Cargando…

Prognostic model construction and immune microenvironment analysis of esophageal cancer based on gene expression data and microRNA target genes

BACKGROUND: Accumulating evidence suggests that microRNA-target genes are closely related to tumorigenesis and progression. This study aims to screen the intersection of differentially expressed mRNAs (DEmRNAs) and the target genes of differentially expressed microRNAs (DEmiRNAs), and to construct a...

Descripción completa

Detalles Bibliográficos
Autores principales: Gu, Bingbing, Zhang, Shuai, Fan, Zhe, Che, Jiajing, Li, Shuting, Li, Yunfei, Pan, Keyu, Lv, Jiali, Wang, Cheng, Zhang, Tao, Wang, Jialin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: AME Publishing Company 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10248560/
https://www.ncbi.nlm.nih.gov/pubmed/37304542
http://dx.doi.org/10.21037/tcr-22-2588
_version_ 1785055402789961728
author Gu, Bingbing
Zhang, Shuai
Fan, Zhe
Che, Jiajing
Li, Shuting
Li, Yunfei
Pan, Keyu
Lv, Jiali
Wang, Cheng
Zhang, Tao
Wang, Jialin
author_facet Gu, Bingbing
Zhang, Shuai
Fan, Zhe
Che, Jiajing
Li, Shuting
Li, Yunfei
Pan, Keyu
Lv, Jiali
Wang, Cheng
Zhang, Tao
Wang, Jialin
author_sort Gu, Bingbing
collection PubMed
description BACKGROUND: Accumulating evidence suggests that microRNA-target genes are closely related to tumorigenesis and progression. This study aims to screen the intersection of differentially expressed mRNAs (DEmRNAs) and the target genes of differentially expressed microRNAs (DEmiRNAs), and to construct a prognostic gene model of esophageal cancer (EC). METHODS: Gene expression, microRNA expression, somatic mutation, and clinical information data of EC from The Cancer Genome Atlas (TCGA) database were used. The intersection of DEmRNAs and the target genes of DEmiRNAs predicted by the Targetscan database and microRNA Data Integration Portal (mirDIP) database were screened. The screened genes were used to construct a prognostic model of EC. Then, the molecular and immune signatures of these genes were explored. Finally, the GSE53625 dataset from the Gene Expression Omnibus (GEO) database was further used as a validation cohort to confirm the prognostic value of the genes. RESULTS: Six genes on the grounds of the intersection of DEmiRNAs target genes and DEmRNAs were identified as prognostic genes, including ARHGAP11A, H1.4, HMGB3, LRIG1, PRR11, and COL4A1. Based on the median risk score calculated for these genes, EC patients were divided into a high-risk group (n=72) and a low-risk group (n=72). Survival analysis showed that the high-risk group had a significantly shorter survival time than the low-risk group (TCGA and GEO, P<0.001). The nomogram evaluation showed high reliability in predicting the 1-year, 2-year, and 3-year survival probability of EC patients. Compared to low-risk group, higher expression level of M2 macrophages was found in high-risk group of EC patient (P<0.05), while STAT3 checkpoints showed attenuated expression level in high-risk group. CONCLUSIONS: A panel of differential genes was identified as potential EC prognostic biomarkers and showed great clinical significance in EC prognosis.
format Online
Article
Text
id pubmed-10248560
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher AME Publishing Company
record_format MEDLINE/PubMed
spelling pubmed-102485602023-06-09 Prognostic model construction and immune microenvironment analysis of esophageal cancer based on gene expression data and microRNA target genes Gu, Bingbing Zhang, Shuai Fan, Zhe Che, Jiajing Li, Shuting Li, Yunfei Pan, Keyu Lv, Jiali Wang, Cheng Zhang, Tao Wang, Jialin Transl Cancer Res Original Article BACKGROUND: Accumulating evidence suggests that microRNA-target genes are closely related to tumorigenesis and progression. This study aims to screen the intersection of differentially expressed mRNAs (DEmRNAs) and the target genes of differentially expressed microRNAs (DEmiRNAs), and to construct a prognostic gene model of esophageal cancer (EC). METHODS: Gene expression, microRNA expression, somatic mutation, and clinical information data of EC from The Cancer Genome Atlas (TCGA) database were used. The intersection of DEmRNAs and the target genes of DEmiRNAs predicted by the Targetscan database and microRNA Data Integration Portal (mirDIP) database were screened. The screened genes were used to construct a prognostic model of EC. Then, the molecular and