Identification of a minority population of LMO2+breast cancer cells that integrate into the vasculature and initiate metastasis

Author:

Sikandar Shaheen S.1ORCID,Gulati Gunsagar S.2,Antony Jane2ORCID,Fetter Isobel1ORCID,Kuo Angera H.2ORCID,Ho William Hai Dang2ORCID,Haro-Acosta Veronica1ORCID,Das Soumyashree3,Steen Chloé B.4ORCID,Pereira Thiago Almeida2ORCID,Qian Dalong2ORCID,Beachy Philip A.2ORCID,Dirbas Fredrick5ORCID,Red-Horse Kristy236ORCID,Rabbitts Terence H.7ORCID,Thiery Jean Paul8,Newman Aaron M.29ORCID,Clarke Michael F.210ORCID

Affiliation:

1. Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.

2. Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, School of Medicine, Stanford, CA 94305, USA.

3. Department of Biology, Stanford University, Stanford, CA 94305, USA.

4. Division of Oncology, Department of Medicine, Stanford Cancer Institute, Stanford University, Stanford, CA, USA.

5. Department of Surgery, Stanford Cancer Institute, Stanford University School of Medicine, 875 Blake Wilbur Drive, Rm CC2235, Stanford, CA 94305, USA.

6. Howard Hughes Medical Institute, Stanford, CA 94305, USA.

7. Division of Cancer Therapeutics, Institute of Cancer Research, London SM2 5NG, UK.

8. Guangzhou Laboratory, International Biological Island, Guangzhou, Guangdong 510005, China.

9. Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA.

10. Department of Medicine, Stanford University, Stanford, CA 94305, USA.

Abstract

Metastasis is responsible for most breast cancer–related deaths; however, identifying the cellular determinants of metastasis has remained challenging. Here, we identified a minority population of immatureTHY1+/VEGFA+tumor epithelial cells in human breast tumor biopsies that display angiogenic features and are marked by the expression of the oncogene,LMO2. Higher abundance ofLMO2+basal cells correlated with tumor endothelial content and predicted poor distant recurrence–free survival in patients. UsingMMTV-PyMT/Lmo2CreERT2mice, we demonstrated thatLmo2lineage–traced cells integrate into the vasculature and have a higher propensity to metastasize. LMO2 knockdown in human breast tumors reduced lung metastasis by impairing intravasation, leading to a reduced frequency of circulating tumor cells. Mechanistically, we find that LMO2 binds to STAT3 and is required for STAT3 activation by tumor necrosis factor–α and interleukin-6. Collectively, our study identifies a population of metastasis-initiating cells with angiogenic features and establishes the LMO2-STAT3 signaling axis as a therapeutic target in breast cancer metastasis.

Publisher

American Association for the Advancement of Science (AAAS)

Subject

Multidisciplinary

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