Cargando…

Tumor-associated macrophage-derived exosomes promote EGFR-TKI resistance in non-small cell lung cancer by regulating the AKT, ERK1/2 and STAT3 signaling pathways

The evolutionary properties of organisms lead to the issue of targeted drug resistance. Numerous clinical trials have shown that tumor-associated macrophages (TAMs) in patients with lung cancer adversely affect the clinical efficacy of epithelial growth factor receptor (EGFR) tyrosine kinase inhibit...

Descripción completa

Detalles Bibliográficos
Autores principales: Yuan, Shiyang, Chen, Wenjun, Yang, Jian, Zheng, Yuanhai, Ye, Wen, Xie, Hui, Dong, Lie, Xie, Junping
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9478622/
https://www.ncbi.nlm.nih.gov/pubmed/36168315
http://dx.doi.org/10.3892/ol.2022.13476
_version_ 1784790613320794112
author Yuan, Shiyang
Chen, Wenjun
Yang, Jian
Zheng, Yuanhai
Ye, Wen
Xie, Hui
Dong, Lie
Xie, Junping
author_facet Yuan, Shiyang
Chen, Wenjun
Yang, Jian
Zheng, Yuanhai
Ye, Wen
Xie, Hui
Dong, Lie
Xie, Junping
author_sort Yuan, Shiyang
collection PubMed
description The evolutionary properties of organisms lead to the issue of targeted drug resistance. Numerous clinical trials have shown that tumor-associated macrophages (TAMs) in patients with lung cancer adversely affect the clinical efficacy of epithelial growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs). However, the mechanism by which TAMs influence the tumor cell response to TKIs remains unclear. The aim of the present study was to investigate the influence of TAM-derived exosomes on the sensitivity of PC9 and HCC827 lung adenocarcinoma cells to the EGFR inhibitor gefitinib. Multiple cytokines were used to induce the differentiation of THP-1 human leukemia monocytes into macrophages in vitro. The obtained cells were identified as TAMs by cytomorphology and flow cytometry. Exosomes were extracted from the TAM culture supernatants and identified using electron microscopy and nanoparticle tracking analysis. Flow cytometry was used to examine the apoptosis of lung adenocarcinoma cells when treated with gefitinib and/or TAM-derived exosomes. In addition, western blotting was used to detect the expression of the key proteins of the AKT, ERK1/2 and STAT3 signaling pathways. TAM-derived exosomes were successfully obtained. The TAM-derived exosomes were shown to affect the proliferation and apoptosis of lung adenocarcinoma cells. Furthermore, the killing effect of gefitinib on the tumor cells was attenuated. The mechanism underlying the effects of the TAM-derived exosomes may be associated with reactivation of the AKT, ERK1/2 and STAT3 signaling pathways. In conclusion, the findings indicate that TAM-derived exosomes promote resistance to gefitinib in non-small cell lung cancer (NSCLC), and the mechanism may be associated with reactivation of the AKT, ERK1/2 and STAT3 signaling pathways. This study may serve as a reference in the exploration of alternative strategies for NSCLC following the development of resistance to EGFR-targeted drugs.
format Online
Article
Text
id pubmed-9478622
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-94786222022-09-26 Tumor-associated macrophage-derived exosomes promote EGFR-TKI resistance in non-small cell lung cancer by regulating the AKT, ERK1/2 and STAT3 signaling pathways Yuan, Shiyang Chen, Wenjun Yang, Jian Zheng, Yuanhai Ye, Wen Xie, Hui Dong, Lie Xie, Junping Oncol Lett Articles The evolutionary properties of organisms lead to the issue of targeted drug resistance. Numerous clinical trials have shown that tumor-associated macrophages (TAMs) in patients with lung cancer adversely affect the clinical efficacy of epithelial growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs). However, the mechanism by which TAMs influence the tumor cell response to TKIs remains unclear. The aim of the present study was to investigate the influence of TAM-derived exosomes on the sensitivity of PC9 and HCC827 lung adenocarcinoma cells to the EGFR inhibitor gefitinib. Multiple cytokines were used to induce the differentiation of THP-1 human leukemia monocytes into