Reciprocal crosstalk between Th17 and mesothelial cells promotes metastasis‐associated adhesion of ovarian cancer cells

Author:

Neuhaus Felix12,Lieber Sonja1,Shinkevich Veronika3,Steitz Anna Mary2,Raifer Hartmann14,Roth Kathrin5,Finkernagel Florian6,Worzfeld Thomas37,Burchert Andreas8,Keber Corinna9,Nist Andrea10,Stiewe Thorsten10,Reinartz Silke2ORCID,Beutgen Vanessa M.11ORCID,Graumann Johannes11ORCID,Pauck Kim12,Garn Holger12,Gaida Matthias131415ORCID,Müller Rolf2ORCID,Huber Magdalena1

Affiliation:

1. Institute of Systems Immunology Center for Tumor Biology and Immunology (ZTI) Philipps University Marburg Germany

2. Department of Translational Oncology Center for Tumor Biology and Immunology (ZTI) Philipps University Marburg Germany

3. Institute of Pharmacology Philipps University Marburg Germany

4. FACS Core Facility Center for Tumor Biology and Immunology (ZTI) Philipps University Marburg Germany

5. Cell Imaging Core Facility, Center for Tumor Biology and Immunology (ZTI) Philipps University Marburg Germany

6. Bioinformatics Core Facility, Center for Tumor Biology and Immunology (ZTI) Philipps University Marburg Germany

7. Department of Pharmacology Max Planck Institute for Heart and Lung Research Bad Nauheim Germany

8. Department of Hematology Oncology and Immunology University Hospital Giessen and Marburg Marburg Germany

9. Comprehensive Biomaterial Bank Marburg (CBBMR) and Institute of Pathology Philipps University Marburg Germany

10. Genomics Core Facility Institute of Molecular Oncology Member of the German Center for Lung Research (DZL) Philipps University Marburg Germany

11. Institute of Translational Proteomics and Translational Proteomics Core Facility Biochemical Pharmacological Centre Philipps University Marburg Germany

12. Translational Inflammation Research Division and Core Facility for Single Cell Multiomics Philipps University Marburg Germany

13. Institute of Pathology University Medical Center Mainz, Johannes Gutenberg University Mainz Germany

14. TRON, Translational Oncology at the University Medical Center Johannes Gutenberg University Mainz Germany

15. Research Center for Immunotherapy University Medical Center Mainz, Johannes Gutenberg University Mainz Germany

Abstract

AbstractBackgroundIL‐17A and TNF synergistically promote inflammation and tumorigenesis. Their interplay and impact on ovarian carcinoma (OC) progression are, however, poorly understood. We addressed this question focusing on mesothelial cells, whose interaction with tumor cells is known to play a pivotal role in transcoelomic metastasis formation.MethodsFlow‐cytometry and immunohistochemistry experiments were employed to identify cellular sources of IL‐17A and TNF. Changes in transcriptomes and secretomes were determined by bulk and single cell RNA sequencing as well as affinity proteomics. Functional consequences were investigated by microscopic analyses and tumor cell adhesion assays. Potential clinical implications were assessed by immunohistochemistry and survival analyses.ResultsWe identified Th17 cells as the main population of IL‐17A‐ and TNF producers in ascites and detected their accumulation in early omental metastases. Both IL‐17A and its receptor subunit IL‐17RC were associated with short survival of OC patients, pointing to a role in clinical progression. IL‐17A and TNF synergistically induced the reprogramming of mesothelial cells towards a pro‐inflammatory mesenchymal phenotype, concomitantly with a loss of tight junctions and an impairment of mesothelial monolayer integrity, thereby promoting cancer cell adhesion. IL‐17A and TNF synergistically induced the Th17‐promoting cytokines IL‐6 and IL‐1β as well as the Th17‐attracting chemokine CCL20 in mesothelial cells, indicating a reciprocal crosstalk that potentiates the tumor‐promoting role of Th17 cells in OC.ConclusionsOur findings reveal a novel function for Th17 cells in the OC microenvironment, which entails the IL‐17A/TNF‐mediated induction of mesothelial‐mesenchymal transition, disruption of mesothelial layer integrity and consequently promotion of OC cell adhesion. These effects are potentiated by a positive feedback loop between mesothelial and Th17 cells. Together with the observed clinical associations and accumulation of Th17 cells in omental micrometastases, our observations point to a potential role in early metastases formation and thus to new therapeutic options.

Funder

Deutsche Forschungsgemeinschaft

Publisher

Wiley

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