Cargando…

Fangchinoline Inhibits Human Esophageal Cancer by Transactivating ATF4 to Trigger Both Noxa-Dependent Intrinsic and DR5-Dependent Extrinsic Apoptosis

Esophageal squamous cell carcinoma (ESCC) is a recalcitrant cancer. The Chinese herbal monomer fangchinoline (FCL) has been reported to have anti-tumor activity in several human cancer cell types. However, the therapeutic efficacy and underlying mechanism on ESCC remain to be elucidated. In the pres...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Yunjing, Wang, Shiwen, Chen, Yukun, Zhang, Junqian, Yang, Jing, Xian, Jingrong, Li, Lihui, Zhao, Hu, Hoffman, Robert M., Zhang, Yanmei, Jia, Lijun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8236818/
https://www.ncbi.nlm.nih.gov/pubmed/34195076
http://dx.doi.org/10.3389/fonc.2021.666549
_version_ 1783714622332731392
author Zhang, Yunjing
Wang, Shiwen
Chen, Yukun
Zhang, Junqian
Yang, Jing
Xian, Jingrong
Li, Lihui
Zhao, Hu
Hoffman, Robert M.
Zhang, Yanmei
Jia, Lijun
author_facet Zhang, Yunjing
Wang, Shiwen
Chen, Yukun
Zhang, Junqian
Yang, Jing
Xian, Jingrong
Li, Lihui
Zhao, Hu
Hoffman, Robert M.
Zhang, Yanmei
Jia, Lijun
author_sort Zhang, Yunjing
collection PubMed
description Esophageal squamous cell carcinoma (ESCC) is a recalcitrant cancer. The Chinese herbal monomer fangchinoline (FCL) has been reported to have anti-tumor activity in several human cancer cell types. However, the therapeutic efficacy and underlying mechanism on ESCC remain to be elucidated. In the present study, for the first time, we demonstrated that FCL significantly suppressed the growth of ESCC both in vitro and in vivo. Mechanistic studies revealed that FCL-induced G1 phase cell-cycle arrest in ESCC which is dependent on p21 and p27. Moreover, we found that FCL coordinatively triggered Noxa-dependent intrinsic apoptosis and DR5-dependent extrinsic apoptosis by transactivating ATF4, which is a novel mechanism. Our findings elucidated the tumor-suppressive efficacy and mechanisms of FCL and demonstrated FCL is a potential anti-ESCC agent.
format Online
Article
Text
id pubmed-8236818
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-82368182021-06-29 Fangchinoline Inhibits Human Esophageal Cancer by Transactivating ATF4 to Trigger Both Noxa-Dependent Intrinsic and DR5-Dependent Extrinsic Apoptosis Zhang, Yunjing Wang, Shiwen Chen, Yukun Zhang, Junqian Yang, Jing Xian, Jingrong Li, Lihui Zhao, Hu Hoffman, Robert M. Zhang, Yanmei Jia, Lijun Front Oncol Oncology Esophageal squamous cell carcinoma (ESCC) is a recalcitrant cancer. The Chinese herbal monomer fangchinoline (FCL) has been reported to have anti-tumor activity in several human cancer cell types. However, the therapeutic efficacy and underlying mechanism on ESCC remain to be elucidated. In the present study, for the first time, we demonstrated that FCL significantly suppressed the growth of ESCC both in vitro and in vivo. Mechanistic studies revealed that FCL-induced G1 phase cell-cycle arrest in ESCC which is dependent on p21 and p27. Moreover, we found that FCL coordinatively triggered Noxa-dependent intrinsic apoptosis and DR5-dependent extrinsic apoptosis by transactivating ATF4, which is a novel mechanism. Our findings elucidated the tumor-suppressive efficacy and mechanisms of FCL and demonstrated FCL is a potential anti-ESCC agent. Frontiers Media S.A. 2021-06-14 /pmc/articles/PMC8236818/ /pubmed/34195076 http://dx.doi.org/10.3389/fonc.2021.666549 Text en Copyright © 2021 Zhang, Wang, Chen, Zhang, Yang, Xian, Li, Zhao, Hoffman, Zhang and Jia https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Oncology
Zhang, Yunjing
Wang, Shiwen
Chen, Yukun
Zhang, Junqian
Yang, Jing
Xian, Jingrong
Li, Lihui
Zhao, Hu
Hoffman, Robert M.
