Metabolism-associated marker gene-based predictive model for prognosis, targeted therapy, and immune landscape in ovarian cancer: an integrative analysis of single-cell and bulk RNA sequencing with spatial transcriptomics.

IF 2.4 3区 医学 Q2 OBSTETRICS & GYNECOLOGY
Lele Ling, Bingrong Li, Boliang Ke, Yinjie Hu, Kaiyong Zhang, Siwen Li, Te Liu, Peng Liu, Bimeng Zhang
{"title":"Metabolism-associated marker gene-based predictive model for prognosis, targeted therapy, and immune landscape in ovarian cancer: an integrative analysis of single-cell and bulk RNA sequencing with spatial transcriptomics.","authors":"Lele Ling, Bingrong Li, Boliang Ke, Yinjie Hu, Kaiyong Zhang, Siwen Li, Te Liu, Peng Liu, Bimeng Zhang","doi":"10.1186/s12905-025-03750-y","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Ovarian cancer (OC) is a formidable gynecological tumor marked with the highest mortality rate. The lack of effective biomarkers and treatment drugs places a substantial proportion of patients with OC at significant risk of mortality, primarily due to metastasis. Glycolysis metabolism, lipid metabolism, choline metabolism, and sphingolipid metabolism are closely intertwined with the occurrence and progression of OC. Thus, it is of utmost significance to identify potent prognostic biomarkers and delve into the exploration of novel therapeutic drugs and targets, in pursuit of advancing the treatment of OC.</p><p><strong>Methods: </strong>Single-cell RNA sequencing (scRNA-seq) data related to OC were analyzed using AUCell scores to identify subpopulations at the single-cell level. The \"AddModuleScore\" function of the \"Seurat\" package was adopted to score and select marker genes from four gene sets: glycolysis metabolism, lipid metabolism, choline metabolism, and sphingolipid metabolism. A prognostic model for metabolism-related genes (MRGs) was constructed and validated using OC-related marker genes selected from bulk RNAseq data. The MRG-based prognostic model was further utilized for functional analysis of the model gene set, pan-cancer analysis of genomic variations, spatial transcriptomics analysis, as well as GO and KEGG enrichment analysis. CIBERSORT and ESTIMATE algorithms were utilized for assessing the immune microenvironment of TCGA-ovarian serous cystadenocarcinoma (OV) samples. Furthermore, the Tracking Tumor Immunophenotype (TIP) database was employed to examine the anti-cancer immune response in patients with OC. To gain a more in-depth understanding of the process, the frequency of somatic mutations and different types of mutated genes were visualized through the somatic mutation profile of the TCGA database. Moreover, the benefits of immune checkpoint inhibitor (ICI) therapy in individuals with OC were predicted in the TIDE database. In addition, the CMap database was used to predict small-molecule drugs for the treatment of OC. Furthermore, immunohistochemistry, RT-qPCR, CCK-8, Transwell assay, and in vivo tumor xenograft experiments were conducted to validate the prognostic ability of the MRG Triggering Receptor Expressed on Myeloid Cells-1 (TREM1) in OC.</p><p><strong>Results: </strong>Monocytes were selected using AUCell scoring, and two subpopulations of monocytes, marked by the expression of C1QC<sup>+</sup> tumor-associated macrophages (TAMs) and FCN1<sup>+</sup> resident tissue macrophages (RTMs), were identified as marker genes for OC. Subsequently, a prognostic model consisting of 12 MRGs was constructed and validated. Genomic exploration of the prognostic model unveiled an array of biological functions linked with metabolism. Furthermore, copy number variation (CNV), mRNA expression, single nucleotide variation (SNV), and methylation were significantly different across diverse tumors. Analysis of the TIP database demonstrated that the low-risk group, as determined by the MRG-based prognostic model, exhibited significantly higher anti-cancer immune activity relative to the high-risk group. Furthermore, predictions from the TIDE database revealed that individuals in the high-risk group were more prone to immune evasion when treated with ICIs. The resulting data identified candesartan and PD-123319 as potential therapeutic drugs for OC, possibly acting on the target ATGR2. In vitro and in vivo experiments elucidated that the targeted downregulation of TREM1 effectively inhibited the proliferation and migration of OC cells.</p><p><strong>Conclusion: </strong>The MRG-based prognostic model constructed through the combined analysis of glycolysis metabolism, lipid metabolism, choline metabolism, and sphingolipid metabolism is potentially effective as a prognostic biomarker. Furthermore, candesartan and PD-123319 may be potential therapeutic drugs for OC, possibly acting on the target ATGR2.</p>","PeriodicalId":9204,"journal":{"name":"BMC Women's Health","volume":"25 1","pages":"233"},"PeriodicalIF":2.4000,"publicationDate":"2025-05-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12084907/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"BMC Women's Health","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s12905-025-03750-y","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"OBSTETRICS & GYNECOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Ovarian cancer (OC) is a formidable gynecological tumor marked with the highest mortality rate. The lack of effective biomarkers and treatment drugs places a substantial proportion of patients with OC at significant risk of mortality, primarily due to metastasis. Glycolysis metabolism, lipid metabolism, choline metabolism, and sphingolipid metabolism are closely intertwined with the occurrence and progression of OC. Thus, it is of utmost significance to identify potent prognostic biomarkers and delve into the exploration of novel therapeutic drugs and targets, in pursuit of advancing the treatment of OC.

