Cargando…

RA and ω-3 PUFA co-treatment activates autophagy in cancer cells

Retinoic acid (RA), is a promising therapeutic agent for the treatment of breast cancer. However, metabolic disorders and drug resistance reduce the efficacy of RA. In this study, we found that RA and ω-3 polyunsaturated fatty acids (ω-3 PUFAs) synergistically induced cell death in vitro and in vivo...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhu, Shenglong, Lin, Guangxiao, Song, Ci, Wu, Yikuan, Feng, Ninghan, Chen, Wei, He, Zhao, Chen, Yong Q.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5752509/
https://www.ncbi.nlm.nih.gov/pubmed/29312596
http://dx.doi.org/10.18632/oncotarget.22629
_version_ 1783290120256880640
author Zhu, Shenglong
Lin, Guangxiao
Song, Ci
Wu, Yikuan
Feng, Ninghan
Chen, Wei
He, Zhao
Chen, Yong Q.
author_facet Zhu, Shenglong
Lin, Guangxiao
Song, Ci
Wu, Yikuan
Feng, Ninghan
Chen, Wei
He, Zhao
Chen, Yong Q.
author_sort Zhu, Shenglong
collection PubMed
description Retinoic acid (RA), is a promising therapeutic agent for the treatment of breast cancer. However, metabolic disorders and drug resistance reduce the efficacy of RA. In this study, we found that RA and ω-3 polyunsaturated fatty acids (ω-3 PUFAs) synergistically induced cell death in vitro and in vivo and autophagy activation. Moreover, RA-induced hypercholesterolemia was completely corrected by ω-3 PUFA supplementation. In addition, we demonstrated that the effects of this combination on the autophagic flux were independent of the two major canonic regulatory complexes controlling autophagic vesicle formation. The treatment activated Gαq-p38 MAPK signaling pathways, which resulted in autophagy of breast cancer cells. Knockdown of Gαq or P38 expression prevented RA and ω-3 PUFAs from inducing autophagy. Data indicated that Gαq-p38activation was mediated by the co-activation of GPR40 and RARα in lipid rafts, rather than by the activation of GPR120, RARβ, or RARγ. The results of this study suggest that hyperlipidemic drug side effects may be ameliorated by the administration of ω-3 PUFAs. Thus, the therapeutic indexes of the corresponding drugs may be increased.
format Online
Article
Text
id pubmed-5752509
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-57525092018-01-08 RA and ω-3 PUFA co-treatment activates autophagy in cancer cells Zhu, Shenglong Lin, Guangxiao Song, Ci Wu, Yikuan Feng, Ninghan Chen, Wei He, Zhao Chen, Yong Q. Oncotarget Research Paper Retinoic acid (RA), is a promising therapeutic agent for the treatment of breast cancer. However, metabolic disorders and drug resistance reduce the efficacy of RA. In this study, we found that RA and ω-3 polyunsaturated fatty acids (ω-3 PUFAs) synergistically induced cell death in vitro and in vivo and autophagy activation. Moreover, RA-induced hypercholesterolemia was completely corrected by ω-3 PUFA supplementation. In addition, we demonstrated that the effects of this combination on the autophagic flux were independent of the two major canonic regulatory complexes controlling autophagic vesicle formation. The treatment activated Gαq-p38 MAPK signaling pathways, which resulted in autophagy of breast cancer cells. Knockdown of Gαq or P38 expression prevented RA and ω-3 PUFAs from inducing autophagy. Data indicated that Gαq-p38activation was mediated by the co-activation of GPR40 and RARα in lipid rafts, rather than by the activation of GPR120, RARβ, or RARγ. The results of this study suggest that hyperlipidemic drug side effects may be ameliorated by the administration of ω-3 PUFAs. Thus, the therapeutic indexes of the corresponding drugs may be increased. Impact Journals LLC 2017-11-22 /pmc/articles/PMC5752509/ /pubmed/29312596 http://dx.doi.org/10.18632/oncotarget.22629 Text en Copyright: © 2017 Zhu et al. http://creativecommons.org/licenses/by/3.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0/) 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Zhu, Shenglong
Lin, Guangxiao
Song, Ci
Wu, Yikuan
Feng, Ninghan
Chen, Wei
He, Zhao
Chen, Yong Q.
RA and ω-3 PUFA co-treatment activates autophagy in cancer cells
title RA and ω-3 PUFA co-treatment activates autophagy in cancer cells
title_full RA and ω-3 PUFA co-treatment activates autophagy in cancer cells
title_fullStr RA and ω-3 PUFA co-treatment activates autophagy in cancer cells
title_full_unstemmed RA and ω-3 PUFA co-treatment activates autophagy in cancer cells
title_short RA and ω-3 PUFA co-treatment activates autophagy in cancer cells
title_sort ra and ω-3 pufa co-treatment activates autophagy in cancer cells
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5752509/
https://www.ncbi.nlm.nih.gov/pubmed/29312596
http://dx.doi.org/10.18632/oncotarget.22629
work_keys_str_mv AT zhushenglong raandō3pufacotreatmentactivatesautophagyincancercells
AT linguangxiao raandō3pufacotreatmentactivatesautophagyincancercells
AT songci raandō3pufacotreatmentactivatesautophagyincancercells
AT wuyikuan raandō3pufacotreatmentactivatesautophagyincancercells
AT fengninghan raandō3pufacotreatmentactivatesautophagyincancercells
AT chenwei raandō3pufacotreatmentactivatesautophagyincancercells
AT hezhao raandō3pufacotreatmentactivatesautophagyincancercells
AT chenyongq raandō3pufacotreatmentactivatesautophagyincancercells