Cargando…

Why MUC16 mutations lead to a better prognosis: A study based on The Cancer Genome Atlas gastric cancer cohort

BACKGROUND: MUC16, encoding cancer antigen 125, is a frequently mutated gene in gastric cancer. In addition, MUC16 mutations seem to result in a better prognosis in gastric cancer. However, the mechanisms that lead to a better prognosis by MUC16 mutations have not yet been clarified. AIM: To delve d...

Descripción completa

Detalles Bibliográficos
Autores principales: Huang, Yu-Jie, Cao, Zhi-Fei, Wang, Jie, Yang, Jian, Wei, Yi-Jun, Tang, Yu-Chen, Cheng, Yin-Xiang, Zhou, Jian, Zhang, Zi-Xiang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Baishideng Publishing Group Inc 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8173414/
https://www.ncbi.nlm.nih.gov/pubmed/34141777
http://dx.doi.org/10.12998/wjcc.v9.i17.4143
_version_ 1783702719947603968
author Huang, Yu-Jie
Cao, Zhi-Fei
Wang, Jie
Yang, Jian
Wei, Yi-Jun
Tang, Yu-Chen
Cheng, Yin-Xiang
Zhou, Jian
Zhang, Zi-Xiang
author_facet Huang, Yu-Jie
Cao, Zhi-Fei
Wang, Jie
Yang, Jian
Wei, Yi-Jun
Tang, Yu-Chen
Cheng, Yin-Xiang
Zhou, Jian
Zhang, Zi-Xiang
author_sort Huang, Yu-Jie
collection PubMed
description BACKGROUND: MUC16, encoding cancer antigen 125, is a frequently mutated gene in gastric cancer. In addition, MUC16 mutations seem to result in a better prognosis in gastric cancer. However, the mechanisms that lead to a better prognosis by MUC16 mutations have not yet been clarified. AIM: To delve deeper into the underlying mechanisms that explain why MUC16 mutations signal a better prognosis in gastric cancer. METHODS: We used multi-omics data, including mRNA, simple nucleotide variation, copy number variation and methylation data from The Cancer Genome Atlas, to explore the relationship between MUC16 mutations and prognosis. Cox regression and random survival forest algorithms were applied to search for hub genes. Gene set enrichment analysis was used to elucidate the molecular mechanisms. Single-sample gene set enrichment analysis and “EpiDISH” were used to assess immune cells infiltration, and “ESTIMATE” for analysis of the tumor microenvironment. RESULTS: Our study found that compared to the wild-type group, the mutation group had a better prognosis. Additional analysis indicated that the MUC16 mutations appear to activate the DNA repair and p53 pathways to act as an anti-tumor agent. We also identified a key gene, NPY1R (neuropeptide Y receptor Y1), which was significantly more highly expressed in the MUC16 mutations group than in the MUC16 wild-type group. The high expression of NPY1R predicted a poorer prognosis, which was also confirmed in a separate Gene Expression Omnibus cohort. Further susceptibility analysis revealed that NPY1R might be a potential drug target for gastric cancer. Furthermore, in the analysis of the tumor microenvironment, we found that immune cells in the mutation group exhibited higher anti-tumor effects. In addition, the tumor mutation burden and cancer stem cells index were also higher in the mutation group than in the wild-type group. CONCLUSION: We speculated that the MUC16 mutations might activate the p53 pathway and DNA repair pathway: alternatively, the tumor microenvironment may be involved.
format Online
Article
Text
id pubmed-8173414
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Baishideng Publishing Group Inc
record_format MEDLINE/PubMed
spelling pubmed-81734142021-06-16 Why MUC16 mutations lead to a better prognosis: A study based on The Cancer Genome Atlas gastric cancer cohort Huang, Yu-Jie Cao, Zhi-Fei Wang, Jie Yang, Jian Wei, Yi-Jun Tang, Yu-Chen Cheng, Yin-Xiang Zhou, Jian Zhang, Zi-Xiang World J Clin Cases Retrospective Study BACKGROUND: MUC16, encoding cancer antigen 125, is a frequently mutated gene in gastric cancer. In addition, MUC16 mutations seem to result in a better prognosis in gastric cancer. However, the mechanisms that lead to a better prognosis by MUC16 mutations have not yet been clarified. AIM: To delve deeper into the underlying mechanisms that explain why MUC16 mutations signal a better prognosis in