Galectin-1 Mediates Chronic STING Activation in Tumors to Promote Metastasis through MDSC Recruitment

Author:

Nambiar Dhanya K.1ORCID,Viswanathan Vignesh1ORCID,Cao Hongbin1ORCID,Zhang Weiruo2ORCID,Guan Li1ORCID,Chamoli Manish3ORCID,Holmes Brittany4ORCID,Kong Christina4ORCID,Hildebrand Rachel1ORCID,Koong Amanda Jeanette1ORCID,von Eyben Rie1ORCID,Plevritis Sylvia25ORCID,Li Lingyin6ORCID,Giaccia Amato17ORCID,Engleman Edgar4ORCID,Le Quynh Thu1ORCID

Affiliation:

1. 1Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.

2. 2Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California.

3. 3Buck Institute for Research on Aging, Novato, California.

4. 4Department of Pathology, Stanford University School of Medicine, Stanford, California.

5. 5Department of Radiology, Stanford University School of Medicine, Stanford, California.

6. 6Department of Biochemistry; Program in Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, California.

7. 7MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom.

Abstract

Abstract The immune system plays a crucial role in the regulation of metastasis. Tumor cells systemically change immune functions to facilitate metastatic progression. Through this study, we deciphered how tumoral galectin-1 (Gal1) expression shapes the systemic immune environment to promote metastasis in head and neck cancer (HNC). In multiple preclinical models of HNC and lung cancer in immunogenic mice, Gal1 fostered the establishment of a premetastatic niche through polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC), which altered the local microenvironment to support metastatic spread. RNA sequencing of MDSCs from premetastatic lungs in these models demonstrated the role of PMN-MDSCs in collagen and extracellular matrix remodeling in the premetastatic compartment. Gal1 promoted MDSC accumulation in the premetastatic niche through the NF-κB signaling axis, triggering enhanced CXCL2-mediated MDSC migration. Mechanistically, Gal1 sustained NF-κB activation in tumor cells by enhancing stimulator of interferon gene (STING) protein stability, leading to prolonged inflammation-driven MDSC expansion. These findings suggest an unexpected protumoral role of STING activation in metastatic progression and establish Gal1 as an endogenous-positive regulator of STING in advanced-stage cancers. Significance: Galectin-1 increases STING stability in cancer cells that activates NF-κB signaling and CXCL2 expression to promote MDSC trafficking, which stimulates the generation of a premetastatic niche and facilitates metastatic progression.

Funder

NCI, USA

National Institute of Dental and Craniofacial Research

HHS NIH

NIH

Publisher

American Association for Cancer Research (AACR)

Subject

Cancer Research,Oncology

Reference55 articles.

1. Cancer statistics, 2021;Siegel;CA Cancer J Clin,2021

2. Emerging biological principles of metastasis;Lambert;Cell,2017

3. Pre-metastatic niches: organ-specific homes for metastases;Peinado;Nat Rev Cancer,2017

4. The immune microenvironment and cancer metastasis;El-Kenawi;Cold Spring Harb Perspect Med,2020

5. Systemic immunity in cancer;Hiam-Galvez;Nat Rev Cancer,2021

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

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