磷酸甲羟戊酸激酶产生的 4-乙酰氨基丁酸的增加抑制了 CD8+ T 细胞的活化并使肿瘤免疫逃逸得以实现。

IF 14.3 1区 材料科学 Q1 CHEMISTRY, MULTIDISCIPLINARY
Xinyi Zhou, Zhiqiang Chen, Yijiang Yu, Mengjiao Li, Yu Cao, Edward V Prochownik, Youjun Li
{"title":"磷酸甲羟戊酸激酶产生的 4-乙酰氨基丁酸的增加抑制了 CD8+ T 细胞的活化并使肿瘤免疫逃逸得以实现。","authors":"Xinyi Zhou, Zhiqiang Chen, Yijiang Yu, Mengjiao Li, Yu Cao, Edward V Prochownik, Youjun Li","doi":"10.1002/advs.202403629","DOIUrl":null,"url":null,"abstract":"<p><p>Certain metabolites in the tumor microenvironment (TME) can alter innate immunity. Here, it is shown how phosphomevalonate kinase (PMVK) allows hepatocellular carcinoma (HCC) cells to overcome the anti-tumor immunity mediated by CD8<sup>+</sup> T cells. In HCCs, depletion of PMVK is required to facilitate CD8<sup>+</sup> T cell activation and their subsequent suppression of tumor growth. Mechanistically, PMVK phosphorylates and stabilizes glutamate decarboxylase 1 (GAD1), thus increasing the synthesis of γ-aminobutyric acid (GABA), which normally functions as a neurotransmitter. However, PMVK also recruits acetyl-CoA acetyltransferase 1 (ACAT1) and allows it to convert GABA, to 4-acetaminobutyric acid (4-Ac-GABA), which is released into the TME. There, 4-Ac-GABA activates the GABAA receptor (GABAAR) on CD8<sup>+</sup> T cells, which inhibits AKT1 signaling. This in turn suppresses CD8<sup>+</sup> T cell activation, intratumoral infiltration, and the anti-tumor response. Inhibiting PMVK or GABAAR in HCC mouse models overcomes resistance to anti-PD-1 immune checkpoint therapy. These findings reveal non-canonical and cooperative functions among the key metabolic enzymes PMVK, GAD1, and ACAT1 that reprogram glutamine metabolism to synthesize a potent CD8<sup>+</sup> T cell inhibitor 4-Ac-GABA. Blocking 4-Ac-GABA signaling in CD8<sup>+</sup> T cells, particularly when combined with immune checkpoint inhibition, potentially represents a new and potent form of immunotherapy.</p>","PeriodicalId":117,"journal":{"name":"Advanced Science","volume":null,"pages":null},"PeriodicalIF":14.3000,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Increases in 4-Acetaminobutyric Acid Generated by Phosphomevalonate Kinase Suppress CD8<sup>+</sup> T Cell Activation and Allow Tumor Immune Escape.\",\"authors\":\"Xinyi Zhou, Zhiqiang Chen, Yijiang Yu, Mengjiao Li, Yu Cao, Edward V Prochownik, Youjun Li\",\"doi\":\"10.1002/advs.202403629\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Certain metabolites in the tumor microenvironment (TME) can alter innate immunity. Here, it is shown how phosphomevalonate kinase (PMVK) allows hepatocellular carcinoma (HCC) cells to overcome the anti-tumor immunity mediated by CD8<sup>+</sup> T cells. In HCCs, depletion of PMVK is required to facilitate CD8<sup>+</sup> T cell activation and their subsequent suppression of tumor growth. Mechanistically, PMVK phosphorylates and stabilizes glutamate decarboxylase 1 (GAD1), thus increasing the synthesis of γ-aminobutyric acid (GABA), which normally functions as a neurotransmitter. However, PMVK also recruits acetyl-CoA acetyltransferase 1 (ACAT1) and allows it to convert GABA, to 4-acetaminobutyric acid (4-Ac-GABA), which is released into the TME. There, 4-Ac-GABA activates the GABAA receptor (GABAAR) on CD8<sup>+</sup> T cells, which inhibits AKT1 signaling. This in turn suppresses CD8<sup>+</sup> T cell activation, intratumoral infiltration, and the anti-tumor response. Inhibiting PMVK or GABAAR in HCC mouse models overcomes resistance to anti-PD-1 immune checkpoint therapy. These findings reveal non-canonical and cooperative functions among the key metabolic enzymes PMVK, GAD1, and ACAT1 that reprogram glutamine metabolism to synthesize a potent CD8<sup>+</sup> T cell inhibitor 4-Ac-GABA. Blocking 4-Ac-GABA signaling in CD8<sup>+</sup> T cells, particularly when combined with immune checkpoint inhibition, potentially represents a new and potent form of immunotherapy.</p>\",\"PeriodicalId\":117,\"journal\":{\"name\":\"Advanced Science\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":14.3000,\"publicationDate\":\"2024-09-26\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Advanced Science\",\"FirstCategoryId\":\"88\",\"ListUrlMain\":\"https://doi.org/10.1002/advs.202403629\",\"RegionNum\":1,\"RegionCategory\":\"材料科学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"CHEMISTRY, MULTIDISCIPLINARY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Advanced Science","FirstCategoryId":"88","ListUrlMain":"https://doi.org/10.1002/advs.202403629","RegionNum":1,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CHEMISTRY, MULTIDISCIPLINARY","Score":null,"Total":0}
引用次数: 0

