Cargando…

Cancer-Associated Fibroblasts Hinder Lung Squamous Cell Carcinoma Oxidative Stress-Induced Apoptosis via METTL3 Mediated m(6)A Methylation of COL10A1

BACKGROUND: Cancer-associated fibroblasts (CAFs) within the tumor microenvironment are key players in tumorigenesis and tumor development. Nevertheless, the regulatory mechanisms of CAFs on lung squamous cell carcinoma- (LUSC-) associated remain poorly elucidated. METHODS: The microarray dataset GSE...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Yuchan, Li, Xiaoxue, Deng, Muwen, Ye, Changda, Peng, Yuanhong, Lu, Yan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Hindawi 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9560815/
https://www.ncbi.nlm.nih.gov/pubmed/36246404
http://dx.doi.org/10.1155/2022/4320809
_version_ 1784807835023966208
author Li, Yuchan
Li, Xiaoxue
Deng, Muwen
Ye, Changda
Peng, Yuanhong
Lu, Yan
author_facet Li, Yuchan
Li, Xiaoxue
Deng, Muwen
Ye, Changda
Peng, Yuanhong
Lu, Yan
author_sort Li, Yuchan
collection PubMed
description BACKGROUND: Cancer-associated fibroblasts (CAFs) within the tumor microenvironment are key players in tumorigenesis and tumor development. Nevertheless, the regulatory mechanisms of CAFs on lung squamous cell carcinoma- (LUSC-) associated remain poorly elucidated. METHODS: The microarray dataset GSE22874, containing 30 specimens of primary culture of normal fibroblasts (NFs) and 8 specimens of cancer-associated fibroblasts (CAFs) samples derived from LUSC, was retrieved from the Gene Expression Omnibus (GEO) database and then calculated by using the R language (limma package) to identify differentially expressed genes (DEGs). CAF-conditioned medium (CAF-CM) was collected and used to culture LUSC cells, followed by assessment of cell proliferation, apoptosis, and oxidative stress levels by using CCK-8, annexin V-FITC/PI double staining and ELISA assays. Subsequently, COL10A1 was knocked down in CAFs to assess the role of COL10A1 in CAF regulation of LUSC behavior. Bioinformatics online analysis and MeRIP were applied to predict and test the m(6)A modification of COL10A1 mRNA and the regulatory relationship with METTL3. Rescue experiments were next performed to explore the effects of METTL3 and COL10A1 in CAFs on LUSC cell proliferation, apoptosis, and oxidative stress. LUSC tumor cells with or without (COL10A1-silenced) CAFs were subcutaneously inoculated in nude mice to evaluate the effect of COL10A1 in CAFs on LUSC tumor growth. RESULTS: Elevated expression of COL10A1 was found in LUSC-derived CAFs by GSE22874 dataset analysis. We discovered that COL10A1 and METTL3 was expressed in both LUSC cells and matched CAFs, while COL10A1 expression was prominently higher in CAFs than in LUSC cells. CAF-CM memorably encouraged LUSC cell proliferation and suppressed apoptosis-induced oxidative stress, which was reversed by interfering with COL10A1 expression in CAFs, suggesting that COL10A1 might be secreted by CAFs into the culture medium to exert its effects inside LUSC cells. Global m(6)A modification was decreased in METTL3 knocked down CAFs. M(6)A modification, expression levels, and stability of COL10A1 mRNA were impaired upon METTL3 knockdown in CAFs. Overexpression of COL10A1 in CAFs partially reversed the effect of METTL3 knockdown on the malignant behavior of LUSC cells. In vivo studies confirmed that CAFs accelerated LUSC tumor growth, and this effect was counteracted by COL10A1 silencing. CONCLUSIONS: COL10A1 secreted by CAFs could facilitate LUSC cell proliferation and repress apoptosis-induced oxidative stress, and the mechanism was due to elevated expression mediated by METTL3 promoting its mRNA m(6)A modification, thereby accelerating tumor growth.
