Cargando…

aPKCι promotes gallbladder cancer tumorigenesis and gemcitabine resistance by competing with Nrf2 for binding to Keap1

Gallbladder cancer (GBC) is a highly malignant bile duct cancer with poor prognosis characterized by its insensitivity to chemotherapy. Emerging evidence indicates that cytoprotective antioxidation is involved in drug resistance of various cancers; however, the underlying molecular mechanisms remain...

Descripción completa

Detalles Bibliográficos
Autores principales: Tian, Li, Lu, Yun, Yang, Tao, Deng, Zhengdong, Xu, Lei, Yao, Wei, Ma, Chaoqun, Li, Xiangyu, Zhang, Jian, Liu, Yan, Wang, Jianming
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Elsevier 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6395946/
https://www.ncbi.nlm.nih.gov/pubmed/30822690
http://dx.doi.org/10.1016/j.redox.2019.101149
_version_ 1783399173431754752
author Tian, Li
Lu, Yun
Yang, Tao
Deng, Zhengdong
Xu, Lei
Yao, Wei
Ma, Chaoqun
Li, Xiangyu
Zhang, Jian
Liu, Yan
Wang, Jianming
author_facet Tian, Li
Lu, Yun
Yang, Tao
Deng, Zhengdong
Xu, Lei
Yao, Wei
Ma, Chaoqun
Li, Xiangyu
Zhang, Jian
Liu, Yan
Wang, Jianming
author_sort Tian, Li
collection PubMed
description Gallbladder cancer (GBC) is a highly malignant bile duct cancer with poor prognosis characterized by its insensitivity to chemotherapy. Emerging evidence indicates that cytoprotective antioxidation is involved in drug resistance of various cancers; however, the underlying molecular mechanisms remain obscure. Here, we demonstrated that atypical protein kinase Cι (aPKCι) mediated reactive oxygen species (ROS) inhibition in a kinase-independent manner, which played a crucial role in tumorigenesis and chemoresistance. Mechanistically, we found that aPKCι facilitated nuclear factor erythroid 2-related factor 2 (Nrf2) accumulation, nuclear translocation and activated its target genes by competing with Nrf2 for binding to Kelch-like ECH-associated protein 1 (Keap1) through a highly conserved DLL motif. In addition, the aPKCι-Keap1 interaction was required for antioxidant effect, cell growth and gemcitabine resistance in GBC. Importantly, we further confirmed that aPKCι was frequently upregulated and correlated with poor prognosis in patients with GBC. Collectively, our findings suggested that aPKCι positively modulated the Keap1-Nrf2 pathway to enhance GBC growth and gemcitabine resistance, implying that the aPKCι-Keap1-Nrf2 axis may be a potential approach to overcome the drug resistance for the treatment of GBC.
format Online
Article
Text
id pubmed-6395946
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Elsevier
record_format MEDLINE/PubMed
spelling pubmed-63959462019-03-11 aPKCι promotes gallbladder cancer tumorigenesis and gemcitabine resistance by competing with Nrf2 for binding to Keap1 Tian, Li Lu, Yun Yang, Tao Deng, Zhengdong Xu, Lei Yao, Wei Ma, Chaoqun Li, Xiangyu Zhang, Jian Liu, Yan Wang, Jianming Redox Biol Research Paper Gallbladder cancer (GBC) is a highly malignant bile duct cancer with poor prognosis characterized by its insensitivity to chemotherapy. Emerging evidence indicates that cytoprotective antioxidation is involved in drug resistance of various cancers; however, the underlying molecular mechanisms remain obscure. Here, we demonstrated that atypical protein kinase Cι (aPKCι) mediated reactive oxygen species (ROS) inhibition in a kinase-independent manner, which played a crucial role in tumorigenesis and chemoresistance. Mechanistically, we found that aPKCι facilitated