Fusion of Invasive Tumor Cells with Infiltrating Macrophages Fuels Epithelial-Mesenchymal Transition and Adaptive Immune Evasion

Author:

Shao Rong1,Han Xiao1,Zhu Bowen1,Luo Jing2,Zhang Xuemei1,Chen Hui1,Brown Christine3,Burnside Amy3,Fu Guohui1,Zhao Faxue1,Chen Hongzhuan1,Oh Dennis4,Schwartz Lawrence M5,Liu Yingbin1

Affiliation:

1. Shanghai Jiao Tong University School of Medicine

2. shanghai Jiao University School of Medicine

3. University of Massachusetts Amherst

4. University of Massachusetts School of Medicine

5. University of Massachusetts, Amherst

Abstract

Abstract Heterotypic interaction between tumor cells and adjacent stromal cells mediates tumor development. However, how tumor heterogeneity commits tumors to the malignant transformation and evasion of immunity against metastasis is poorly understood. Here, we have investigated the fusogenicity of human invasive glioblastoma, triple negative breast cancer and gallbladder cancer cells that are all characterized by mesenchymal cell plasticity. These cells displayed the rigorous ability to fuse with macrophages and augment epithelial-mesenchymal transition (EMT), transforming the fused cells into highly invasive hybrids. YKL-40 (Chitinase-3-like-1), known to promote inflammation and serve as an EMT marker, was essential and sufficient for both cell fusion and the invasiveness of tumor cells that express EMT and tumor-associated macrophage markers. Intriguingly, differential gene profiling of single clones from the hybrids demonstrated that YKL-40 and immune checkpoint protein B7-2 (CD86) were elevated and functioned to independently suppress anti-tumor immune factor levels of CD8+-cytotoxic T lymphocytes (CTL); thus resulting in escape of immune surveillance. YKL-40 and B7-2 dual shRNA abrogated YKL-40-mediated cell fusion and restored CTL anti-tumor immunity, compromising tumor development in xenografts. Clinically, we found tumor hybrids were present in mesenchymal types of glioblastomas, gallbladder cancer and breast cancer. In addition, YKL-40 expression in glioblastoma was correlated with decreased disease-free survival in patients. Collectively, these data offer novel cellular and molecular mechanisms underpinning immune evasion and tumor malignancy, and suggest a new immunotherapeutic intervention strategy by targeting both YKL-40 and B7-2 in cancer.

Publisher

Research Square Platform LLC

Reference43 articles.

1. An analysis of genetic heterogeneity in untreated cancers;Reiter JG;Nat Rev Cancer.,2019

2. Continuous cultures of fused cells secreting antibody of predefined specificity;Köhler G;Nature.,1975

3. Tumour cell hybridization and metastasis revisited;Pawelek JM;Melanoma Res.,2000

4. vivo hybridisation of human tumour and normal hamster cells;Goldenberg DM;Nature.,1974

5. Relevance of spontaneous in vivo tumor-host cell fusion to tumor progression and metastasis evaluated using a series of lectin-resistant mutant tumor sublines;Kerbel RS;Symp Fundam Cancer Res.,1983

Cited by 1 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

1. Intrinsic signalling factors associated with cancer cell-cell fusion;Cell Communication and Signaling;2023-04-04

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3