Cargando…

Breast cancer cell-derived extracellular vesicles transfer miR-182-5p and promote breast carcinogenesis via the CMTM7/EGFR/AKT axis

BACKGROUND: Extracellular vesicles (EVs) derived from tumor cells are implicated in the progression of malignancies through the transfer of molecular cargo microRNAs (miRNAs or miRs). We aimed to explore the role of EVs derived from breast cancer cells carrying miR-182-5p in the occurrence and devel...

Descripción completa

Detalles Bibliográficos
Autores principales: Lu, Chong, Zhao, Yu, Wang, Jing, Shi, Wei, Dong, Fang, Xin, Yue, Zhao, Xiangwang, Liu, Chunping
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8296627/
https://www.ncbi.nlm.nih.gov/pubmed/34294040
http://dx.doi.org/10.1186/s10020-021-00338-8
_version_ 1783725681295753216
author Lu, Chong
Zhao, Yu
Wang, Jing
Shi, Wei
Dong, Fang
Xin, Yue
Zhao, Xiangwang
Liu, Chunping
author_facet Lu, Chong
Zhao, Yu
Wang, Jing
Shi, Wei
Dong, Fang
Xin, Yue
Zhao, Xiangwang
Liu, Chunping
author_sort Lu, Chong
collection PubMed
description BACKGROUND: Extracellular vesicles (EVs) derived from tumor cells are implicated in the progression of malignancies through the transfer of molecular cargo microRNAs (miRNAs or miRs). We aimed to explore the role of EVs derived from breast cancer cells carrying miR-182-5p in the occurrence and development of breast cancer. METHODS: Differentially expressed miRNAs and their downstream target genes related to breast cancer were screened through GEO and TCGA databases. miR-182-5p expression was examined in cancer tissues and adjacent normal tissues from patients with breast cancer. EVs were isolated from breast cancer cell line MDA-MB-231 cells and identified. The gain- and loss-of function approaches of miR-182-5p and CKLF-like MARVEL transmembrane domain-containing 7 (CMTM7) were performed in MDA-MB-231 cells and the isolated EVs. Human umbilical vein endothelial cells (HUVECs) were subjected to co-culture with MDA-MB-231 cell-derived EVs and biological behaviors were detected by CCK-8 assay, flow cytometry, immunohistochemical staining, Transwell assay and vessel-like tube formation in vitro. A xenograft mouse model in nude mice was established to observe the tumorigenesis and metastasis of breast cancer cells in vivo. RESULTS: miR-182-5p was highly expressed in breast cancer tissues and cells, and this high expression was associated with poor prognosis of breast cancer patients. miR-182-5p overexpression was shown to promote tumor angiogenesis in breast cancer. Moreover, our data indicated that miR-182-5p was highly enriched in EVs from MDA-MD-231 cells and then ultimately enhanced the proliferation, migration, and angiogenesis of HUVECs in vitro and in vivo. Moreover, we found that CMTM7 is a target of miR-182-5p. EVs-miR-182-5p promotes tumorigenesis and metastasis of breast cancer cells by regulating the CMTM7/EGFR/AKT signaling axis. CONCLUSIONS: Taken altogether, our findings demonstrates that EVs secreted by breast cancer cells could carry miR-182-5p to aggravate breast cancer through downregulating CMTM7 expression and activating the EGFR/AKT signaling pathway. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s10020-021-00338-8.
format Online
Article
Text
id pubmed-8296627
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-82966272021-07-22 Breast cancer cell-derived extracellular vesicles transfer miR-182-5p and promote breast carcinogenesis via the CMTM7/EGFR/AKT axis Lu, Chong Zhao, Yu Wang, Jing Shi, Wei Dong, Fang Xin, Yue Zhao, Xiangwang Liu, Chunping Mol Med Research Article BACKGROUND: Extracellular vesicles (EVs) derived from tumor cells are implicated in the progression of malignancies through the transfer of molecular cargo microRNAs (miRNAs or miRs). We aimed to explore the role of EVs derived from breast cancer cells carrying miR-182-5p in the occurrence and development of breast cancer. METHODS: Differentially expressed miRNAs and their downstream target genes related to breast cancer were screened through GEO and TCGA databases. miR-182-5p expression was examined in cancer