Advanced Science (Nov 2020)

Nidogen 1‐Enriched Extracellular Vesicles Facilitate Extrahepatic Metastasis of Liver Cancer by Activating Pulmonary Fibroblasts to Secrete Tumor Necrosis Factor Receptor 1

  • Xiaowen Mao,
  • Sze Keong Tey,
  • Cherlie Lot Sum Yeung,
  • Ernest Man Lok Kwong,
  • Yi Man Eva Fung,
  • Clive Yik Sham Chung,
  • Lung‐Yi Mak,
  • Danny Ka Ho Wong,
  • Man‐Fung Yuen,
  • James Chung Man Ho,
  • Herbert Pang,
  • Maria Pik Wong,
  • Carmen Oi‐Ning Leung,
  • Terence Kin Wah Lee,
  • Victor Ma,
  • William Chi‐Shing Cho,
  • Peihua Cao,
  • Xiaoping Xu,
  • Yi Gao,
  • Judy Wai Ping Yam

DOI
https://doi.org/10.1002/advs.202002157
Journal volume & issue
Vol. 7, no. 21
pp. n/a – n/a

Abstract

Read online

Abstract In hepatocellular carcinoma (HCC) patients with extrahepatic metastasis, the lung is the most frequent site of metastasis. However, how the lung microenvironment favors disseminated cells remains unclear. Here, it is found that nidogen 1 (NID1) in metastatic HCC cell‐derived extracellular vesicles (EVs) promotes pre‐metastatic niche formation in the lung by enhancing angiogenesis and pulmonary endothelial permeability to facilitate colonization of tumor cells and extrahepatic metastasis. EV‐NID1 also activates fibroblasts, which secrete tumor necrosis factor receptor 1 (TNFR1), facilitate lung colonization of tumor cells, and augment HCC cell growth and motility. Administration of anti‐TNFR1 antibody effectively diminishes lung metastasis induced by the metastatic HCC cell‐derived EVs in mice. In the clinical perspective, analysis of serum EV‐NID1 and TNFR1 in HCC patients reveals their positive correlation and association with tumor stages suggesting the potential of these molecules as noninvasive biomarkers for the early detection of HCC. In conclusion, these results demonstrate the interplay of HCC EVs and activated fibroblasts in pre‐metastatic niche formation and how blockage of their functions inhibits distant metastasis to the lungs. This study offers promise for the new direction of HCC treatment by targeting oncogenic EV components and their mediated pathways.

Keywords