肌红蛋白表达改善t细胞代谢和抗肿瘤效应功能。

IF 10.6 1区 医学 Q1 IMMUNOLOGY
Julia Werner, Haifeng C Xu, Georgios Theodorakis, Ichiro Katahira, Mitrajit Ghosh, Michal Gorzkiewicz, Luisa de Sousa Santos, Ann Kathrin Bergmann, Max Anstötz, Anne Busch, Diran Herebian, Sascha Dietrich, Carsten Berndt, Ertan Mayatepek, Aleksandra A Pandyra, Dirk Brenner, Philipp A Lang
{"title":"肌红蛋白表达改善t细胞代谢和抗肿瘤效应功能。","authors":"Julia Werner, Haifeng C Xu, Georgios Theodorakis, Ichiro Katahira, Mitrajit Ghosh, Michal Gorzkiewicz, Luisa de Sousa Santos, Ann Kathrin Bergmann, Max Anstötz, Anne Busch, Diran Herebian, Sascha Dietrich, Carsten Berndt, Ertan Mayatepek, Aleksandra A Pandyra, Dirk Brenner, Philipp A Lang","doi":"10.1136/jitc-2025-011503","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>The tumor microenvironment is frequently hypoxic and characterized by a scarcity of nutritional resources including a shortage of glucose. As effector T cells have high energy demands, tumor metabolism can contribute to T-cell dysfunction and exhaustion.</p><p><strong>Methods: </strong>In this study, we determined hypoxia in spleen and tumor tissue from tumor-bearing C57BL/6J mice using reverse transcription polymerase chain reaction (RT-PCR), histology and flow cytometry. Next, CD8<sup>+</sup> T cells isolated from C57BL6J mice or P14<sup>+</sup> mice were transduced with Thy1.1 (Control) or Thy1.1-Myoglobin (Mb) packaged retrovirus. Expression of Mb was confirmed with RT-PCR and western blot. Cellular metabolism was determined by flow cytometry, transmission electron microscopy, focused ion beam scanning electron microscopy, Seahorse, metabolomics and luminescence assays. Mb expressing or control P14<sup>+</sup> or OT-I<sup>+</sup> T cells were transferred in B16F10-gp33 or MC38-ova tumor-bearing mice respectively and analyzed using flow cytometry and histology. B16F10-gp33 tumor-bearing mice were additionally treated with anti-programmed cell death protein-1 (PD-1) checkpoint inhibitor.</p><p><strong>Results: </strong>Here we demonstrate that expression of the oxygen-binding protein myoglobin in T cells can boost their mitochondrial and glycolytic metabolic functions. Metabolites and tricarboxylic acid compounds were highly increased in the presence of myoglobin (Mb), which was associated with increased ATP levels. Mb-expressing T cells exhibited low expression of hypoxia-inducible factor-1α after activation and during infiltration into the tumor microenvironment (TME). Accordingly, Mb expression increased effector T-cell function against tumor cells in vitro with concomitant reductions in superoxide levels. Following adoptive transfer into tumor-bearing mice, Mb expression facilitated increased infiltration into the TME. Although T cells expressing Mb exhibited increased expression of effector cytokines, PD-1 was still detected and targetable by anti-PD-1 monoclonal antibodies, which in combination with transfer of Mb-expressing T cells demonstrated maximal efficacy in delaying tumor growth.</p><p><strong>Conclusion: </strong>Taken together, we show that expression of Mb in T cells can increase their metabolism, infiltration into the tumor tissue, and effector function against cancer cells.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 6","pages":""},"PeriodicalIF":10.6000,"publicationDate":"2025-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12142173/pdf/","citationCount":"0","resultStr":"{\"title\":\"Myoglobin expression improves T-cell metabolism and antitumor effector function.\",\"authors\":\"Julia Werner, Haifeng C Xu, Georgios Theodorakis, Ichiro Katahira, Mitrajit Ghosh, Michal Gorzkiewicz, Luisa de Sousa Santos, Ann Kathrin Bergmann, Max Anstötz, Anne Busch, Diran Herebian, Sascha Dietrich, Carsten Berndt, Ertan Mayatepek, Aleksandra A Pandyra, Dirk Brenner, Philipp A Lang\",\"doi\":\"10.1136/jitc-2025-011503\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>The tumor microenvironment is frequently hypoxic and characterized by a scarcity of nutritional resources including a shortage of glucose. As effector T cells have high energy demands, tumor metabolism can contribute to T-cell dysfunction and exhaustion.