Autophagy-dependent cell cycle arrest in esophageal cancer cells exposed to dihydroartemisinin

Abstract Background Dihydroartemisinin (DHA), a derivate of artemisinin, is an effective antimalarial agent. DHA has been shown to exert anticancer activities to numerous cancer cells in the past few years, while the exact molecular mechanisms remain to be elucidated, especially in esophageal cancer...

Full description

Bibliographic Details
Main Authors: Qiang Ma, Hebin Liao, Lei Xu, Qingrong Li, Jiang Zou, Ru Sun, Dan Xiao, Chang Liu, Wenjie Pu, Jibing Cheng, Xi Zhou, Guangcheng Huang, Lihua Yao, Xiaowu Zhong, Xiaolan Guo
Format: Article
Language:English
Published: BMC 2020-04-01
Series:Chinese Medicine
Subjects:
Online Access:http://link.springer.com/article/10.1186/s13020-020-00318-w
_version_ 1811331663667396608
author Qiang Ma
Hebin Liao
Lei Xu
Qingrong Li
Jiang Zou
Ru Sun
Dan Xiao
Chang Liu
Wenjie Pu
Jibing Cheng
Xi Zhou
Guangcheng Huang
Lihua Yao
Xiaowu Zhong
Xiaolan Guo
author_facet Qiang Ma
Hebin Liao
Lei Xu
Qingrong Li
Jiang Zou
Ru Sun
Dan Xiao
Chang Liu
Wenjie Pu
Jibing Cheng
Xi Zhou
Guangcheng Huang
Lihua Yao
Xiaowu Zhong
Xiaolan Guo
author_sort Qiang Ma
collection DOAJ
description Abstract Background Dihydroartemisinin (DHA), a derivate of artemisinin, is an effective antimalarial agent. DHA has been shown to exert anticancer activities to numerous cancer cells in the past few years, while the exact molecular mechanisms remain to be elucidated, especially in esophageal cancer. Methods Crystal violet assay was conducted to determine the cell viability of human esophageal cancer cell line Eca109 treated with DHA. Tumor-bearing nude mice were employed to evaluate the anticancer effect of DHA in vivo. Soft agar and crystal violet assays were used to measure the tumorigenicity of Eca109 cells. Flow cytometry was performed to evaluate ROS or cell cycle distribution. GFP-LC3 plasmids were delivered into Eca109 cells to visualize autophagy induced by DHA under a fluorescence microscope. The mRNA and protein levels of each gene were tested by qRT-PCR and western blot, respectively. Results Our results proved that DHA significantly reduced the viability of Eca109 cells in a dose- and time-dependent manner. Further investigation showed that DHA evidently induced cell cycle arrest at the G2/M phase in Eca109 cells. Mechanistically, DHA induced intracellular ROS generation and autophagy in Eca109 cells, while blocking ROS by an antioxidant NAC obviously inhibited autophagy. Furthermore, we found that telomere shelterin component TRF2 was down-regulated in Eca109 cells exposed to DHA through autophagy-dependent degradation, which could be rescued after autophagy was blocked by ROS inhibition. Moreover, the DNA damage response (DDR) was induced obviously in DHA treated cells. To further explore whether ROS or autophagy played a vital role in DHA induced cell cycle arrest, the cell cycle distribution of Eca109 cells was evaluated after ROS or autophagy blocking, and the results showed that autophagy, but not ROS, was essential for cell cycle arrest in DHA treated cells. Conclusion Taken together, DHA showed anticancer effect on esophageal cancer cells through autophagy-dependent cell cycle arrest at the G2/M phase, which unveiled a novel mechanism of DHA as a chemotherapeutic agent, and the degradation of TRF2 followed by DDR might be responsible for this cell phenotype.
first_indexed 2024-04-13T16:24:05Z
format Article
id doaj.art-94a1c04b700e42069a59ef5f28b4f8d0
institution Directory Open Access Journal
issn 1749-8546
language English
last_indexed 2024-04-13T16:24:05Z
publishDate 2020-04-01
publisher BMC
record_format Article
series Chinese Medicine
spelling doaj.art-94a1c04b700e42069a59ef5f28b4f8d02022-12-22T02:39:48ZengBMCChinese Medicine1749-85462020-04-0115111310.1186/s13020-020-00318-wAutophagy-dependent cell cycle arrest in esophageal cancer cells exposed to dihydroartemisininQiang Ma0Hebin Liao1Lei Xu2Qingrong Li3Jiang Zou4Ru Sun5Dan Xiao6Chang Liu7Wenjie Pu8Jibing Cheng9Xi Zhou10Guangcheng Huang11Lihua Yao12Xiaowu Zhong13Xiaolan Guo14Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical CollegeTranslational Medicine Research Center, North Sichuan Medical CollegeTranslational Medicine Research Center, North Sichuan Medical CollegeDepartment of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical CollegeDepartment of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical CollegeDepartment of Laboratory Medicine, North Sichuan Medical CollegeDepartment of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical CollegeDepartment of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical CollegeDepartment of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical CollegeDepartment of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical CollegeDepartment of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical CollegeDepartment of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical CollegeDepartment of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical CollegeDepartment of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical CollegeDepartment of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical CollegeAbstract Background Dihydroartemisinin (DHA), a derivate of artemisinin, is an effective antimalarial agent. DHA has been shown to exert anticancer activities to numerous cancer cells in the past few years, while the exact molecular mechanisms remain to be elucidated, especially in esophageal cancer. Methods Crystal violet assay was conducted to determine the cell viability of human esophageal cancer cell line Eca109 treated with DHA. Tumor-bearing nude mice were employed to evaluate the anticancer effect of DHA in vivo. Soft agar and crystal violet assays were used to measure the tumorigenicity of Eca109 cells. Flow cytometry was performed to evaluate ROS or cell cycle distribution. GFP-LC3 plasmids were delivered into Eca109 cells to visualize autophagy induced by DHA under a fluorescence microscope. The mRNA and protein levels of each gene were tested by qRT-PCR and western blot, respectively. Results Our results proved that DHA significantly reduced the viability of Eca109 cells in a dose- and time-dependent manner. Further investigation showed that DHA evidently induced cell cycle arrest at the G2/M phase in Eca109 cells. Mechanistically, DHA induced intracellular ROS generation and autophagy in Eca109 cells, while blocking ROS by an antioxidant NAC obviously inhibited autophagy. Furthermore, we found that telomere shelterin component TRF2 was down-regulated in Eca109 cells exposed to DHA through autophagy-dependent degradation, which could be rescued after autophagy was blocked by ROS inhibition. Moreover, the DNA damage response (DDR) was induced obviously in DHA treated cells. To further explore whether ROS or autophagy played a vital role in DHA induced cell cycle arrest, the cell cycle distribution of Eca109 cells was evaluated after ROS or autophagy blocking, and the results showed that autophagy, but not ROS, was essential for cell cycle arrest in DHA treated cells. Conclusion Taken together, DHA showed anticancer effect on esophageal cancer cells through autophagy-dependent cell cycle arrest at the G2/M phase, which unveiled a novel mechanism of DHA as a chemotherapeutic agent, and the degradation of TRF2 followed by DDR might be responsible for this cell phenotype.http://link.springer.com/article/10.1186/s13020-020-00318-wCell cycle arrestAutophagyTRF2DihydroartemisininEsophageal cancer
spellingShingle Qiang Ma
Hebin Liao
Lei Xu
Qingrong Li
Jiang Zou
Ru Sun
Dan Xiao
Chang Liu
Wenjie Pu
Jibing Cheng
Xi Zhou
Guangcheng Huang
Lihua Yao
Xiaowu Zhong
Xiaolan Guo
Autophagy-dependent cell cycle arrest in esophageal cancer cells exposed to dihydroartemisinin
Chinese Medicine
Cell cycle arrest
Autophagy
TRF2
Dihydroartemisinin
Esophageal cancer
title Autophagy-dependent cell cycle arrest in esophageal cancer cells exposed to dihydroartemisinin
title_full Autophagy-dependent cell cycle arrest in esophageal cancer cells exposed to dihydroartemisinin
title_fullStr Autophagy-dependent cell cycle arrest in esophageal cancer cells exposed to dihydroartemisinin
title_full_unstemmed Autophagy-dependent cell cycle arrest in esophageal cancer cells exposed to dihydroartemisinin
title_short Autophagy-dependent cell cycle arrest in esophageal cancer cells exposed to dihydroartemisinin
title_sort autophagy dependent cell cycle arrest in esophageal cancer cells exposed to dihydroartemisinin
topic Cell cycle arrest
Autophagy
TRF2
Dihydroartemisinin
Esophageal cancer
url http://link.springer.com/article/10.1186/s13020-020-00318-w
work_keys_str_mv AT qiangma autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT hebinliao autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT leixu autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT qingrongli autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT jiangzou autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT rusun autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT danxiao autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT changliu autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT wenjiepu autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT jibingcheng autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT xizhou autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT guangchenghuang autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT lihuayao autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT xiaowuzhong autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin
AT xiaolanguo autophagydependentcellcyclearrestinesophagealcancercellsexposedtodihydroartemisinin