tissues and adjacent normal tissues from patients with breast cancer. EVs were isolated from breast cancer cell line MDA-MB-231 cells and identified. The gain- and loss-of function approaches of miR-182-5p and CKLF-like MARVEL transmembrane domain-containing 7 (CMTM7) were performed in MDA-MB-231 cells and the isolated EVs. Human umbilical vein endothelial cells (HUVECs) were subjected to co-culture with MDA-MB-231 cell-derived EVs and biological behaviors were detected by CCK-8 assay, flow cytometry, immunohistochemical staining, Transwell assay and vessel-like tube formation in vitro. A xenograft mouse model in nude mice was established to observe the tumorigenesis and metastasis of breast cancer cells in vivo. RESULTS: miR-182-5p was highly expressed in breast cancer tissues and cells, and this high expression was associated with poor prognosis of breast cancer patients. miR-182-5p overexpression was shown to promote tumor angiogenesis in breast cancer. Moreover, our data indicated that miR-182-5p was highly enriched in EVs from MDA-MD-231 cells and then ultimately enhanced the proliferation, migration, and angiogenesis of HUVECs in vitro and in vivo. Moreover, we found that CMTM7 is a target of miR-182-5p. EVs-miR-182-5p promotes tumorigenesis and metastasis of breast cancer cells by regulating the CMTM7/EGFR/AKT signaling axis. CONCLUSIONS: Taken altogether, our findings demonstrates that EVs secreted by breast cancer cells could carry miR-182-5p to aggravate breast cancer through downregulating CMTM7 expression and activating the EGFR/AKT signaling pathway. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s10020-021-00338-8. BioMed Central 2021-07-16 /pmc/articles/PMC8296627/ /pubmed/34294040 http://dx.doi.org/10.1186/s10020-021-00338-8 Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Research Article
Lu, Chong
Zhao, Yu
Wang, Jing
Shi, Wei
Dong, Fang
Xin, Yue
Zhao, Xiangwang
Liu, Chunping
Breast cancer cell-derived extracellular vesicles transfer miR-182-5p and promote breast carcinogenesis via the CMTM7/EGFR/AKT axis
title Breast cancer cell-derived extracellular vesicles transfer miR-182-5p and promote breast carcinogenesis via the CMTM7/EGFR/AKT axis
title_full Breast cancer cell-derived extracellular vesicles transfer miR-182-5p and promote breast carcinogenesis via the CMTM7/EGFR/AKT axis
title_fullStr Breast cancer cell-derived extracellular vesicles transfer miR-182-5p and promote breast carcinogenesis via the CMTM7/EGFR/AKT axis
title_full_unstemmed Breast cancer cell-derived extracellular vesicles transfer miR-182-5p and promote breast carcinogenesis via the CMTM7/EGFR/AKT axis
title_short Breast cancer cell-derived extracellular vesicles transfer miR-182-5p and promote breast carcinogenesis via the CMTM7/EGFR/AKT axis
title_sort breast cancer cell-derived extracellular vesicles transfer mir-182-5p and promote breast carcinogenesis via the cmtm7/egfr/akt axis
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8296627/
https://www.ncbi.nlm.nih.gov/pubmed/34294040
http://dx.doi.org/10.1186/s10020-021-00338-8
work_keys_str_mv AT luchong breastcancercellderivedextracellularvesiclestransfermir1825pandpromotebreastcarcinogenesisviathecmtm7egfraktaxis
AT zhaoyu breastcancercellderivedextracellularvesiclestransfermir1825pandpromotebreastcarcinogenesisviathecmtm7egfraktaxis
AT wangjing breastcancercellderivedextracellularvesiclestransfermir1825pandpromotebreastcarcinogenesisviathecmtm7egfraktaxis
AT shiwei breastcancercellderivedextracellularvesiclestransfermir1825pandpromotebreastcarcinogenesisviathecmtm7egfraktaxis
AT dongfang breastcancercellderivedextracellularvesiclestransfermir1825pandpromotebreastcarcinogenesisviathecmtm7egfraktaxis
AT xinyue breastcancercellderivedextracellularvesiclestransfermir1825pandpromotebreastcarcinogenesisviathecmtm7egfraktaxis
AT zhaoxiangwang breastcancercellderivedextracellularvesiclestransfermir1825pandpromotebreastcarcinogenesisviathecmtm7egfraktaxis
AT liuchunping breastcancercellderivedextracellularvesiclestransfermir1825pandpromotebreastcarcinogenesisviathecmtm7egfraktaxis