Cargando…

Upregulation of RIP3 promotes necroptosis via a ROS-dependent NF-κB pathway to induce chronic inflammation in HK-2 cells

Tubular atrophy/interstitial fibrosis (TA/IF) is a major cause of late allograft loss, and inflammation within areas of TA/IF is associated with adverse outcomes in kidney transplantation. However, there is currently no satisfactory method to suppress this inflammation to improve TA/IF. The present...

Descripción completa

Detalles Bibliográficos
Autores principales: Wei, Junjun, Chen, Liangliang, Wang, Duidui, Tang, Li, Xie, Zhenhua, Chen, Weifeng, Zhang, Shuwei, Weng, Guobin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8441977/
https://www.ncbi.nlm.nih.gov/pubmed/34498705
http://dx.doi.org/10.3892/mmr.2021.12423
_version_ 1783752932394532864
author Wei, Junjun
Chen, Liangliang
Wang, Duidui
Tang, Li
Xie, Zhenhua
Chen, Weifeng
Zhang, Shuwei
Weng, Guobin
author_facet Wei, Junjun
Chen, Liangliang
Wang, Duidui
Tang, Li
Xie, Zhenhua
Chen, Weifeng
Zhang, Shuwei
Weng, Guobin
author_sort Wei, Junjun
collection PubMed
description Tubular atrophy/interstitial fibrosis (TA/IF) is a major cause of late allograft loss, and inflammation within areas of TA/IF is associated with adverse outcomes in kidney transplantation. However, there is currently no satisfactory method to suppress this inflammation to improve TA/IF. The present study aimed to determine the proinflammatory role of receptor-interacting protein 3 (RIP3) in TA/IF to discover a novel therapeutic target. Reverse transcription-quantitative PCR and western blotting were performed to detect the expression of RIP3 and inflammation-associated factors. Lactate dehydrogenase release assay was used to determine necroptosis. Fluorescent 2,7-dichlorodihydrofluorescein diacetate was used to detect the levels of reactive oxygen species (ROS). The results demonstrated that patients with chronic TA/IF exhibited upregulated receptor-interacting protein 3 (RIP3) expression compared with the patients who had a favorable recovery after renal transplant. Therefore, the current study used normal renal tubular epithelial cells HK-2 to establish a cellular model with a high expression level of RIP3 in order to investigate the effect of RIP3 on renal epithelial cells after transplantation. The western blotting results demonstrated that overexpression of RIP3 could significantly increase the phosphorylation level of the necroptosis executive molecule mixed lineage kinase domain-like protein. Lactate dehydrogenase release, a key feature of necroptosis, was also markedly improved by RIP3 overexpression. Moreover, a higher inflammatory response was detected in HK-2 cells with RIP3 overexpression, and this elevated inflammation could be restored by the necroptosis inhibitor necrosulfonamide. Of note, it was found that overexpression of RIP3 activated the NF-κB signaling pathway via the excessive accumulation of ROS to induce necroptosis, which ultimately led to inflammation. Collectively, these findings indicated that overexpression of RIP3 promoted necroptosis via a ROS-dependent NF-κB pathway to induce chronic inflammation, suggesting that RIP3 may have the potential to be a therapeutic target against inflammation in TA/IF.
format Online
Article
Text
id pubmed-8441977
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-84419772021-09-17 Upregulation of RIP3 promotes necroptosis via a ROS-dependent NF-κB pathway to induce chronic inflammation in HK-2 cells Wei, Junjun Chen, Liangliang Wang, Duidui Tang, Li Xie, Zhenhua Chen, Weifeng Zhang, Shuwei Weng, Guobin Mol Med Rep Articles Tubular atrophy/interstitial fibrosis (TA/IF) is a major cause of late allograft loss, and inflammation within areas of TA/IF is associated with adverse outcomes in kidney transplantation. However, there is currently no satisfactory method to suppress this inflammation to improve TA/IF. The present study aimed to determine the proinflammatory role of receptor-interacting protein 3 (RIP3) in TA/IF to discover a novel therapeutic target. Reverse