Cargando…

LncRNA‐ATB regulates epithelial‐mesenchymal transition progression in pulmonary fibrosis via sponging miR‐29b‐2‐5p and miR‐34c‐3p

Dysregulation of non‐coding RNAs (ncRNAs) has been proved to play pivotal roles in epithelial‐mesenchymal transition (EMT) and fibrosis. We have previously demonstrated the crucial function of long non‐coding RNA (lncRNA) ATB in silica‐induced pulmonary fibrosis‐related EMT progression. However, the...

Descripción completa

Detalles Bibliográficos
Autores principales: Xu, Qi, Cheng, Demin, Liu, Yi, Pan, Honghong, Li, Guanru, Li, Ping, Li, Yan, Sun, Wenqing, Ma, Dongyu, Ni, Chunhui
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8335671/
https://www.ncbi.nlm.nih.gov/pubmed/34180127
http://dx.doi.org/10.1111/jcmm.16758
_version_ 1783733160215838720
author Xu, Qi
Cheng, Demin
Liu, Yi
Pan, Honghong
Li, Guanru
Li, Ping
Li, Yan
Sun, Wenqing
Ma, Dongyu
Ni, Chunhui
author_facet Xu, Qi
Cheng, Demin
Liu, Yi
Pan, Honghong
Li, Guanru
Li, Ping
Li, Yan
Sun, Wenqing
Ma, Dongyu
Ni, Chunhui
author_sort Xu, Qi
collection PubMed
description Dysregulation of non‐coding RNAs (ncRNAs) has been proved to play pivotal roles in epithelial‐mesenchymal transition (EMT) and fibrosis. We have previously demonstrated the crucial function of long non‐coding RNA (lncRNA) ATB in silica‐induced pulmonary fibrosis‐related EMT progression. However, the underlying molecular mechanism has not been fully elucidated. Here, we verified miR‐29b‐2‐5p and miR‐34c‐3p as two vital downstream targets of lncRNA‐ATB. As opposed to lncRNA‐ATB, a significant reduction of both miR‐29b‐2‐5p and miR‐34c‐3p was observed in lung epithelial cells treated with TGF‐β1 and a murine silicosis model. Overexpression miR‐29b‐2‐5p or miR‐34c‐3p inhibited EMT process and abrogated the pro‐fibrotic effects of lncRNA‐ATB in vitro. Further, the ectopic expression of miR‐29b‐2‐5p and miR‐34c‐3p with chemotherapy attenuated silica‐induced pulmonary fibrosis in vivo. Mechanistically, TGF‐β1‐induced lncRNA‐ATB accelerated EMT as a sponge of miR‐29b‐2‐5p and miR‐34c‐3p and shared miRNA response elements with MEKK2 and NOTCH2, thus relieving these two molecules from miRNA‐mediated translational repression. Interestingly, the co‐transfection of miR‐29b‐2‐5p and miR‐34c‐3p showed a synergistic suppression effect on EMT in vitro. Furthermore, the co‐expression of these two miRNAs by using adeno‐associated virus (AAV) better alleviated silica‐induced fibrogenesis than single miRNA. Approaches aiming at lncRNA‐ATB and its downstream effectors may represent new effective therapeutic strategies in pulmonary fibrosis.
format Online
Article
Text
id pubmed-8335671
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-83356712021-08-09 LncRNA‐ATB regulates epithelial‐mesenchymal transition progression in pulmonary fibrosis via sponging miR‐29b‐2‐5p and miR‐34c‐3p Xu, Qi Cheng, Demin Liu, Yi Pan, Honghong Li, Guanru Li, Ping Li, Yan Sun, Wenqing Ma, Dongyu Ni, Chunhui J Cell Mol Med Original Articles Dysregulation of non‐coding RNAs (ncRNAs) has been proved to play pivotal roles in epithelial‐mesenchymal transition (EMT) and fibrosis. We have previously demonstrated the crucial function of long non‐coding RNA (lncRNA) ATB in silica‐induced pulmonary fibrosis‐related EMT progression. However, the underlying molecular mechanism has not been fully elucidated. Here, we verified miR‐29b‐2‐5p and miR‐34c‐3p as two vital downstream targets of lncRNA‐ATB. As opposed to lncRNA‐ATB, a significant reduction of both miR‐29b‐2‐5p and miR‐34c‐3p was observed in lung epithelial cells treated with TGF‐β1 and a murine silicosis model. Overexpression miR‐29b‐2‐5p