format Online
Article
Text
id pubmed-9560815
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Hindawi
record_format MEDLINE/PubMed
spelling pubmed-95608152022-10-14 Cancer-Associated Fibroblasts Hinder Lung Squamous Cell Carcinoma Oxidative Stress-Induced Apoptosis via METTL3 Mediated m(6)A Methylation of COL10A1 Li, Yuchan Li, Xiaoxue Deng, Muwen Ye, Changda Peng, Yuanhong Lu, Yan Oxid Med Cell Longev Research Article BACKGROUND: Cancer-associated fibroblasts (CAFs) within the tumor microenvironment are key players in tumorigenesis and tumor development. Nevertheless, the regulatory mechanisms of CAFs on lung squamous cell carcinoma- (LUSC-) associated remain poorly elucidated. METHODS: The microarray dataset GSE22874, containing 30 specimens of primary culture of normal fibroblasts (NFs) and 8 specimens of cancer-associated fibroblasts (CAFs) samples derived from LUSC, was retrieved from the Gene Expression Omnibus (GEO) database and then calculated by using the R language (limma package) to identify differentially expressed genes (DEGs). CAF-conditioned medium (CAF-CM) was collected and used to culture LUSC cells, followed by assessment of cell proliferation, apoptosis, and oxidative stress levels by using CCK-8, annexin V-FITC/PI double staining and ELISA assays. Subsequently, COL10A1 was knocked down in CAFs to assess the role of COL10A1 in CAF regulation of LUSC behavior. Bioinformatics online analysis and MeRIP were applied to predict and test the m(6)A modification of COL10A1 mRNA and the regulatory relationship with METTL3. Rescue experiments were next performed to explore the effects of METTL3 and COL10A1 in CAFs on LUSC cell proliferation, apoptosis, and oxidative stress. LUSC tumor cells with or without (COL10A1-silenced) CAFs were subcutaneously inoculated in nude mice to evaluate the effect of COL10A1 in CAFs on LUSC tumor growth. RESULTS: Elevated expression of COL10A1 was found in LUSC-derived CAFs by GSE22874 dataset analysis. We discovered that COL10A1 and METTL3 was expressed in both LUSC cells and matched CAFs, while COL10A1 expression was prominently higher in CAFs than in LUSC cells. CAF-CM memorably encouraged LUSC cell proliferation and suppressed apoptosis-induced oxidative stress, which was reversed by interfering with COL10A1 expression in CAFs, suggesting that COL10A1 might be secreted by CAFs into the culture medium to exert its effects inside LUSC cells. Global m(6)A modification was decreased in METTL3 knocked down CAFs. M(6)A modification, expression levels, and stability of COL10A1 mRNA were impaired upon METTL3 knockdown in CAFs. Overexpression of COL10A1 in CAFs partially reversed the effect of METTL3 knockdown on the malignant behavior of LUSC cells. In vivo studies confirmed that CAFs accelerated LUSC tumor growth, and this effect was counteracted by COL10A1 silencing. CONCLUSIONS: COL10A1 secreted by CAFs could facilitate LUSC cell proliferation and repress apoptosis-induced oxidative stress, and the mechanism was due to elevated expression mediated by METTL3 promoting its mRNA m(6)A modification, thereby accelerating tumor growth. Hindawi 2022-10-06 /pmc/articles/PMC9560815/ /pubmed/36246404 http://dx.doi.org/10.1155/2022/4320809 Text en Copyright © 2022 Yuchan Li et al. https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Article
Li, Yuchan
Li, Xiaoxue
Deng, Muwen
Ye, Changda
Peng, Yuanhong
Lu, Yan
Cancer-Associated Fibroblasts Hinder Lung Squamous Cell Carcinoma Oxidative Stress-Induced Apoptosis via METTL3 Mediated m(6)A Methylation of COL10A1
title Cancer-Associated Fibroblasts Hinder Lung Squamous Cell Carcinoma Oxidative Stress-Induced Apoptosis via METTL3 Mediated m(6)A Methylation of COL10A1
title_full Cancer-Associated Fibroblasts Hinder Lung Squamous Cell Carcinoma Oxidative Stress-Induced Apoptosis via METTL3 Mediated m(6)A Methylation of COL10A1
title_fullStr Cancer-Associated Fibroblasts Hinder Lung Squamous Cell Carcinoma Oxidative Stress-Induced Apoptosis via METTL3 Mediated m(6)A Methylation of COL10A1
title_full_unstemmed Cancer-Associated Fibroblasts Hinder Lung Squamous Cell Carcinoma Oxidative Stress-Induced Apoptosis via METTL3 Mediated m(6)A Methylation of COL10A1
title_short Cancer-Associated Fibroblasts Hinder Lung Squamous Cell Carcinoma Oxidative Stress-Induced Apoptosis via METTL3 Mediated m(6)A Methylation of COL10A1
title_sort cancer-associated fibroblasts hinder lung squamous cell carcinoma oxidative stress-induced apoptosis via mettl3 mediated m(6)a methylation of col10a1
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9560815/
https://www.ncbi.nlm.nih.gov/pubmed/36246404
http://dx.doi.org/10.1155/2022/4320809
work_keys_str_mv AT liyuchan cancerassociatedfibroblastshinderlungsquamouscellcarcinomaoxidativestressinducedapoptosisviamettl3mediatedm6amethylationofcol10a1
AT lixiaoxue cancerassociatedfibroblastshinderlungsquamouscellcarcinomaoxidativestressinducedapoptosisviamettl3mediatedm6amethylationofcol10a1
AT dengmuwen cancerassociatedfibroblastshinderlungsquamouscellcarcinomaoxidativestressinducedapoptosisviamettl3mediatedm6amethylationofcol10a1
AT yechangda cancerassociatedfibroblastshinderlungsquamouscellcarcinomaoxidativestressinducedapoptosisviamettl3mediatedm6amethylationofcol10a1
AT pengyuanhong cancerassociatedfibroblastshinderlungsquamouscellcarcinomaoxidativestressinducedapoptosisviamettl3mediatedm6amethylationofcol10a1
AT luyan cancerassociatedfibroblastshinderlungsquamouscellcarcinomaoxidativestressinducedapoptosisviamettl3mediatedm6amethylationofcol10a1