Cargando…

Inhibition of lung cancer growth and metastasis by DHA and its metabolite, RvD1, through miR-138-5p/FOXC1 pathway

BACKGROUND: Non small cell lung cancer (NSCLC) is one of the most common cancers in the world. DHA is known to be capable of suppressing NSCLC cell proliferation and metastasis. However, the mechanisms by which DHA exhibits its antitumor effects are unknown. Here we aimed to identify the effects and...

Descripción completa

Detalles Bibliográficos
Autores principales: Bai, Xiaoming, Shao, Jiaofang, Zhou, Sujin, Zhao, Zhenggang, Li, Fanghong, Xiang, Rong, Zhao, Allan Z., Pan, Jinshun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6884860/
https://www.ncbi.nlm.nih.gov/pubmed/31783879
http://dx.doi.org/10.1186/s13046-019-1478-3
_version_ 1783474635459788800
author Bai, Xiaoming
Shao, Jiaofang
Zhou, Sujin
Zhao, Zhenggang
Li, Fanghong
Xiang, Rong
Zhao, Allan Z.
Pan, Jinshun
author_facet Bai, Xiaoming
Shao, Jiaofang
Zhou, Sujin
Zhao, Zhenggang
Li, Fanghong
Xiang, Rong
Zhao, Allan Z.
Pan, Jinshun
author_sort Bai, Xiaoming
collection PubMed
description BACKGROUND: Non small cell lung cancer (NSCLC) is one of the most common cancers in the world. DHA is known to be capable of suppressing NSCLC cell proliferation and metastasis. However, the mechanisms by which DHA exhibits its antitumor effects are unknown. Here we aimed to identify the effects and mechanisms of DHA and its metabolites on lung cancer cell growth and invasion. METHODS: As measures of cell proliferation and invasion ability, the cell viability and transwell assays were used in vitro. Transgenic mfat-1 mice, which convert ω-6 PUFAs to ω-3 PUFAs, were used to detect the effect of endogenous DHA on tumor transplantation. An LC − MS/MS analysis identified the elevation of several eicosanoid metabolites of DHA. By using qPCR miRNA microarray, online prediction software, luciferase reporter assays and Western blot analysis, we further elucidated the mechanisms. RESULTS: Addition of exogenous DHA inhibited the growth and invasion in NSCLC cells in vitro. Endogenously produced DHA attenuated LLC-derived tumor growth and metastasis in the transgenic mfat-1 mice. Among the elevation of DHA metabolites, resolvin D1 (RvD1) significantly contributed to the inhibition in cell growth and invasion. MiRNA microarray revealed that the level of miR-138-5p was significantly increased after RvD1 treatment. MiR-138-5p mimics decreased cell viability and invasion; while miR-138-5p inhibitor abolished RvD1-mediated suppression of cell viability and invasion. The expression of FOXC1 was significantly reduced upon overexpression of miR-138-5p while luciferase reporter assay showed that FOXC1 was a direct target of miR-138-5p. In vivo, endogenous DHA by the mfat-1 transgene enhanced miR-138-5p expression and decreased FOXC1 expression. Furthermore, overexpression of FOXC1 reversed the inhibition in cell viability and invasion induced by RvD1 treatment. CONCLUSIONS: These data identified the RvD1/miR-138-5p/FOXC1 pathway as a novel mechanism by DHA and its metabolite, RvD1, and the potential of targeting such pathway as a therapeutic strategy in treating NSCLC.
format Online
Article
Text
id pubmed-6884860
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-68848602019-12-03 Inhibition of lung cancer growth and metastasis by DHA and its metabolite, RvD1, through miR-138-5p/FOXC1 pathway Bai, Xiaoming Shao, Jiaofang Zhou, Sujin Zhao, Zhenggang Li, Fanghong Xiang, Rong Zhao, Allan Z. Pan, Jinshun J Exp Clin Cancer Res Research BACKGROUND: Non small cell lung cancer (NSCLC) is one of the most common cancers in the world. DHA is known to be capable of suppressing NSCLC cell proliferation and metastasis. However, the mechanisms by which DHA exhibits its antitumor effects are unknown. Here we aimed to identify the effects and mechanisms of DHA and its metabolites on lung cancer cell