4.8 Article

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

期刊

SCIENCE ADVANCES
卷 8, 期 45, 页码 -

出版社

AMER ASSOC ADVANCEMENT SCIENCE
DOI: 10.1126/sciadv.abm3548

关键词

-

资金

  1. NIH/NCI [U01CA154209-01, P01 CA139490-05]
  2. Breast Cancer Research Foundation
  3. U.S. Department of Defense [W81XWH-11-1-0287, W81XWH-13-1-0281, W81XWH-12-1-0020]
  4. National Cancer Institute [R0490CA187192-03, R01CA255450, 5R01CA100225-09]
  5. National Cancer Institute (PHS) [CA09302]
  6. Stanford Bio-X Interdisciplinary Initiatives Seed Grants Program (IIP)
  7. Virginia and D.K. Ludwig Fund for Cancer Research
  8. Stinehart-Reed Foundation
  9. Donald E. and Delia B. Baxter Foundation
  10. Stanford School of Medicine Dean's Fellowship
  11. Stanford Bio-X Bowes Graduate Student Fellowship
  12. Stanford Medical Science Training Program
  13. NIH/NHLBI [R01-HL128503]

向作者/读者索取更多资源

A minority population of immature tumor epithelial cells with angiogenic features, marked by the oncogene LMO2, has been identified in human breast tumor biopsies. Higher abundance of LMO2(+) basal cells is correlated with tumor endothelial content and predicts poor prognosis. LMO2 binds to STAT3 and is required for STAT3 activation, promoting breast cancer metastasis.
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 immature THY1(+)/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 of LMO2(+) basal cells correlated with tumor endothelial content and predicted poor distant recurrence-free survival in patients. Using MMTV-PyMT/Lmo2(CreERT2) mice, we demonstrated that Lmo2 lineage-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-alpha 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.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.8
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据