Cargando…

MAPK signaling mediates sinomenine hydrochloride-induced human breast cancer cell death via both reactive oxygen species-dependent and -independent pathways: an in vitro and in vivo study

Sinomenine, the main alkaloid extracted from the medicinal plant Sinomenium acutum, is known for its anti-inflammatory effects. Recent studies have suggested its anti-cancer effect in synovial sarcoma, lung cancer and hepatic cancer. However, the underlying molecular mechanism for its anti-cancer ef...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, X, Wang, K, Ren, Y, Zhang, L, Tang, X-J, Zhang, H-M, Zhao, C-Q, Liu, P-J, Zhang, J-M, He, J-J
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4123109/
https://www.ncbi.nlm.nih.gov/pubmed/25077542
http://dx.doi.org/10.1038/cddis.2014.321
_version_ 1782329459540492288
author Li, X
Wang, K
Ren, Y
Zhang, L
Tang, X-J
Zhang, H-M
Zhao, C-Q
Liu, P-J
Zhang, J-M
He, J-J
author_facet Li, X
Wang, K
Ren, Y
Zhang, L
Tang, X-J
Zhang, H-M
Zhao, C-Q
Liu, P-J
Zhang, J-M
He, J-J
author_sort Li, X
collection PubMed
description Sinomenine, the main alkaloid extracted from the medicinal plant Sinomenium acutum, is known for its anti-inflammatory effects. Recent studies have suggested its anti-cancer effect in synovial sarcoma, lung cancer and hepatic cancer. However, the underlying molecular mechanism for its anti-cancer effect still remains unclear. This study investigated the anti-tumor activity of sinomenine hydrochloride (SH), a hydrochloride form of sinomenine, in human breast cancer cells in vitro and in vivo. We found that SH potently inhibited cell viability of a broad panel of breast cancer cell lines. Two representative breast cancer cell lines, namely ER(−)/PR(−) MDA-MB-231 and ER(+)/PR(+) MCF-7, were used for further investigation. The results showed that SH induced G1/S cell cycle arrest, caused apoptosis and induced ATM/Chk2- and ATR/Chk1-mediated DNA-damage response in MDA-MB-231 and MCF-7. The anti-cancer effect of SH was regulated by increased expression levels of p-ERK, p-JNK and p-38 MAPK. Further studies showed that SH resulted in an increase in reactive oxygen species (ROS) and inhibition of ROS by N-acetyl-L-cysteine (NAC) almost blocked SH-induced DNA damage but only mitigated SH-induced MAPK expression changes, suggesting that both ROS-dependent and -independent pathways were involved in MAPK-mediated SH-induced breast cancer cell death. The in vivo study demonstrated that SH effectively inhibited tumor growth without showing significant toxicity. In conclusion, SH induced breast cancer cell death through ROS-dependent and -independent pathways with an upregulation of MAPKs, indicating that SH may be a potential anti-tumor drug for breast cancer treatment.
format Online
Article
Text
id pubmed-4123109
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-41231092014-08-15 MAPK signaling mediates sinomenine hydrochloride-induced human breast cancer cell death via both reactive oxygen species-dependent and -independent pathways: an in vitro and in vivo study Li, X Wang, K Ren, Y Zhang, L Tang, X-J Zhang, H-M Zhao, C-Q Liu, P-J Zhang, J-M He, J-J Cell Death Dis Original Article Sinomenine, the main alkaloid extracted from the medicinal plant Sinomenium acutum, is known for its anti-inflammatory effects. Recent studies have suggested its anti-cancer effect in synovial sarcoma, lung cancer and hepatic cancer. However, the underlying molecular mechanism for its anti-cancer effect still remains unclear. This study investigated the anti-tumor activity of sinomenine hydrochloride (SH), a hydrochloride form of sinomenine, in human breast cancer cells in vitro and in vivo. We found that SH potently inhibited cell viability of a broad panel of breast cancer cell lines. Two representative breast cancer cell lines, namely ER(−)/PR(−) MDA-MB-231 and ER(+)/PR(+) MCF-7, were used for further investigation. The results showed that SH induced G1/S cell cycle arrest, caused apoptosis and induced ATM/Chk2- and ATR/Chk1-mediated DNA-damage response in MDA-MB-231 and MCF-7. The anti-cancer effect of SH was regulated by increased expression levels of p-ERK, p-JNK and p-38 MAPK. Further studies showed that SH resulted in an increase in reactive oxygen species (ROS) and inhibition of ROS by N-acetyl-L-cysteine (NAC) almost blocked SH-induced DNA damage but only mitigated SH-induced