文献检索文档翻译深度研究
Suppr Zotero 插件Zotero 插件
邀请有礼套餐&价格历史记录

新学期,新优惠

限时优惠:9月1日-9月22日

30天高级会员仅需29元

1天体验卡首发特惠仅需5.99元

了解详情
不再提醒
插件&应用
Suppr Zotero 插件Zotero 插件浏览器插件Mac 客户端Windows 客户端微信小程序
高级版
套餐订阅购买积分包
AI 工具
文献检索文档翻译深度研究
关于我们
关于 Suppr公司介绍联系我们用户协议隐私条款
关注我们

Suppr 超能文献

核心技术专利:CN118964589B侵权必究
粤ICP备2023148730 号-1Suppr @ 2025

来自嗜黏蛋白阿克曼氏菌的Amuc_1434通过抑制结直肠癌中的PD-L1增强CD8 + T细胞介导的抗肿瘤免疫。

Amuc_1434 From Akkermansia muciniphila Enhances CD8+ T Cell-Mediated Anti-Tumor Immunity by Suppressing PD-L1 in Colorectal Cancer.

作者信息

Zhu Jiahao, Qin Shaolei, Gu Ruike, Ji Shengjun, Wu Gang, Gu Ke

机构信息

Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, People's Republic of China.

Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, People's Republic of China.

出版信息

FASEB J. 2025 Apr 30;39(8):e70540. doi: 10.1096/fj.202403295RR.


DOI:10.1096/fj.202403295RR
PMID:40231387
Abstract

Colorectal cancer (CRC) shows a limited response to programmed death-ligand 1 (PD-L1) immunotherapies. Akkermansia muciniphila (AKK) may enhance tumor immunity. This study examines how its Outer Membrane Vesicles (OMVs) and Amuc_1434 influence PD-L1 expression and CD8+ T cell activity in CRC. OMVs were isolated and their characteristics were examined through transmission electron microscopy and Western blotting. PD-L1 expression was quantified via Western blot, while CD8+ T cell proliferation was measured using flow cytometry. Cytokine production (interferon-gamma (IFN-γ) and interleukin-2 (IL-2)) was evaluated using ELISA. A CRC mouse model was employed to examine its impact on tumor growth and immune cell infiltration. In CRC cells, treatment with AKK-derived OMVs (AKK-OMVs) significantly downregulated PD-L1 expression (p < 0.05) and markedly increased CD8+ T cell proliferation and the levels of IFN-γ and IL-2 (p < 0.01). Amuc_1434 was identified as the key protein mediating these effects. In vivo, AKK-OMVs treatment substantially reduced tumor volume (p < 0.01) and significantly enhanced CD8+ T cell infiltration into the tumor microenvironment (p < 0.01). Additionally, AKK-OMVs-treated mice showed increased expression of immune activation markers within the tumor tissue, further indicating enhanced antitumor immunity. This study reveals that AKK-OMVs, particularly those containing Amuc_1434, can modulate PD-L1 expression and potentiate CD8+ T cell-mediated antitumor immunity in CRC. These findings suggest a novel approach to overcoming resistance to immune checkpoint inhibitors in CRC.

摘要

结直肠癌(CRC)对程序性死亡配体1(PD-L1)免疫疗法的反应有限。嗜黏蛋白阿克曼氏菌(AKK)可能会增强肿瘤免疫力。本研究探讨了其外膜囊泡(OMV)和Amuc_1434如何影响CRC中PD-L1的表达和CD8 + T细胞活性。分离出OMV,并通过透射电子显微镜和蛋白质免疫印迹法检测其特征。通过蛋白质免疫印迹法定量PD-L1表达,同时使用流式细胞术测量CD8 + T细胞增殖。使用酶联免疫吸附测定法评估细胞因子产生(γ干扰素(IFN-γ)和白细胞介素-2(IL-2))。采用CRC小鼠模型检查其对肿瘤生长和免疫细胞浸润的影响。在CRC细胞中,用源自AKK的OMV(AKK-OMV)处理可显著下调PD-L1表达(p <0.05),并显著增加CD8 + T细胞增殖以及IFN-γ和IL-2水平(p <0.01)。Amuc_1434被确定为介导这些作用的关键蛋白。在体内,AKK-OMV处理可显著减小肿瘤体积(p <0.01),并显著增强CD8 + T细胞向肿瘤微环境的浸润(p <0.01)。此外,经AKK-OMV处理的小鼠肿瘤组织内免疫激活标志物的表达增加,进一步表明抗肿瘤免疫力增强。本研究表明,AKK-OMV,特别是那些含有Amuc_1434的OMV,可以调节PD-L1表达并增强CRC中CD8 + T细胞介导的抗肿瘤免疫力。这些发现提示了一种克服CRC对免疫检查点抑制剂耐药性的新方法。

相似文献

[1]
Amuc_1434 From Akkermansia muciniphila Enhances CD8+ T Cell-Mediated Anti-Tumor Immunity by Suppressing PD-L1 in Colorectal Cancer.

FASEB J. 2025-4-30

[2]
Akkermansia muciniphila outer membrane protein regulates recruitment of CD8 T cells in lung adenocarcinoma and through JAK-STAT signalling pathway.

Microb Biotechnol. 2024-7

[3]
5-Fluorouracil upregulates cell surface B7-H1 (PD-L1) expression in gastrointestinal cancers.

J Immunother Cancer. 2016-10-18

[4]
Landscape of tumoral ecosystem for enhanced anti-PD-1 immunotherapy by gut Akkermansia muciniphila.

Cell Rep. 2024-6-25

[5]
Cancer cells accelerate exhaustion of persistently activated mouse CD4 T cells.

Oncoimmunology. 2025-12

[6]
The combination of and mitigates DSS-induced colitis and attenuates colitis-associated tumorigenesis by modulating gut microbiota and reducing CD8 T cells in mice.

mSystems. 2025-2-18

[7]
Correction to "Amuc_1434 from Akkermansia muciniphila Enhances CD8+ T Cell-Mediated Anti-Tumor Immunity by Suppressing PD-L1 in Colorectal Cancer".

FASEB J. 2025-6-30

[8]
Extracellular Vesicles from Elicit Antitumor Immunity Against Prostate Cancer via Modulation of CD8 T Cells and Macrophages.

Int J Nanomedicine. 2021

[9]
NAT10 regulates tumor progression and immune microenvironment in pancreatic ductal adenocarcinoma via the N4-acetylated LAMB3-mediated FAK/ERK pathway.

Cancer Commun (Lond). 2025-6-20

[10]
PMN-MDSCs are responsible for immune suppression in anti-PD-1 treated TAP1 defective melanoma.

Clin Transl Oncol. 2025-1-18

文献AI研究员

20分钟写一篇综述,助力文献阅读效率提升50倍

立即体验

用中文搜PubMed

大模型驱动的PubMed中文搜索引擎

马上搜索

推荐工具

医学文档翻译智能文献检索