UBR1 is a prognostic biomarker and therapeutic target associated with immune cell infiltration in gastric cancer.

IF 3.9 3区 医学 Q2 CELL BIOLOGY Aging-Us Pub Date : 2024-08-23 DOI:10.18632/aging.206079
Weiwei Yuan, Jianye Han, Chen Chen, Yue Qiu, Yuanmin Xu, Yang Huang, Zhangming Chen, Aman Xu, Minzhi Sun
{"title":"UBR1 is a prognostic biomarker and therapeutic target associated with immune cell infiltration in gastric cancer.","authors":"Weiwei Yuan, Jianye Han, Chen Chen, Yue Qiu, Yuanmin Xu, Yang Huang, Zhangming Chen, Aman Xu, Minzhi Sun","doi":"10.18632/aging.206079","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Ubiquitination is a targeted protein modification process mediated by intracellular molecules. UBR1 encodes a protein that binds to unstable N-terminal residues of substrate proteins and contributes to the formation of substrate-linked polyubiquitin chains. However, the function and cellular pathways of UBR1 in tumors have received inadequate attention. This study aimed to investigate the potential of UBR1 as a prognostic biomarker and immunotherapy target for stomach adenocarcinoma (STAD) as well as its biological function and molecular mechanism in relation to the disease.</p><p><strong>Methods: </strong>Differential expression and pan-cancer gene set enrichment analysis (GSEA) were conducted using The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and Genotype-Tissue Expression (GTEx) datasets. The Human Protein Atlas (HPA) database was utilized to identify UBR1-enriched pathways in AGS cells and to compare immunohistochemical differences between cancerous and adjacent non-cancerous tissues in gastric cancer. Quantitative Polymerase Chain Reaction (QPCR) and Western blot (WB) analyses were employed to validate these findings in both cancerous and adjacent non-cancerous tissues of gastric cancer. UBR1 expression in GES-1 and four gastric cancer cell lines was assessed using QPCR and WB. Kaplan-Meier curves, univariate and multivariate Cox regression analyses, and receiver operating characteristic (ROC) curve analyses were performed to evaluate the prognostic and diagnostic roles of UBR1. Additionally, the correlation between UBR1 expression and clinical parameters was analyzed using TCGA and GEO databases. UBR1 mutation data were obtained from the cBioPortal database. The mutation landscape, mutation-associated genes, protein structure, tumor mutation burden (TMB), and microsatellite instability (MSI) correlations were analyzed and illustrated. The biological functions of UBR1 were investigated using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. The correlation between UBR1 and immune infiltration was assessed using TIMER and EPIC computational methods. Protein expression levels of UBR1 in gastric cancer cell lines were determined by immunohistochemistry (IHC) and WB analysis. Quantitative real-time PCR (qRT-PCR) was employed to analyze mRNA expression. Immunoprecipitation (IP) assays were conducted to detect protein-protein interactions between UBR1 and PDL1, while cellular immunofluorescence was used to observe the co-localization of these proteins. Cell proliferation was evaluated using CCK8 and colony formation assays. Cell migration was assessed using Transwell and wound healing assays. Finally, apoptosis was analyzed using flow cytometry, and WB was used to detect changes in apoptotic proteins and NF-κB P65 pathway proteins.</p><p><strong>Results: </strong>UBR1 was upregulated in 28 cancer types, including STAD, and its overexpression was validated in gastric cancer cell lines and tissues. UBR1 expression was associated with advanced pathological characteristics. High UBR1 expression was linked to poor prognostic outcomes, including overall survival (OS), progression-free interval (PFI), disease-specific survival (DSS), as well as responses to surgery, chemotherapy, and HER2 expression. UBR1 expression showed significant correlations with clinical parameters such as age, gender, TNM stage, pathological stage, tumor resection, and anti-reflux therapy. Amplifications and deletions were the most frequent genetic alterations associated with UBR1. According to KEGG and GSEA analyses, UBR1 was significantly associated with several cancer pathways, oxidative phosphorylation, and the TNF-NFκB pathway. UBR1 also exhibited a significant correlation with immune cell infiltration and immunotherapy, including a direct interaction with PDL1. Knockdown of UBR1 inhibited the proliferation, migration, and invasion of STAD cells and promoted apoptosis.</p><p><strong>Conclusions: </strong>UBR1 is overexpressed in STAD, promoting its progression and positively correlating with immune cell infiltration and immunotherapeutic responses. Therefore, UBR1 could be a promising biomarker for the prognosis and immunotherapy of STAD.</p>","PeriodicalId":55547,"journal":{"name":"Aging-Us","volume":null,"pages":null},"PeriodicalIF":3.9000,"publicationDate":"2024-08-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11386912/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Aging-Us","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.18632/aging.206079","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Ubiquitination is a targeted protein modification process mediated by intracellular molecules. UBR1 encodes a protein that binds to unstable N-terminal residues of substrate proteins and contributes to the formation of substrate-linked polyubiquitin chains. However, the function and cellular pathways of UBR1 in tumors have received inadequate attention. This study aimed to investigate the potential of UBR1 as a prognostic biomarker and immunotherapy target for stomach adenocarcinoma (STAD) as well as its biological function and molecular mechanism in relation to the disease.

