4 overexpression. Collectively, for the first time, a novel aspect of NEP was explored in kidney fibrosis through ACSL4‐mediated tubular epithelial cell ferroptosis. Our data further confirmed that NEP inhibition exerted a promising therapeutic against fibrotic kidney diseases. John Wiley and Sons Inc. 2023-07-14 /pmc/articles/PMC10349188/ /pubmed/37457659 http://dx.doi.org/10.1002/mco2.330 Text en © 2023 The Authors. MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd. https://creativecommons.org/licenses/by/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Articles
Lai, Weijing
Huang, Rongshuang
Wang, Bo
Shi, Min
Guo, Fan
Li, Lingzhi
Ren, Qian
Tao, Sibei
Fu, Ping
Ma, Liang
Novel aspect of neprilysin in kidney fibrosis via ACSL4‐mediated ferroptosis of tubular epithelial cells
title Novel aspect of neprilysin in kidney fibrosis via ACSL4‐mediated ferroptosis of tubular epithelial cells
title_full Novel aspect of neprilysin in kidney fibrosis via ACSL4‐mediated ferroptosis of tubular epithelial cells
title_fullStr Novel aspect of neprilysin in kidney fibrosis via ACSL4‐mediated ferroptosis of tubular epithelial cells
title_full_unstemmed Novel aspect of neprilysin in kidney fibrosis via ACSL4‐mediated ferroptosis of tubular epithelial cells
title_short Novel aspect of neprilysin in kidney fibrosis via ACSL4‐mediated ferroptosis of tubular epithelial cells
title_sort novel aspect of neprilysin in kidney fibrosis via acsl4‐mediated ferroptosis of tubular epithelial cells
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10349188/
https://www.ncbi.nlm.nih.gov/pubmed/37457659
http://dx.doi.org/10.1002/mco2.330
work_keys_str_mv AT laiweijing novelaspectofneprilysininkidneyfibrosisviaacsl4mediatedferroptosisoftubularepithelialcells
AT huangrongshuang novelaspectofneprilysininkidneyfibrosisviaacsl4mediatedferroptosisoftubularepithelialcells
AT wangbo novelaspectofneprilysininkidneyfibrosisviaacsl4mediatedferroptosisoftubularepithelialcells
AT shimin novelaspectofneprilysininkidneyfibrosisviaacsl4mediatedferroptosisoftubularepithelialcells
AT guofan novelaspectofneprilysininkidneyfibrosisviaacsl4mediatedferroptosisoftubularepithelialcells
AT lilingzhi novelaspectofneprilysininkidneyfibrosisviaacsl4mediatedferroptosisoftubularepithelialcells
AT renqian novelaspectofneprilysininkidneyfibrosisviaacsl4mediatedferroptosisoftubularepithelialcells
AT taosibei novelaspectofneprilysininkidneyfibrosisviaacsl4mediatedferroptosisoftubularepithelialcells
AT fuping novelaspectofneprilysininkidneyfibrosisviaacsl4mediatedferroptosisoftubularepithelialcells
AT maliang novelaspectofneprilysininkidneyfibrosisviaacsl4mediatedferroptosisoftubularepithelialcells