gastric cancer. METHODS: We used multi-omics data, including mRNA, simple nucleotide variation, copy number variation and methylation data from The Cancer Genome Atlas, to explore the relationship between MUC16 mutations and prognosis. Cox regression and random survival forest algorithms were applied to search for hub genes. Gene set enrichment analysis was used to elucidate the molecular mechanisms. Single-sample gene set enrichment analysis and “EpiDISH” were used to assess immune cells infiltration, and “ESTIMATE” for analysis of the tumor microenvironment. RESULTS: Our study found that compared to the wild-type group, the mutation group had a better prognosis. Additional analysis indicated that the MUC16 mutations appear to activate the DNA repair and p53 pathways to act as an anti-tumor agent. We also identified a key gene, NPY1R (neuropeptide Y receptor Y1), which was significantly more highly expressed in the MUC16 mutations group than in the MUC16 wild-type group. The high expression of NPY1R predicted a poorer prognosis, which was also confirmed in a separate Gene Expression Omnibus cohort. Further susceptibility analysis revealed that NPY1R might be a potential drug target for gastric cancer. Furthermore, in the analysis of the tumor microenvironment, we found that immune cells in the mutation group exhibited higher anti-tumor effects. In addition, the tumor mutation burden and cancer stem cells index were also higher in the mutation group than in the wild-type group. CONCLUSION: We speculated that the MUC16 mutations might activate the p53 pathway and DNA repair pathway: alternatively, the tumor microenvironment may be involved. Baishideng Publishing Group Inc 2021-06-16 2021-06-16 /pmc/articles/PMC8173414/ /pubmed/34141777 http://dx.doi.org/10.12998/wjcc.v9.i17.4143 Text en ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved. https://creativecommons.org/licenses/by-nc/4.0/This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
spellingShingle Retrospective Study
Huang, Yu-Jie
Cao, Zhi-Fei
Wang, Jie
Yang, Jian
Wei, Yi-Jun
Tang, Yu-Chen
Cheng, Yin-Xiang
Zhou, Jian
Zhang, Zi-Xiang
Why MUC16 mutations lead to a better prognosis: A study based on The Cancer Genome Atlas gastric cancer cohort
title Why MUC16 mutations lead to a better prognosis: A study based on The Cancer Genome Atlas gastric cancer cohort
title_full Why MUC16 mutations lead to a better prognosis: A study based on The Cancer Genome Atlas gastric cancer cohort
title_fullStr Why MUC16 mutations lead to a better prognosis: A study based on The Cancer Genome Atlas gastric cancer cohort
title_full_unstemmed Why MUC16 mutations lead to a better prognosis: A study based on The Cancer Genome Atlas gastric cancer cohort
title_short Why MUC16 mutations lead to a better prognosis: A study based on The Cancer Genome Atlas gastric cancer cohort
title_sort why muc16 mutations lead to a better prognosis: a study based on the cancer genome atlas gastric cancer cohort
topic Retrospective Study
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8173414/
https://www.ncbi.nlm.nih.gov/pubmed/34141777
http://dx.doi.org/10.12998/wjcc.v9.i17.4143
work_keys_str_mv AT huangyujie whymuc16mutationsleadtoabetterprognosisastudybasedonthecancergenomeatlasgastriccancercohort
AT caozhifei whymuc16mutationsleadtoabetterprognosisastudybasedonthecancergenomeatlasgastriccancercohort
AT wangjie whymuc16mutationsleadtoabetterprognosisastudybasedonthecancergenomeatlasgastriccancercohort
AT yangjian whymuc16mutationsleadtoabetterprognosisastudybasedonthecancergenomeatlasgastriccancercohort
AT weiyijun whymuc16mutationsleadtoabetterprognosisastudybasedonthecancergenomeatlasgastriccancercohort
AT tangyuchen whymuc16mutationsleadtoabetterprognosisastudybasedonthecancergenomeatlasgastriccancercohort
AT chengyinxiang whymuc16mutationsleadtoabetterprognosisastudybasedonthecancergenomeatlasgastriccancercohort
AT zhoujian whymuc16mutationsleadtoabetterprognosisastudybasedonthecancergenomeatlasgastriccancercohort
AT zhangzixiang whymuc16mutationsleadtoabetterprognosisastudybasedonthecancergenomeatlasgastriccancercohort