Extracellular Vesicles From Gastric Cancer Cells Induce PD-L1 Expression on Neutrophils to Suppress T-Cell Immunity

Neutrophils are prominent components of solid tumors and exhibit distinct phenotypes in different tumor milieu. We have previously shown that tumor extracellular vesicles (EVs) could induce pro-tumor activation of neutrophils; however, the role of tumor EV-elicited neutrophils in tumor immunity rema...

Full description

Bibliographic Details
Main Authors: Yinghong Shi, Jiahui Zhang, Zheying Mao, Han Jiang, Wei Liu, Hui Shi, Runbi Ji, Wenrong Xu, Hui Qian, Xu Zhang
Format: Article
Language:English
Published: Frontiers Media S.A. 2020-05-01
Series:Frontiers in Oncology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fonc.2020.00629/full
id doaj-3f410703a63d43e2b7533088e6894844
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Yinghong Shi
Yinghong Shi
Jiahui Zhang
Jiahui Zhang
Zheying Mao
Zheying Mao
Han Jiang
Han Jiang
Wei Liu
Wei Liu
Hui Shi
Hui Shi
Runbi Ji
Runbi Ji
Wenrong Xu
Wenrong Xu
Hui Qian
Hui Qian
Xu Zhang
Xu Zhang
spellingShingle Yinghong Shi
Yinghong Shi
Jiahui Zhang
Jiahui Zhang
Zheying Mao
Zheying Mao
Han Jiang
Han Jiang
Wei Liu
Wei Liu
Hui Shi
Hui Shi
Runbi Ji
Runbi Ji
Wenrong Xu
Wenrong Xu
Hui Qian
Hui Qian
Xu Zhang
Xu Zhang
Extracellular Vesicles From Gastric Cancer Cells Induce PD-L1 Expression on Neutrophils to Suppress T-Cell Immunity
Frontiers in Oncology
extracellular vesicles
programmed death-ligand 1 (PD-L1)
neutrophils
immune suppression
gastric cancer
author_facet Yinghong Shi
Yinghong Shi
Jiahui Zhang
Jiahui Zhang
Zheying Mao
Zheying Mao
Han Jiang
Han Jiang
Wei Liu
Wei Liu
Hui Shi
Hui Shi
Runbi Ji
Runbi Ji
Wenrong Xu
Wenrong Xu
Hui Qian
Hui Qian
Xu Zhang
Xu Zhang
author_sort Yinghong Shi
title Extracellular Vesicles From Gastric Cancer Cells Induce PD-L1 Expression on Neutrophils to Suppress T-Cell Immunity
title_short Extracellular Vesicles From Gastric Cancer Cells Induce PD-L1 Expression on Neutrophils to Suppress T-Cell Immunity
title_full Extracellular Vesicles From Gastric Cancer Cells Induce PD-L1 Expression on Neutrophils to Suppress T-Cell Immunity
title_fullStr Extracellular Vesicles From Gastric Cancer Cells Induce PD-L1 Expression on Neutrophils to Suppress T-Cell Immunity
title_full_unstemmed Extracellular Vesicles From Gastric Cancer Cells Induce PD-L1 Expression on Neutrophils to Suppress T-Cell Immunity
title_sort extracellular vesicles from gastric cancer cells induce pd-l1 expression on neutrophils to suppress t-cell immunity
publisher Frontiers Media S.A.
series Frontiers in Oncology
issn 2234-943X
publishDate 2020-05-01
description Neutrophils are prominent components of solid tumors and exhibit distinct phenotypes in different tumor milieu. We have previously shown that tumor extracellular vesicles (EVs) could induce pro-tumor activation of neutrophils; however, the role of tumor EV-elicited neutrophils in tumor immunity remains unclear. Herein, we reported that gastric cancer cell-derived EVs (GC-EVs) induced the expression of programmed death-ligand 1 (PD-L1) on neutrophils. GC-EVs transported high-mobility group box-1 (HMGB1) to activate signal transducer and activator of transcription 3 (STAT3) and upregulate PD-L1 gene expression in neutrophils. Blocking STAT3 pathway and silencing HMGB1 reversed GC-EV-induced PD-L1 expression on neutrophils. GC-EV-elicited neutrophils suppressed T cell proliferation, activation, and function in vitro, which could be antagonized by a specific PD-L1 antibody. Furthermore, GC tissue-derived EVs also showed similar effects. Taken together, our results indicate that EVs from the GC microenvironment induce PD-L1 expression on neutrophils to suppress T-cell immunity, which provides a new insight into the pro-tumor roles of neutrophils in GC and sheds light on the multifaceted roles of EVs in orchestrating an immunosuppressive microenvironment.
topic extracellular vesicles
programmed death-ligand 1 (PD-L1)
neutrophils
immune suppression
gastric cancer
url https://www.frontiersin.org/article/10.3389/fonc.2020.00629/full
