Cargando…

Andrographolide induces apoptosis in human osteosarcoma cells via the ROS/JNK pathway

Osteosarcoma is the most common primary malignant tumor of the bone and the long-term survival of patients with this disease has remained unsatisfactory over the past several decades. Andrographolide, a traditional drug used in Chinese medicine, has been found to exert a significant antitumor effect...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Shengdong, Li, Hengyuan, Chen, Shi, Wang, Zenan, Yao, Yuhong, Chen, Tao, Ye, Zhaoming, Lin, Peng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7170044/
https://www.ncbi.nlm.nih.gov/pubmed/32236589
http://dx.doi.org/10.3892/ijo.2020.5032
_version_ 1783523834981253120
author Wang, Shengdong
Li, Hengyuan
Chen, Shi
Wang, Zenan
Yao, Yuhong
Chen, Tao
Ye, Zhaoming
Lin, Peng
author_facet Wang, Shengdong
Li, Hengyuan
Chen, Shi
Wang, Zenan
Yao, Yuhong
Chen, Tao
Ye, Zhaoming
Lin, Peng
author_sort Wang, Shengdong
collection PubMed
description Osteosarcoma is the most common primary malignant tumor of the bone and the long-term survival of patients with this disease has remained unsatisfactory over the past several decades. Andrographolide, a traditional drug used in Chinese medicine, has been found to exert a significant antitumor effect against several types of cancer. However, relatively little is known about the effect of andrographolide on osteosarcoma and the underlying mechanisms. In the present study, it was shown that andrographolide inhibited osteosarcoma cell proliferation by arresting the cell cycle at the G2/M phase and increasing caspase-mediated apoptosis. Furthermore, treatment with andrographolide induced JNK activation and increased production of reactive oxygen species (ROS). The andrographolide-triggered apoptosis in osteosarcoma cells was partly abrogated by a JNK inhibitor and completely reversed by a ROS scavenger. Additionally, JNK activation and cell cycle arrest at the G2/M phase were prevented by administration of an ROS scavenger. In vivo, it was also found that andrographolide inhibited tumor growth by increasing the levels of ROS and activating JNK; thus inducing cytotoxicity in primary osteosarcoma cells. Together, the results of the present study suggest that andrographolide caused G2/M arrest and induced cell apoptosis via regulation of the ROS/JNK signaling pathway in osteosarcoma cells. Thus, andrographolide may serve as a promising antitumor therapeutic agent against osteosarcoma.
format Online
Article
Text
id pubmed-7170044
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-71700442020-04-23 Andrographolide induces apoptosis in human osteosarcoma cells via the ROS/JNK pathway Wang, Shengdong Li, Hengyuan Chen, Shi Wang, Zenan Yao, Yuhong Chen, Tao Ye, Zhaoming Lin, Peng Int J Oncol Articles Osteosarcoma is the most common primary malignant tumor of the bone and the long-term survival of patients with this disease has remained unsatisfactory over the past several decades. Andrographolide, a traditional drug used in Chinese medicine, has been found to exert a significant antitumor effect against several types of cancer. However, relatively little is known about the effect of andrographolide on osteosarcoma and the underlying mechanisms. In the present study, it was shown that andrographolide inhibited osteosarcoma cell proliferation by arresting the cell cycle at the G2/M phase and increasing caspase-mediated apoptosis. Furthermore, treatment with andrographolide induced JNK activation and increased production of reactive oxygen species (ROS). The andrographolide-triggered apoptosis in osteosarcoma cells was partly abrogated by a JNK inhibitor and completely reversed by a ROS scavenger. Additionally, JNK activation and cell cycle arrest at the G2/M phase were prevented by administration of an ROS scavenger. In vivo, it was also found that andrographolide inhibited tumor growth by increasing the levels of ROS and activating JNK; thus inducing cytotoxicity in primary osteosarcoma cells. Together, the results of the present study suggest that andrographolide caused G2/M arrest and induced cell apoptosis via regulation of the ROS/JNK signaling pathway in osteosarcoma cells. Thus, andrographolide may serve as a promising antitumor therapeutic agent against osteosarcoma. D.A. Spandidos 2020-03-30 /pmc/articles/PMC7170044/ /pubmed/32236589 http://dx.doi.org/10.3892/ijo.2020.5032 Text en Copyright: © Wang et al. 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
Wang, Shengdong
Li, Hengyuan
Chen, Shi
Wang, Zenan
Yao, Yuhong
Chen, Tao
Ye, Zhaoming
Lin, Peng
Andrographolide induces apoptosis in human osteosarcoma cells via the ROS/JNK pathway
title Andrographolide induces apoptosis in human osteosarcoma cells via the ROS/JNK pathway
title_full Andrographolide induces apoptosis in human osteosarcoma cells via the ROS/JNK pathway
title_fullStr Andrographolide induces apoptosis in human osteosarcoma cells via the ROS/JNK pathway
title_full_unstemmed Andrographolide induces apoptosis in human osteosarcoma cells via the ROS/JNK pathway
title_short Andrographolide induces apoptosis in human osteosarcoma cells via the ROS/JNK pathway
title_sort andrographolide induces apoptosis in human osteosarcoma cells via the ros/jnk pathway
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7170044/
https://www.ncbi.nlm.nih.gov/pubmed/32236589
http://dx.doi.org/10.3892/ijo.2020.5032
work_keys_str_mv AT wangshengdong andrographolideinducesapoptosisinhumanosteosarcomacellsviatherosjnkpathway
AT lihengyuan andrographolideinducesapoptosisinhumanosteosarcomacellsviatherosjnkpathway
AT chenshi andrographolideinducesapoptosisinhumanosteosarcomacellsviatherosjnkpathway
AT wangzenan andrographolideinducesapoptosisinhumanosteosarcomacellsviatherosjnkpathway
AT yaoyuhong andrographolideinducesapoptosisinhumanosteosarcomacellsviatherosjnkpathway
AT chentao andrographolideinducesapoptosisinhumanosteosarcomacellsviatherosjnkpathway
AT yezhaoming andrographolideinducesapoptosisinhumanosteosarcomacellsviatherosjnkpathway
AT linpeng andrographolideinducesapoptosisinhumanosteosarcomacellsviatherosjnkpathway