or miR‐34c‐3p inhibited EMT process and abrogated the pro‐fibrotic effects of lncRNA‐ATB in vitro. Further, the ectopic expression of miR‐29b‐2‐5p and miR‐34c‐3p with chemotherapy attenuated silica‐induced pulmonary fibrosis in vivo. Mechanistically, TGF‐β1‐induced lncRNA‐ATB accelerated EMT as a sponge of miR‐29b‐2‐5p and miR‐34c‐3p and shared miRNA response elements with MEKK2 and NOTCH2, thus relieving these two molecules from miRNA‐mediated translational repression. Interestingly, the co‐transfection of miR‐29b‐2‐5p and miR‐34c‐3p showed a synergistic suppression effect on EMT in vitro. Furthermore, the co‐expression of these two miRNAs by using adeno‐associated virus (AAV) better alleviated silica‐induced fibrogenesis than single miRNA. Approaches aiming at lncRNA‐ATB and its downstream effectors may represent new effective therapeutic strategies in pulmonary fibrosis. John Wiley and Sons Inc. 2021-06-27 2021-08 /pmc/articles/PMC8335671/ /pubmed/34180127 http://dx.doi.org/10.1111/jcmm.16758 Text en © 2021 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd. https://creativecommons.org/licenses/by/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Articles
Xu, Qi
Cheng, Demin
Liu, Yi
Pan, Honghong
Li, Guanru
Li, Ping
Li, Yan
Sun, Wenqing
Ma, Dongyu
Ni, Chunhui
LncRNA‐ATB regulates epithelial‐mesenchymal transition progression in pulmonary fibrosis via sponging miR‐29b‐2‐5p and miR‐34c‐3p
title LncRNA‐ATB regulates epithelial‐mesenchymal transition progression in pulmonary fibrosis via sponging miR‐29b‐2‐5p and miR‐34c‐3p
title_full LncRNA‐ATB regulates epithelial‐mesenchymal transition progression in pulmonary fibrosis via sponging miR‐29b‐2‐5p and miR‐34c‐3p
title_fullStr LncRNA‐ATB regulates epithelial‐mesenchymal transition progression in pulmonary fibrosis via sponging miR‐29b‐2‐5p and miR‐34c‐3p
title_full_unstemmed LncRNA‐ATB regulates epithelial‐mesenchymal transition progression in pulmonary fibrosis via sponging miR‐29b‐2‐5p and miR‐34c‐3p
title_short LncRNA‐ATB regulates epithelial‐mesenchymal transition progression in pulmonary fibrosis via sponging miR‐29b‐2‐5p and miR‐34c‐3p
title_sort lncrna‐atb regulates epithelial‐mesenchymal transition progression in pulmonary fibrosis via sponging mir‐29b‐2‐5p and mir‐34c‐3p
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8335671/
https://www.ncbi.nlm.nih.gov/pubmed/34180127
http://dx.doi.org/10.1111/jcmm.16758
work_keys_str_mv AT xuqi lncrnaatbregulatesepithelialmesenchymaltransitionprogressioninpulmonaryfibrosisviaspongingmir29b25pandmir34c3p
AT chengdemin lncrnaatbregulatesepithelialmesenchymaltransitionprogressioninpulmonaryfibrosisviaspongingmir29b25pandmir34c3p
AT liuyi lncrnaatbregulatesepithelialmesenchymaltransitionprogressioninpulmonaryfibrosisviaspongingmir29b25pandmir34c3p
AT panhonghong lncrnaatbregulatesepithelialmesenchymaltransitionprogressioninpulmonaryfibrosisviaspongingmir29b25pandmir34c3p
AT liguanru lncrnaatbregulatesepithelialmesenchymaltransitionprogressioninpulmonaryfibrosisviaspongingmir29b25pandmir34c3p
AT liping lncrnaatbregulatesepithelialmesenchymaltransitionprogressioninpulmonaryfibrosisviaspongingmir29b25pandmir34c3p
AT liyan lncrnaatbregulatesepithelialmesenchymaltransitionprogressioninpulmonaryfibrosisviaspongingmir29b25pandmir34c3p
AT sunwenqing lncrnaatbregulatesepithelialmesenchymaltransitionprogressioninpulmonaryfibrosisviaspongingmir29b25pandmir34c3p
AT madongyu lncrnaatbregulatesepithelialmesenchymaltransitionprogressioninpulmonaryfibrosisviaspongingmir29b25pandmir34c3p
AT nichunhui lncrnaatbregulatesepithelialmesenchymaltransitionprogressioninpulmonaryfibrosisviaspongingmir29b25pandmir34c3p