Cargando…

Ibrutinib, a Bruton’s tyrosine kinase inhibitor, exhibits antitumoral activity and induces autophagy in glioblastoma

BACKGROUND: Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. Ibrutinib, a Bruton’s tyrosine kinase (BTK) inhibitor, is a novel anticancer drug used for treating several types of cancers. In this study, we aimed to determine the role of ibrutinib on GBM. METHODS: Ce...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Jin, Liu, Xiaoyang, Hong, Yongzhi, Wang, Songtao, Chen, Pin, Gu, Aihua, Guo, Xiaoyuan, Zhao, Peng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5513110/
https://www.ncbi.nlm.nih.gov/pubmed/28716053
http://dx.doi.org/10.1186/s13046-017-0549-6
_version_ 1783250598326435840
author Wang, Jin
Liu, Xiaoyang
Hong, Yongzhi
Wang, Songtao
Chen, Pin
Gu, Aihua
Guo, Xiaoyuan
Zhao, Peng
author_facet Wang, Jin
Liu, Xiaoyang
Hong, Yongzhi
Wang, Songtao
Chen, Pin
Gu, Aihua
Guo, Xiaoyuan
Zhao, Peng
author_sort Wang, Jin
collection PubMed
description BACKGROUND: Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. Ibrutinib, a Bruton’s tyrosine kinase (BTK) inhibitor, is a novel anticancer drug used for treating several types of cancers. In this study, we aimed to determine the role of ibrutinib on GBM. METHODS: Cell proliferation was determined by using cell viability, colony formation, and 5-ethynyl-2′-deoxyuridine (EdU) assays. Cell cycle and cell apoptosis were analyzed by flow cytometry. Cell migratory ability was evaluated by wound healing assays and trans-well migration assays. ATG7 expression was knocked-down by transfection with Atg7-specific small interfering RNA. Overexpression of active Akt protein was achieved by transfecting the cells with a plasmid expressing constitutively active Akt (CA-Akt). Transmission electron microscopy was performed to examine the formation of autophagosomes in cells. Immunofluorescence and western blot analyses were used to analyze protein expression. Tumor xenografts in nude mice and immunohistochemistry were performed to evaluate the effect of ibrutinib on tumor growth in vivo. RESULTS: Ibrutinib inhibited cellular proliferation and migration, and induced apoptosis and autophagy in LN229 and U87 cells. Overexpression of the active Akt protein decreased ibrutinib-induced autophagy, while inhibiting Akt by LY294002 treatment enhanced ibrutinib-induced autophagy. Specific inhibition of autophagy by 3-methyladenine (3MA) or Atg7 targeting with small interfering RNA (si-Atg7) enhanced the anti-GBM effect of ibrutinib in vitro and in vivo. CONCLUSIONS: Our results indicate that ibrutinib exerts a profound antitumor effect and induces autophagy through Akt/mTOR signaling pathway in GBM cells. Autophagy inhibition promotes the antitumor activity of ibrutinib in GBM. Our findings provide important insights into the action of an anticancer agent combining with autophagy inhibitor for malignant glioma. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s13046-017-0549-6) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-5513110
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-55131102017-07-19 Ibrutinib, a Bruton’s tyrosine kinase inhibitor, exhibits antitumoral activity and induces autophagy in glioblastoma Wang, Jin Liu, Xiaoyang Hong, Yongzhi Wang, Songtao Chen, Pin Gu, Aihua Guo, Xiaoyuan Zhao, Peng J Exp Clin Cancer Res Research BACKGROUND: Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults. Ibrutinib, a Bruton’s tyrosine kinase (BTK) inhibitor, is a novel anticancer drug used for treating several types of cancers. In this study, we aimed to determine the role of ibrutinib on GBM. METHODS: Cell proliferation was determined by using cell viability, colony formation, and 5-ethynyl-2′-deoxyuridine (EdU) assays. Cell cycle and cell apoptosis were analyzed by flow cytometry. Cell migratory ability was