Cargando…

Research on the pathological mechanism of rectal adenocarcinoma based on DNA methylation

Colorectal cancer is one of the 3 most common cancers worldwide. In this study, a weighted network-based analysis method was proposed to explore the pathological mechanisms and prognostic targets of rectal adenocarcinoma (READ) at the deoxyribonucleic acid (DNA) methylation level. In this study, we...

Descripción completa

Detalles Bibliográficos
Autores principales: Pan, Xiaoqiang, Yi, Xingcheng, Lan, Maozhuo, Su, Xiaoyun, Zhou, Fang, Wu, Wei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Lippincott Williams & Wilkins 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9876001/
https://www.ncbi.nlm.nih.gov/pubmed/36705386
http://dx.doi.org/10.1097/MD.0000000000032763
_version_ 1784878072288247808
author Pan, Xiaoqiang
Yi, Xingcheng
Lan, Maozhuo
Su, Xiaoyun
Zhou, Fang
Wu, Wei
author_facet Pan, Xiaoqiang
Yi, Xingcheng
Lan, Maozhuo
Su, Xiaoyun
Zhou, Fang
Wu, Wei
author_sort Pan, Xiaoqiang
collection PubMed
description Colorectal cancer is one of the 3 most common cancers worldwide. In this study, a weighted network-based analysis method was proposed to explore the pathological mechanisms and prognostic targets of rectal adenocarcinoma (READ) at the deoxyribonucleic acid (DNA) methylation level. In this study, we downloaded clinical information and DNA methylation data from The Cancer Genome Atlas database. Differentially methylated gene analysis was used to identify the differential methylated genes in READ. Canonical correlation analysis was used to construct the weighted gene regulatory network for READ. Multilevel analysis and association analyses between gene modules and clinical information were used to mine key modules related to tumor metastasis evaluation. Genetic significance analysis was used to identify methylation sites in key modules. Finally, the importance of these methylation sites was confirmed using survival analysis. DNA methylation datasets from 90 cancer tissue samples and 6 paracancerous tissue samples were selected. A weighted gene regulatory network was constructed, and a multilevel algorithm was used to divide the gene co-expression network into 20 modules. From gene ontology enrichment analysis, characteristic M was related to biological processes such as the chemotaxis of fibroblast growth factors and the activation and regulation of immune cells etc and characteristic N was associated with the regulation of cytoskeleton formation, mainly microtubules and flagella, regulation of synapses, and regulation of cell mitosis. Based on the results of survival analysis, 7 key methylation sites were found closely correlated to the survival rate of READ, such as cg04441191 (microtubule-associated protein 4 [MAP4]), cg05658717 (KSR2), cg09622330 (GRIN2A), cg10698404 (YWHAG), cg17047993 (SPAG9), cg24504843 (CEP135), and cg24531267 (CEP250). Mutational and transcriptomic level studies revealed significant differences in DNA methylation, single nucleotide polymorphism, and transcript levels between YWHAG and MAP4 in normal tissues compared to tumor tissues, and differential expression of the 2 proteins in immunohistochemistry. Therefore, potential targeting drugs were screened for these 2 proteins for molecular docking, and artenimol was found to bind to MAP4 protein and 27-hydroxycholesterol to YWHAG. Our study found that key methylation sites played an important role in tumor metastasis and were associated with the prognosis of READ. Mutations and methylation may jointly regulate the transcription and translation of related genes, which in turn affect cancer progression. This may provide some new potential therapeutic targets and thoughts for the prognosis of READ.
format Online
Article
Text
id pubmed-9876001
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Lippincott Williams & Wilkins
record_format MEDLINE/PubMed
