Cargando…

Monitoring methylation-driven genes as prognostic biomarkers in patients with lung squamous cell cancer

Aberrant DNA methylations have been reported to be significantly associated with lung squamous cell cancer (LUSC). The aim of this study was to investigate the DNA methylation-driven genes in LUSC by integrative bioinformatics analysis. In the present study, methylation-driven genes in LUSC were scr...

Descripción completa

Detalles Bibliográficos
Autores principales: Han, Pengkai, Liu, Qiping, Xiang, Jianhua
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6924172/
https://www.ncbi.nlm.nih.gov/pubmed/31897186
http://dx.doi.org/10.3892/ol.2019.11163
_version_ 1783481677088030720
author Han, Pengkai
Liu, Qiping
Xiang, Jianhua
author_facet Han, Pengkai
Liu, Qiping
Xiang, Jianhua
author_sort Han, Pengkai
collection PubMed
description Aberrant DNA methylations have been reported to be significantly associated with lung squamous cell cancer (LUSC). The aim of this study was to investigate the DNA methylation-driven genes in LUSC by integrative bioinformatics analysis. In the present study, methylation-driven genes in LUSC were screened out, and survival analysis related to these genes was performed to confirm their value in prognostic assessment. Gene expression and methylation data were downloaded from The Cancer Genome Atlas (TCGA), and the MethylMix algorithm was used to identify methylation-driven genes. ConsensusPathDB was used to perform Gene Ontology and pathway enrichment analysis of methylation-driven genes. Survival analysis was performed to investigate the correlation with prognosis. In total, 52 differentially expressed methylation-driven genes were identified in LUSC and adjacent tissues. Survival analysis showed that DQX1, GPR75, STX12, and TRIM61 could serve as independent prognostic biomarkers. In addition, the combined methylation and gene expression survival analysis revealed that the combined expression level of the genes ALG1L, DQX1, and ZNF418 alone can be used as a prognostic marker or drug target. Methylation of four sites of gene ZNF418, four sites of ZNF701, two sites of DQX1, and four sites of DCAF4L2 was significantly associated with survival. The present study provides an important bioinformatic and relevant theoretical basis for subsequent early diagnosis and prognostic assessment of LUSC.
format Online
Article
Text
id pubmed-6924172
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-69241722020-01-02 Monitoring methylation-driven genes as prognostic biomarkers in patients with lung squamous cell cancer Han, Pengkai Liu, Qiping Xiang, Jianhua Oncol Lett Articles Aberrant DNA methylations have been reported to be significantly associated with lung squamous cell cancer (LUSC). The aim of this study was to investigate the DNA methylation-driven genes in LUSC by integrative bioinformatics analysis. In the present study, methylation-driven genes in LUSC were screened out, and survival analysis related to these genes was performed to confirm their value in prognostic assessment. Gene expression and methylation data were downloaded from The Cancer Genome Atlas (TCGA), and the MethylMix algorithm was used to identify methylation-driven genes. ConsensusPathDB was used to perform Gene Ontology and pathway enrichment analysis of methylation-driven genes. Survival analysis was performed to investigate the correlation with prognosis. In total, 52 differentially expressed methylation-driven genes were identified in LUSC and adjacent tissues. Survival analysis showed that DQX1, GPR75, STX12, and TRIM61 could serve as independent prognostic biomarkers. In addition, the combined methylation and gene expression survival analysis revealed that the combined expression level of the genes ALG1L, DQX1, and ZNF418 alone can be used as a prognostic marker or drug target. Methylation of four sites of gene ZNF418, four sites of ZNF701, two sites of DQX1, and four sites of DCAF4L2 was significantly associated with survival. The present study provides an important bioinformatic and relevant theoretical basis for subsequent early diagnosis and prognostic assessment of LUSC. D.A. Spandidos 2020-01 2019-11-29 /pmc/articles/PMC6924172/ /pubmed/31897186 http://dx.doi.org/10.3892/ol.2019.11163 Text en Copyright: © Han 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
Han, Pengkai
Liu, Qiping
Xiang, Jianhua
Monitoring methylation-driven genes as prognostic biomarkers in patients with lung squamous cell cancer
title Monitoring methylation-driven genes as prognostic biomarkers in patients with lung squamous cell cancer
title_full Monitoring methylation-driven genes as prognostic biomarkers in patients with lung squamous cell cancer
title_fullStr Monitoring methylation-driven genes as prognostic biomarkers in patients with lung squamous cell cancer
title_full_unstemmed Monitoring methylation-driven genes as prognostic biomarkers in patients with lung squamous cell cancer
title_short Monitoring methylation-driven genes as prognostic biomarkers in patients with lung squamous cell cancer
title_sort monitoring methylation-driven genes as prognostic biomarkers in patients with lung squamous cell cancer
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6924172/
https://www.ncbi.nlm.nih.gov/pubmed/31897186
http://dx.doi.org/10.3892/ol.2019.11163
work_keys_str_mv AT hanpengkai monitoringmethylationdrivengenesasprognosticbiomarkersinpatientswithlungsquamouscellcancer
AT liuqiping monitoringmethylationdrivengenesasprognosticbiomarkersinpatientswithlungsquamouscellcancer
AT xiangjianhua monitoringmethylationdrivengenesasprognosticbiomarkersinpatientswithlungsquamouscellcancer