Single-cell RNA sequencing highlights the immunosuppression of IDO1+ macrophages in the malignant transformation of oral leukoplakia.

IF 12.4 1区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL
Theranostics Pub Date : 2024-08-12 eCollection Date: 2024-01-01 DOI:10.7150/thno.99112
Yu Zhang, Jie Zhang, Simin Zhao, Yan Xu, Yingying Huang, Shaopeng Liu, Peng Su, Caijiao Wang, Yahui Li, Hao Li, Pishan Yang, Chengzhe Yang
{"title":"Single-cell RNA sequencing highlights the immunosuppression of IDO1<sup>+</sup> macrophages in the malignant transformation of oral leukoplakia.","authors":"Yu Zhang, Jie Zhang, Simin Zhao, Yan Xu, Yingying Huang, Shaopeng Liu, Peng Su, Caijiao Wang, Yahui Li, Hao Li, Pishan Yang, Chengzhe Yang","doi":"10.7150/thno.99112","DOIUrl":null,"url":null,"abstract":"<p><p><b>Rationale</b>: Immunosuppressive tumor microenvironment (iTME) plays an important role in carcinogenesis, and some macrophage subsets are associated with iTME generation. However, the sub-population characterization of macrophages in oral carcinogenesis remains largely unclear. Here, we investigated the immunosuppressive status with focus on function of a macrophage subset that expressed indoleamine 2,3 dioxygenase 1 (Macro-IDO1) in oral carcinogenesis. <b>Methods</b>: We built a single cell transcriptome atlas from 3 patients simultaneously containing oral squamous cell carcinoma (OSCC), precancerous oral leukoplakia (preca-OLK) and paracancerous tissue (PCA). Through single-cell RNA sequencing and further validation using multicolor immunofluorescence staining and the <i>in vitro</i>/<i>in vivo</i> experiments, the immunosuppressive cell profiles were built and the role of a macrophage subset that expressed indoleamine 2,3 dioxygenase 1 (Macro-IDO1) in the malignant transformation of oral leukoplakia was evaluated. <b>Results</b>: The iTME formed at preca-OLK stage, as evidenced by increased exhausted T cells, Tregs and some special subsets of macrophages and fibroblasts. Macro-IDO1 was predominantly enriched in preca-OLK and OSCC, distributed near exhausted T cells and possessed tumor associated macrophage transformation potentials. Functional analysis revealed the established immunosuppressive role of Macro-IDO1 in preca-OLK and OSCC: enriching the immunosuppression related genes; having an established level of immune checkpoint score; exerting strong immunosuppressive interaction with T cells; positively correlating with the CD8-exhausted. The immunosuppression related gene expression of macrophages also increased in preca-OLK/OSCC compared to PCA. The use of the IDO1 inhibitor reduced 4NQO induced oral carcinogenesis in mice. Mechanistically, IFN-γ-JAK-STAT pathway was associated with IDO1 upregulation in OLK and OSCC. <b>Conclusions</b>: These results highlight that Macro-IDO1-enriched in preca-OLK possesses a strong immunosuppressive role and contributes to oral carcinogenesis, providing a potential target for preventing precancerous legions from transformation into OSCC.</p>","PeriodicalId":22932,"journal":{"name":"Theranostics","volume":null,"pages":null},"PeriodicalIF":12.4000,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11373622/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Theranostics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.7150/thno.99112","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/1/1 0:00:00","PubModel":"eCollection","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

