Cargando…

Exosomal transfer of microRNA-590-3p between renal tubular epithelial cells after renal ischemia-reperfusion injury regulates autophagy by targeting TRAF6

BACKGROUND: Acute kidney injury (AKI) is a common complication in patients, especially elderly patients, who undergo cardiac surgery with cardiopulmonary bypass. Studies have indicated a protective role of autophagy in AKI. However, the mechanisms underlying the regulatory effect of autophagy in AKI...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Yimeng, Zhang, Congya, Du, Yingjie, Yang, Xiying, Liu, Min, Yang, Wenjing, Lei, Guiyu, Wang, Guyan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Lippincott Williams & Wilkins 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9945297/
https://www.ncbi.nlm.nih.gov/pubmed/36449688
http://dx.doi.org/10.1097/CM9.0000000000002377
_version_ 1784892111217229824
author Chen, Yimeng
Zhang, Congya
Du, Yingjie
Yang, Xiying
Liu, Min
Yang, Wenjing
Lei, Guiyu
Wang, Guyan
author_facet Chen, Yimeng
Zhang, Congya
Du, Yingjie
Yang, Xiying
Liu, Min
Yang, Wenjing
Lei, Guiyu
Wang, Guyan
author_sort Chen, Yimeng
collection PubMed
description BACKGROUND: Acute kidney injury (AKI) is a common complication in patients, especially elderly patients, who undergo cardiac surgery with cardiopulmonary bypass. Studies have indicated a protective role of autophagy in AKI. However, the mechanisms underlying the regulatory effect of autophagy in AKI among patients undergoing cardiac surgeries are poorly understood. In this study, we aimed to test the hypothesis that exosomal microRNAs (miRNAs) regulate autophagy in tubular epithelial cells after AKI. METHODS: Plasma exosomal RNA was extracted from young and elderly AKI patients undergoing cardiac surgery, and the miRNAs expression during the perioperative period were analyzed using next-generation sequencing. The screened miRNAs and their target genes were subjected to gene oncology function and Kyoto Encyclopedia of Genes and Genome enrichment analyses. Renal tubular epithelial cell line (HK-2 cells) was cultured and hypoxia/reoxygenation (H/R) model was established, which is an in vitro renal ischemia/reperfusion (I/R) model. We used Western blot analysis, cell viability assay, transfection, luciferase assay to investigate the mechanisms underlying the observed increases in the levels of renal I/R injury-mediated exosomal miRNAs and their roles in regulating HK-2 cells autophagy. RESULTS: miR-590-3p was highly enriched in the plasma exosomes of young AKI patients after cardiac surgery. Increased levels of miR-590-3p led to the increases in the expression of autophagy marker proteins, including Beclin-1 and microtubule associated protein 1 light chain 3 beta (LC3II), and prolonged the autophagic response in HK-2 cells after H/R treatment. These effects were achieved mainly via increases in the exosomal miR-590-3p levels, and the tumor necrosis factor receptor-associated factor 6 protein was shown to play a key role in I/R injury-mediated autophagy induction. CONCLUSION: Exosomes released from HK-2 cells after renal I/R injury regulate autophagy by transferring miR-590-3p in a paracrine manner, which suggests that increasing the miR-590-3p levels in HK-2 cell-derived exosomes may increase autophagy and protect against kidney injury after renal I/R injury.
