Cargando…

DFIQ, a Novel Quinoline Derivative, Shows Anticancer Potential by Inducing Apoptosis and Autophagy in NSCLC Cell and In Vivo Zebrafish Xenograft Models

Lung cancer is one of the deadliest cancers worldwide due to chemoresistance in patients with late-stage disease. Quinoline derivatives show biological activity against HIV, malaria, bacteriuria, and cancer. DFIQ is a novel synthetic quinoline derivative that induces cell death in both in vitro and...

Descripción completa

Detalles Bibliográficos
Autores principales: Huang, Hurng-Wern, Bow, Yung-Ding, Wang, Chia-Yih, Chen, Yen-Chun, Fu, Pei-Rong, Chang, Kuo-Feng, Wang, Tso-Wen, Tseng, Chih-Hua, Chen, Yeh-Long, Chiu, Chien-Chih
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7281296/
https://www.ncbi.nlm.nih.gov/pubmed/32466291
http://dx.doi.org/10.3390/cancers12051348
_version_ 1783543890252398592
author Huang, Hurng-Wern
Bow, Yung-Ding
Wang, Chia-Yih
Chen, Yen-Chun
Fu, Pei-Rong
Chang, Kuo-Feng
Wang, Tso-Wen
Tseng, Chih-Hua
Chen, Yeh-Long
Chiu, Chien-Chih
author_facet Huang, Hurng-Wern
Bow, Yung-Ding
Wang, Chia-Yih
Chen, Yen-Chun
Fu, Pei-Rong
Chang, Kuo-Feng
Wang, Tso-Wen
Tseng, Chih-Hua
Chen, Yeh-Long
Chiu, Chien-Chih
author_sort Huang, Hurng-Wern
collection PubMed
description Lung cancer is one of the deadliest cancers worldwide due to chemoresistance in patients with late-stage disease. Quinoline derivatives show biological activity against HIV, malaria, bacteriuria, and cancer. DFIQ is a novel synthetic quinoline derivative that induces cell death in both in vitro and in vivo zebrafish xenograft models. DFIQ induced cell death, including apoptosis, and the IC(50) values were 4.16 and 2.31 μM at 24 and 48 h, respectively. DFIQ was also found to induce apoptotic protein cleavage and DNA damage, reduce cell cycle-associated protein expression, and disrupt reactive oxygen species (ROS) reduction, thus resulting in the accumulation of superoxide radicals. Autophagy is also a necessary process associated with chemotherapy-induced cell death. Lysosome accumulation and lysosome-associated membrane protein-2 (LAMP2) depletion were observed after DFIQ treatment, and cell death induction was restored upon treatment with the autophagy inhibitor 3-methyladenine (3-MA). Nevertheless, ROS production was found to be involved in DFIQ-induced autophagy activation and LAMP2 depletion. Our data provide the first evidence for developing DFIQ for clinical usage and show the regulatory mechanism by which DFIQ affects ROS, autophagy, and apoptosis.
format Online
Article
Text
id pubmed-7281296
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-72812962020-06-19 DFIQ, a Novel Quinoline Derivative, Shows Anticancer Potential by Inducing Apoptosis and Autophagy in NSCLC Cell and In Vivo Zebrafish Xenograft Models Huang, Hurng-Wern Bow, Yung-Ding Wang, Chia-Yih Chen, Yen-Chun Fu, Pei-Rong Chang, Kuo-Feng Wang, Tso-Wen Tseng, Chih-Hua Chen, Yeh-Long Chiu, Chien-Chih Cancers (Basel) Article Lung cancer is one of the deadliest cancers worldwide due to chemoresistance in patients with late-stage disease. Quinoline derivatives show biological activity against HIV, malaria, bacteriuria, and cancer. DFIQ is a novel synthetic quinoline derivative that induces cell death in both in vitro and in vivo zebrafish xenograft models. DFIQ induced cell death, including apoptosis, and the IC(50) values were 4.16 and 2.31 μM at 24 and 48 h, respectively. DFIQ was also found to induce apoptotic protein cleavage and DNA damage, reduce cell cycle-associated protein expression, and disrupt reactive oxygen species (ROS) reduction, thus resulting in the accumulation of superoxide