以UQCRFS1为代表的高氧化磷酸化标志着弥漫性大b细胞淋巴瘤中CD8 +肿瘤浸润淋巴细胞的衰竭。

IF 4.9 2区 生物学 Q1 BIOLOGY
Yiming Yang, Yaqi Shu, Zujun Qin, Yi Zeng, Kexin Chen, Xin Liu, Shunhai Jian, Qiqi Zhu
{"title":"以UQCRFS1为代表的高氧化磷酸化标志着弥漫性大b细胞淋巴瘤中CD8 +肿瘤浸润淋巴细胞的衰竭。","authors":"Yiming Yang, Yaqi Shu, Zujun Qin, Yi Zeng, Kexin Chen, Xin Liu, Shunhai Jian, Qiqi Zhu","doi":"10.1186/s13062-025-00684-1","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Metabolic alterations are closely associated with the exhaustion and immune deficiency of CD8<sup>+</sup> tumor-infiltrating lymphocytes (TILs), while little is known about diffuse large B-cell lymphoma (DLBCL). This study aimed to elucidate the significance of the metabolic alterations in exhausted CD8<sup>+</sup>TILs and its underlying regulatory mechanism in DLBCL.</p><p><strong>Methods: </strong>The metabolic alterations in exhausted CD8<sup>+</sup>TILs in DLBCL were evaluated through single-cell RNA sequencing (scRNA-seq). The crucial metabolic pathway and its significance in the biological function of exhausted CD8<sup>+</sup>TILs were investigated by scRNA-seq and RNA sequencing. The marker gene in crucial metabolic pathway, and its correlations between exhaustion status, the tumor microenvironment (TME) composition, clinicopathological characteristics, prognosis, and immune checkpoint blockade (ICB) therapy efficacy were evaluated by scRNA-seq, RNA sequencing, immunohistochemistry, and RT-qPCR. Furthermore, the underlying regulatory mechanism involved in the metabolic alteration related to CD8<sup>+</sup>TILs exhaustion was explored through scRNA-seq, RNA sequencing, and somatic mutation analysis.</p><p><strong>Results: </strong>Our study illustrated the metabolic heterogeneity in CD8<sup>+</sup>TILs, and demonstrated that oxidative phosphorylation (OXPHOS) was the crucial pathway in CD8<sup>+</sup>TILs exhaustion. The high OXPHOS activity indicated the immune deficiency in exhausted CD8<sup>+</sup>TILs, and UQCRFS1 was identified as a marker gene. High UQCRFS1 indicated the immunosuppressive TME, severe clinicopathological characteristics, including activated B-cell-like subtype, high IPI and PS score, advanced stage, dismal prognosis, and resistance to ICB therapy. Furthermore, MYC-related signaling and P2RY8 mutation in DLBCL may regulate the UQCRFS1 expression in exhausted CD8<sup>+</sup>TILs.</p><p><strong>Conclusions: </strong>Our study highlights the importance of OXPHOS activity in CD8<sup>+</sup>TILs exhaustion and suggests its possible regulatory mechanism, which is feasible in clinical evaluation and beneficial for novel immunotherapeutic approaches in DLBCL.</p>","PeriodicalId":9164,"journal":{"name":"Biology Direct","volume":"20 1","pages":"92"},"PeriodicalIF":4.9000,"publicationDate":"2025-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12345122/pdf/","citationCount":"0","resultStr":"{\"title\":\"High oxidative phosphorylation represented by UQCRFS1 marks CD8 + tumor-infiltrating lymphocytes exhaustion in diffuse large B-cell lymphoma.\",\"authors\":\"Yiming Yang, Yaqi Shu, Zujun Qin, Yi Zeng, Kexin Chen, Xin Liu, Shunhai Jian, Qiqi Zhu\",\"doi\":\"10.1186/s13062-025-00684-1\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Metabolic alterations are closely associated with the exhaustion and immune deficiency of CD8<sup>+</sup> tumor-infiltrating lymphocytes (TILs), while little is known about diffuse large B-cell lymphoma (DLBCL). This study aimed to elucidate the significance of the metabolic alterations in exhausted CD8<sup>+</sup>TILs and its underlying regulatory mechanism in DLBCL.</p><p><strong>Methods: </strong>The metabolic alterations in exhausted CD8<sup>+</sup>TILs in DLBCL were evaluated through single-cell RNA sequencing (scRNA-seq). The crucial metabolic pathway and its significance in the biological function of exhausted CD8<sup>+</sup>TILs were investigated by scRNA-seq and RNA sequencing. The marker gene in crucial metabolic pathway, and its correlations between exhaustion status, the tumor microenvironment (TME) composition, clinicopathological characteristics, prognosis, and immune checkpoint blockade (ICB) therapy efficacy were evaluated by scRNA-seq, RNA sequencing, immunohistochemistry, and RT-qPCR. Furthermore, the underlying regulatory mechanism involved in the metabolic alteration related to CD8<sup>+</sup>TILs exhaustion was explored through scRNA-seq, RNA sequencing, and somatic mutation analysis.</p><p><strong>Results: </strong>Our study illustrated the metabolic heterogeneity in CD8<sup>+</sup>TILs, and demonstrated that oxidative phosphorylation (OXPHOS) was the crucial pathway in CD8<sup>+</sup>TILs exhaustion. The high OXPHOS activity indicated the immune deficiency in exhausted CD8<sup>+</sup>TILs, and UQCRFS1 was identified as a marker gene. High UQCRFS1 indicated the immunosuppressive TME, severe clinicopathological characteristics, including activated B-cell-like subtype, high IPI and PS score, advanced stage, dismal prognosis, and resistance to ICB therapy. Furthermore, MYC-related signaling and P2RY8 mutation in DLBCL may regulate the UQCRFS1 expression in exhausted CD8<sup>+</sup>TILs.</p><p><strong>Conclusions: </strong>Our study highlights the importance of OXPHOS activity in CD8<sup>+</sup>TILs exhaustion and suggests its possible regulatory mechanism, which is feasible in clinical evaluation and beneficial for novel immunotherapeutic approaches in DLBCL.</p>\",\"PeriodicalId\":9164,\"journal\":{\"name\":\"Biology Direct\",\"volume\":\"20 1\",\"pages\":\"92\"},\"PeriodicalIF\":4.9000,\"publicationDate\":\"2025-08-13\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12345122/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Biology Direct\",\"FirstCategoryId\":\"99\",\"ListUrlMain\":\"https://doi.org/10.1186/s13062-025-00684-1\",\"RegionNum\":2,\"RegionCategory\":\"生物学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"BIOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Biology Direct","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1186/s13062-025-00684-1","RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"BIOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:代谢改变与CD8+肿瘤浸润淋巴细胞(til)耗竭和免疫缺陷密切相关,而对弥漫性大b细胞淋巴瘤(DLBCL)知之甚少。本研究旨在阐明耗尽的CD8+TILs代谢改变在DLBCL中的意义及其潜在的调节机制。方法:采用单细胞RNA测序(scRNA-seq)技术评价DLBCL中耗竭CD8+TILs的代谢变化。通过scRNA-seq和RNA测序研究了耗尽CD8+TILs的关键代谢途径及其在生物学功能中的意义。通过scRNA-seq、RNA测序、免疫组织化学和RT-qPCR评估关键代谢途径标记基因及其与衰竭状态、肿瘤微环境(TME)组成、临床病理特征、预后和免疫检查点阻断(ICB)治疗效果的相关性。此外,通过scRNA-seq、RNA测序和体细胞突变分析,探讨了与CD8+TILs耗竭相关的代谢改变的潜在调控机制。结果:我们的研究揭示了CD8+TILs的代谢异质性,并证明氧化磷酸化(OXPHOS)是CD8+TILs耗竭的关键途径。高OXPHOS活性提示耗尽的CD8+TILs存在免疫缺陷,UQCRFS1被确定为标记基因。高UQCRFS1提示TME免疫抑制,临床病理特征严重,包括活化的b细胞样亚型,IPI和PS评分高,晚期,预后差,对ICB治疗耐药。此外,DLBCL中myc相关信号通路和P2RY8突变可能调节枯竭CD8+TILs中UQCRFS1的表达。结论:我们的研究突出了OXPHOS活性在CD8+TILs耗竭中的重要性,并提示了其可能的调节机制,这在临床评估中是可行的,并有利于DLBCL的新型免疫治疗方法。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
High oxidative phosphorylation represented by UQCRFS1 marks CD8 + tumor-infiltrating lymphocytes exhaustion in diffuse large B-cell lymphoma.

