金属离子转运体slc39a14介导的铁凋亡和糖基化调节肿瘤免疫微环境:泛癌症多组学治疗潜力探索。

IF 6 2区 医学 Q1 ONCOLOGY
Yi-Chun Chiang, Chih-Yang Wang, Sachin Kumar, Chung-Bao Hsieh, Kai-Fu Chang, Ching-Chung Ko, Chih-Hsuan Chang, Hui-Ru Lin, Chi-Jen Wu, Chien-Han Yuan, Do Thi Minh Xuan, Juan Lorell Ngadio, Dahlak Daniel Solomon, Fitria Sari Wulandari, Hung-Yun Lin, Shun-Fa Yang, Yung-Kuo Lee
{"title":"金属离子转运体slc39a14介导的铁凋亡和糖基化调节肿瘤免疫微环境:泛癌症多组学治疗潜力探索。","authors":"Yi-Chun Chiang, Chih-Yang Wang, Sachin Kumar, Chung-Bao Hsieh, Kai-Fu Chang, Ching-Chung Ko, Chih-Hsuan Chang, Hui-Ru Lin, Chi-Jen Wu, Chien-Han Yuan, Do Thi Minh Xuan, Juan Lorell Ngadio, Dahlak Daniel Solomon, Fitria Sari Wulandari, Hung-Yun Lin, Shun-Fa Yang, Yung-Kuo Lee","doi":"10.1186/s12935-025-04003-6","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a pivotal mechanism in cancer progression and therapeutic resistance. Concurrently, glycosylation, as a key post-translational modification, plays a critical role in regulating cell signaling, immune evasion, and metastasis. Although both processes are independently implicated in tumor biology, the intersection between ferroptosis and glycosylation remains largely unexplored.</p><p><strong>Methods: </strong>We performed a comprehensive pan-cancer multi-omics analysis, integrating bulk transcriptomics, epigenomics, and single-cell RNA sequencing datasets. Ferroptosis and glycosylation-related genes were curated from The Molecular Signatures Database (MSigDB), leading to the identification of the metal ion transporter, SLC39A14 (solute carrier family 39 member 14), as a key intersecting gene. A ferroptosis-related gene signature was constructed using machine learning and Cox regression models, followed by survival analyses, immune microenvironment profiling, and a pathway enrichment analysis across The Cancer Genome Atlas (TCGA) cohort.</p><p><strong>Results: </strong>SLC39A14 was found to be significantly upregulated across multiple tumor types and associated with a poor prognosis, immune-stromal infiltration, and ferroptosis resistance. Among all cancer types analyzed, glioblastoma multiforme (GBM) and kidney renal cell carcinoma (KIRC) exhibited the significantly prognostic associations and the most pronounced differential expression of SLC39A14. Single-cell analysis revealed that SLC39A14 expression was enriched between stromal and immune populations, hypoxic perivascular niches, confirming its microenvironment-specific functions. These findings were corroborated by DNA methylation data showing promoter hypomethylation of SLC39A14 in tumors compared to normal tissues. Functionally, SLC39A14 was highly enriched in pathways related to angiogenesis, the epithelial-to-mesenchymal transition, cytokine signaling, and oxidative stress adaptation including vascular endothelial growth factor (VEGF) and cell metabolism-related signaling. A nomogram integrating SLC39A14 expression with clinical parameters improved overall survival predictions.</p><p><strong>Conclusions: </strong>In this study, we identified SLC39A14 as a dual regulator at the interface of ferroptosis and glycosylation, with significant impacts on tumor microenvironmental remodeling and therapeutic resistance. By leveraging multi-omics and single-cell transcriptomic data, we establish SLC39A14 as a promising prognostic biomarker and therapeutic target, particularly in brain and kidney cancers where ferroptosis modulation could offer novel clinical strategies.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"363"},"PeriodicalIF":6.0000,"publicationDate":"2025-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Metal ion transporter SLC39A14-mediated ferroptosis and glycosylation modulate the tumor immune microenvironment: pan-cancer multi-omics exploration of therapeutic potential.\",\"authors\":\"Yi-Chun Chiang, Chih-Yang Wang, Sachin Kumar, Chung-Bao Hsieh, Kai-Fu Chang, Ching-Chung Ko, Chih-Hsuan Chang, Hui-Ru Lin, Chi-Jen Wu, Chien-Han Yuan, Do Thi Minh Xuan, Juan Lorell Ngadio, Dahlak Daniel Solomon, Fitria Sari Wulandari, Hung-Yun Lin, Shun-Fa Yang, Yung-Kuo Lee\",\"doi\":\"10.1186/s12935-025-04003-6\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a pivotal mechanism in cancer progression and therapeutic resistance. Concurrently, glycosylation, as a key post-translational modification, plays a critical role in regulating cell signaling, immune evasion, and metastasis. Although both processes are independently implicated in tumor biology, the intersection between ferroptosis and glycosylation remains largely unexplored.