transcription-quantitative PCR and western blotting were performed to detect the expression of RIP3 and inflammation-associated factors. Lactate dehydrogenase release assay was used to determine necroptosis. Fluorescent 2,7-dichlorodihydrofluorescein diacetate was used to detect the levels of reactive oxygen species (ROS). The results demonstrated that patients with chronic TA/IF exhibited upregulated receptor-interacting protein 3 (RIP3) expression compared with the patients who had a favorable recovery after renal transplant. Therefore, the current study used normal renal tubular epithelial cells HK-2 to establish a cellular model with a high expression level of RIP3 in order to investigate the effect of RIP3 on renal epithelial cells after transplantation. The western blotting results demonstrated that overexpression of RIP3 could significantly increase the phosphorylation level of the necroptosis executive molecule mixed lineage kinase domain-like protein. Lactate dehydrogenase release, a key feature of necroptosis, was also markedly improved by RIP3 overexpression. Moreover, a higher inflammatory response was detected in HK-2 cells with RIP3 overexpression, and this elevated inflammation could be restored by the necroptosis inhibitor necrosulfonamide. Of note, it was found that overexpression of RIP3 activated the NF-κB signaling pathway via the excessive accumulation of ROS to induce necroptosis, which ultimately led to inflammation. Collectively, these findings indicated that overexpression of RIP3 promoted necroptosis via a ROS-dependent NF-κB pathway to induce chronic inflammation, suggesting that RIP3 may have the potential to be a therapeutic target against inflammation in TA/IF. D.A. Spandidos 2021-11 2021-09-07 /pmc/articles/PMC8441977/ /pubmed/34498705 http://dx.doi.org/10.3892/mmr.2021.12423 Text en Copyright: © Wei et al. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Wei, Junjun
Chen, Liangliang
Wang, Duidui
Tang, Li
Xie, Zhenhua
Chen, Weifeng
Zhang, Shuwei
Weng, Guobin
Upregulation of RIP3 promotes necroptosis via a ROS-dependent NF-κB pathway to induce chronic inflammation in HK-2 cells
title Upregulation of RIP3 promotes necroptosis via a ROS-dependent NF-κB pathway to induce chronic inflammation in HK-2 cells
title_full Upregulation of RIP3 promotes necroptosis via a ROS-dependent NF-κB pathway to induce chronic inflammation in HK-2 cells
title_fullStr Upregulation of RIP3 promotes necroptosis via a ROS-dependent NF-κB pathway to induce chronic inflammation in HK-2 cells
title_full_unstemmed Upregulation of RIP3 promotes necroptosis via a ROS-dependent NF-κB pathway to induce chronic inflammation in HK-2 cells
title_short Upregulation of RIP3 promotes necroptosis via a ROS-dependent NF-κB pathway to induce chronic inflammation in HK-2 cells
title_sort upregulation of rip3 promotes necroptosis via a ros-dependent nf-κb pathway to induce chronic inflammation in hk-2 cells
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8441977/
https://www.ncbi.nlm.nih.gov/pubmed/34498705
http://dx.doi.org/10.3892/mmr.2021.12423
work_keys_str_mv AT weijunjun upregulationofrip3promotesnecroptosisviaarosdependentnfkbpathwaytoinducechronicinflammationinhk2cells
AT chenliangliang upregulationofrip3promotesnecroptosisviaarosdependentnfkbpathwaytoinducechronicinflammationinhk2cells
AT wangduidui upregulationofrip3promotesnecroptosisviaarosdependentnfkbpathwaytoinducechronicinflammationinhk2cells
AT tangli upregulationofrip3promotesnecroptosisviaarosdependentnfkbpathwaytoinducechronicinflammationinhk2cells
AT xiezhenhua upregulationofrip3promotesnecroptosisviaarosdependentnfkbpathwaytoinducechronicinflammationinhk2cells
AT chenweifeng upregulationofrip3promotesnecroptosisviaarosdependentnfkbpathwaytoinducechronicinflammationinhk2cells
AT zhangshuwei upregulationofrip3promotesnecroptosisviaarosdependentnfkbpathwaytoinducechronicinflammationinhk2cells
AT wengguobin upregulationofrip3promotesnecroptosisviaarosdependentnfkbpathwaytoinducechronicinflammationinhk2cells