</p><p><strong>Methods: </strong>In this study, we determined hypoxia in spleen and tumor tissue from tumor-bearing C57BL/6J mice using reverse transcription polymerase chain reaction (RT-PCR), histology and flow cytometry. Next, CD8<sup>+</sup> T cells isolated from C57BL6J mice or P14<sup>+</sup> mice were transduced with Thy1.1 (Control) or Thy1.1-Myoglobin (Mb) packaged retrovirus. Expression of Mb was confirmed with RT-PCR and western blot. Cellular metabolism was determined by flow cytometry, transmission electron microscopy, focused ion beam scanning electron microscopy, Seahorse, metabolomics and luminescence assays. Mb expressing or control P14<sup>+</sup> or OT-I<sup>+</sup> T cells were transferred in B16F10-gp33 or MC38-ova tumor-bearing mice respectively and analyzed using flow cytometry and histology. B16F10-gp33 tumor-bearing mice were additionally treated with anti-programmed cell death protein-1 (PD-1) checkpoint inhibitor.</p><p><strong>Results: </strong>Here we demonstrate that expression of the oxygen-binding protein myoglobin in T cells can boost their mitochondrial and glycolytic metabolic functions. Metabolites and tricarboxylic acid compounds were highly increased in the presence of myoglobin (Mb), which was associated with increased ATP levels. Mb-expressing T cells exhibited low expression of hypoxia-inducible factor-1α after activation and during infiltration into the tumor microenvironment (TME). Accordingly, Mb expression increased effector T-cell function against tumor cells in vitro with concomitant reductions in superoxide levels. Following adoptive transfer into tumor-bearing mice, Mb expression facilitated increased infiltration into the TME. Although T cells expressing Mb exhibited increased expression of effector cytokines, PD-1 was still detected and targetable by anti-PD-1 monoclonal antibodies, which in combination with transfer of Mb-expressing T cells demonstrated maximal efficacy in delaying tumor growth.</p><p><strong>Conclusion: </strong>Taken together, we show that expression of Mb in T cells can increase their metabolism, infiltration into the tumor tissue, and effector function against cancer cells.</p>\",\"PeriodicalId\":14820,\"journal\":{\"name\":\"Journal for Immunotherapy of Cancer\",\"volume\":\"13 6\",\"pages\":\"\"},\"PeriodicalIF\":10.6000,\"publicationDate\":\"2025-06-03\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12142173/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Journal for Immunotherapy of Cancer\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1136/jitc-2025-011503\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"IMMUNOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal for Immunotherapy of Cancer","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1136/jitc-2025-011503","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:肿瘤微环境经常是缺氧的,其特征是营养资源的缺乏,包括葡萄糖的缺乏。由于效应T细胞具有高能量需求,肿瘤代谢可导致T细胞功能障碍和衰竭。方法:采用逆转录聚合酶链反应(RT-PCR)、组织学和流式细胞术检测荷瘤C57BL/6J小鼠脾脏和肿瘤组织缺氧情况。接下来,从C57BL6J小鼠或P14+小鼠分离的CD8+ T细胞被Thy1.1(对照)或Thy1.1-肌红蛋白(Mb)包装的逆转录病毒转导。采用RT-PCR和western blot方法证实Mb的表达。采用流式细胞术、透射电镜、聚焦离子束扫描电镜、海马、代谢组学和发光法测定细胞代谢。将表达或对照P14+或OT-I+ T细胞的Mb分别转移到B16F10-gp33或MC38-ova荷瘤小鼠体内,用流式细胞术和组织学方法进行分析。在B16F10-gp33荷瘤小鼠的基础上添加抗程序性细胞死亡蛋白-1 (PD-1)检查点抑制剂。结果:在这里,我们证明了氧结合蛋白肌红蛋白在T细胞中的表达可以增强其线粒体和糖酵解代谢功能。代谢产物和三羧酸化合物在肌红蛋白(Mb)存在时高度增加,这与ATP水平增加有关。表达mb的T细胞在激活后和浸润到肿瘤微环境(TME)过程中表现出低表达的缺氧诱导因子-1α。因此,Mb的表达增加了体外效应t细胞对肿瘤细胞的功能,同时降低了超氧化物水平。在过继转移到荷瘤小鼠后,Mb的表达促进了TME的浸润增加。虽然表达Mb的T细胞表现出效应细胞因子的表达增加,但PD-1仍然被抗PD-1单克隆抗体检测和靶向,与表达Mb的T细胞转移联合,PD-1单克隆抗体在延缓肿瘤生长方面显示出最大的功效。结论:综上所述,我们发现Mb在T细胞中的表达可以增加T细胞的代谢,促进其向肿瘤组织的浸润,并具有对癌细胞的效应功能。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Myoglobin expression improves T-cell metabolism and antitumor effector function.

