M2 macrophage-secreted exosomes promote metastasis and increase vascular permeability in hepatocellular carcinoma

Abstract Background Metastasis is a key feature of malignant tumors and significantly contributes to their high mortality, particularly in hepatocellular carcinoma (HCC). Therefore, it is imperative to explore the mechanism of tumor metastasis. Recently, tumor-associated macrophages (TAMs) have been...

Full description

Bibliographic Details
Main Authors: Yiwei Lu, Guoyong Han, Yao Zhang, Long Zhang, Zhi Li, Qingyuan Wang, Zhiqiang Chen, Xuehao Wang, Jindao Wu
Format: Article
Language:English
Published: BMC 2023-10-01
Series:Cell Communication and Signaling
Subjects:
Online Access:https://doi.org/10.1186/s12964-022-00872-w
_version_ 1797636738933325824
author Yiwei Lu
Guoyong Han
Yao Zhang
Long Zhang
Zhi Li
Qingyuan Wang
Zhiqiang Chen
Xuehao Wang
Jindao Wu
author_facet Yiwei Lu
Guoyong Han
Yao Zhang
Long Zhang
Zhi Li
Qingyuan Wang
Zhiqiang Chen
Xuehao Wang
Jindao Wu
author_sort Yiwei Lu
collection DOAJ
description Abstract Background Metastasis is a key feature of malignant tumors and significantly contributes to their high mortality, particularly in hepatocellular carcinoma (HCC). Therefore, it is imperative to explore the mechanism of tumor metastasis. Recently, tumor-associated macrophages (TAMs) have been demonstrated to promote tumor progression, while TAM-derived molecules involved in HCC metastasis warrant further investigation. Methods THP-1 was treated with IL-4 (Interleukin-4) and IL-13 (Interleukin-13) for M2 polarized macrophages. Exosomes derived from M2 macrophages were characterized. Then, HCC cells or human umbilical vein endothelial cells (HUVECs) were co-cultured with M2 macrophages or treated with M2 macrophage-secreted exosomes. Next, Transwell®, Scratch assay, tube formation, and endothelial permeability assays were performed. Moreover, RT-PCR, western blotting, immunofluorescence, and ELISA were used to assess mRNA and protein expression levels. Finally, the miRNA expression profiles of exosomes derived from M2 and M0 macrophages were analyzed. Results M2 macrophage infiltration was correlated with metastasis and a poor prognosis in HCC patients. M2-derived exosomes were absorbed by HCC and HUVEC cells and promoted the epithelial-mesenchymal transition (EMT), vascular permeability, and angiogenesis. Notably, MiR-23a-3p levels were significantly higher in M2-derived exosomes and hnRNPA1 mediated miR-23a-3p packaging into exosomes. Phosphatase and tensin homolog (PTEN) and tight junction protein 1 (TJP1) were the targets of miR-23a-3p, as confirmed by luciferase reporter assays. Lastly, HCC cells co-cultured with M2-derived exosomes secreted more GM-CSF, VEGF, G-CSF, MCP-1, and IL-4, which in turn further recruited M2 macrophages. Conclusions Our findings suggest that M2 macrophage-derived miR-23a-3p enhances HCC metastasis by promoting EMT and angiogenesis, as well as increasing vascular permeability. Video Abstract
first_indexed 2024-03-11T12:39:24Z
format Article
id doaj.art-281196f5ab4f431bbff7b5a80e087eff
institution Directory Open Access Journal
issn 1478-811X
language English
last_indexed 2024-03-11T12:39:24Z
publishDate 2023-10-01
publisher BMC
record_format Article
series Cell Communication and Signaling
spelling doaj.art-281196f5ab4f431bbff7b5a80e087eff2023-11-05T12:25:04ZengBMCCell Communication and Signaling1478-811X2023-10-0121111610.1186/s12964-022-00872-wM2 macrophage-secreted exosomes promote metastasis and increase vascular permeability in hepatocellular carcinomaYiwei Lu0Guoyong Han1Yao Zhang2Long Zhang3Zhi Li4Qingyuan Wang5Zhiqiang Chen6Xuehao Wang7Jindao Wu8Department of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical UniversityDepartment of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical UniversityDepartment of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical UniversityDepartment of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical UniversityDepartment of General Surgery, The First Affiliated Hospital of Nanjing Medical UniversityDepartment of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical UniversityDepartment of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical UniversityDepartment of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical UniversityDepartment of Hepatobiliary Surgery, The First