</p><p><strong>Methods: </strong>We performed a comprehensive pan-cancer multi-omics analysis, integrating bulk transcriptomics, epigenomics, and single-cell RNA sequencing datasets. Ferroptosis and glycosylation-related genes were curated from The Molecular Signatures Database (MSigDB), leading to the identification of the metal ion transporter, SLC39A14 (solute carrier family 39 member 14), as a key intersecting gene. A ferroptosis-related gene signature was constructed using machine learning and Cox regression models, followed by survival analyses, immune microenvironment profiling, and a pathway enrichment analysis across The Cancer Genome Atlas (TCGA) cohort.</p><p><strong>Results: </strong>SLC39A14 was found to be significantly upregulated across multiple tumor types and associated with a poor prognosis, immune-stromal infiltration, and ferroptosis resistance. Among all cancer types analyzed, glioblastoma multiforme (GBM) and kidney renal cell carcinoma (KIRC) exhibited the significantly prognostic associations and the most pronounced differential expression of SLC39A14. Single-cell analysis revealed that SLC39A14 expression was enriched between stromal and immune populations, hypoxic perivascular niches, confirming its microenvironment-specific functions. These findings were corroborated by DNA methylation data showing promoter hypomethylation of SLC39A14 in tumors compared to normal tissues. Functionally, SLC39A14 was highly enriched in pathways related to angiogenesis, the epithelial-to-mesenchymal transition, cytokine signaling, and oxidative stress adaptation including vascular endothelial growth factor (VEGF) and cell metabolism-related signaling. A nomogram integrating SLC39A14 expression with clinical parameters improved overall survival predictions.</p><p><strong>Conclusions: </strong>In this study, we identified SLC39A14 as a dual regulator at the interface of ferroptosis and glycosylation, with significant impacts on tumor microenvironmental remodeling and therapeutic resistance. By leveraging multi-omics and single-cell transcriptomic data, we establish SLC39A14 as a promising prognostic biomarker and therapeutic target, particularly in brain and kidney cancers where ferroptosis modulation could offer novel clinical strategies.</p>\",\"PeriodicalId\":9385,\"journal\":{\"name\":\"Cancer Cell International\",\"volume\":\"25 1\",\"pages\":\"363\"},\"PeriodicalIF\":6.0000,\"publicationDate\":\"2025-10-21\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Cancer Cell International\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1186/s12935-025-04003-6\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"ONCOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Cell International","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s12935-025-04003-6","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:铁凋亡是一种由脂质过氧化驱动的铁依赖性细胞死亡形式,已成为癌症进展和治疗耐药的关键机制。同时,糖基化作为一种关键的翻译后修饰,在调节细胞信号传导、免疫逃避和转移中起着关键作用。虽然这两个过程都独立地涉及肿瘤生物学,但铁下垂和糖基化之间的交叉仍然在很大程度上未被探索。方法:我们进行了全面的泛癌症多组学分析,整合了大量转录组学、表观基因组学和单细胞RNA测序数据集。从分子特征数据库(MSigDB)中筛选铁中毒和糖基化相关基因,鉴定出金属离子转运体SLC39A14(溶质载体家族39成员14)是一个关键的交叉基因。研究人员利用机器学习和Cox回归模型构建了嗜铁相关基因特征,随后进行了生存分析、免疫微环境分析和跨癌症基因组图谱(TCGA)队列的途径富集分析。结果:SLC39A14在多种肿瘤类型中显著上调,并与预后不良、免疫间质浸润和铁垂抵抗相关。在分析的所有癌症类型中,多形性胶质母细胞瘤(GBM)和肾肾细胞癌(KIRC)表现出显著的预后相关性,SLC39A14的差异表达最为明显。单细胞分析显示,SLC39A14在基质和免疫群体、缺氧血管周围生态位之间表达丰富,证实了其微环境特异性功能。DNA甲基化数据证实了这些发现,显示与正常组织相比,肿瘤中SLC39A14的启动子甲基化程度较低。在功能上,SLC39A14在血管生成、上皮-间质转化、细胞因子信号传导和氧化应激适应(包括血管内皮生长因子(VEGF)和细胞代谢相关信号传导)相关的通路中高度富集。结合SLC39A14表达与临床参数的nomogram改善了总体生存预测。结论:在本研究中,我们鉴定出SLC39A14作为铁死亡和糖基化界面的双重调节因子,对肿瘤微环境重塑和治疗耐药性有显著影响。通过利用多组学和单细胞转录组学数据,我们建立了SLC39A14作为一个有前景的预后生物标志物和治疗靶点,特别是在脑癌和肾癌中,铁死亡调节可以提供新的临床策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Metal ion transporter SLC39A14-mediated ferroptosis and glycosylation modulate the tumor immune microenvironment: pan-cancer multi-omics exploration of therapeutic potential.