Background: The tumor microenvironment is frequently hypoxic and characterized by a scarcity of nutritional resources including a shortage of glucose. As effector T cells have high energy demands, tumor metabolism can contribute to T-cell dysfunction and exhaustion.

Methods: In this study, we determined hypoxia in spleen and tumor tissue from tumor-bearing C57BL/6J mice using reverse transcription polymerase chain reaction (RT-PCR), histology and flow cytometry. Next, CD8+ T cells isolated from C57BL6J mice or P14+ mice were transduced with Thy1.1 (Control) or Thy1.1-Myoglobin (Mb) packaged retrovirus. Expression of Mb was confirmed with RT-PCR and western blot. Cellular metabolism was determined by flow cytometry, transmission electron microscopy, focused ion beam scanning electron microscopy, Seahorse, metabolomics and luminescence assays. Mb expressing or control P14+ or OT-I+ T cells were transferred in B16F10-gp33 or MC38-ova tumor-bearing mice respectively and analyzed using flow cytometry and histology. B16F10-gp33 tumor-bearing mice were additionally treated with anti-programmed cell death protein-1 (PD-1) checkpoint inhibitor.

Results: Here we demonstrate that expression of the oxygen-binding protein myoglobin in T cells can boost their mitochondrial and glycolytic metabolic functions. Metabolites and tricarboxylic acid compounds were highly increased in the presence of myoglobin (Mb), which was associated with increased ATP levels. Mb-expressing T cells exhibited low expression of hypoxia-inducible factor-1α after activation and during infiltration into the tumor microenvironment (TME). Accordingly, Mb expression increased effector T-cell function against tumor cells in vitro with concomitant reductions in superoxide levels. Following adoptive transfer into tumor-bearing mice, Mb expression facilitated increased infiltration into the TME. Although T cells expressing Mb exhibited increased expression of effector cytokines, PD-1 was still detected and targetable by anti-PD-1 monoclonal antibodies, which in combination with transfer of Mb-expressing T cells demonstrated maximal efficacy in delaying tumor growth.

Conclusion: Taken together, we show that expression of Mb in T cells can increase their metabolism, infiltration into the tumor tissue, and effector function against cancer cells.

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
Journal for Immunotherapy of Cancer
Journal for Immunotherapy of Cancer Biochemistry, Genetics and Molecular Biology-Molecular Medicine
CiteScore
17.70
自引率
4.60%
发文量
522
审稿时长
18 weeks
期刊介绍: The Journal for ImmunoTherapy of Cancer (JITC) is a peer-reviewed publication that promotes scientific exchange and deepens knowledge in the constantly evolving fields of tumor immunology and cancer immunotherapy. With an open access format, JITC encourages widespread access to its findings. The journal covers a wide range of topics, spanning from basic science to translational and clinical research. Key areas of interest include tumor-host interactions, the intricate tumor microenvironment, animal models, the identification of predictive and prognostic immune biomarkers, groundbreaking pharmaceutical and cellular therapies, innovative vaccines, combination immune-based treatments, and the study of immune-related toxicity.
×
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