radicals. Autophagy is also a necessary process associated with chemotherapy-induced cell death. Lysosome accumulation and lysosome-associated membrane protein-2 (LAMP2) depletion were observed after DFIQ treatment, and cell death induction was restored upon treatment with the autophagy inhibitor 3-methyladenine (3-MA). Nevertheless, ROS production was found to be involved in DFIQ-induced autophagy activation and LAMP2 depletion. Our data provide the first evidence for developing DFIQ for clinical usage and show the regulatory mechanism by which DFIQ affects ROS, autophagy, and apoptosis. MDPI 2020-05-25 /pmc/articles/PMC7281296/ /pubmed/32466291 http://dx.doi.org/10.3390/cancers12051348 Text en © 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Huang, Hurng-Wern
Bow, Yung-Ding
Wang, Chia-Yih
Chen, Yen-Chun
Fu, Pei-Rong
Chang, Kuo-Feng
Wang, Tso-Wen
Tseng, Chih-Hua
Chen, Yeh-Long
Chiu, Chien-Chih
DFIQ, a Novel Quinoline Derivative, Shows Anticancer Potential by Inducing Apoptosis and Autophagy in NSCLC Cell and In Vivo Zebrafish Xenograft Models
title DFIQ, a Novel Quinoline Derivative, Shows Anticancer Potential by Inducing Apoptosis and Autophagy in NSCLC Cell and In Vivo Zebrafish Xenograft Models
title_full DFIQ, a Novel Quinoline Derivative, Shows Anticancer Potential by Inducing Apoptosis and Autophagy in NSCLC Cell and In Vivo Zebrafish Xenograft Models
title_fullStr DFIQ, a Novel Quinoline Derivative, Shows Anticancer Potential by Inducing Apoptosis and Autophagy in NSCLC Cell and In Vivo Zebrafish Xenograft Models
title_full_unstemmed DFIQ, a Novel Quinoline Derivative, Shows Anticancer Potential by Inducing Apoptosis and Autophagy in NSCLC Cell and In Vivo Zebrafish Xenograft Models
title_short DFIQ, a Novel Quinoline Derivative, Shows Anticancer Potential by Inducing Apoptosis and Autophagy in NSCLC Cell and In Vivo Zebrafish Xenograft Models
title_sort dfiq, a novel quinoline derivative, shows anticancer potential by inducing apoptosis and autophagy in nsclc cell and in vivo zebrafish xenograft models
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7281296/
https://www.ncbi.nlm.nih.gov/pubmed/32466291
http://dx.doi.org/10.3390/cancers12051348
work_keys_str_mv AT huanghurngwern dfiqanovelquinolinederivativeshowsanticancerpotentialbyinducingapoptosisandautophagyinnsclccellandinvivozebrafishxenograftmodels
AT bowyungding dfiqanovelquinolinederivativeshowsanticancerpotentialbyinducingapoptosisandautophagyinnsclccellandinvivozebrafishxenograftmodels
AT wangchiayih dfiqanovelquinolinederivativeshowsanticancerpotentialbyinducingapoptosisandautophagyinnsclccellandinvivozebrafishxenograftmodels
AT chenyenchun dfiqanovelquinolinederivativeshowsanticancerpotentialbyinducingapoptosisandautophagyinnsclccellandinvivozebrafishxenograftmodels
AT fupeirong dfiqanovelquinolinederivativeshowsanticancerpotentialbyinducingapoptosisandautophagyinnsclccellandinvivozebrafishxenograftmodels
AT changkuofeng dfiqanovelquinolinederivativeshowsanticancerpotentialbyinducingapoptosisandautophagyinnsclccellandinvivozebrafishxenograftmodels
AT wangtsowen dfiqanovelquinolinederivativeshowsanticancerpotentialbyinducingapoptosisandautophagyinnsclccellandinvivozebrafishxenograftmodels
AT tsengchihhua dfiqanovelquinolinederivativeshowsanticancerpotentialbyinducingapoptosisandautophagyinnsclccellandinvivozebrafishxenograftmodels
AT chenyehlong dfiqanovelquinolinederivativeshowsanticancerpotentialbyinducingapoptosisandautophagyinnsclccellandinvivozebrafishxenograftmodels
AT chiuchienchih dfiqanovelquinolinederivativeshowsanticancerpotentialbyinducingapoptosisandautophagyinnsclccellandinvivozebrafishxenograftmodels