Methods: Single-cell RNA sequencing (scRNA-seq) data related to OC were analyzed using AUCell scores to identify subpopulations at the single-cell level. The "AddModuleScore" function of the "Seurat" package was adopted to score and select marker genes from four gene sets: glycolysis metabolism, lipid metabolism, choline metabolism, and sphingolipid metabolism. A prognostic model for metabolism-related genes (MRGs) was constructed and validated using OC-related marker genes selected from bulk RNAseq data. The MRG-based prognostic model was further utilized for functional analysis of the model gene set, pan-cancer analysis of genomic variations, spatial transcriptomics analysis, as well as GO and KEGG enrichment analysis. CIBERSORT and ESTIMATE algorithms were utilized for assessing the immune microenvironment of TCGA-ovarian serous cystadenocarcinoma (OV) samples. Furthermore, the Tracking Tumor Immunophenotype (TIP) database was employed to examine the anti-cancer immune response in patients with OC. To gain a more in-depth understanding of the process, the frequency of somatic mutations and different types of mutated genes were visualized through the somatic mutation profile of the TCGA database. Moreover, the benefits of immune checkpoint inhibitor (ICI) therapy in individuals with OC were predicted in the TIDE database. In addition, the CMap database was used to predict small-molecule drugs for the treatment of OC. Furthermore, immunohistochemistry, RT-qPCR, CCK-8, Transwell assay, and in vivo tumor xenograft experiments were conducted to validate the prognostic ability of the MRG Triggering Receptor Expressed on Myeloid Cells-1 (TREM1) in OC.

Results: Monocytes were selected using AUCell scoring, and two subpopulations of monocytes, marked by the expression of C1QC+ tumor-associated macrophages (TAMs) and FCN1+ resident tissue macrophages (RTMs), were identified as marker genes for OC. Subsequently, a prognostic model consisting of 12 MRGs was constructed and validated. Genomic exploration of the prognostic model unveiled an array of biological functions linked with metabolism. Furthermore, copy number variation (CNV), mRNA expression, single nucleotide variation (SNV), and methylation were significantly different across diverse tumors. Analysis of the TIP database demonstrated that the low-risk group, as determined by the MRG-based prognostic model, exhibited significantly higher anti-cancer immune activity relative to the high-risk group. Furthermore, predictions from the TIDE database revealed that individuals in the high-risk group were more prone to immune evasion when treated with ICIs. The resulting data identified candesartan and PD-123319 as potential therapeutic drugs for OC, possibly acting on the target ATGR2. In vitro and in vivo experiments elucidated that the targeted downregulation of TREM1 effectively inhibited the proliferation and migration of OC cells.

