Cargando…

Kidney tubular epithelial cells control interstitial fibroblast fate by releasing TNFAIP8-encapsulated exosomes

Kidney fibrosis, characterized by the activation and expansion of the matrix-producing fibroblasts, is the common outcome of chronic kidney disease (CKD). While fibroblast proliferation is well studied in CKD, little is known about the regulation and mechanism of fibroblast depletion. Here, we show...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Xi, Liu, Zhao, Wang, Cong, Miao, Jinhua, Zhou, Shan, Ren, Qian, Jia, Nan, Zhou, Lili, Liu, Youhua
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10570316/
https://www.ncbi.nlm.nih.gov/pubmed/37828075
http://dx.doi.org/10.1038/s41419-023-06209-w
_version_ 1785119738081312768
author Liu, Xi
Liu, Zhao
Wang, Cong
Miao, Jinhua
Zhou, Shan
Ren, Qian
Jia, Nan
Zhou, Lili
Liu, Youhua
author_facet Liu, Xi
Liu, Zhao
Wang, Cong
Miao, Jinhua
Zhou, Shan
Ren, Qian
Jia, Nan
Zhou, Lili
Liu, Youhua
author_sort Liu, Xi
collection PubMed
description Kidney fibrosis, characterized by the activation and expansion of the matrix-producing fibroblasts, is the common outcome of chronic kidney disease (CKD). While fibroblast proliferation is well studied in CKD, little is known about the regulation and mechanism of fibroblast depletion. Here, we show that exosomes derived from stressed/injured tubules play a pivotal role in dictating fibroblast apoptosis and fate. When human kidney tubular cells (HK-2) were stimulated with TGF-β1, they produced and released increased amounts of exosomes (TGFβ-Exo), which prevented renal interstitial fibroblasts from apoptosis. In vivo, injections of TGFβ-Exo promoted renal fibroblast survival, whereas blockade of exosome secretion accelerated fibroblast apoptosis in obstructive nephropathy. Proteomics profiling identified the tumor necrosis factor-α-induced protein 8 (TNFAIP8) as a key component enriched in TGFβ-Exo. TNFAIP8 was induced in renal tubular epithelium and enriched in the exosomes from fibrotic kidneys. Knockdown of TNFAIP8 in tubular cells abolished the ability of TGFβ-Exo to prevent fibroblast apoptosis. In vivo, gain- or loss- of TNFAIP8 prevented or aggravated renal fibroblast apoptosis after obstructive injury. Mechanistically, exosomal-TNFAIP8 promoted p53 ubiquitination leading to its degradation, thereby inhibiting fibroblasts apoptosis and inducing their proliferation. Collectively, these results indicate that tubule-derived exosomes play a critical role in controlling the size of fibroblast population during renal fibrogenesis through shuttling TNFAIP8 to block p53 signaling. Strategies to target exosomes may be effective strategies for the therapy of fibrotic CKD.
format Online
Article
Text
id pubmed-10570316
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-105703162023-10-14 Kidney tubular epithelial cells control interstitial fibroblast fate by releasing TNFAIP8-encapsulated exosomes Liu, Xi Liu, Zhao Wang, Cong Miao, Jinhua Zhou, Shan Ren, Qian Jia, Nan Zhou, Lili Liu, Youhua Cell Death Dis Article Kidney fibrosis, characterized by the activation and expansion of the matrix-producing fibroblasts, is the common outcome of chronic kidney disease (CKD). While fibroblast proliferation is well studied in CKD, little is known about the regulation and mechanism of fibroblast depletion. Here, we show that exosomes derived from stressed/injured tubules play a pivotal role in dictating fibroblast apoptosis and fate. When human kidney tubular cells (HK-2) were stimulated with TGF-β1, they produced and released increased amounts of exosomes (TGFβ-Exo), which prevented renal interstitial fibroblasts from apoptosis. In vivo, injections of TGFβ-Exo promoted renal fibroblast survival, whereas blockade of exosome secretion accelerated fibroblast apoptosis in obstructive nephropathy. Proteomics profiling identified the tumor necrosis factor-α-induced protein 8 (TNFAIP8) as a key component enriched in TGFβ-Exo. TNFAIP8 was induced in renal tubular epithelium and enriched in the exosomes from fibrotic kidneys. Knockdown of TNFAIP8 in tubular cells abolished the ability of TGFβ-Exo to prevent fibroblast apoptosis. In vivo, gain- or loss- of TNFAIP8 prevented or aggravated renal fibroblast apoptosis after obstructive injury. Mechanistically, exosomal-TNFAIP8 promoted p53 ubiquitination leading to its degradation, thereby inhibiting fibroblasts apoptosis and inducing their proliferation. Collectively, these results indicate that tubule-derived exosomes play a critical role in controlling the size of fibroblast population during renal fibrogenesis through shuttling TNFAIP8 to block p53 signaling. Strategies to target exosomes may be effective strategies for the therapy of fibrotic CKD. Nature Publishing Group UK 2023-10-12 /pmc/articles/PMC10570316/ /pubmed/37828075 http://dx.doi.org/10.1038/s41419-023-06209-w Text en © The Author(s) 2023 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
Liu, Xi
Liu, Zhao
Wang, Cong
Miao, Jinhua
Zhou, Shan
Ren, Qian
Jia, Nan
Zhou, Lili
Liu, Youhua
Kidney tubular epithelial cells control interstitial fibroblast fate by releasing TNFAIP8-encapsulated exosomes
title Kidney tubular epithelial cells control interstitial fibroblast fate by releasing TNFAIP8-encapsulated exosomes
title_full Kidney tubular epithelial cells control interstitial fibroblast fate by releasing TNFAIP8-encapsulated exosomes
title_fullStr Kidney tubular epithelial cells control interstitial fibroblast fate by releasing TNFAIP8-encapsulated exosomes
title_full_unstemmed Kidney tubular epithelial cells control interstitial fibroblast fate by releasing TNFAIP8-encapsulated exosomes
title_short Kidney tubular epithelial cells control interstitial fibroblast fate by releasing TNFAIP8-encapsulated exosomes
title_sort kidney tubular epithelial cells control interstitial fibroblast fate by releasing tnfaip8-encapsulated exosomes
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10570316/
https://www.ncbi.nlm.nih.gov/pubmed/37828075
http://dx.doi.org/10.1038/s41419-023-06209-w
work_keys_str_mv AT liuxi kidneytubularepithelialcellscontrolinterstitialfibroblastfatebyreleasingtnfaip8encapsulatedexosomes
AT liuzhao kidneytubularepithelialcellscontrolinterstitialfibroblastfatebyreleasingtnfaip8encapsulatedexosomes
AT wangcong kidneytubularepithelialcellscontrolinterstitialfibroblastfatebyreleasingtnfaip8encapsulatedexosomes
AT miaojinhua kidneytubularepithelialcellscontrolinterstitialfibroblastfatebyreleasingtnfaip8encapsulatedexosomes
AT zhoushan kidneytubularepithelialcellscontrolinterstitialfibroblastfatebyreleasingtnfaip8encapsulatedexosomes
AT renqian kidneytubularepithelialcellscontrolinterstitialfibroblastfatebyreleasingtnfaip8encapsulatedexosomes
AT jianan kidneytubularepithelialcellscontrolinterstitialfibroblastfatebyreleasingtnfaip8encapsulatedexosomes
AT zhoulili kidneytubularepithelialcellscontrolinterstitialfibroblastfatebyreleasingtnfaip8encapsulatedexosomes
AT liuyouhua kidneytubularepithelialcellscontrolinterstitialfibroblastfatebyreleasingtnfaip8encapsulatedexosomes