Affiliated Hospital of Nanjing Medical UniversityAbstract Background Metastasis is a key feature of malignant tumors and significantly contributes to their high mortality, particularly in hepatocellular carcinoma (HCC). Therefore, it is imperative to explore the mechanism of tumor metastasis. Recently, tumor-associated macrophages (TAMs) have been demonstrated to promote tumor progression, while TAM-derived molecules involved in HCC metastasis warrant further investigation. Methods THP-1 was treated with IL-4 (Interleukin-4) and IL-13 (Interleukin-13) for M2 polarized macrophages. Exosomes derived from M2 macrophages were characterized. Then, HCC cells or human umbilical vein endothelial cells (HUVECs) were co-cultured with M2 macrophages or treated with M2 macrophage-secreted exosomes. Next, Transwell®, Scratch assay, tube formation, and endothelial permeability assays were performed. Moreover, RT-PCR, western blotting, immunofluorescence, and ELISA were used to assess mRNA and protein expression levels. Finally, the miRNA expression profiles of exosomes derived from M2 and M0 macrophages were analyzed. Results M2 macrophage infiltration was correlated with metastasis and a poor prognosis in HCC patients. M2-derived exosomes were absorbed by HCC and HUVEC cells and promoted the epithelial-mesenchymal transition (EMT), vascular permeability, and angiogenesis. Notably, MiR-23a-3p levels were significantly higher in M2-derived exosomes and hnRNPA1 mediated miR-23a-3p packaging into exosomes. Phosphatase and tensin homolog (PTEN) and tight junction protein 1 (TJP1) were the targets of miR-23a-3p, as confirmed by luciferase reporter assays. Lastly, HCC cells co-cultured with M2-derived exosomes secreted more GM-CSF, VEGF, G-CSF, MCP-1, and IL-4, which in turn further recruited M2 macrophages. Conclusions Our findings suggest that M2 macrophage-derived miR-23a-3p enhances HCC metastasis by promoting EMT and angiogenesis, as well as increasing vascular permeability. Video Abstracthttps://doi.org/10.1186/s12964-022-00872-wM2 macrophagesExosomesEMTAngiogenesisVascular permeabilitymiR-23a-3p
spellingShingle Yiwei Lu
Guoyong Han
Yao Zhang
Long Zhang
Zhi Li
Qingyuan Wang
Zhiqiang Chen
Xuehao Wang
Jindao Wu
M2 macrophage-secreted exosomes promote metastasis and increase vascular permeability in hepatocellular carcinoma
Cell Communication and Signaling
M2 macrophages
Exosomes
EMT
Angiogenesis
Vascular permeability
miR-23a-3p
title M2 macrophage-secreted exosomes promote metastasis and increase vascular permeability in hepatocellular carcinoma
title_full M2 macrophage-secreted exosomes promote metastasis and increase vascular permeability in hepatocellular carcinoma
title_fullStr M2 macrophage-secreted exosomes promote metastasis and increase vascular permeability in hepatocellular carcinoma
title_full_unstemmed M2 macrophage-secreted exosomes promote metastasis and increase vascular permeability in hepatocellular carcinoma
title_short M2 macrophage-secreted exosomes promote metastasis and increase vascular permeability in hepatocellular carcinoma
title_sort m2 macrophage secreted exosomes promote metastasis and increase vascular permeability in hepatocellular carcinoma
topic M2 macrophages
Exosomes
EMT
Angiogenesis
Vascular permeability
miR-23a-3p
url https://doi.org/10.1186/s12964-022-00872-w
work_keys_str_mv AT yiweilu m2macrophagesecretedexosomespromotemetastasisandincreasevascularpermeabilityinhepatocellularcarcinoma
AT guoyonghan m2macrophagesecretedexosomespromotemetastasisandincreasevascularpermeabilityinhepatocellularcarcinoma
AT yaozhang m2macrophagesecretedexosomespromotemetastasisandincreasevascularpermeabilityinhepatocellularcarcinoma
AT longzhang m2macrophagesecretedexosomespromotemetastasisandincreasevascularpermeabilityinhepatocellularcarcinoma
AT zhili m2macrophagesecretedexosomespromotemetastasisandincreasevascularpermeabilityinhepatocellularcarcinoma
AT qingyuanwang m2macrophagesecretedexosomespromotemetastasisandincreasevascularpermeabilityinhepatocellularcarcinoma
AT zhiqiangchen m2macrophagesecretedexosomespromotemetastasisandincreasevascularpermeabilityinhepatocellularcarcinoma
AT xuehaowang m2macrophagesecretedexosomespromotemetastasisandincreasevascularpermeabilityinhepatocellularcarcinoma
AT jindaowu m2macrophagesecretedexosomespromotemetastasisandincreasevascularpermeabilityinhepatocellularcarcinoma