Zhang, Yanmei
Jia, Lijun
Fangchinoline Inhibits Human Esophageal Cancer by Transactivating ATF4 to Trigger Both Noxa-Dependent Intrinsic and DR5-Dependent Extrinsic Apoptosis
title Fangchinoline Inhibits Human Esophageal Cancer by Transactivating ATF4 to Trigger Both Noxa-Dependent Intrinsic and DR5-Dependent Extrinsic Apoptosis
title_full Fangchinoline Inhibits Human Esophageal Cancer by Transactivating ATF4 to Trigger Both Noxa-Dependent Intrinsic and DR5-Dependent Extrinsic Apoptosis
title_fullStr Fangchinoline Inhibits Human Esophageal Cancer by Transactivating ATF4 to Trigger Both Noxa-Dependent Intrinsic and DR5-Dependent Extrinsic Apoptosis
title_full_unstemmed Fangchinoline Inhibits Human Esophageal Cancer by Transactivating ATF4 to Trigger Both Noxa-Dependent Intrinsic and DR5-Dependent Extrinsic Apoptosis
title_short Fangchinoline Inhibits Human Esophageal Cancer by Transactivating ATF4 to Trigger Both Noxa-Dependent Intrinsic and DR5-Dependent Extrinsic Apoptosis
title_sort fangchinoline inhibits human esophageal cancer by transactivating atf4 to trigger both noxa-dependent intrinsic and dr5-dependent extrinsic apoptosis
topic Oncology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8236818/
https://www.ncbi.nlm.nih.gov/pubmed/34195076
http://dx.doi.org/10.3389/fonc.2021.666549
work_keys_str_mv AT zhangyunjing fangchinolineinhibitshumanesophagealcancerbytransactivatingatf4totriggerbothnoxadependentintrinsicanddr5dependentextrinsicapoptosis
AT wangshiwen fangchinolineinhibitshumanesophagealcancerbytransactivatingatf4totriggerbothnoxadependentintrinsicanddr5dependentextrinsicapoptosis
AT chenyukun fangchinolineinhibitshumanesophagealcancerbytransactivatingatf4totriggerbothnoxadependentintrinsicanddr5dependentextrinsicapoptosis
AT zhangjunqian fangchinolineinhibitshumanesophagealcancerbytransactivatingatf4totriggerbothnoxadependentintrinsicanddr5dependentextrinsicapoptosis
AT yangjing fangchinolineinhibitshumanesophagealcancerbytransactivatingatf4totriggerbothnoxadependentintrinsicanddr5dependentextrinsicapoptosis
AT xianjingrong fangchinolineinhibitshumanesophagealcancerbytransactivatingatf4totriggerbothnoxadependentintrinsicanddr5dependentextrinsicapoptosis
AT lilihui fangchinolineinhibitshumanesophagealcancerbytransactivatingatf4totriggerbothnoxadependentintrinsicanddr5dependentextrinsicapoptosis
AT zhaohu fangchinolineinhibitshumanesophagealcancerbytransactivatingatf4totriggerbothnoxadependentintrinsicanddr5dependentextrinsicapoptosis
AT hoffmanrobertm fangchinolineinhibitshumanesophagealcancerbytransactivatingatf4totriggerbothnoxadependentintrinsicanddr5dependentextrinsicapoptosis
AT zhangyanmei fangchinolineinhibitshumanesophagealcancerbytransactivatingatf4totriggerbothnoxadependentintrinsicanddr5dependentextrinsicapoptosis
AT jialijun fangchinolineinhibitshumanesophagealcancerbytransactivatingatf4totriggerbothnoxadependentintrinsicanddr5dependentextrinsicapoptosis