spelling pubmed-98760012023-01-27 Research on the pathological mechanism of rectal adenocarcinoma based on DNA methylation Pan, Xiaoqiang Yi, Xingcheng Lan, Maozhuo Su, Xiaoyun Zhou, Fang Wu, Wei Medicine (Baltimore) 5700 Colorectal cancer is one of the 3 most common cancers worldwide. In this study, a weighted network-based analysis method was proposed to explore the pathological mechanisms and prognostic targets of rectal adenocarcinoma (READ) at the deoxyribonucleic acid (DNA) methylation level. In this study, we downloaded clinical information and DNA methylation data from The Cancer Genome Atlas database. Differentially methylated gene analysis was used to identify the differential methylated genes in READ. Canonical correlation analysis was used to construct the weighted gene regulatory network for READ. Multilevel analysis and association analyses between gene modules and clinical information were used to mine key modules related to tumor metastasis evaluation. Genetic significance analysis was used to identify methylation sites in key modules. Finally, the importance of these methylation sites was confirmed using survival analysis. DNA methylation datasets from 90 cancer tissue samples and 6 paracancerous tissue samples were selected. A weighted gene regulatory network was constructed, and a multilevel algorithm was used to divide the gene co-expression network into 20 modules. From gene ontology enrichment analysis, characteristic M was related to biological processes such as the chemotaxis of fibroblast growth factors and the activation and regulation of immune cells etc and characteristic N was associated with the regulation of cytoskeleton formation, mainly microtubules and flagella, regulation of synapses, and regulation of cell mitosis. Based on the results of survival analysis, 7 key methylation sites were found closely correlated to the survival rate of READ, such as cg04441191 (microtubule-associated protein 4 [MAP4]), cg05658717 (KSR2), cg09622330 (GRIN2A), cg10698404 (YWHAG), cg17047993 (SPAG9), cg24504843 (CEP135), and cg24531267 (CEP250). Mutational and transcriptomic level studies revealed significant differences in DNA methylation, single nucleotide polymorphism, and transcript levels between YWHAG and MAP4 in normal tissues compared to tumor tissues, and differential expression of the 2 proteins in immunohistochemistry. Therefore, potential targeting drugs were screened for these 2 proteins for molecular docking, and artenimol was found to bind to MAP4 protein and 27-hydroxycholesterol to YWHAG. Our study found that key methylation sites played an important role in tumor metastasis and were associated with the prognosis of READ. Mutations and methylation may jointly regulate the transcription and translation of related genes, which in turn affect cancer progression. This may provide some new potential therapeutic targets and thoughts for the prognosis of READ. Lippincott Williams & Wilkins 2023-01-27 /pmc/articles/PMC9876001/ /pubmed/36705386 http://dx.doi.org/10.1097/MD.0000000000032763 Text en Copyright © 2023 the Author(s). Published by Wolters Kluwer Health, Inc. https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the Creative Commons Attribution License 4.0 (CCBY) (https://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle 5700
Pan, Xiaoqiang
Yi, Xingcheng
Lan, Maozhuo
Su, Xiaoyun
Zhou, Fang
Wu, Wei
Research on the pathological mechanism of rectal adenocarcinoma based on DNA methylation
title Research on the pathological mechanism of rectal adenocarcinoma based on DNA methylation
title_full Research on the pathological mechanism of rectal adenocarcinoma based on DNA methylation
title_fullStr Research on the pathological mechanism of rectal adenocarcinoma based on DNA methylation
title_full_unstemmed Research on the pathological mechanism of rectal adenocarcinoma based on DNA methylation
title_short Research on the pathological mechanism of rectal adenocarcinoma based on DNA methylation
title_sort research on the pathological mechanism of rectal adenocarcinoma based on dna methylation
topic 5700
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9876001/
https://www.ncbi.nlm.nih.gov/pubmed/36705386
http://dx.doi.org/10.1097/MD.0000000000032763
work_keys_str_mv AT panxiaoqiang researchonthepathologicalmechanismofrectaladenocarcinomabasedondnamethylation
AT yixingcheng researchonthepathologicalmechanismofrectaladenocarcinomabasedondnamethylation
AT lanmaozhuo researchonthepathologicalmechanismofrectaladenocarcinomabasedondnamethylation
AT suxiaoyun researchonthepathologicalmechanismofrectaladenocarcinomabasedondnamethylation
AT zhoufang researchonthepathologicalmechanismofrectaladenocarcinomabasedondnamethylation
AT wuwei researchonthepathologicalmechanismofrectaladenocarcinomabasedondnamethylation