growth and invasion. METHODS: As measures of cell proliferation and invasion ability, the cell viability and transwell assays were used in vitro. Transgenic mfat-1 mice, which convert ω-6 PUFAs to ω-3 PUFAs, were used to detect the effect of endogenous DHA on tumor transplantation. An LC − MS/MS analysis identified the elevation of several eicosanoid metabolites of DHA. By using qPCR miRNA microarray, online prediction software, luciferase reporter assays and Western blot analysis, we further elucidated the mechanisms. RESULTS: Addition of exogenous DHA inhibited the growth and invasion in NSCLC cells in vitro. Endogenously produced DHA attenuated LLC-derived tumor growth and metastasis in the transgenic mfat-1 mice. Among the elevation of DHA metabolites, resolvin D1 (RvD1) significantly contributed to the inhibition in cell growth and invasion. MiRNA microarray revealed that the level of miR-138-5p was significantly increased after RvD1 treatment. MiR-138-5p mimics decreased cell viability and invasion; while miR-138-5p inhibitor abolished RvD1-mediated suppression of cell viability and invasion. The expression of FOXC1 was significantly reduced upon overexpression of miR-138-5p while luciferase reporter assay showed that FOXC1 was a direct target of miR-138-5p. In vivo, endogenous DHA by the mfat-1 transgene enhanced miR-138-5p expression and decreased FOXC1 expression. Furthermore, overexpression of FOXC1 reversed the inhibition in cell viability and invasion induced by RvD1 treatment. CONCLUSIONS: These data identified the RvD1/miR-138-5p/FOXC1 pathway as a novel mechanism by DHA and its metabolite, RvD1, and the potential of targeting such pathway as a therapeutic strategy in treating NSCLC. BioMed Central 2019-11-29 /pmc/articles/PMC6884860/ /pubmed/31783879 http://dx.doi.org/10.1186/s13046-019-1478-3 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
Bai, Xiaoming
Shao, Jiaofang
Zhou, Sujin
Zhao, Zhenggang
Li, Fanghong
Xiang, Rong
Zhao, Allan Z.
Pan, Jinshun
Inhibition of lung cancer growth and metastasis by DHA and its metabolite, RvD1, through miR-138-5p/FOXC1 pathway
title Inhibition of lung cancer growth and metastasis by DHA and its metabolite, RvD1, through miR-138-5p/FOXC1 pathway
title_full Inhibition of lung cancer growth and metastasis by DHA and its metabolite, RvD1, through miR-138-5p/FOXC1 pathway
title_fullStr Inhibition of lung cancer growth and metastasis by DHA and its metabolite, RvD1, through miR-138-5p/FOXC1 pathway
title_full_unstemmed Inhibition of lung cancer growth and metastasis by DHA and its metabolite, RvD1, through miR-138-5p/FOXC1 pathway
title_short Inhibition of lung cancer growth and metastasis by DHA and its metabolite, RvD1, through miR-138-5p/FOXC1 pathway
title_sort inhibition of lung cancer growth and metastasis by dha and its metabolite, rvd1, through mir-138-5p/foxc1 pathway
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6884860/
https://www.ncbi.nlm.nih.gov/pubmed/31783879
http://dx.doi.org/10.1186/s13046-019-1478-3
work_keys_str_mv AT baixiaoming inhibitionoflungcancergrowthandmetastasisbydhaanditsmetabolitervd1throughmir1385pfoxc1pathway
AT shaojiaofang inhibitionoflungcancergrowthandmetastasisbydhaanditsmetabolitervd1throughmir1385pfoxc1pathway
AT zhousujin inhibitionoflungcancergrowthandmetastasisbydhaanditsmetabolitervd1throughmir1385pfoxc1pathway
AT zhaozhenggang inhibitionoflungcancergrowthandmetastasisbydhaanditsmetabolitervd1throughmir1385pfoxc1pathway
AT lifanghong inhibitionoflungcancergrowthandmetastasisbydhaanditsmetabolitervd1throughmir1385pfoxc1pathway
AT xiangrong inhibitionoflungcancergrowthandmetastasisbydhaanditsmetabolitervd1throughmir1385pfoxc1pathway
AT zhaoallanz inhibitionoflungcancergrowthandmetastasisbydhaanditsmetabolitervd1throughmir1385pfoxc1pathway
AT panjinshun inhibitionoflungcancergrowthandmetastasisbydhaanditsmetabolitervd1throughmir1385pfoxc1pathway