摘要

肿瘤微环境(TME)中的某些代谢物可改变先天性免疫。本文展示了磷酸甲羟戊酸激酶(PMVK)如何使肝癌(HCC)细胞克服由CD8+ T细胞介导的抗肿瘤免疫。在 HCC 中,需要消耗 PMVK 来促进 CD8+ T 细胞的活化及其随后对肿瘤生长的抑制。从机理上讲,PMVK 可使谷氨酸脱羧酶 1(GAD1)磷酸化并使其稳定,从而增加γ-氨基丁酸(GABA)的合成,而γ-氨基丁酸通常是一种神经递质。不过,PMVK 还能招募乙酰-CoA 乙酰转移酶 1(ACAT1),使其将 GABA 转化为 4-乙酰氨基丁酸(4-Ac-GABA),释放到 TME 中。在那里,4-Ac-GABA 会激活 CD8+ T 细胞上的 GABAA 受体(GABAAR),从而抑制 AKT1 信号传导。这反过来又抑制了 CD8+ T 细胞的活化、瘤内浸润和抗肿瘤反应。在HCC小鼠模型中抑制PMVK或GABAAR可以克服抗PD-1免疫检查点疗法的耐药性。这些发现揭示了关键代谢酶 PMVK、GAD1 和 ACAT1 之间的非规范合作功能,它们重新规划谷氨酰胺代谢,合成一种强效的 CD8+ T 细胞抑制剂 4-Ac-GABA。阻断 CD8+ T 细胞中的 4-Ac-GABA 信号传导,尤其是与免疫检查点抑制相结合时,有可能成为一种新的强效免疫疗法。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Increases in 4-Acetaminobutyric Acid Generated by Phosphomevalonate Kinase Suppress CD8+ T Cell Activation and Allow Tumor Immune Escape.

Certain metabolites in the tumor microenvironment (TME) can alter innate immunity. Here, it is shown how phosphomevalonate kinase (PMVK) allows hepatocellular carcinoma (HCC) cells to overcome the anti-tumor immunity mediated by CD8+ T cells. In HCCs, depletion of PMVK is required to facilitate CD8+ T cell activation and their subsequent suppression of tumor growth. Mechanistically, PMVK phosphorylates and stabilizes glutamate decarboxylase 1 (GAD1), thus increasing the synthesis of γ-aminobutyric acid (GABA), which normally functions as a neurotransmitter. However, PMVK also recruits acetyl-CoA acetyltransferase 1 (ACAT1) and allows it to convert GABA, to 4-acetaminobutyric acid (4-Ac-GABA), which is released into the TME. There, 4-Ac-GABA activates the GABAA receptor (GABAAR) on CD8+ T cells, which inhibits AKT1 signaling. This in turn suppresses CD8+ T cell activation, intratumoral infiltration, and the anti-tumor response. Inhibiting PMVK or GABAAR in HCC mouse models overcomes resistance to anti-PD-1 immune checkpoint therapy. These findings reveal non-canonical and cooperative functions among the key metabolic enzymes PMVK, GAD1, and ACAT1 that reprogram glutamine metabolism to synthesize a potent CD8+ T cell inhibitor 4-Ac-GABA. Blocking 4-Ac-GABA signaling in CD8+ T cells, particularly when combined with immune checkpoint inhibition, potentially represents a new and potent form of immunotherapy.

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
Advanced Science
Advanced Science CHEMISTRY, MULTIDISCIPLINARYNANOSCIENCE &-NANOSCIENCE & NANOTECHNOLOGY
CiteScore
18.90
自引率
2.60%
发文量
1602
审稿时长
1.9 months
期刊介绍: Advanced Science is a prestigious open access journal that focuses on interdisciplinary research in materials science, physics, chemistry, medical and life sciences, and engineering. The journal aims to promote cutting-edge research by employing a rigorous and impartial review process. It is committed to presenting research articles with the highest quality production standards, ensuring maximum accessibility of top scientific findings. With its vibrant and innovative publication platform, Advanced Science seeks to revolutionize the dissemination and organization of scientific knowledge.
×
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:481959085
Book学术官方微信