Abstract

Cancer-associated fibroblasts (CAFs) are major component of tumor microenvironment (TME), which plays crucial roles in tumor growth, invasion and metastasis; however, the underling mechanism is not fully elucidated. Despite many studies are focused on the tumor promoting effect of CAFs-derived cytokines, the upstream regulators of cytokine release in CAFs is largely unknown. Here we found that miR-101-3p was downregulated in primary lung cancer-associated CAFs compared to normal fibroblasts (NFs). Ectopic overexpression of miR-101-3p suppressed CAFs activation, and abrogated the promoting effect of CAFs on migration and invasion of non-small cell lung cancer cells (NSCLC), through attenuating CAFs’ effect on epithelial mesenchymal transition (EMT) process, metastasis-related genes (MMP9, TWIST1) and AKT/endothelial nitric oxide synthase (eNOS) signaling pathway. Further study indicated that vascular endothelial growth factor A (VEGFA) was a novel target of miR-101-3p, and CAFs-derived VEGFA mediated the effect of miR-101-3p on migration and invasion of lung cancer cells, demonstrated by using recombinant VEGFA and VEGFA neutralizing antibody. Interestingly, the analysis of the Cancer Genome Atlas (TCGA) database revealed that lung cancer tissues expressed lower level of miR-101-3p than non-cancerous tissues, and low/medium-expression of miR-101-3p was associated with poor overall survival (OS) rate. Moreover, the mouse xenograft experiment also showed that CAFs accelerated tumor growth whereas miR-101-3p diminished CAFs’ effect. These findings revealed a novel mechanism that CAFs facilitated lung cancer metastasis potential via miR-101-3p/VEGFA/AKT signaling pathway, suggesting miR-101-3p as a potential candidate for metastasis therapy.

Highlights

  • Lung cancer is the main course of cancer-related mortality worldwide

  • We further demonstrated that downregulation of miR-101-3p in Cancer-associated fibroblasts (CAFs) increased vascular endothelial growth factor A (VEGFA) secretion, facilitating the metastasis potential of lung cancer cells via activation of Akt/endothelial nitric oxide synthase (eNOS) signaling pathway

  • CAFs are major components of tumor stroma, in the present study, we established the culture of CAFs derived from human lung cancer tissues, and investigated their effects on metastasis potential of lung cancer cells

Read more

Summary

Introduction

Lung cancer is the main course of cancer-related mortality worldwide. According to the report of the World Health Organization (WHO), there were estimated 2.2 million new lung cancer cases and 1.8 million deaths in 2020 (Sung et al, 2021). CAFs are one of the major stromal cells in TME. CAFs support and promote tumor progression via secreting cytokines and growth factors. CAFs upregulated CXCR4, β-catenin, PPARδ, and enhanced invasiveness of lung adenocarcinoma by secretion of stromal cell-derived factor (SDF-1) (Wang Y. et al, 2021). Zhang et al (2019) reported that human colorectal cancer-derived CAFs stimulated adhesion of colorectal cancer cells to endothelial cells via releasing hepatocyte growth factor (HGF). Our previous studies showed that CAFs facilitate metastasis and chemoresistance of lung cancer cells through IL-6 and ANXA3 secretion (Wang et al, 2017; Wang et al, 2019). Despite many studies are focused on the promoting effect of CAFs-derived cytokines on cancer cells, the upstream regulators of cytokine release in CAFs is largely unknown

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call