nuclear factor erythroid 2-related factor 2 (Nrf2) accumulation, nuclear translocation and activated its target genes by competing with Nrf2 for binding to Kelch-like ECH-associated protein 1 (Keap1) through a highly conserved DLL motif. In addition, the aPKCι-Keap1 interaction was required for antioxidant effect, cell growth and gemcitabine resistance in GBC. Importantly, we further confirmed that aPKCι was frequently upregulated and correlated with poor prognosis in patients with GBC. Collectively, our findings suggested that aPKCι positively modulated the Keap1-Nrf2 pathway to enhance GBC growth and gemcitabine resistance, implying that the aPKCι-Keap1-Nrf2 axis may be a potential approach to overcome the drug resistance for the treatment of GBC. Elsevier 2019-02-21 /pmc/articles/PMC6395946/ /pubmed/30822690 http://dx.doi.org/10.1016/j.redox.2019.101149 Text en © 2019 The Authors. Published by Elsevier B.V. http://creativecommons.org/licenses/by-nc-nd/4.0/ This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Research Paper
Tian, Li
Lu, Yun
Yang, Tao
Deng, Zhengdong
Xu, Lei
Yao, Wei
Ma, Chaoqun
Li, Xiangyu
Zhang, Jian
Liu, Yan
Wang, Jianming
aPKCι promotes gallbladder cancer tumorigenesis and gemcitabine resistance by competing with Nrf2 for binding to Keap1
title aPKCι promotes gallbladder cancer tumorigenesis and gemcitabine resistance by competing with Nrf2 for binding to Keap1
title_full aPKCι promotes gallbladder cancer tumorigenesis and gemcitabine resistance by competing with Nrf2 for binding to Keap1
title_fullStr aPKCι promotes gallbladder cancer tumorigenesis and gemcitabine resistance by competing with Nrf2 for binding to Keap1
title_full_unstemmed aPKCι promotes gallbladder cancer tumorigenesis and gemcitabine resistance by competing with Nrf2 for binding to Keap1
title_short aPKCι promotes gallbladder cancer tumorigenesis and gemcitabine resistance by competing with Nrf2 for binding to Keap1
title_sort apkcι promotes gallbladder cancer tumorigenesis and gemcitabine resistance by competing with nrf2 for binding to keap1
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6395946/
https://www.ncbi.nlm.nih.gov/pubmed/30822690
http://dx.doi.org/10.1016/j.redox.2019.101149
work_keys_str_mv AT tianli apkcipromotesgallbladdercancertumorigenesisandgemcitabineresistancebycompetingwithnrf2forbindingtokeap1
AT luyun apkcipromotesgallbladdercancertumorigenesisandgemcitabineresistancebycompetingwithnrf2forbindingtokeap1
AT yangtao apkcipromotesgallbladdercancertumorigenesisandgemcitabineresistancebycompetingwithnrf2forbindingtokeap1
AT dengzhengdong apkcipromotesgallbladdercancertumorigenesisandgemcitabineresistancebycompetingwithnrf2forbindingtokeap1
AT xulei apkcipromotesgallbladdercancertumorigenesisandgemcitabineresistancebycompetingwithnrf2forbindingtokeap1
AT yaowei apkcipromotesgallbladdercancertumorigenesisandgemcitabineresistancebycompetingwithnrf2forbindingtokeap1
AT machaoqun apkcipromotesgallbladdercancertumorigenesisandgemcitabineresistancebycompetingwithnrf2forbindingtokeap1
AT lixiangyu apkcipromotesgallbladdercancertumorigenesisandgemcitabineresistancebycompetingwithnrf2forbindingtokeap1
AT zhangjian apkcipromotesgallbladdercancertumorigenesisandgemcitabineresistancebycompetingwithnrf2forbindingtokeap1
AT liuyan apkcipromotesgallbladdercancertumorigenesisandgemcitabineresistancebycompetingwithnrf2forbindingtokeap1
AT wangjianming apkcipromotesgallbladdercancertumorigenesisandgemcitabineresistancebycompetingwithnrf2forbindingtokeap1