Cargando…

LncRNA IRAR regulates chemokines production in tubular epithelial cells thus promoting kidney ischemia-reperfusion injury

Increasing evidence demonstrates that long noncoding RNAs (lncRNAs) play an important role in several pathogenic processes of the kidney. However, functions of lncRNAs in ischemic acute kidney injury (AKI) remain undefined. In this study, global lncRNA profiling indicated that many lncRNA transcript...

Descripción completa

Detalles Bibliográficos
Autores principales: Jia, Ping, Xu, Sujuan, Ren, Ting, Pan, Tianyi, Wang, Xiaoyan, Zhang, Yunlu, Zou, Zhouping, Guo, Man, Zeng, Qi, Shen, Bo, Ding, Xiaoqiang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9217935/
https://www.ncbi.nlm.nih.gov/pubmed/35732633
http://dx.doi.org/10.1038/s41419-022-05018-x
_version_ 1784731768738283520
author Jia, Ping
Xu, Sujuan
Ren, Ting
Pan, Tianyi
Wang, Xiaoyan
Zhang, Yunlu
Zou, Zhouping
Guo, Man
Zeng, Qi
Shen, Bo
Ding, Xiaoqiang
author_facet Jia, Ping
Xu, Sujuan
Ren, Ting
Pan, Tianyi
Wang, Xiaoyan
Zhang, Yunlu
Zou, Zhouping
Guo, Man
Zeng, Qi
Shen, Bo
Ding, Xiaoqiang
author_sort Jia, Ping
collection PubMed
description Increasing evidence demonstrates that long noncoding RNAs (lncRNAs) play an important role in several pathogenic processes of the kidney. However, functions of lncRNAs in ischemic acute kidney injury (AKI) remain undefined. In this study, global lncRNA profiling indicated that many lncRNA transcripts were deregulated in kidney after ischemia reperfusion (IR). Among them, we identified IRAR (ischemia-reperfusion injury associated RNA) as a potential lncRNA candidate, which was mostly expressed by the tubular epithelial cells (TECs) after IR, involved in the development of AKI. GapmeR-mediated silencing and viral-based overexpression of IRAR were carried out to assess its function and contribution to IR-induced AKI. The results revealed that in vivo silencing of IRAR significantly reduced IR-induced proinflammatory cells infiltration and AKI. IRAR overexpression induced chemokine CCL2, CXCL1 and CXCL2 expression both in mRNA and protein levels in TECs, while, silencing of IRAR resulted in downregulation of these chemokines. RNA immunoprecipitation and RNA pulldown assay validated the association between IRAR and CCL2, CXCL1/2. Further examination revealed that specific ablation of CCL2 in TECs reduced macrophages infiltration and proinflammatory cytokine production, attenuated renal dysfunction in IR mice. Inhibition of CXC chemokine receptor 2 (receptor of CXCL1/2) reduced neutrofils infiltration, but had no overt effect on kidney function. To explore the mechanism of IRAR upregulation in kidney during IR, we analyzed promoter region of IRAR and predicted a potential binding site for transcription factor C/EBP β on IRAR promoter. Silencing of C/EBP β reduced IRAR expression in TECs. A dual-luciferase reporter assay and chromatin immunoprecipitation (ChIP) confirmed that IRAR was a transcriptional target of the C/EBP β. Altogether, our findings identify IRAR as a new player in the development of ischemic AKI through regulating chemokine production and immune cells infiltration, suggesting that IRAR is a potential target for prevention and/or attenuation of AKI.
format Online
Article
Text
id pubmed-9217935
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-92179352022-06-24 LncRNA IRAR regulates chemokines production in tubular epithelial cells thus promoting kidney ischemia-reperfusion injury Jia, Ping Xu, Sujuan Ren, Ting Pan, Tianyi Wang, Xiaoyan Zhang, Yunlu Zou, Zhouping Guo, Man Zeng, Qi Shen, Bo Ding, Xiaoqiang Cell Death Dis Article Increasing evidence demonstrates that long noncoding RNAs (lncRNAs) play an important role in several pathogenic processes of the kidney. However, functions of lncRNAs in ischemic acute kidney injury (AKI) remain undefined. In this study, global lncRNA profiling indicated that many lncRNA transcripts were deregulated in kidney after ischemia reperfusion (IR). Among them, we identified IRAR (ischemia-reperfusion injury associated RNA) as a potential lncRNA candidate, which was mostly expressed by the tubular epithelial cells (TECs) after IR, involved in the development of AKI. GapmeR-mediated