Back to Search Start Over

Loss of endothelial EMCN drives tumor lung metastasis through the premetastatic niche.

Authors :
Zhang, Guoxin
Li, Mengyuan
Zhou, Dandan
Yang, Xingjiu
Zhang, Wenlong
Gao, Ran
Source :
Journal of Translational Medicine. 10/2/2022, Vol. 20 Issue 1, p1-18. 18p.
Publication Year :
2022

Abstract

<bold>Background: </bold>Metastasis is the primary cause of cancer-related mortality. Metastasis involves a complex multistep process during which individual tumor cells spread primarily through destruction of the endothelial barrier, entering the circulatory system to colonize distant organs. However, the role of the endothelial barrier as the rate-limiting process in tumor metastasis and how these processes affect the regulation of the host microenvironment at the molecular level are poorly understood.<bold>Methods: </bold>Here, we analyzed differentially expressed genes in breast cancer and lung adenocarcinoma, including metastatic and recurrent specimens, using TCGA dataset. The effects of EMCN on endothelial cells in vitro and in vivo were analyzed by assessing angiogenesis and vascular permeability, respectively. We established a syngeneic mouse model of endothelial cell-specific knockout of EMCN (EMCNecko) to study the role of EMCN in tumor growth and metastasis. Transcriptome sequencing, Western blotting, qPCR and immunofluorescence confirmed important factors in the premetastatic niche. A mouse model of allograft tumor resection with lung metastasis was established to confirm the therapeutic effect of a notch inhibitor combined with an anti-TGF-β antibody.<bold>Results: </bold>We found a strong correlation of EMCN deficiency with tumor recurrence and metastasis. Comparative experiments in WT and EMCNecko mice revealed that endothelial EMCN deficiency did not affect primary tumor growth significantly but strongly promoted spontaneous metastasis. EMCN deficiency was associated with gene profiles that regulate cell junctions in vitro and enhance vascular permeability in vivo. Mechanistically, EMCN deficiency mainly affected the host microenvironment and led to the formation of a lung premetastatic niche by recruiting Ly6G+ neutrophils and upregulating MMP9, S100A8/A9 and TGF-β expression. Anti-TGF-β antibody effectively eliminated TGF-β-induced neutrophil polarization, thereby reducing lung metastasis. Notably, the combination of a Notch inhibitor and an anti-TGF-β antibody effectively inhibited tumor growth and lung metastasis and prolonged the survival time of mice.<bold>Conclusions: </bold>We present a new translational strategy of EMCN as a new key player in tumor lung metastasis by affecting the host microenvironment. These findings could provide a sound theoretical basis for clinical treatment. [ABSTRACT FROM AUTHOR]

Details

Language :
English
ISSN :
14795876
Volume :
20
Issue :
1
Database :
Academic Search Index
Journal :
Journal of Translational Medicine
Publication Type :
Academic Journal
Accession number :
159440610
Full Text :
https://doi.org/10.1186/s12967-022-03649-4