work_keys_str_mv AT yinghongshi extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT yinghongshi extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT jiahuizhang extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT jiahuizhang extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT zheyingmao extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT zheyingmao extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT hanjiang extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT hanjiang extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT weiliu extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT weiliu extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT huishi extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT huishi extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT runbiji extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT runbiji extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT wenrongxu extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT wenrongxu extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT huiqian extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT huiqian extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT xuzhang extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
AT xuzhang extracellularvesiclesfromgastriccancercellsinducepdl1expressiononneutrophilstosuppresstcellimmunity
_version_ 1724679581693116416
spelling doaj-3f410703a63d43e2b7533088e68948442020-11-25T03:04:50ZengFrontiers Media S.A.Frontiers in Oncology2234-943X2020-05-011010.3389/fonc.2020.00629531984Extracellular Vesicles From Gastric Cancer Cells Induce PD-L1 Expression on Neutrophils to Suppress T-Cell ImmunityYinghong Shi0Yinghong Shi1Jiahui Zhang2Jiahui Zhang3Zheying Mao4Zheying Mao5Han Jiang6Han Jiang7Wei Liu8Wei Liu9Hui Shi10Hui Shi11Runbi Ji12Runbi Ji13Wenrong Xu14Wenrong Xu15Hui Qian16Hui Qian17Xu Zhang18Xu Zhang19Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, ChinaZhenjiang Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang, ChinaJiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, ChinaZhenjiang Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang, ChinaJiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, ChinaZhenjiang Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang, ChinaJiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, ChinaZhenjiang Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang, ChinaJiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, ChinaZhenjiang Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang, ChinaJiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, ChinaZhenjiang Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang, ChinaZhenjiang Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang, ChinaDepartment of Clinical Laboratory, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, ChinaJiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, ChinaZhenjiang Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang, ChinaJiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, ChinaZhenjiang Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang, ChinaJiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, ChinaZhenjiang Key Laboratory of High Technology for Basic and Translational Research on Exosomes, Zhenjiang, ChinaNeutrophils are prominent components of solid tumors and exhibit distinct phenotypes in different tumor milieu. We have previously shown that tumor extracellular vesicles (EVs) could induce pro-tumor activation of neutrophils; however, the role of tumor EV-elicited neutrophils in tumor immunity remains unclear. Herein, we reported that gastric cancer cell-derived EVs (GC-EVs) induced the expression of programmed death-ligand 1 (PD-L1) on neutrophils. GC-EVs transported high-mobility group box-1 (HMGB1) to activate signal transducer and activator of transcription 3 (STAT3) and upregulate PD-L1 gene expression in neutrophils. Blocking STAT3 pathway and silencing HMGB1 reversed GC-EV-induced PD-L1 expression on neutrophils. GC-EV-elicited neutrophils suppressed T cell proliferation, activation, and function in vitro, which could be antagonized by a specific PD-L1 antibody. Furthermore, GC tissue-derived EVs also showed similar effects. Taken together, our results indicate that EVs from the GC microenvironment induce PD-L1 expression on neutrophils to suppress T-cell immunity, which provides a new insight into the pro-tumor roles of neutrophils in GC and sheds light on the multifaceted roles of EVs in orchestrating an immunosuppressive microenvironment.https://www.frontiersin.org/article/10.3389/fonc.2020.00629/fullextracellular vesiclesprogrammed death-ligand 1 (PD-L1)neutrophilsimmune suppressiongastric cancer