macrophages in vitro. The obtained cells were identified as TAMs by cytomorphology and flow cytometry. Exosomes were extracted from the TAM culture supernatants and identified using electron microscopy and nanoparticle tracking analysis. Flow cytometry was used to examine the apoptosis of lung adenocarcinoma cells when treated with gefitinib and/or TAM-derived exosomes. In addition, western blotting was used to detect the expression of the key proteins of the AKT, ERK1/2 and STAT3 signaling pathways. TAM-derived exosomes were successfully obtained. The TAM-derived exosomes were shown to affect the proliferation and apoptosis of lung adenocarcinoma cells. Furthermore, the killing effect of gefitinib on the tumor cells was attenuated. The mechanism underlying the effects of the TAM-derived exosomes may be associated with reactivation of the AKT, ERK1/2 and STAT3 signaling pathways. In conclusion, the findings indicate that TAM-derived exosomes promote resistance to gefitinib in non-small cell lung cancer (NSCLC), and the mechanism may be associated with reactivation of the AKT, ERK1/2 and STAT3 signaling pathways. This study may serve as a reference in the exploration of alternative strategies for NSCLC following the development of resistance to EGFR-targeted drugs. D.A. Spandidos 2022-08-24 /pmc/articles/PMC9478622/ /pubmed/36168315 http://dx.doi.org/10.3892/ol.2022.13476 Text en Copyright: © Yuan et al. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Yuan, Shiyang
Chen, Wenjun
Yang, Jian
Zheng, Yuanhai
Ye, Wen
Xie, Hui
Dong, Lie
Xie, Junping
Tumor-associated macrophage-derived exosomes promote EGFR-TKI resistance in non-small cell lung cancer by regulating the AKT, ERK1/2 and STAT3 signaling pathways
title Tumor-associated macrophage-derived exosomes promote EGFR-TKI resistance in non-small cell lung cancer by regulating the AKT, ERK1/2 and STAT3 signaling pathways
title_full Tumor-associated macrophage-derived exosomes promote EGFR-TKI resistance in non-small cell lung cancer by regulating the AKT, ERK1/2 and STAT3 signaling pathways
title_fullStr Tumor-associated macrophage-derived exosomes promote EGFR-TKI resistance in non-small cell lung cancer by regulating the AKT, ERK1/2 and STAT3 signaling pathways
title_full_unstemmed Tumor-associated macrophage-derived exosomes promote EGFR-TKI resistance in non-small cell lung cancer by regulating the AKT, ERK1/2 and STAT3 signaling pathways
title_short Tumor-associated macrophage-derived exosomes promote EGFR-TKI resistance in non-small cell lung cancer by regulating the AKT, ERK1/2 and STAT3 signaling pathways
title_sort tumor-associated macrophage-derived exosomes promote egfr-tki resistance in non-small cell lung cancer by regulating the akt, erk1/2 and stat3 signaling pathways
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9478622/
https://www.ncbi.nlm.nih.gov/pubmed/36168315
http://dx.doi.org/10.3892/ol.2022.13476
work_keys_str_mv AT yuanshiyang tumorassociatedmacrophagederivedexosomespromoteegfrtkiresistanceinnonsmallcelllungcancerbyregulatingtheakterk12andstat3signalingpathways
AT chenwenjun tumorassociatedmacrophagederivedexosomespromoteegfrtkiresistanceinnonsmallcelllungcancerbyregulatingtheakterk12andstat3signalingpathways
AT yangjian tumorassociatedmacrophagederivedexosomespromoteegfrtkiresistanceinnonsmallcelllungcancerbyregulatingtheakterk12andstat3signalingpathways
AT zhengyuanhai tumorassociatedmacrophagederivedexosomespromoteegfrtkiresistanceinnonsmallcelllungcancerbyregulatingtheakterk12andstat3signalingpathways
AT yewen tumorassociatedmacrophagederivedexosomespromoteegfrtkiresistanceinnonsmallcelllungcancerbyregulatingtheakterk12andstat3signalingpathways
AT xiehui tumorassociatedmacrophagederivedexosomespromoteegfrtkiresistanceinnonsmallcelllungcancerbyregulatingtheakterk12andstat3signalingpathways
AT donglie tumorassociatedmacrophagederivedexosomespromoteegfrtkiresistanceinnonsmallcelllungcancerbyregulatingtheakterk12andstat3signalingpathways
AT xiejunping tumorassociatedmacrophagederivedexosomespromoteegfrtkiresistanceinnonsmallcelllungcancerbyregulatingtheakterk12andstat3signalingpathways