Abstract

Rationale: Immunosuppressive tumor microenvironment (iTME) plays an important role in carcinogenesis, and some macrophage subsets are associated with iTME generation. However, the sub-population characterization of macrophages in oral carcinogenesis remains largely unclear. Here, we investigated the immunosuppressive status with focus on function of a macrophage subset that expressed indoleamine 2,3 dioxygenase 1 (Macro-IDO1) in oral carcinogenesis. Methods: We built a single cell transcriptome atlas from 3 patients simultaneously containing oral squamous cell carcinoma (OSCC), precancerous oral leukoplakia (preca-OLK) and paracancerous tissue (PCA). Through single-cell RNA sequencing and further validation using multicolor immunofluorescence staining and the in vitro/in vivo experiments, the immunosuppressive cell profiles were built and the role of a macrophage subset that expressed indoleamine 2,3 dioxygenase 1 (Macro-IDO1) in the malignant transformation of oral leukoplakia was evaluated. Results: The iTME formed at preca-OLK stage, as evidenced by increased exhausted T cells, Tregs and some special subsets of macrophages and fibroblasts. Macro-IDO1 was predominantly enriched in preca-OLK and OSCC, distributed near exhausted T cells and possessed tumor associated macrophage transformation potentials. Functional analysis revealed the established immunosuppressive role of Macro-IDO1 in preca-OLK and OSCC: enriching the immunosuppression related genes; having an established level of immune checkpoint score; exerting strong immunosuppressive interaction with T cells; positively correlating with the CD8-exhausted. The immunosuppression related gene expression of macrophages also increased in preca-OLK/OSCC compared to PCA. The use of the IDO1 inhibitor reduced 4NQO induced oral carcinogenesis in mice. Mechanistically, IFN-γ-JAK-STAT pathway was associated with IDO1 upregulation in OLK and OSCC. Conclusions: These results highlight that Macro-IDO1-enriched in preca-OLK possesses a strong immunosuppressive role and contributes to oral carcinogenesis, providing a potential target for preventing precancerous legions from transformation into OSCC.

单细胞 RNA 测序突显了 IDO1+ 巨噬细胞在口腔白斑病恶性转化过程中的免疫抑制作用。
理由:免疫抑制性肿瘤微环境(iTME)在癌变过程中发挥着重要作用,而某些巨噬细胞亚群与 iTME 的产生有关。然而,巨噬细胞在口腔癌发生中的亚群特征在很大程度上仍不清楚。在此,我们研究了表达吲哚胺 2,3 二氧合酶 1(Macro-IDO1)的巨噬细胞亚群在口腔癌发生中的免疫抑制状态,并重点研究了其功能。研究方法我们建立了一个单细胞转录组图谱,该图谱来自同时包含口腔鳞状细胞癌(OSCC)、癌前口腔白斑(preca-OLK)和癌旁组织(PCA)的3名患者。通过单细胞 RNA 测序以及多色免疫荧光染色和体内外实验的进一步验证,建立了免疫抑制细胞图谱,并评估了表达吲哚胺 2,3 二氧合酶 1(Macro-IDO1)的巨噬细胞亚群在口腔白斑病恶性转化中的作用。结果显示iTME形成于preca-OLK阶段,表现为衰竭T细胞、Tregs以及巨噬细胞和成纤维细胞的一些特殊亚群的增加。巨噬细胞-IDO1主要富集于preca-OLK和OSCC,分布在衰竭T细胞附近,并具有肿瘤相关巨噬细胞转化潜能。功能分析显示,Macro-IDO1在preca-OLK和OSCC中具有既定的免疫抑制作用:富集免疫抑制相关基因;具有既定的免疫检查点评分水平;与T细胞产生强烈的免疫抑制相互作用;与CD8耗竭正相关。与 PCA 相比,preca-OLK/OSC 中巨噬细胞的免疫抑制相关基因表达也有所增加。使用 IDO1 抑制剂可减少 4NQO 诱导的小鼠口腔癌发生。从机制上讲,IFN-γ-JAK-STAT 通路与 OLK 和 OSCC 中 IDO1 的上调有关。结论:这些结果表明,Preca-OLK中富集的巨-IDO1具有很强的免疫抑制作用,有助于口腔癌的发生,为防止癌前军团转化为OSCC提供了一个潜在的靶点。
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 求助全文
来源期刊
Theranostics
Theranostics MEDICINE, RESEARCH & EXPERIMENTAL-
CiteScore
25.40
自引率
1.60%
发文量
433
审稿时长
1 months
期刊介绍: Theranostics serves as a pivotal platform for the exchange of clinical and scientific insights within the diagnostic and therapeutic molecular and nanomedicine community, along with allied professions engaged in integrating molecular imaging and therapy. As a multidisciplinary journal, Theranostics showcases innovative research articles spanning fields such as in vitro diagnostics and prognostics, in vivo molecular imaging, molecular therapeutics, image-guided therapy, biosensor technology, nanobiosensors, bioelectronics, system biology, translational medicine, point-of-care applications, and personalized medicine. Encouraging a broad spectrum of biomedical research with potential theranostic applications, the journal rigorously peer-reviews primary research, alongside publishing reviews, news, and commentary that aim to bridge the gap between the laboratory, clinic, and biotechnology industries.
×
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