Cargando…

A Novel Microtubule-Disrupting Agent Induces Endoplasmic Reticular Stress-Mediated Cell Death in Human Hepatocellular Carcinoma Cells

Here, we present evidence of a novel microtubule-disrupting agent, N-deacetyl-N-(chromone-2-carbonyl)-thiocolchicine (TCD), exhibiting potent antitumor activity (with IC(50) values in the nanomolar range) against hepatocellular carcinoma cell lines. Cell cycle analysis revealed that TCD induced G(2)...

Descripción completa

Detalles Bibliográficos
Autores principales: Ho, Chun-Te, Chang, Yu-Jia, Yang, Li-Xi, Wei, Po-Li, Liu, Tsan-Zon, Liu, Jun-Jen
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4565632/
https://www.ncbi.nlm.nih.gov/pubmed/26355599
http://dx.doi.org/10.1371/journal.pone.0136340
_version_ 1782389600020332544
author Ho, Chun-Te
Chang, Yu-Jia
Yang, Li-Xi
Wei, Po-Li
Liu, Tsan-Zon
Liu, Jun-Jen
author_facet Ho, Chun-Te
Chang, Yu-Jia
Yang, Li-Xi
Wei, Po-Li
Liu, Tsan-Zon
Liu, Jun-Jen
author_sort Ho, Chun-Te
collection PubMed
description Here, we present evidence of a novel microtubule-disrupting agent, N-deacetyl-N-(chromone-2-carbonyl)-thiocolchicine (TCD), exhibiting potent antitumor activity (with IC(50) values in the nanomolar range) against hepatocellular carcinoma cell lines. Cell cycle analysis revealed that TCD induced G(2)/M cell-cycle arrest in a dose- and time-dependent manner in both Hep-J5 and Mahlavu HCC cell lines. TCD also induced a decrease in mitochondrial membrane potential (ΔΨm) and caused DNA damage. Mechanistically, TCD activated protein kinase RNA-like endoplasmic reticular kinase and several transcription factors, including activating transcription factor (ATF) 6, ATF4, ATF3, and the CCAAT-enhancer binding protein homologous protein. These data clearly demonstrate that the antitumor activity of TCD is mechanistically linked to its capacity to trigger both intrinsic and extrinsic apoptotic cell death via endoplasmic reticular stress pathway. The potent antitumor activity of TCD was similarly demonstrated in a hepatocellular carcinoma xenograft model, where 5 and 10 mg/kg doses of TCD significantly arrested Hep-J5 and Mahlavu tumor growth. Our finding suggests that TCD is a promising therapeutic agent against hepatocellular carcinoma; further translational assessment of its clinical usage is warranted.
format Online
Article
Text
id pubmed-4565632
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-45656322015-09-18 A Novel Microtubule-Disrupting Agent Induces Endoplasmic Reticular Stress-Mediated Cell Death in Human Hepatocellular Carcinoma Cells Ho, Chun-Te Chang, Yu-Jia Yang, Li-Xi Wei, Po-Li Liu, Tsan-Zon Liu, Jun-Jen PLoS One Research Article Here, we present evidence of a novel microtubule-disrupting agent, N-deacetyl-N-(chromone-2-carbonyl)-thiocolchicine (TCD), exhibiting potent antitumor activity (with IC(50) values in the nanomolar range) against hepatocellular carcinoma cell lines. Cell cycle analysis revealed that TCD induced G(2)/M cell-cycle arrest in a dose- and time-dependent manner in both Hep-J5 and Mahlavu HCC cell lines. TCD also induced a decrease in mitochondrial membrane potential (ΔΨm) and caused DNA damage. Mechanistically, TCD activated protein kinase RNA-like endoplasmic reticular kinase and several transcription factors, including activating transcription factor (ATF) 6, ATF4, ATF3, and the CCAAT-enhancer binding protein homologous protein. These data clearly demonstrate that the antitumor activity of TCD is mechanistically linked