Cargando…

Identification of DNA methylation-driven genes by integrative analysis of DNA methylation and transcriptome data in pancreatic adenocarcinoma

Pancreatic adenocarcinoma (PAAD) is a painful and fatal disease that undoubtedly remains a health care priority and offers significant therapeutic challenges. The significance of epigenetic modifications, including DNA methylation in tumor development, has gained the attention of researchers. Identi...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Wei, Shang, Shuai, Yang, Yingying, Lu, Peiyao, Wang, Teng, Cui, Xinyi, Tang, Xuexi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7086284/
https://www.ncbi.nlm.nih.gov/pubmed/32256782
http://dx.doi.org/10.3892/etm.2020.8554
_version_ 1783509095845724160
author Zhang, Wei
Shang, Shuai
Yang, Yingying
Lu, Peiyao
Wang, Teng
Cui, Xinyi
Tang, Xuexi
author_facet Zhang, Wei
Shang, Shuai
Yang, Yingying
Lu, Peiyao
Wang, Teng
Cui, Xinyi
Tang, Xuexi
author_sort Zhang, Wei
collection PubMed
description Pancreatic adenocarcinoma (PAAD) is a painful and fatal disease that undoubtedly remains a health care priority and offers significant therapeutic challenges. The significance of epigenetic modifications, including DNA methylation in tumor development, has gained the attention of researchers. Identifying DNA methylation-driven genes and investigating the mechanisms underlying the tumorigenesis of PAAD are of substantial importance for developing methods of physiological evaluation, treatment planning and prognostic prediction for PAAD. In the present study, a comprehensive analysis of DNA methylation and gene expression data from 188 clinical samples was performed to identify DNA methylation-driven genes in PAAD. In addition, the diagnostic and prognostic value of DNA methylation-driven genes was evaluated using receiver operating characteristic curve, survival and recurrence analyses. A total of 7 DNA methylation-driven genes, namely zinc finger protein 208 (ZNF208), eomesodermin (EOMES), prostaglandin D2 receptor (PTGDR), chromosome 12 open reading frame 42 (C12orf42), integrin subunit α 4 (ITGA4), dedicator of cytokinesis 8 and protein phosphatase 1 regulatory inhibitor subunit 14D (PPP1R14D), were identified. All of them may be used to diagnose PAAD with excellent specificity and sensitivity (area under curve, >0.8). Of the 7 DNA methylation-driven genes, 6 were significantly associated with overall survival (OS) and recurrence-free survival (RFS) P<0.05). Among them, ZNF208, EOMES, PTGDR, C12orf42 and ITGA4 were significantly negatively associated with the OS rate and positively associated with the recurrence rate, while PPP1R14D was significantly positively associated with the OS rate and negatively associated with the recurrence rate. The present study provides novel insight into the epigenetic alterations associated with the occurrence and progression of PAAD, thereby increasing the mechanistic understanding of this disease, offering potential novel molecular biomarkers and contributing to the development of therapeutic targets for PAAD.