immune signatures of these genes were explored. Finally, the GSE53625 dataset from the Gene Expression Omnibus (GEO) database was further used as a validation cohort to confirm the prognostic value of the genes. RESULTS: Six genes on the grounds of the intersection of DEmiRNAs target genes and DEmRNAs were identified as prognostic genes, including ARHGAP11A, H1.4, HMGB3, LRIG1, PRR11, and COL4A1. Based on the median risk score calculated for these genes, EC patients were divided into a high-risk group (n=72) and a low-risk group (n=72). Survival analysis showed that the high-risk group had a significantly shorter survival time than the low-risk group (TCGA and GEO, P<0.001). The nomogram evaluation showed high reliability in predicting the 1-year, 2-year, and 3-year survival probability of EC patients. Compared to low-risk group, higher expression level of M2 macrophages was found in high-risk group of EC patient (P<0.05), while STAT3 checkpoints showed attenuated expression level in high-risk group. CONCLUSIONS: A panel of differential genes was identified as potential EC prognostic biomarkers and showed great clinical significance in EC prognosis. AME Publishing Company 2023-04-13 2023-05-31 /pmc/articles/PMC10248560/ /pubmed/37304542 http://dx.doi.org/10.21037/tcr-22-2588 Text en 2023 Translational Cancer Research. All rights reserved. https://creativecommons.org/licenses/by-nc-nd/4.0/Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0 (https://creativecommons.org/licenses/by-nc-nd/4.0/) .
spellingShingle Original Article
Gu, Bingbing
Zhang, Shuai
Fan, Zhe
Che, Jiajing
Li, Shuting
Li, Yunfei
Pan, Keyu
Lv, Jiali
Wang, Cheng
Zhang, Tao
Wang, Jialin
Prognostic model construction and immune microenvironment analysis of esophageal cancer based on gene expression data and microRNA target genes
title Prognostic model construction and immune microenvironment analysis of esophageal cancer based on gene expression data and microRNA target genes
title_full Prognostic model construction and immune microenvironment analysis of esophageal cancer based on gene expression data and microRNA target genes
title_fullStr Prognostic model construction and immune microenvironment analysis of esophageal cancer based on gene expression data and microRNA target genes
title_full_unstemmed Prognostic model construction and immune microenvironment analysis of esophageal cancer based on gene expression data and microRNA target genes
title_short Prognostic model construction and immune microenvironment analysis of esophageal cancer based on gene expression data and microRNA target genes
title_sort prognostic model construction and immune microenvironment analysis of esophageal cancer based on gene expression data and microrna target genes
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10248560/
https://www.ncbi.nlm.nih.gov/pubmed/37304542
http://dx.doi.org/10.21037/tcr-22-2588
work_keys_str_mv AT gubingbing prognosticmodelconstructionandimmunemicroenvironmentanalysisofesophagealcancerbasedongeneexpressiondataandmicrornatargetgenes
AT zhangshuai prognosticmodelconstructionandimmunemicroenvironmentanalysisofesophagealcancerbasedongeneexpressiondataandmicrornatargetgenes
AT fanzhe prognosticmodelconstructionandimmunemicroenvironmentanalysisofesophagealcancerbasedongeneexpressiondataandmicrornatargetgenes
AT chejiajing prognosticmodelconstructionandimmunemicroenvironmentanalysisofesophagealcancerbasedongeneexpressiondataandmicrornatargetgenes
AT lishuting prognosticmodelconstructionandimmunemicroenvironmentanalysisofesophagealcancerbasedongeneexpressiondataandmicrornatargetgenes
AT liyunfei prognosticmodelconstructionandimmunemicroenvironmentanalysisofesophagealcancerbasedongeneexpressiondataandmicrornatargetgenes
AT pankeyu prognosticmodelconstructionandimmunemicroenvironmentanalysisofesophagealcancerbasedongeneexpressiondataandmicrornatargetgenes
AT lvjiali prognosticmodelconstructionandimmunemicroenvironmentanalysisofesophagealcancerbasedongeneexpressiondataandmicrornatargetgenes
AT wangcheng prognosticmodelconstructionandimmunemicroenvironmentanalysisofesophagealcancerbasedongeneexpressiondataandmicrornatargetgenes
AT zhangtao prognosticmodelconstructionandimmunemicroenvironmentanalysisofesophagealcancerbasedongeneexpressiondataandmicrornatargetgenes
AT wangjialin prognosticmodelconstructionandimmunemicroenvironmentanalysisofesophagealcancerbasedongeneexpressiondataandmicrornatargetgenes