Conclusion: The MRG-based prognostic model constructed through the combined analysis of glycolysis metabolism, lipid metabolism, choline metabolism, and sphingolipid metabolism is potentially effective as a prognostic biomarker. Furthermore, candesartan and PD-123319 may be potential therapeutic drugs for OC, possibly acting on the target ATGR2.

基于代谢相关标记基因的卵巢癌预后、靶向治疗和免疫景观预测模型:单细胞和大量RNA测序与空间转录组学的综合分析
背景:卵巢癌是一种死亡率最高的妇科恶性肿瘤。由于缺乏有效的生物标志物和治疗药物,很大一部分OC患者主要由于转移而面临严重的死亡风险。糖酵解代谢、脂质代谢、胆碱代谢、鞘脂代谢与OC的发生发展密切相关。因此,寻找有效的预后生物标志物,深入探索新的治疗药物和靶点,对推进OC的治疗具有重要意义。方法:使用AUCell评分分析与OC相关的单细胞RNA测序(scRNA-seq)数据,以确定单细胞水平的亚群。采用“Seurat”软件包的“AddModuleScore”功能,从糖酵解代谢、脂质代谢、胆碱代谢和鞘脂代谢四个基因组中对标记基因进行评分和选择。利用从大量RNAseq数据中选择的oc相关标记基因,构建并验证了代谢相关基因(MRGs)的预后模型。基于核磁共振成像的预后模型进一步用于模型基因集的功能分析、基因组变异的泛癌分析、空间转录组学分析以及GO和KEGG富集分析。采用CIBERSORT和ESTIMATE算法评估tcga -卵巢浆液性囊腺癌(OV)样本的免疫微环境。此外,采用肿瘤免疫表型追踪(TIP)数据库检测OC患者的抗癌免疫反应。为了更深入地了解这一过程,通过TCGA数据库的体细胞突变谱,可视化了体细胞突变的频率和不同类型的突变基因。此外,在TIDE数据库中预测了免疫检查点抑制剂(ICI)治疗对OC患者的益处。此外,CMap数据库用于预测治疗OC的小分子药物。此外,通过免疫组织化学、RT-qPCR、CCK-8、Transwell实验和体内肿瘤异种移植实验来验证骨髓细胞上表达的MRG触发受体-1 (TREM1)在OC中的预后能力。结果:使用AUCell评分法选择单核细胞,鉴定出C1QC+肿瘤相关巨噬细胞(tam)和FCN1+常驻组织巨噬细胞(RTMs)表达的两个单核细胞亚群作为OC的标记基因。随后,构建并验证了由12个mrg组成的预后模型。对预后模型的基因组探索揭示了一系列与代谢相关的生物学功能。此外,拷贝数变异(CNV)、mRNA表达、单核苷酸变异(SNV)和甲基化在不同肿瘤中存在显著差异。TIP数据库的分析表明,低风险组,根据基于核磁共振成像的预后模型,相对于高风险组,表现出更高的抗癌免疫活性。此外,TIDE数据库的预测显示,高风险群体的个体在接受ICIs治疗时更容易免疫逃避。结果数据确定坎地沙坦和PD-123319是潜在的OC治疗药物,可能作用于靶点ATGR2。体外和体内实验表明,靶向下调TREM1可有效抑制OC细胞的增殖和迁移。结论:通过糖酵解代谢、脂质代谢、胆碱代谢和鞘脂代谢的联合分析构建的基于mri的预后模型是一种潜在有效的预后生物标志物。此外,坎地沙坦和PD-123319可能是潜在的OC治疗药物,可能作用于靶点ATGR2。
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 求助全文
来源期刊
BMC Women's Health
BMC Women's Health OBSTETRICS & GYNECOLOGY-
CiteScore
3.40
自引率
4.00%
发文量
444
审稿时长
>12 weeks
期刊介绍: BMC Women''s Health is an open access, peer-reviewed journal that considers articles on all aspects of the health and wellbeing of adolescent girls and women, with a particular focus on the physical, mental, and emotional health of women in developed and developing nations. The journal welcomes submissions on women''s public health issues, health behaviours, breast cancer, gynecological diseases, mental health and health promotion.
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信