Cargando…

Upregulation of allograft inflammatory factor-1 expression and secretion by macrophages stimulated with aldosterone promotes renal fibroblasts to a profibrotic phenotype

Macrophages have been identified as a key cell type in the pathogenesis of renal interstitial fibrosis (RIF). However, the mechanism through which macrophages drive fibrosis remains unclear. The current study focuses on the effects and possible underlying mechanism of allograft inflammatory factor-1...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Yushu, Wang, Xingzhi, Zhang, Lei, Yuan, Xueying, Hao, Jianbing, Ni, Jie, Hao, Lirong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6034929/
https://www.ncbi.nlm.nih.gov/pubmed/29749461
http://dx.doi.org/10.3892/ijmm.2018.3667
_version_ 1783337966918172672
author Li, Yushu
Wang, Xingzhi
Zhang, Lei
Yuan, Xueying
Hao, Jianbing
Ni, Jie
Hao, Lirong
author_facet Li, Yushu
Wang, Xingzhi
Zhang, Lei
Yuan, Xueying
Hao, Jianbing
Ni, Jie
Hao, Lirong
author_sort Li, Yushu
collection PubMed
description Macrophages have been identified as a key cell type in the pathogenesis of renal interstitial fibrosis (RIF). However, the mechanism through which macrophages drive fibrosis remains unclear. The current study focuses on the effects and possible underlying mechanism of allograft inflammatory factor-1 (AIF-1), an inflammation-responsive scaffold protein expressed and secreted by macrophages, in promoting fibroblasts to a profibrotic phenotype. In vivo experiments indicated that AIF-1, CD68 and α-smooth muscle actin (α-SMA) were upregulated in kidney tissues of mice subjected to unilateral ureteric obstruction, while their expressions were inhibited by an aldosterone receptor antagonist, spironolactone. Double immunofluorescence staining revealed that AIF-1 expression co-localized with CD68-positive macrophages in the renal interstitium, indicating that AIF-1 expression in macrophages was increased in the RIF animal model. Furthermore, to identify the role of AIF-1 in promoting fibrosis, its expression and secretion by the RAW264.7 macrophage cell line were detected in vitro. The expression levels of α-SMA, phosphorylated p38 (p-p38) and fibronectin (FN) in fibroblasts were examined subsequent to co-culture with macrophages. The increase in AIF-1 expression and secretion was confirmed in RAW264.7 cells in response to aldosterone. After 72 h of co-culture between fibroblasts and macrophages stimulated with aldosterone, the α-SMA expression was induced in fibroblasts, with significantly increased expression levels of FN and p-p38 observed. In addition, AIF-1 expression was reduced by stable transfection of RAW264.7 cells with AIF-1 small interfering RNA, resulting in significantly reduced expression levels of α-SMA, p-p38 and FN in fibroblasts co-cultured with macrophages as compared with normal macrophages. These findings indicate that the expression of AIF-1 in macrophages is critical for the activation of renal fibroblasts to a profibrotic phenotype. AIF-1 expression was upregulated in macrophages, and may be a novel mechanism linking macrophages to the promotion of RIF via the p38 signaling pathway.
