Loss of Trefoil Factor 1 Accelerates the Immune Response to Colorectal Cancer.

IF 1.6 4区 医学 Q4 ONCOLOGY Anticancer research Pub Date : 2024-09-01 DOI:10.21873/anticanres.17200
Takanori Jinno, Junpei Yamaguchi, Atsushi Ogura, Toshio Kokuryo, Yukihiro Yokoyama, Masaki Sunagawa, Taisuke Baba, Yuki Murata, Tomoki Ebata
{"title":"Loss of Trefoil Factor 1 Accelerates the Immune Response to Colorectal Cancer.","authors":"Takanori Jinno, Junpei Yamaguchi, Atsushi Ogura, Toshio Kokuryo, Yukihiro Yokoyama, Masaki Sunagawa, Taisuke Baba, Yuki Murata, Tomoki Ebata","doi":"10.21873/anticanres.17200","DOIUrl":null,"url":null,"abstract":"<p><strong>Background/aim: </strong>Recent studies suggest that PD-L1 expression in immune cells, rather than tumor cells, plays a key role in tumor immunity. Trefoil factor family 1 (TFF1) is a secreted protein expressed mainly by the gastrointestinal epithelium and is related to the development of malignant disease. This study investigated the effects of TFF1 on tumor immunity in a xenograft mouse model of colorectal cancer (CRC).</p><p><strong>Materials and methods: </strong>MC38 cells were implanted in wild-type (WT) and TFF1KO mice, and the tumor micro-environment was investigated using immunohistochemistry. The circulating immune cells were analyzed using flow cytometry.</p><p><strong>Results: </strong>Tumor growth was suppressed in TFF1KO mice. In the tumor microenvironment, CD8- and CD4-positive T cells and CD11c-positive dendritic cells (DCs) were frequently found in TFF1KO mice. When an immune checkpoint inhibitor was administered to these mice, almost half of the tumors in TFF1KO mice showed a complete response. The number of circulating PD-L1/DCs was markedly associated with tumor volume, with TFF1 deletion accelerating this effect and its injection decreasing it. These findings indicate that loss of TFF1 activates tumor immunity via frequent T-cell priming by DCs, and eventually suppresses tumor growth in CRC. In addition, the number of circulating PD-L1/DCs was identified as a predictive marker of checkpoint-inhibiting therapy efficacy.</p><p><strong>Conclusion: </strong>Loss of TFF1 resulted in accelerated immune response to colorectal cancer. Further studies are needed to investigate the precise mechanisms of TFF1 in immunotolerance and develop a novel TFF1-inhibiting immunotherapeutic strategy for CRC.</p>","PeriodicalId":8072,"journal":{"name":"Anticancer research","volume":null,"pages":null},"PeriodicalIF":1.6000,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Anticancer research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.21873/anticanres.17200","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q4","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background/aim: Recent studies suggest that PD-L1 expression in immune cells, rather than tumor cells, plays a key role in tumor immunity. Trefoil factor family 1 (TFF1) is a secreted protein expressed mainly by the gastrointestinal epithelium and is related to the development of malignant disease. This study investigated the effects of TFF1 on tumor immunity in a xenograft mouse model of colorectal cancer (CRC).

Materials and methods: MC38 cells were implanted in wild-type (WT) and TFF1KO mice, and the tumor micro-environment was investigated using immunohistochemistry. The circulating immune cells were analyzed using flow cytometry.

Results: Tumor growth was suppressed in TFF1KO mice. In the tumor microenvironment, CD8- and CD4-positive T cells and CD11c-positive dendritic cells (DCs) were frequently found in TFF1KO mice. When an immune checkpoint inhibitor was administered to these mice, almost half of the tumors in TFF1KO mice showed a complete response. The number of circulating PD-L1/DCs was markedly associated with tumor volume, with TFF1 deletion accelerating this effect and its injection decreasing it. These findings indicate that loss of TFF1 activates tumor immunity via frequent T-cell priming by DCs, and eventually suppresses tumor growth in CRC. In addition, the number of circulating PD-L1/DCs was identified as a predictive marker of checkpoint-inhibiting therapy efficacy.

Conclusion: Loss of TFF1 resulted in accelerated immune response to colorectal cancer. Further studies are needed to investigate the precise mechanisms of TFF1 in immunotolerance and develop a novel TFF1-inhibiting immunotherapeutic strategy for CRC.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
三叶因子 1 的缺失会加速结直肠癌的免疫反应
背景/目的:最新研究表明,免疫细胞而非肿瘤细胞中的 PD-L1 表达在肿瘤免疫中起着关键作用。三叶草因子家族 1(TFF1)是一种主要由胃肠道上皮表达的分泌蛋白,与恶性疾病的发生发展有关。本研究在大肠癌(CRC)异种移植小鼠模型中研究了TFF1对肿瘤免疫的影响:将 MC38 细胞植入野生型(WT)和 TFF1KO 小鼠体内,用免疫组化方法研究肿瘤微环境。使用流式细胞术分析循环免疫细胞:结果:TFF1KO小鼠的肿瘤生长受到抑制。在肿瘤微环境中,TFF1KO小鼠经常发现CD8和CD4阳性T细胞以及CD11c阳性树突状细胞(DCs)。当给这些小鼠注射免疫检查点抑制剂时,TFF1KO 小鼠近一半的肿瘤出现了完全反应。循环中PD-L1/DCs的数量与肿瘤体积明显相关,TFF1缺失会加速这种效应,而注射TFF1则会降低这种效应。这些研究结果表明,TFF1的缺失会通过DC对T细胞的频繁引诱激活肿瘤免疫,并最终抑制肿瘤在CRC中的生长。此外,循环中PD-L1/DCs的数量被确定为检查点抑制疗法疗效的预测标志物:结论:TFF1的缺失会加速结直肠癌的免疫反应。结论:TFF1的缺失会导致结直肠癌免疫反应的加速,需要进一步研究TFF1在免疫耐受中的确切机制,并开发出治疗CRC的新型TFF1抑制免疫治疗策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
Anticancer research
Anticancer research 医学-肿瘤学
CiteScore
3.70
自引率
10.00%
发文量
566
审稿时长
2 months
期刊介绍: ANTICANCER RESEARCH is an independent international peer-reviewed journal devoted to the rapid publication of high quality original articles and reviews on all aspects of experimental and clinical oncology. Prompt evaluation of all submitted articles in confidence and rapid publication within 1-2 months of acceptance are guaranteed. ANTICANCER RESEARCH was established in 1981 and is published monthly (bimonthly until the end of 2008). Each annual volume contains twelve issues and index. Each issue may be divided into three parts (A: Reviews, B: Experimental studies, and C: Clinical and Epidemiological studies). Special issues, presenting the proceedings of meetings or groups of papers on topics of significant progress, will also be included in each volume. There is no limitation to the number of pages per issue.
期刊最新文献
Bioinformatics Analysis of the Expression and Prognostic Value of PLAU Gene in Wilms' Tumor. Change in Liver Function in Durvalumab Plus Tremelimumab Treatment for Unresectable Hepatocellular Carcinoma. Clinical Significance of Body Weight Loss During Chemotherapy for Advanced Gastric Cancer Undergoing Conversion Surgery. Corrigendum. Effect of Chemoradiotherapy on Locoregional Recurrence After Pancreatectomy for Pancreatic Cancer.
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1