evaluated by wound healing assays and trans-well migration assays. ATG7 expression was knocked-down by transfection with Atg7-specific small interfering RNA. Overexpression of active Akt protein was achieved by transfecting the cells with a plasmid expressing constitutively active Akt (CA-Akt). Transmission electron microscopy was performed to examine the formation of autophagosomes in cells. Immunofluorescence and western blot analyses were used to analyze protein expression. Tumor xenografts in nude mice and immunohistochemistry were performed to evaluate the effect of ibrutinib on tumor growth in vivo. RESULTS: Ibrutinib inhibited cellular proliferation and migration, and induced apoptosis and autophagy in LN229 and U87 cells. Overexpression of the active Akt protein decreased ibrutinib-induced autophagy, while inhibiting Akt by LY294002 treatment enhanced ibrutinib-induced autophagy. Specific inhibition of autophagy by 3-methyladenine (3MA) or Atg7 targeting with small interfering RNA (si-Atg7) enhanced the anti-GBM effect of ibrutinib in vitro and in vivo. CONCLUSIONS: Our results indicate that ibrutinib exerts a profound antitumor effect and induces autophagy through Akt/mTOR signaling pathway in GBM cells. Autophagy inhibition promotes the antitumor activity of ibrutinib in GBM. Our findings provide important insights into the action of an anticancer agent combining with autophagy inhibitor for malignant glioma. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s13046-017-0549-6) contains supplementary material, which is available to authorized users. BioMed Central 2017-07-17 /pmc/articles/PMC5513110/ /pubmed/28716053 http://dx.doi.org/10.1186/s13046-017-0549-6 Text en © The Author(s). 2017 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Wang, Jin
Liu, Xiaoyang
Hong, Yongzhi
Wang, Songtao
Chen, Pin
Gu, Aihua
Guo, Xiaoyuan
Zhao, Peng
Ibrutinib, a Bruton’s tyrosine kinase inhibitor, exhibits antitumoral activity and induces autophagy in glioblastoma
title Ibrutinib, a Bruton’s tyrosine kinase inhibitor, exhibits antitumoral activity and induces autophagy in glioblastoma
title_full Ibrutinib, a Bruton’s tyrosine kinase inhibitor, exhibits antitumoral activity and induces autophagy in glioblastoma
title_fullStr Ibrutinib, a Bruton’s tyrosine kinase inhibitor, exhibits antitumoral activity and induces autophagy in glioblastoma
title_full_unstemmed Ibrutinib, a Bruton’s tyrosine kinase inhibitor, exhibits antitumoral activity and induces autophagy in glioblastoma
title_short Ibrutinib, a Bruton’s tyrosine kinase inhibitor, exhibits antitumoral activity and induces autophagy in glioblastoma
title_sort ibrutinib, a bruton’s tyrosine kinase inhibitor, exhibits antitumoral activity and induces autophagy in glioblastoma
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5513110/
https://www.ncbi.nlm.nih.gov/pubmed/28716053
http://dx.doi.org/10.1186/s13046-017-0549-6
work_keys_str_mv AT wangjin ibrutinibabrutonstyrosinekinaseinhibitorexhibitsantitumoralactivityandinducesautophagyinglioblastoma
AT liuxiaoyang ibrutinibabrutonstyrosinekinaseinhibitorexhibitsantitumoralactivityandinducesautophagyinglioblastoma
AT hongyongzhi ibrutinibabrutonstyrosinekinaseinhibitorexhibitsantitumoralactivityandinducesautophagyinglioblastoma
AT wangsongtao ibrutinibabrutonstyrosinekinaseinhibitorexhibitsantitumoralactivityandinducesautophagyinglioblastoma
AT chenpin ibrutinibabrutonstyrosinekinaseinhibitorexhibitsantitumoralactivityandinducesautophagyinglioblastoma
AT guaihua ibrutinibabrutonstyrosinekinaseinhibitorexhibitsantitumoralactivityandinducesautophagyinglioblastoma
AT guoxiaoyuan ibrutinibabrutonstyrosinekinaseinhibitorexhibitsantitumoralactivityandinducesautophagyinglioblastoma
AT zhaopeng ibrutinibabrutonstyrosinekinaseinhibitorexhibitsantitumoralactivityandinducesautophagyinglioblastoma