silencing and viral-based overexpression of IRAR were carried out to assess its function and contribution to IR-induced AKI. The results revealed that in vivo silencing of IRAR significantly reduced IR-induced proinflammatory cells infiltration and AKI. IRAR overexpression induced chemokine CCL2, CXCL1 and CXCL2 expression both in mRNA and protein levels in TECs, while, silencing of IRAR resulted in downregulation of these chemokines. RNA immunoprecipitation and RNA pulldown assay validated the association between IRAR and CCL2, CXCL1/2. Further examination revealed that specific ablation of CCL2 in TECs reduced macrophages infiltration and proinflammatory cytokine production, attenuated renal dysfunction in IR mice. Inhibition of CXC chemokine receptor 2 (receptor of CXCL1/2) reduced neutrofils infiltration, but had no overt effect on kidney function. To explore the mechanism of IRAR upregulation in kidney during IR, we analyzed promoter region of IRAR and predicted a potential binding site for transcription factor C/EBP β on IRAR promoter. Silencing of C/EBP β reduced IRAR expression in TECs. A dual-luciferase reporter assay and chromatin immunoprecipitation (ChIP) confirmed that IRAR was a transcriptional target of the C/EBP β. Altogether, our findings identify IRAR as a new player in the development of ischemic AKI through regulating chemokine production and immune cells infiltration, suggesting that IRAR is a potential target for prevention and/or attenuation of AKI. Nature Publishing Group UK 2022-06-22 /pmc/articles/PMC9217935/ /pubmed/35732633 http://dx.doi.org/10.1038/s41419-022-05018-x Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Jia, Ping
Xu, Sujuan
Ren, Ting
Pan, Tianyi
Wang, Xiaoyan
Zhang, Yunlu
Zou, Zhouping
Guo, Man
Zeng, Qi
Shen, Bo
Ding, Xiaoqiang
LncRNA IRAR regulates chemokines production in tubular epithelial cells thus promoting kidney ischemia-reperfusion injury
title LncRNA IRAR regulates chemokines production in tubular epithelial cells thus promoting kidney ischemia-reperfusion injury
title_full LncRNA IRAR regulates chemokines production in tubular epithelial cells thus promoting kidney ischemia-reperfusion injury
title_fullStr LncRNA IRAR regulates chemokines production in tubular epithelial cells thus promoting kidney ischemia-reperfusion injury
title_full_unstemmed LncRNA IRAR regulates chemokines production in tubular epithelial cells thus promoting kidney ischemia-reperfusion injury
title_short LncRNA IRAR regulates chemokines production in tubular epithelial cells thus promoting kidney ischemia-reperfusion injury
title_sort lncrna irar regulates chemokines production in tubular epithelial cells thus promoting kidney ischemia-reperfusion injury
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9217935/
https://www.ncbi.nlm.nih.gov/pubmed/35732633
http://dx.doi.org/10.1038/s41419-022-05018-x
work_keys_str_mv AT jiaping lncrnairarregulateschemokinesproductionintubularepithelialcellsthuspromotingkidneyischemiareperfusioninjury
AT xusujuan lncrnairarregulateschemokinesproductionintubularepithelialcellsthuspromotingkidneyischemiareperfusioninjury
AT renting lncrnairarregulateschemokinesproductionintubularepithelialcellsthuspromotingkidneyischemiareperfusioninjury
AT pantianyi lncrnairarregulateschemokinesproductionintubularepithelialcellsthuspromotingkidneyischemiareperfusioninjury
AT wangxiaoyan lncrnairarregulateschemokinesproductionintubularepithelialcellsthuspromotingkidneyischemiareperfusioninjury
AT zhangyunlu lncrnairarregulateschemokinesproductionintubularepithelialcellsthuspromotingkidneyischemiareperfusioninjury
AT zouzhouping lncrnairarregulateschemokinesproductionintubularepithelialcellsthuspromotingkidneyischemiareperfusioninjury
AT guoman lncrnairarregulateschemokinesproductionintubularepithelialcellsthuspromotingkidneyischemiareperfusioninjury
AT zengqi lncrnairarregulateschemokinesproductionintubularepithelialcellsthuspromotingkidneyischemiareperfusioninjury
AT shenbo lncrnairarregulateschemokinesproductionintubularepithelialcellsthuspromotingkidneyischemiareperfusioninjury
AT dingxiaoqiang lncrnairarregulateschemokinesproductionintubularepithelialcellsthuspromotingkidneyischemiareperfusioninjury