Methods: Differential expression and pan-cancer gene set enrichment analysis (GSEA) were conducted using The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and Genotype-Tissue Expression (GTEx) datasets. The Human Protein Atlas (HPA) database was utilized to identify UBR1-enriched pathways in AGS cells and to compare immunohistochemical differences between cancerous and adjacent non-cancerous tissues in gastric cancer. Quantitative Polymerase Chain Reaction (QPCR) and Western blot (WB) analyses were employed to validate these findings in both cancerous and adjacent non-cancerous tissues of gastric cancer. UBR1 expression in GES-1 and four gastric cancer cell lines was assessed using QPCR and WB. Kaplan-Meier curves, univariate and multivariate Cox regression analyses, and receiver operating characteristic (ROC) curve analyses were performed to evaluate the prognostic and diagnostic roles of UBR1. Additionally, the correlation between UBR1 expression and clinical parameters was analyzed using TCGA and GEO databases. UBR1 mutation data were obtained from the cBioPortal database. The mutation landscape, mutation-associated genes, protein structure, tumor mutation burden (TMB), and microsatellite instability (MSI) correlations were analyzed and illustrated. The biological functions of UBR1 were investigated using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. The correlation between UBR1 and immune infiltration was assessed using TIMER and EPIC computational methods. Protein expression levels of UBR1 in gastric cancer cell lines were determined by immunohistochemistry (IHC) and WB analysis. Quantitative real-time PCR (qRT-PCR) was employed to analyze mRNA expression. Immunoprecipitation (IP) assays were conducted to detect protein-protein interactions between UBR1 and PDL1, while cellular immunofluorescence was used to observe the co-localization of these proteins. Cell proliferation was evaluated using CCK8 and colony formation assays. Cell migration was assessed using Transwell and wound healing assays. Finally, apoptosis was analyzed using flow cytometry, and WB was used to detect changes in apoptotic proteins and NF-κB P65 pathway proteins.

Results: UBR1 was upregulated in 28 cancer types, including STAD, and its overexpression was validated in gastric cancer cell lines and tissues. UBR1 expression was associated with advanced pathological characteristics. High UBR1 expression was linked to poor prognostic outcomes, including overall survival (OS), progression-free interval (PFI), disease-specific survival (DSS), as well as responses to surgery, chemotherapy, and HER2 expression. UBR1 expression showed significant correlations with clinical parameters such as age, gender, TNM stage, pathological stage, tumor resection, and anti-reflux therapy. Amplifications and deletions were the most frequent genetic alterations associated with UBR1. According to KEGG and GSEA analyses, UBR1 was significantly associated with several cancer pathways, oxidative phosphorylation, and the TNF-NFκB pathway. UBR1 also exhibited a significant correlation with immune cell infiltration and immunotherapy, including a direct interaction with PDL1. Knockdown of UBR1 inhibited the proliferation, migration, and invasion of STAD cells and promoted apoptosis.