format Online
Article
Text
id pubmed-7086284
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-70862842020-04-02 Identification of DNA methylation-driven genes by integrative analysis of DNA methylation and transcriptome data in pancreatic adenocarcinoma Zhang, Wei Shang, Shuai Yang, Yingying Lu, Peiyao Wang, Teng Cui, Xinyi Tang, Xuexi Exp Ther Med Articles Pancreatic adenocarcinoma (PAAD) is a painful and fatal disease that undoubtedly remains a health care priority and offers significant therapeutic challenges. The significance of epigenetic modifications, including DNA methylation in tumor development, has gained the attention of researchers. Identifying DNA methylation-driven genes and investigating the mechanisms underlying the tumorigenesis of PAAD are of substantial importance for developing methods of physiological evaluation, treatment planning and prognostic prediction for PAAD. In the present study, a comprehensive analysis of DNA methylation and gene expression data from 188 clinical samples was performed to identify DNA methylation-driven genes in PAAD. In addition, the diagnostic and prognostic value of DNA methylation-driven genes was evaluated using receiver operating characteristic curve, survival and recurrence analyses. A total of 7 DNA methylation-driven genes, namely zinc finger protein 208 (ZNF208), eomesodermin (EOMES), prostaglandin D2 receptor (PTGDR), chromosome 12 open reading frame 42 (C12orf42), integrin subunit α 4 (ITGA4), dedicator of cytokinesis 8 and protein phosphatase 1 regulatory inhibitor subunit 14D (PPP1R14D), were identified. All of them may be used to diagnose PAAD with excellent specificity and sensitivity (area under curve, >0.8). Of the 7 DNA methylation-driven genes, 6 were significantly associated with overall survival (OS) and recurrence-free survival (RFS) P<0.05). Among them, ZNF208, EOMES, PTGDR, C12orf42 and ITGA4 were significantly negatively associated with the OS rate and positively associated with the recurrence rate, while PPP1R14D was significantly positively associated with the OS rate and negatively associated with the recurrence rate. The present study provides novel insight into the epigenetic alterations associated with the occurrence and progression of PAAD, thereby increasing the mechanistic understanding of this disease, offering potential novel molecular biomarkers and contributing to the development of therapeutic targets for PAAD. D.A. Spandidos 2020-04 2020-02-26 /pmc/articles/PMC7086284/ /pubmed/32256782 http://dx.doi.org/10.3892/etm.2020.8554 Text en Copyright: © Zhang 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
Zhang, Wei
Shang, Shuai
Yang, Yingying
Lu, Peiyao
Wang, Teng
Cui, Xinyi
Tang, Xuexi
Identification of DNA methylation-driven genes by integrative analysis of DNA methylation and transcriptome data in pancreatic adenocarcinoma
title Identification of DNA methylation-driven genes by integrative analysis of DNA methylation and transcriptome data in pancreatic adenocarcinoma
title_full Identification of DNA methylation-driven genes by integrative analysis of DNA methylation and transcriptome data in pancreatic adenocarcinoma
title_fullStr Identification of DNA methylation-driven genes by integrative analysis of DNA methylation and transcriptome data in pancreatic adenocarcinoma
title_full_unstemmed Identification of DNA methylation-driven genes by integrative analysis of DNA methylation and transcriptome data in pancreatic adenocarcinoma
title_short Identification of DNA methylation-driven genes by integrative analysis of DNA methylation and transcriptome data in pancreatic adenocarcinoma
title_sort identification of dna methylation-driven genes by integrative analysis of dna methylation and transcriptome data in pancreatic adenocarcinoma
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7086284/
https://www.ncbi.nlm.nih.gov/pubmed/32256782
http://dx.doi.org/10.3892/etm.2020.8554
work_keys_str_mv AT zhangwei identificationofdnamethylationdrivengenesbyintegrativeanalysisofdnamethylationandtranscriptomedatainpancreaticadenocarcinoma
AT shangshuai identificationofdnamethylationdrivengenesbyintegrativeanalysisofdnamethylationandtranscriptomedatainpancreaticadenocarcinoma
AT yangyingying identificationofdnamethylationdrivengenesbyintegrativeanalysisofdnamethylationandtranscriptomedatainpancreaticadenocarcinoma
AT lupeiyao identificationofdnamethylationdrivengenesbyintegrativeanalysisofdnamethylationandtranscriptomedatainpancreaticadenocarcinoma
AT wangteng identificationofdnamethylationdrivengenesbyintegrativeanalysisofdnamethylationandtranscriptomedatainpancreaticadenocarcinoma
AT cuixinyi identificationofdnamethylationdrivengenesbyintegrativeanalysisofdnamethylationandtranscriptomedatainpancreaticadenocarcinoma
AT tangxuexi identificationofdnamethylationdrivengenesbyintegrativeanalysisofdnamethylationandtranscriptomedatainpancreaticadenocarcinoma