Cargando…

Hypoxic exosomes facilitate angiogenesis and metastasis in esophageal squamous cell carcinoma through altering the phenotype and transcriptome of endothelial cells

BACKGROUND: In cancer progression, hypoxia, or low oxygen tension, is a major regulator of tumor aggressiveness and metastasis. However, how cancer cells adapt to the hypoxia and communicate with other mesenchymal cells in microenvironment during tumor development remains to be elucidated. Here, we...

Descripción completa

Detalles Bibliográficos
Autores principales: Mao, Yu, Wang, Yimin, Dong, Lixin, Zhang, Yunjie, Zhang, Yanqiu, Wang, Chao, Zhang, Qiang, Yang, Sen, Cao, Liyan, Zhang, Xinyuan, Li, Xin, Fu, Zhanzhao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6727585/
https://www.ncbi.nlm.nih.gov/pubmed/31488217
http://dx.doi.org/10.1186/s13046-019-1384-8
_version_ 1783449284055662592
author Mao, Yu
Wang, Yimin
Dong, Lixin
Zhang, Yunjie
Zhang, Yanqiu
Wang, Chao
Zhang, Qiang
Yang, Sen
Cao, Liyan
Zhang, Xinyuan
Li, Xin
Fu, Zhanzhao
author_facet Mao, Yu
Wang, Yimin
Dong, Lixin
Zhang, Yunjie
Zhang, Yanqiu
Wang, Chao
Zhang, Qiang
Yang, Sen
Cao, Liyan
Zhang, Xinyuan
Li, Xin
Fu, Zhanzhao
author_sort Mao, Yu
collection PubMed
description BACKGROUND: In cancer progression, hypoxia, or low oxygen tension, is a major regulator of tumor aggressiveness and metastasis. However, how cancer cells adapt to the hypoxia and communicate with other mesenchymal cells in microenvironment during tumor development remains to be elucidated. Here, we investigated the involvement of exosomes in modulating angiogenesis and enhancing metastasis in esophageal squamous cell carcinoma (ESCC). METHODS: Differential centrifugation, transmission electron microscopy and nanoparticle tracking analysis were used to isolate and characterize exosomes. Colony formation and transwell assay were performed to assess the proliferation, migration and invasion of human umbilical vein endothelial cells (HUVECs). The tube formation assay and matrigel plug assay were used to evaluate the vascular formation ability of HUVECs in vitro and in vivo respectively. An in vivo nude mice model was established to detect the regulatory role of exosomes in ESCC progression. Microarray analysis was performed to analyze the transcriptome profiles in HUVECs. RESULTS: Exosomes derived from ESCC cells cultured under hypoxia played a better role in promoting proliferation, migration, invasion and tube formation of HUVECs in vitro and in vivo than exosomes from ESCC cells cultured under normoxia. Moreover, hypoxic exosomes significantly enhanced the tumor growth and lung metastasis compared with normoxic exosomes in nude mice models. Interestingly, endothelial cells were programmed by hypoxic and normoxic exosomes from ESCC cells which altered the transcriptome profile of HUVECs. CONCLUSIONS: Taken together, our data identified an angiogenic role of exosomes from ESCC cells which shed light on the further application of exosomes as valuable therapeutic target for ESCC. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s13046-019-1384-8) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-6727585
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-67275852019-09-12 Hypoxic exosomes facilitate angiogenesis and metastasis in esophageal squamous cell carcinoma through altering the phenotype and transcriptome of endothelial cells Mao, Yu Wang, Yimin Dong, Lixin Zhang, Yunjie Zhang, Yanqiu Wang, Chao Zhang, Qiang Yang, Sen Cao, Liyan Zhang, Xinyuan Li, Xin Fu, Zhanzhao J Exp Clin Cancer Res Research BACKGROUND: In cancer progression, hypoxia, or low oxygen tension, is a major regulator of tumor aggressiveness and metastasis. However, how cancer cells adapt to the hypoxia and communicate with other mesenchymal cells in microenvironment during tumor development remains to be elucidated. Here, we investigated the involvement of exosomes in modulating angiogenesis and enhancing metastasis in esophageal squamous cell carcinoma (ESCC). METHODS: Differential centrifugation, transmission electron microscopy and nanoparticle tracking analysis were used to isolate and characterize exosomes. Colony formation and transwell assay were performed to assess the proliferation, migration and invasion of human umbilical vein endothelial cells (HUVECs). The tube formation assay and matrigel plug assay were used to evaluate the vascular formation ability of HUVECs in vitro and in vivo respectively. An in vivo nude mice model was established to detect the regulatory role of exosomes in ESCC