Conclusions: UBR1 is overexpressed in STAD, promoting its progression and positively correlating with immune cell infiltration and immunotherapeutic responses. Therefore, UBR1 could be a promising biomarker for the prognosis and immunotherapy of STAD.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
UBR1 是与胃癌免疫细胞浸润相关的预后生物标志物和治疗靶点。
背景:泛素化是一种由细胞内分子介导的靶向蛋白质修饰过程。UBR1 编码的蛋白能与底物蛋白不稳定的 N 端残基结合,并促进底物连接的多泛素链的形成。然而,UBR1 在肿瘤中的功能和细胞通路并未受到足够重视。本研究旨在探讨UBR1作为胃腺癌(STAD)预后生物标志物和免疫治疗靶点的潜力及其生物学功能和分子机制:方法:利用癌症基因组图谱(TCGA)、基因表达总库(GEO)和基因型-组织表达(GTEx)数据集进行差异表达和泛癌症基因组富集分析(GSEA)。人类蛋白质图谱(HPA)数据库用于鉴定AGS细胞中富集UBR1的通路,并比较胃癌癌组织与邻近非癌组织之间的免疫组化差异。定量聚合酶链反应(QPCR)和免疫印迹(WB)分析被用来验证这些在胃癌癌组织和邻近非癌组织中的发现。利用 QPCR 和 WB 评估了 UBR1 在 GES-1 和四种胃癌细胞系中的表达。对 UBR1 的预后和诊断作用进行了 Kaplan-Meier 曲线、单变量和多变量 Cox 回归分析以及接收器操作特征曲线分析。此外,还利用 TCGA 和 GEO 数据库分析了 UBR1 表达与临床参数之间的相关性。UBR1 基因突变数据来自 cBioPortal 数据库。分析并说明了突变情况、突变相关基因、蛋白质结构、肿瘤突变负荷(TMB)和微卫星不稳定性(MSI)的相关性。利用基因本体(GO)和京都基因组百科全书(KEGG)富集分析研究了UBR1的生物学功能。利用TIMER和EPIC计算方法评估了UBR1与免疫浸润之间的相关性。通过免疫组化(IHC)和WB分析确定了UBR1在胃癌细胞系中的蛋白表达水平。定量实时 PCR(qRT-PCR)用于分析 mRNA 表达。免疫沉淀(IP)试验用于检测 UBR1 和 PDL1 之间的蛋白相互作用,细胞免疫荧光用于观察这些蛋白的共定位。细胞增殖采用 CCK8 和集落形成试验进行评估。使用 Transwell 和伤口愈合试验评估细胞迁移。最后,使用流式细胞术分析细胞凋亡,并使用 WB 检测凋亡蛋白和 NF-κB P65 通路蛋白的变化:结果:UBR1在包括STAD在内的28种癌症中上调,其在胃癌细胞系和组织中的过表达得到了验证。UBR1 的表达与晚期病理特征相关。UBR1 的高表达与预后不良有关,包括总生存期(OS)、无进展间期(PFI)、疾病特异性生存期(DSS)以及对手术、化疗和 HER2 表达的反应。UBR1的表达与年龄、性别、TNM分期、病理分期、肿瘤切除术和抗反流治疗等临床参数有明显的相关性。扩增和缺失是与UBR1相关的最常见基因改变。根据KEGG和GSEA分析,UBR1与几种癌症通路、氧化磷酸化和TNF-NFκB通路显著相关。UBR1 还与免疫细胞浸润和免疫疗法有明显的相关性,包括与 PDL1 的直接相互作用。敲除 UBR1 可抑制 STAD 细胞的增殖、迁移和侵袭,并促进细胞凋亡:结论:UBR1 在 STAD 中过表达,促进了 STAD 的进展,并与免疫细胞浸润和免疫治疗反应呈正相关。因此,UBR1可能是STAD预后和免疫治疗的一个有前途的生物标志物。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
Aging-Us
Aging-Us CELL BIOLOGY-
CiteScore
10.00
自引率
0.00%
发文量
595
审稿时长
6-12 weeks
期刊介绍: Information not localized
期刊最新文献
Correction for: miR-926-3p influences myocardial injury in septic mice through regulation of mTOR signaling pathway by targeting TSC1. Retraction of: Sulfated polysaccharide of Sepiella maindroni ink targets Akt and overcomes resistance to the FGFR inhibitor AZD4547 in bladder cancer. DDIT3 switches osteogenic potential of BMP9 to lipogenic by attenuating Wnt/β-catenin signaling via up-regulating DKK1 in mesenchymal stem cells. Frailty and pre-frailty associated with long-term diminished physical performance and quality of life in breast cancer and hematopoietic cell transplant survivors. Revealing a cancer-associated fibroblast-based risk signature for pancreatic adenocarcinoma through single-cell and bulk RNA-seq analysis.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1