Background: Metabolic alterations are closely associated with the exhaustion and immune deficiency of CD8+ tumor-infiltrating lymphocytes (TILs), while little is known about diffuse large B-cell lymphoma (DLBCL). This study aimed to elucidate the significance of the metabolic alterations in exhausted CD8+TILs and its underlying regulatory mechanism in DLBCL.

Methods: The metabolic alterations in exhausted CD8+TILs in DLBCL were evaluated through single-cell RNA sequencing (scRNA-seq). The crucial metabolic pathway and its significance in the biological function of exhausted CD8+TILs were investigated by scRNA-seq and RNA sequencing. The marker gene in crucial metabolic pathway, and its correlations between exhaustion status, the tumor microenvironment (TME) composition, clinicopathological characteristics, prognosis, and immune checkpoint blockade (ICB) therapy efficacy were evaluated by scRNA-seq, RNA sequencing, immunohistochemistry, and RT-qPCR. Furthermore, the underlying regulatory mechanism involved in the metabolic alteration related to CD8+TILs exhaustion was explored through scRNA-seq, RNA sequencing, and somatic mutation analysis.

Results: Our study illustrated the metabolic heterogeneity in CD8+TILs, and demonstrated that oxidative phosphorylation (OXPHOS) was the crucial pathway in CD8+TILs exhaustion. The high OXPHOS activity indicated the immune deficiency in exhausted CD8+TILs, and UQCRFS1 was identified as a marker gene. High UQCRFS1 indicated the immunosuppressive TME, severe clinicopathological characteristics, including activated B-cell-like subtype, high IPI and PS score, advanced stage, dismal prognosis, and resistance to ICB therapy. Furthermore, MYC-related signaling and P2RY8 mutation in DLBCL may regulate the UQCRFS1 expression in exhausted CD8+TILs.

Conclusions: Our study highlights the importance of OXPHOS activity in CD8+TILs exhaustion and suggests its possible regulatory mechanism, which is feasible in clinical evaluation and beneficial for novel immunotherapeutic approaches in DLBCL.

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
Biology Direct
Biology Direct 生物-生物学
CiteScore
6.40
自引率
10.90%
发文量
32
审稿时长
7 months
期刊介绍: Biology Direct serves the life science research community as an open access, peer-reviewed online journal, providing authors and readers with an alternative to the traditional model of peer review. Biology Direct considers original research articles, hypotheses, comments, discovery notes and reviews in subject areas currently identified as those most conducive to the open review approach, primarily those with a significant non-experimental component.
×
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