progression. Microarray analysis was performed to analyze the transcriptome profiles in HUVECs. RESULTS: Exosomes derived from ESCC cells cultured under hypoxia played a better role in promoting proliferation, migration, invasion and tube formation of HUVECs in vitro and in vivo than exosomes from ESCC cells cultured under normoxia. Moreover, hypoxic exosomes significantly enhanced the tumor growth and lung metastasis compared with normoxic exosomes in nude mice models. Interestingly, endothelial cells were programmed by hypoxic and normoxic exosomes from ESCC cells which altered the transcriptome profile of HUVECs. CONCLUSIONS: Taken together, our data identified an angiogenic role of exosomes from ESCC cells which shed light on the further application of exosomes as valuable therapeutic target for ESCC. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s13046-019-1384-8) contains supplementary material, which is available to authorized users. BioMed Central 2019-09-05 /pmc/articles/PMC6727585/ /pubmed/31488217 http://dx.doi.org/10.1186/s13046-019-1384-8 Text en © The Author(s). 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Mao, Yu
Wang, Yimin
Dong, Lixin
Zhang, Yunjie
Zhang, Yanqiu
Wang, Chao
Zhang, Qiang
Yang, Sen
Cao, Liyan
Zhang, Xinyuan
Li, Xin
Fu, Zhanzhao
Hypoxic exosomes facilitate angiogenesis and metastasis in esophageal squamous cell carcinoma through altering the phenotype and transcriptome of endothelial cells
title Hypoxic exosomes facilitate angiogenesis and metastasis in esophageal squamous cell carcinoma through altering the phenotype and transcriptome of endothelial cells
title_full Hypoxic exosomes facilitate angiogenesis and metastasis in esophageal squamous cell carcinoma through altering the phenotype and transcriptome of endothelial cells
title_fullStr Hypoxic exosomes facilitate angiogenesis and metastasis in esophageal squamous cell carcinoma through altering the phenotype and transcriptome of endothelial cells
title_full_unstemmed Hypoxic exosomes facilitate angiogenesis and metastasis in esophageal squamous cell carcinoma through altering the phenotype and transcriptome of endothelial cells
title_short Hypoxic exosomes facilitate angiogenesis and metastasis in esophageal squamous cell carcinoma through altering the phenotype and transcriptome of endothelial cells
title_sort hypoxic exosomes facilitate angiogenesis and metastasis in esophageal squamous cell carcinoma through altering the phenotype and transcriptome of endothelial cells
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6727585/
https://www.ncbi.nlm.nih.gov/pubmed/31488217
http://dx.doi.org/10.1186/s13046-019-1384-8
work_keys_str_mv AT maoyu hypoxicexosomesfacilitateangiogenesisandmetastasisinesophagealsquamouscellcarcinomathroughalteringthephenotypeandtranscriptomeofendothelialcells
AT wangyimin hypoxicexosomesfacilitateangiogenesisandmetastasisinesophagealsquamouscellcarcinomathroughalteringthephenotypeandtranscriptomeofendothelialcells
AT donglixin hypoxicexosomesfacilitateangiogenesisandmetastasisinesophagealsquamouscellcarcinomathroughalteringthephenotypeandtranscriptomeofendothelialcells
AT zhangyunjie hypoxicexosomesfacilitateangiogenesisandmetastasisinesophagealsquamouscellcarcinomathroughalteringthephenotypeandtranscriptomeofendothelialcells
AT zhangyanqiu hypoxicexosomesfacilitateangiogenesisandmetastasisinesophagealsquamouscellcarcinomathroughalteringthephenotypeandtranscriptomeofendothelialcells
AT wangchao hypoxicexosomesfacilitateangiogenesisandmetastasisinesophagealsquamouscellcarcinomathroughalteringthephenotypeandtranscriptomeofendothelialcells
AT zhangqiang hypoxicexosomesfacilitateangiogenesisandmetastasisinesophagealsquamouscellcarcinomathroughalteringthephenotypeandtranscriptomeofendothelialcells
AT yangsen hypoxicexosomesfacilitateangiogenesisandmetastasisinesophagealsquamouscellcarcinomathroughalteringthephenotypeandtranscriptomeofendothelialcells
AT caoliyan hypoxicexosomesfacilitateangiogenesisandmetastasisinesophagealsquamouscellcarcinomathroughalteringthephenotypeandtranscriptomeofendothelialcells
AT zhangxinyuan hypoxicexosomesfacilitateangiogenesisandmetastasisinesophagealsquamouscellcarcinomathroughalteringthephenotypeandtranscriptomeofendothelialcells
AT lixin hypoxicexosomesfacilitateangiogenesisandmetastasisinesophagealsquamouscellcarcinomathroughalteringthephenotypeandtranscriptomeofendothelialcells
AT fuzhanzhao hypoxicexosomesfacilitateangiogenesisandmetastasisinesophagealsquamouscellcarcinomathroughalteringthephenotypeandtranscriptomeofendothelialcells