Background: Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a pivotal mechanism in cancer progression and therapeutic resistance. Concurrently, glycosylation, as a key post-translational modification, plays a critical role in regulating cell signaling, immune evasion, and metastasis. Although both processes are independently implicated in tumor biology, the intersection between ferroptosis and glycosylation remains largely unexplored.

Methods: We performed a comprehensive pan-cancer multi-omics analysis, integrating bulk transcriptomics, epigenomics, and single-cell RNA sequencing datasets. Ferroptosis and glycosylation-related genes were curated from The Molecular Signatures Database (MSigDB), leading to the identification of the metal ion transporter, SLC39A14 (solute carrier family 39 member 14), as a key intersecting gene. A ferroptosis-related gene signature was constructed using machine learning and Cox regression models, followed by survival analyses, immune microenvironment profiling, and a pathway enrichment analysis across The Cancer Genome Atlas (TCGA) cohort.

Results: SLC39A14 was found to be significantly upregulated across multiple tumor types and associated with a poor prognosis, immune-stromal infiltration, and ferroptosis resistance. Among all cancer types analyzed, glioblastoma multiforme (GBM) and kidney renal cell carcinoma (KIRC) exhibited the significantly prognostic associations and the most pronounced differential expression of SLC39A14. Single-cell analysis revealed that SLC39A14 expression was enriched between stromal and immune populations, hypoxic perivascular niches, confirming its microenvironment-specific functions. These findings were corroborated by DNA methylation data showing promoter hypomethylation of SLC39A14 in tumors compared to normal tissues. Functionally, SLC39A14 was highly enriched in pathways related to angiogenesis, the epithelial-to-mesenchymal transition, cytokine signaling, and oxidative stress adaptation including vascular endothelial growth factor (VEGF) and cell metabolism-related signaling. A nomogram integrating SLC39A14 expression with clinical parameters improved overall survival predictions.

Conclusions: In this study, we identified SLC39A14 as a dual regulator at the interface of ferroptosis and glycosylation, with significant impacts on tumor microenvironmental remodeling and therapeutic resistance. By leveraging multi-omics and single-cell transcriptomic data, we establish SLC39A14 as a promising prognostic biomarker and therapeutic target, particularly in brain and kidney cancers where ferroptosis modulation could offer novel clinical strategies.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
10.90
自引率
1.70%
发文量
360
审稿时长
1 months
期刊介绍: Cancer Cell International publishes articles on all aspects of cancer cell biology, originating largely from, but not limited to, work using cell culture techniques. The journal focuses on novel cancer studies reporting data from biological experiments performed on cells grown in vitro, in two- or three-dimensional systems, and/or in vivo (animal experiments). These types of experiments have provided crucial data in many fields, from cell proliferation and transformation, to epithelial-mesenchymal interaction, to apoptosis, and host immune response to tumors. Cancer Cell International also considers articles that focus on novel technologies or novel pathways in molecular analysis and on epidemiological studies that may affect patient care, as well as articles reporting translational cancer research studies where in vitro discoveries are bridged to the clinic. As such, the journal is interested in laboratory and animal studies reporting on novel biomarkers of tumor progression and response to therapy and on their applicability to human cancers.
×
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