to its capacity to trigger both intrinsic and extrinsic apoptotic cell death via endoplasmic reticular stress pathway. The potent antitumor activity of TCD was similarly demonstrated in a hepatocellular carcinoma xenograft model, where 5 and 10 mg/kg doses of TCD significantly arrested Hep-J5 and Mahlavu tumor growth. Our finding suggests that TCD is a promising therapeutic agent against hepatocellular carcinoma; further translational assessment of its clinical usage is warranted. Public Library of Science 2015-09-10 /pmc/articles/PMC4565632/ /pubmed/26355599 http://dx.doi.org/10.1371/journal.pone.0136340 Text en © 2015 Ho et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Ho, Chun-Te
Chang, Yu-Jia
Yang, Li-Xi
Wei, Po-Li
Liu, Tsan-Zon
Liu, Jun-Jen
A Novel Microtubule-Disrupting Agent Induces Endoplasmic Reticular Stress-Mediated Cell Death in Human Hepatocellular Carcinoma Cells
title A Novel Microtubule-Disrupting Agent Induces Endoplasmic Reticular Stress-Mediated Cell Death in Human Hepatocellular Carcinoma Cells
title_full A Novel Microtubule-Disrupting Agent Induces Endoplasmic Reticular Stress-Mediated Cell Death in Human Hepatocellular Carcinoma Cells
title_fullStr A Novel Microtubule-Disrupting Agent Induces Endoplasmic Reticular Stress-Mediated Cell Death in Human Hepatocellular Carcinoma Cells
title_full_unstemmed A Novel Microtubule-Disrupting Agent Induces Endoplasmic Reticular Stress-Mediated Cell Death in Human Hepatocellular Carcinoma Cells
title_short A Novel Microtubule-Disrupting Agent Induces Endoplasmic Reticular Stress-Mediated Cell Death in Human Hepatocellular Carcinoma Cells
title_sort novel microtubule-disrupting agent induces endoplasmic reticular stress-mediated cell death in human hepatocellular carcinoma cells
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4565632/
https://www.ncbi.nlm.nih.gov/pubmed/26355599
http://dx.doi.org/10.1371/journal.pone.0136340
work_keys_str_mv AT hochunte anovelmicrotubuledisruptingagentinducesendoplasmicreticularstressmediatedcelldeathinhumanhepatocellularcarcinomacells
AT changyujia anovelmicrotubuledisruptingagentinducesendoplasmicreticularstressmediatedcelldeathinhumanhepatocellularcarcinomacells
AT yanglixi anovelmicrotubuledisruptingagentinducesendoplasmicreticularstressmediatedcelldeathinhumanhepatocellularcarcinomacells
AT weipoli anovelmicrotubuledisruptingagentinducesendoplasmicreticularstressmediatedcelldeathinhumanhepatocellularcarcinomacells
AT liutsanzon anovelmicrotubuledisruptingagentinducesendoplasmicreticularstressmediatedcelldeathinhumanhepatocellularcarcinomacells
AT liujunjen anovelmicrotubuledisruptingagentinducesendoplasmicreticularstressmediatedcelldeathinhumanhepatocellularcarcinomacells
AT hochunte novelmicrotubuledisruptingagentinducesendoplasmicreticularstressmediatedcelldeathinhumanhepatocellularcarcinomacells
AT changyujia novelmicrotubuledisruptingagentinducesendoplasmicreticularstressmediatedcelldeathinhumanhepatocellularcarcinomacells
AT yanglixi novelmicrotubuledisruptingagentinducesendoplasmicreticularstressmediatedcelldeathinhumanhepatocellularcarcinomacells
AT weipoli novelmicrotubuledisruptingagentinducesendoplasmicreticularstressmediatedcelldeathinhumanhepatocellularcarcinomacells
AT liutsanzon novelmicrotubuledisruptingagentinducesendoplasmicreticularstressmediatedcelldeathinhumanhepatocellularcarcinomacells
AT liujunjen novelmicrotubuledisruptingagentinducesendoplasmicreticularstressmediatedcelldeathinhumanhepatocellularcarcinomacells