MAPK expression changes, suggesting that both ROS-dependent and -independent pathways were involved in MAPK-mediated SH-induced breast cancer cell death. The in vivo study demonstrated that SH effectively inhibited tumor growth without showing significant toxicity. In conclusion, SH induced breast cancer cell death through ROS-dependent and -independent pathways with an upregulation of MAPKs, indicating that SH may be a potential anti-tumor drug for breast cancer treatment. Nature Publishing Group 2014-07 2014-07-31 /pmc/articles/PMC4123109/ /pubmed/25077542 http://dx.doi.org/10.1038/cddis.2014.321 Text en Copyright © 2014 Macmillan Publishers Limited http://creativecommons.org/licenses/by-nc-nd/3.0/ Cell Death and Disease is an open-access journal published by Nature Publishing Group. This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivs 3.0 Unported License. The images or other third party material in this article are included in the article's Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/3.0/
spellingShingle Original Article
Li, X
Wang, K
Ren, Y
Zhang, L
Tang, X-J
Zhang, H-M
Zhao, C-Q
Liu, P-J
Zhang, J-M
He, J-J
MAPK signaling mediates sinomenine hydrochloride-induced human breast cancer cell death via both reactive oxygen species-dependent and -independent pathways: an in vitro and in vivo study
title MAPK signaling mediates sinomenine hydrochloride-induced human breast cancer cell death via both reactive oxygen species-dependent and -independent pathways: an in vitro and in vivo study
title_full MAPK signaling mediates sinomenine hydrochloride-induced human breast cancer cell death via both reactive oxygen species-dependent and -independent pathways: an in vitro and in vivo study
title_fullStr MAPK signaling mediates sinomenine hydrochloride-induced human breast cancer cell death via both reactive oxygen species-dependent and -independent pathways: an in vitro and in vivo study
title_full_unstemmed MAPK signaling mediates sinomenine hydrochloride-induced human breast cancer cell death via both reactive oxygen species-dependent and -independent pathways: an in vitro and in vivo study
title_short MAPK signaling mediates sinomenine hydrochloride-induced human breast cancer cell death via both reactive oxygen species-dependent and -independent pathways: an in vitro and in vivo study
title_sort mapk signaling mediates sinomenine hydrochloride-induced human breast cancer cell death via both reactive oxygen species-dependent and -independent pathways: an in vitro and in vivo study
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4123109/
https://www.ncbi.nlm.nih.gov/pubmed/25077542
http://dx.doi.org/10.1038/cddis.2014.321
work_keys_str_mv AT lix mapksignalingmediatessinomeninehydrochlorideinducedhumanbreastcancercelldeathviabothreactiveoxygenspeciesdependentandindependentpathwaysaninvitroandinvivostudy
AT wangk mapksignalingmediatessinomeninehydrochlorideinducedhumanbreastcancercelldeathviabothreactiveoxygenspeciesdependentandindependentpathwaysaninvitroandinvivostudy
AT reny mapksignalingmediatessinomeninehydrochlorideinducedhumanbreastcancercelldeathviabothreactiveoxygenspeciesdependentandindependentpathwaysaninvitroandinvivostudy
AT zhangl mapksignalingmediatessinomeninehydrochlorideinducedhumanbreastcancercelldeathviabothreactiveoxygenspeciesdependentandindependentpathwaysaninvitroandinvivostudy
AT tangxj mapksignalingmediatessinomeninehydrochlorideinducedhumanbreastcancercelldeathviabothreactiveoxygenspeciesdependentandindependentpathwaysaninvitroandinvivostudy
AT zhanghm mapksignalingmediatessinomeninehydrochlorideinducedhumanbreastcancercelldeathviabothreactiveoxygenspeciesdependentandindependentpathwaysaninvitroandinvivostudy
AT zhaocq mapksignalingmediatessinomeninehydrochlorideinducedhumanbreastcancercelldeathviabothreactiveoxygenspeciesdependentandindependentpathwaysaninvitroandinvivostudy
AT liupj mapksignalingmediatessinomeninehydrochlorideinducedhumanbreastcancercelldeathviabothreactiveoxygenspeciesdependentandindependentpathwaysaninvitroandinvivostudy
AT zhangjm mapksignalingmediatessinomeninehydrochlorideinducedhumanbreastcancercelldeathviabothreactiveoxygenspeciesdependentandindependentpathwaysaninvitroandinvivostudy
AT hejj mapksignalingmediatessinomeninehydrochlorideinducedhumanbreastcancercelldeathviabothreactiveoxygenspeciesdependentandindependentpathwaysaninvitroandinvivostudy