CD4 T cell-intrinsic STING signaling controls the differentiation and effector functions of TH1 and TH9 cells

Author:

Benoit-Lizon IsisORCID,Jacquin EliseORCID,Rivera Vargas Thaiz,Richard Corentin,Roussey Aurélie,Dal Zuffo Ludivine,Martin Tiffany,Melis Andréa,Vinokurova Daria,Shahoei Sayyed Hamed,Baeza Garcia AlvaroORCID,Pignol Cassandre,Giorgiutti Stéphane,Carapito Raphaël,Boidot Romain,Végran Frédérique,Flavell Richard A,Ryffel Bernhard,Nelson Eric R,Soulas-Sprauel Pauline,Lawrence Toby,Apetoh LionelORCID

Abstract

BackgroundWhile stimulator of interferon genes (STING) activation in innate immune cells of the tumor microenvironment can result in CD8 T cell-dependent antitumor immunity, whether STING signaling affects CD4 T-cell responses remains elusive.MethodsHere, we tested whether STING activation modulated the effector functions of CD4 T cells in vivo by analyzing tumor-infiltrating CD4 T cells and evaluating the contribution of the CD4 T cell-derived cytokines in the antitumor activity of the STING ligand 2′3′-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) in two mouse tumor models. We performed ex vivo experiments to assess the impact of STING activation on CD4 T-cell differentiation and investigate the underlying molecular mechanisms. Finally, we tested whether STING activation enhances TH9 cell antitumor activity against mouse melanoma upon adoptive transfer.ResultsWe found that activation of STING signaling cell-intrinsically enhances the differentiation and antitumor functions of TH1 and TH9 cells by increasing their respective production of interferon gamma (IFN-γ) and interleukin-9. IRF3 and type I interferon receptors (IFNARs) are required for the STING-driven enhancement of TH1 cell differentiation. However, STING activation favors TH9 cell differentiation independently of the IFNARs/IRF3 pathway but through mammalian target of rapamycin (mTOR) signaling, underscoring that STING activation differentially affects the fate of distinct CD4 T-cell subsets. The therapeutic effect of STING activation relies on TH1 and TH9-derived cytokines, and STING activation enhances the antitumor activity of TH9 cells upon adoptive transfer.ConclusionOur results reveal the STING signaling pathway as a therapeutic target to boost CD4 T-cell effector functions and antitumor immunity.

Funder

National Cancer Institute of the National Institutes of Health

Department of Defense

Fondation pour la Recherche Médicale

European Research Council

Fondation ARC

FEDER

ANR

LabEX MabImprove

Agence Nationale de la Recherche

Fondation de France

Publisher

BMJ

Subject

Cancer Research,Pharmacology,Oncology,Molecular Medicine,Immunology,Immunology and Allergy

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