format Online
Article
Text
id pubmed-6034929
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-60349292018-07-09 Upregulation of allograft inflammatory factor-1 expression and secretion by macrophages stimulated with aldosterone promotes renal fibroblasts to a profibrotic phenotype Li, Yushu Wang, Xingzhi Zhang, Lei Yuan, Xueying Hao, Jianbing Ni, Jie Hao, Lirong Int J Mol Med Articles Macrophages have been identified as a key cell type in the pathogenesis of renal interstitial fibrosis (RIF). However, the mechanism through which macrophages drive fibrosis remains unclear. The current study focuses on the effects and possible underlying mechanism of allograft inflammatory factor-1 (AIF-1), an inflammation-responsive scaffold protein expressed and secreted by macrophages, in promoting fibroblasts to a profibrotic phenotype. In vivo experiments indicated that AIF-1, CD68 and α-smooth muscle actin (α-SMA) were upregulated in kidney tissues of mice subjected to unilateral ureteric obstruction, while their expressions were inhibited by an aldosterone receptor antagonist, spironolactone. Double immunofluorescence staining revealed that AIF-1 expression co-localized with CD68-positive macrophages in the renal interstitium, indicating that AIF-1 expression in macrophages was increased in the RIF animal model. Furthermore, to identify the role of AIF-1 in promoting fibrosis, its expression and secretion by the RAW264.7 macrophage cell line were detected in vitro. The expression levels of α-SMA, phosphorylated p38 (p-p38) and fibronectin (FN) in fibroblasts were examined subsequent to co-culture with macrophages. The increase in AIF-1 expression and secretion was confirmed in RAW264.7 cells in response to aldosterone. After 72 h of co-culture between fibroblasts and macrophages stimulated with aldosterone, the α-SMA expression was induced in fibroblasts, with significantly increased expression levels of FN and p-p38 observed. In addition, AIF-1 expression was reduced by stable transfection of RAW264.7 cells with AIF-1 small interfering RNA, resulting in significantly reduced expression levels of α-SMA, p-p38 and FN in fibroblasts co-cultured with macrophages as compared with normal macrophages. These findings indicate that the expression of AIF-1 in macrophages is critical for the activation of renal fibroblasts to a profibrotic phenotype. AIF-1 expression was upregulated in macrophages, and may be a novel mechanism linking macrophages to the promotion of RIF via the p38 signaling pathway. D.A. Spandidos 2018-08 2018-05-10 /pmc/articles/PMC6034929/ /pubmed/29749461 http://dx.doi.org/10.3892/ijmm.2018.3667 Text en Copyright: © Li et al. 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
Li, Yushu
Wang, Xingzhi
Zhang, Lei
Yuan, Xueying
Hao, Jianbing
Ni, Jie
Hao, Lirong
Upregulation of allograft inflammatory factor-1 expression and secretion by macrophages stimulated with aldosterone promotes renal fibroblasts to a profibrotic phenotype
title Upregulation of allograft inflammatory factor-1 expression and secretion by macrophages stimulated with aldosterone promotes renal fibroblasts to a profibrotic phenotype
title_full Upregulation of allograft inflammatory factor-1 expression and secretion by macrophages stimulated with aldosterone promotes renal fibroblasts to a profibrotic phenotype
title_fullStr Upregulation of allograft inflammatory factor-1 expression and secretion by macrophages stimulated with aldosterone promotes renal fibroblasts to a profibrotic phenotype
title_full_unstemmed Upregulation of allograft inflammatory factor-1 expression and secretion by macrophages stimulated with aldosterone promotes renal fibroblasts to a profibrotic phenotype
title_short Upregulation of allograft inflammatory factor-1 expression and secretion by macrophages stimulated with aldosterone promotes renal fibroblasts to a profibrotic phenotype
title_sort upregulation of allograft inflammatory factor-1 expression and secretion by macrophages stimulated with aldosterone promotes renal fibroblasts to a profibrotic phenotype
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6034929/
https://www.ncbi.nlm.nih.gov/pubmed/29749461
http://dx.doi.org/10.3892/ijmm.2018.3667
work_keys_str_mv AT liyushu upregulationofallograftinflammatoryfactor1expressionandsecretionbymacrophagesstimulatedwithaldosteronepromotesrenalfibroblaststoaprofibroticphenotype
AT wangxingzhi upregulationofallograftinflammatoryfactor1expressionandsecretionbymacrophagesstimulatedwithaldosteronepromotesrenalfibroblaststoaprofibroticphenotype
AT zhanglei upregulationofallograftinflammatoryfactor1expressionandsecretionbymacrophagesstimulatedwithaldosteronepromotesrenalfibroblaststoaprofibroticphenotype
AT yuanxueying upregulationofallograftinflammatoryfactor1expressionandsecretionbymacrophagesstimulatedwithaldosteronepromotesrenalfibroblaststoaprofibroticphenotype
AT haojianbing upregulationofallograftinflammatoryfactor1expressionandsecretionbymacrophagesstimulatedwithaldosteronepromotesrenalfibroblaststoaprofibroticphenotype
AT nijie upregulationofallograftinflammatoryfactor1expressionandsecretionbymacrophagesstimulatedwithaldosteronepromotesrenalfibroblaststoaprofibroticphenotype
AT haolirong upregulationofallograftinflammatoryfactor1expressionandsecretionbymacrophagesstimulatedwithaldosteronepromotesrenalfibroblaststoaprofibroticphenotype