Cargando…

Overexpression of FES might inhibit cell proliferation, migration, and invasion of osteosarcoma cells

BACKGROUND: This study aimed to screen osteosarcoma (OS) prognosis relevant genes for methylation dysregulation, and the functional mechanisms of FES overexpression in OS cells were investigated. METHODS: The OS prognosis relevant genes with differentially methylated positions (DMPs) identified from...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhao, Yang, Wang, Zhimeng, Wang, Qian, Sun, Liang, Li, Ming, Ren, Cheng, Xue, Hanzhong, Li, Zhong, Zhang, Kun, Hao, Dingjun, Yang, Na, Song, Zhe, Ma, Teng, Lu, Yao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7106745/
https://www.ncbi.nlm.nih.gov/pubmed/32256211
http://dx.doi.org/10.1186/s12935-020-01181-3
_version_ 1783512677050482688
author Zhao, Yang
Wang, Zhimeng
Wang, Qian
Sun, Liang
Li, Ming
Ren, Cheng
Xue, Hanzhong
Li, Zhong
Zhang, Kun
Hao, Dingjun
Yang, Na
Song, Zhe
Ma, Teng
Lu, Yao
author_facet Zhao, Yang
Wang, Zhimeng
Wang, Qian
Sun, Liang
Li, Ming
Ren, Cheng
Xue, Hanzhong
Li, Zhong
Zhang, Kun
Hao, Dingjun
Yang, Na
Song, Zhe
Ma, Teng
Lu, Yao
author_sort Zhao, Yang
collection PubMed
description BACKGROUND: This study aimed to screen osteosarcoma (OS) prognosis relevant genes for methylation dysregulation, and the functional mechanisms of FES overexpression in OS cells were investigated. METHODS: The OS prognosis relevant genes with differentially methylated positions (DMPs) identified from the GSE36001 and GSE36002 datasets, and the UCSC database, were used as a training set to construct a risk model, while the GSE21257 dataset was used as validation set. The expression levels of several key genes in OS cells after 5-Aza-2′-deoxycytidine treatment were detected by qPCR. The effects of FES overexpression on cell proliferation, cell cycle, migration, and invasion of MNNG/HOS were analyzed by CCK8, flow cytometry, and Transwell assays. RESULTS: A total of 31 candidate genes, corresponding to 36 DMPs, were identified as OS prognosis relevant genes; from these, the top 10 genes were used to construct a risk model. Following validation of the risk model, FES, LYL1, MAP4K1, RIPK3, SLC15A3, and STAT3 showed expression changes between the OS and control samples. qPCR results showed that the expression of FES was significantly downregulated in three OS cell lines and increased after 5-Aza-DC treatment. The proliferation, cell cycle progression, migration, and invasion of MNNG/HOS cells were significantly inhibited after transfection with FES overexpression plasmid, and the protein expression of FYN and β catenin were decreased in MNNG/HOS cells by FES overexpression. CONCLUSIONS: The decrease in FES by hypermethylation was associated with OS prognosis, and might contribute to the proliferation, migration, and invasion of OS cells. FES, and its upstream FYN and β catenin, might coordinately exert a tumor suppressor effect in OS cells.
format Online
Article
Text
id pubmed-7106745
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-71067452020-04-01 Overexpression of FES might inhibit cell proliferation, migration, and invasion of osteosarcoma cells Zhao, Yang Wang, Zhimeng Wang, Qian Sun, Liang Li, Ming Ren, Cheng Xue, Hanzhong Li, Zhong Zhang, Kun Hao, Dingjun Yang, Na Song, Zhe Ma, Teng Lu, Yao Cancer Cell Int Primary Research BACKGROUND: This study aimed to screen osteosarcoma (OS) prognosis relevant genes for methylation dysregulation, and the functional mechanisms of FES overexpression in OS cells were investigated. METHODS: The OS prognosis relevant genes with differentially methylated positions (DMPs) identified from the GSE36001 and GSE36002 datasets, and the UCSC database, were used as a training set to construct a risk model, while the GSE21257 dataset was used as validation set. The expression levels of several key genes in OS cells after 5-Aza-2′-deoxycytidine treatment were detected by qPCR. The effects of FES overexpression on cell proliferation, cell cycle, migration, and invasion of MNNG/HOS were analyzed by CCK8, flow cytometry, and Transwell assays. RESULTS: A total of 31 candidate genes, corresponding to 36 DMPs, were identified as OS prognosis relevant genes; from these, the top 10 genes were used to construct a risk model. Following validation of the risk model, FES, LYL1, MAP4K1, RIPK3, SLC15A3, and STAT3 showed expression changes between the OS and control samples. qPCR results showed that the expression of FES was significantly downregulated in three OS cell lines and increased after 5-Aza-DC treatment. The proliferation, cell cycle progression, migration, and invasion of MNNG/HOS cells were significantly inhibited after transfection with FES overexpression plasmid, and the protein expression of FYN and β catenin were decreased in MNNG/HOS cells by FES overexpression. CONCLUSIONS: The decrease in FES by hypermethylation was associated with OS prognosis, and might contribute to the proliferation, migration, and invasion of OS cells. FES, and its upstream FYN and β catenin, might coordinately exert a tumor suppressor effect in OS cells. BioMed Central 2020-03-30 /pmc/articles/PMC7106745/ /pubmed/32256211 http://dx.doi.org/10.1186/s12935-020-01181-3 Text en © The Author(s) 2020 Open AccessThis 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 licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence 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 licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Primary Research
Zhao, Yang
Wang, Zhimeng
Wang, Qian
Sun, Liang
Li, Ming
Ren, Cheng
Xue, Hanzhong
Li, Zhong
Zhang, Kun
Hao, Dingjun
Yang, Na
Song, Zhe
Ma, Teng
Lu, Yao
Overexpression of FES might inhibit cell proliferation, migration, and invasion of osteosarcoma cells
title Overexpression of FES might inhibit cell proliferation, migration, and invasion of osteosarcoma cells
title_full Overexpression of FES might inhibit cell proliferation, migration, and invasion of osteosarcoma cells
title_fullStr Overexpression of FES might inhibit cell proliferation, migration, and invasion of osteosarcoma cells
title_full_unstemmed Overexpression of FES might inhibit cell proliferation, migration, and invasion of osteosarcoma cells
title_short Overexpression of FES might inhibit cell proliferation, migration, and invasion of osteosarcoma cells
title_sort overexpression of fes might inhibit cell proliferation, migration, and invasion of osteosarcoma cells
topic Primary Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7106745/
https://www.ncbi.nlm.nih.gov/pubmed/32256211
http://dx.doi.org/10.1186/s12935-020-01181-3
work_keys_str_mv AT zhaoyang overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT wangzhimeng overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT wangqian overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT sunliang overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT liming overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT rencheng overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT xuehanzhong overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT lizhong overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT zhangkun overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT haodingjun overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT yangna overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT songzhe overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT mateng overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells
AT luyao overexpressionoffesmightinhibitcellproliferationmigrationandinvasionofosteosarcomacells