format Online
Article
Text
id pubmed-9945297
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Lippincott Williams & Wilkins
record_format MEDLINE/PubMed
spelling pubmed-99452972023-02-23 Exosomal transfer of microRNA-590-3p between renal tubular epithelial cells after renal ischemia-reperfusion injury regulates autophagy by targeting TRAF6 Chen, Yimeng Zhang, Congya Du, Yingjie Yang, Xiying Liu, Min Yang, Wenjing Lei, Guiyu Wang, Guyan Chin Med J (Engl) Original Articles BACKGROUND: Acute kidney injury (AKI) is a common complication in patients, especially elderly patients, who undergo cardiac surgery with cardiopulmonary bypass. Studies have indicated a protective role of autophagy in AKI. However, the mechanisms underlying the regulatory effect of autophagy in AKI among patients undergoing cardiac surgeries are poorly understood. In this study, we aimed to test the hypothesis that exosomal microRNAs (miRNAs) regulate autophagy in tubular epithelial cells after AKI. METHODS: Plasma exosomal RNA was extracted from young and elderly AKI patients undergoing cardiac surgery, and the miRNAs expression during the perioperative period were analyzed using next-generation sequencing. The screened miRNAs and their target genes were subjected to gene oncology function and Kyoto Encyclopedia of Genes and Genome enrichment analyses. Renal tubular epithelial cell line (HK-2 cells) was cultured and hypoxia/reoxygenation (H/R) model was established, which is an in vitro renal ischemia/reperfusion (I/R) model. We used Western blot analysis, cell viability assay, transfection, luciferase assay to investigate the mechanisms underlying the observed increases in the levels of renal I/R injury-mediated exosomal miRNAs and their roles in regulating HK-2 cells autophagy. RESULTS: miR-590-3p was highly enriched in the plasma exosomes of young AKI patients after cardiac surgery. Increased levels of miR-590-3p led to the increases in the expression of autophagy marker proteins, including Beclin-1 and microtubule associated protein 1 light chain 3 beta (LC3II), and prolonged the autophagic response in HK-2 cells after H/R treatment. These effects were achieved mainly via increases in the exosomal miR-590-3p levels, and the tumor necrosis factor receptor-associated factor 6 protein was shown to play a key role in I/R injury-mediated autophagy induction. CONCLUSION: Exosomes released from HK-2 cells after renal I/R injury regulate autophagy by transferring miR-590-3p in a paracrine manner, which suggests that increasing the miR-590-3p levels in HK-2 cell-derived exosomes may increase autophagy and protect against kidney injury after renal I/R injury. Lippincott Williams & Wilkins 2022-10-20 2022-11-30 /pmc/articles/PMC9945297/ /pubmed/36449688 http://dx.doi.org/10.1097/CM9.0000000000002377 Text en Copyright © 2022 The Chinese Medical Association, produced by Wolters Kluwer, Inc. under the CC-BY-NC-ND license. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND), where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially without permission from the journal. http://creativecommons.org/licenses/by-nc-nd/4.0 (https://creativecommons.org/licenses/by-nc-nd/4.0/)
spellingShingle Original Articles
Chen, Yimeng
Zhang, Congya
Du, Yingjie
Yang, Xiying
Liu, Min
Yang, Wenjing
Lei, Guiyu
Wang, Guyan
Exosomal transfer of microRNA-590-3p between renal tubular epithelial cells after renal ischemia-reperfusion injury regulates autophagy by targeting TRAF6
title Exosomal transfer of microRNA-590-3p between renal tubular epithelial cells after renal ischemia-reperfusion injury regulates autophagy by targeting TRAF6
title_full Exosomal transfer of microRNA-590-3p between renal tubular epithelial cells after renal ischemia-reperfusion injury regulates autophagy by targeting TRAF6
title_fullStr Exosomal transfer of microRNA-590-3p between renal tubular epithelial cells after renal ischemia-reperfusion injury regulates autophagy by targeting TRAF6
title_full_unstemmed Exosomal transfer of microRNA-590-3p between renal tubular epithelial cells after renal ischemia-reperfusion injury regulates autophagy by targeting TRAF6
title_short Exosomal transfer of microRNA-590-3p between renal tubular epithelial cells after renal ischemia-reperfusion injury regulates autophagy by targeting TRAF6
title_sort exosomal transfer of microrna-590-3p between renal tubular epithelial cells after renal ischemia-reperfusion injury regulates autophagy by targeting traf6
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9945297/
https://www.ncbi.nlm.nih.gov/pubmed/36449688
http://dx.doi.org/10.1097/CM9.0000000000002377
work_keys_str_mv AT chenyimeng exosomaltransferofmicrorna5903pbetweenrenaltubularepithelialcellsafterrenalischemiareperfusioninjuryregulatesautophagybytargetingtraf6
AT zhangcongya exosomaltransferofmicrorna5903pbetweenrenaltubularepithelialcellsafterrenalischemiareperfusioninjuryregulatesautophagybytargetingtraf6
AT duyingjie exosomaltransferofmicrorna5903pbetweenrenaltubularepithelialcellsafterrenalischemiareperfusioninjuryregulatesautophagybytargetingtraf6
AT yangxiying exosomaltransferofmicrorna5903pbetweenrenaltubularepithelialcellsafterrenalischemiareperfusioninjuryregulatesautophagybytargetingtraf6
AT liumin exosomaltransferofmicrorna5903pbetweenrenaltubularepithelialcellsafterrenalischemiareperfusioninjuryregulatesautophagybytargetingtraf6
AT yangwenjing exosomaltransferofmicrorna5903pbetweenrenaltubularepithelialcellsafterrenalischemiareperfusioninjuryregulatesautophagybytargetingtraf6
AT leiguiyu exosomaltransferofmicrorna5903pbetweenrenaltubularepithelialcellsafterrenalischemiareperfusioninjuryregulatesautophagybytargetingtraf6
AT wangguyan exosomaltransferofmicrorna5903pbetweenrenaltubularepithelialcellsafterrenalischemiareperfusioninjuryregulatesautophagybytargetingtraf6