Cargando…

GSTZ1 deficiency promotes hepatocellular carcinoma proliferation via activation of the KEAP1/NRF2 pathway

BACKGROUND: Glutathione S-transferase zeta 1 (GSTZ1) is the penultimate enzyme in phenylalanine/tyrosine catabolism. GSTZ1 is dysregulated in cancers; however, its role in tumorigenesis and progression of hepatocellular carcinoma (HCC) is largely unknown. We aimed to assess the role of GSTZ1 in HCC...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Jingjing, Wang, Qiujie, Yang, Yi, Lei, Chong, Yang, Fan, Liang, Li, Chen, Chang, Xia, Jie, Wang, Kai, Tang, Ni
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6822483/
https://www.ncbi.nlm.nih.gov/pubmed/31666108
http://dx.doi.org/10.1186/s13046-019-1459-6
_version_ 1783464347223195648
author Li, Jingjing
Wang, Qiujie
Yang, Yi
Lei, Chong
Yang, Fan
Liang, Li
Chen, Chang
Xia, Jie
Wang, Kai
Tang, Ni
author_facet Li, Jingjing
Wang, Qiujie
Yang, Yi
Lei, Chong
Yang, Fan
Liang, Li
Chen, Chang
Xia, Jie
Wang, Kai
Tang, Ni
author_sort Li, Jingjing
collection PubMed
description BACKGROUND: Glutathione S-transferase zeta 1 (GSTZ1) is the penultimate enzyme in phenylalanine/tyrosine catabolism. GSTZ1 is dysregulated in cancers; however, its role in tumorigenesis and progression of hepatocellular carcinoma (HCC) is largely unknown. We aimed to assess the role of GSTZ1 in HCC and to reveal the underlying mechanisms, which may contribute to finding a potential therapeutic strategy against HCC. METHODS: We first analyzed GSTZ1 expression levels in paired human HCC and adjacent normal tissue specimens and the prognostic effect of GSTZ1 on HCC patients. Thereafter, we evaluated the role of GSTZ1 in aerobic glycolysis in HCC cells on the basis of the oxygen consumption rate (OCR) and extracellular acidification rate (ECAR). Furthermore, we assessed the effect of GSTZ1 on HCC proliferation, glutathione (GSH) concentration, levels of reactive oxygen species (ROS), and nuclear factor erythroid 2-related factor 2 (NRF2) signaling via gain- and loss- of GSTZ1 function in vitro. Moreover, we investigated the effect of GSTZ1 on diethylnitrosamine (DEN) and carbon tetrachloride (CCl(4)) induced hepatocarcinogenesis in a mouse model of HCC. RESULTS: GSTZ1 was downregulated in HCC, thus indicating a poor prognosis. GSTZ1 deficiency significantly promoted hepatoma cell proliferation and aerobic glycolysis in HCC cells. Moreover, loss of GSTZ1 function depleted GSH, increased ROS levels, and enhanced lipid peroxidation, thus activating the NRF2-mediated antioxidant pathway. Furthermore, Gstz1 knockout in mice promoted DEN/CCl(4)-induced hepatocarcinogenesis via activation of the NRF2 signaling pathway. Furthermore, the antioxidant agent N-acetylcysteine and NRF2 inhibitor brusatol effectively suppressed the growth of Gstz1-knockout HepG2 cells and HCC progression in Gstz1(−/−) mice. CONCLUSIONS: GSTZ1 serves as a tumor suppressor in HCC. GSH depletion caused by GSTZ1 deficiency elevates oxidative stress, thus constitutively activating the NRF2 antioxidant response pathway and accelerating HCC progression. Targeting the NRF2 signaling pathway may be a promising therapeutic approach for this subset of HCC.
format Online
Article
Text
id pubmed-6822483
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-68224832019-11-06 GSTZ1 deficiency promotes hepatocellular carcinoma proliferation via activation of the KEAP1/NRF2 pathway Li, Jingjing Wang, Qiujie Yang, Yi Lei, Chong Yang, Fan Liang, Li Chen, Chang Xia, Jie Wang, Kai Tang, Ni J Exp Clin Cancer Res Research BACKGROUND: Glutathione S-transferase zeta 1 (GSTZ1) is the penultimate enzyme in phenylalanine/tyrosine catabolism. GSTZ1 is dysregulated in cancers; however, its role in tumorigenesis and progression of hepatocellular carcinoma (HCC) is largely unknown. We aimed to assess the role of GSTZ1 in HCC and to reveal the underlying mechanisms, which may contribute to finding a potential therapeutic strategy against HCC. METHODS: We first analyzed GSTZ1 expression levels in paired human HCC and adjacent normal tissue specimens and the prognostic effect of GSTZ1 on HCC patients. Thereafter, we evaluated the role of GSTZ1 in aerobic glycolysis in HCC cells on the basis of the oxygen consumption rate (OCR) and extracellular acidification rate (ECAR). Furthermore, we assessed the effect of GSTZ1 on HCC proliferation, glutathione (GSH) concentration, levels of reactive oxygen species (ROS), and nuclear factor erythroid 2-related factor 2 (NRF2) signaling via gain- and loss- of GSTZ1 function in vitro. Moreover, we investigated the effect of GSTZ1 on diethylnitrosamine (DEN) and carbon tetrachloride (CCl(4)) induced hepatocarcinogenesis in a mouse model of HCC. RESULTS: GSTZ1 was downregulated in HCC, thus indicating a poor prognosis. GSTZ1 deficiency significantly promoted hepatoma cell proliferation and aerobic glycolysis in HCC cells. Moreover, loss of GSTZ1 function depleted GSH, increased ROS levels, and enhanced lipid peroxidation, thus activating the NRF2-mediated antioxidant pathway. Furthermore, Gstz1 knockout in mice promoted DEN/CCl(4)-induced hepatocarcinogenesis via activation of the NRF2 signaling pathway. Furthermore, the antioxidant agent N-acetylcysteine and NRF2 inhibitor brusatol effectively suppressed the growth of Gstz1-knockout HepG2 cells and HCC progression in Gstz1(−/−) mice. CONCLUSIONS: GSTZ1 serves as a tumor suppressor in HCC. GSH depletion caused by GSTZ1 deficiency elevates oxidative stress, thus constitutively activating the NRF2 antioxidant response pathway and accelerating HCC progression. Targeting the NRF2 signaling pathway may be a promising therapeutic approach for this subset of HCC. BioMed Central 2019-10-30 /pmc/articles/PMC6822483/ /pubmed/31666108 http://dx.doi.org/10.1186/s13046-019-1459-6 Text en © The Author(s). 2019 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
Li, Jingjing
Wang, Qiujie
Yang, Yi
Lei, Chong
Yang, Fan
Liang, Li
Chen, Chang
Xia, Jie
Wang, Kai
Tang, Ni
GSTZ1 deficiency promotes hepatocellular carcinoma proliferation via activation of the KEAP1/NRF2 pathway
title GSTZ1 deficiency promotes hepatocellular carcinoma proliferation via activation of the KEAP1/NRF2 pathway
title_full GSTZ1 deficiency promotes hepatocellular carcinoma proliferation via activation of the KEAP1/NRF2 pathway
title_fullStr GSTZ1 deficiency promotes hepatocellular carcinoma proliferation via activation of the KEAP1/NRF2 pathway
title_full_unstemmed GSTZ1 deficiency promotes hepatocellular carcinoma proliferation via activation of the KEAP1/NRF2 pathway
title_short GSTZ1 deficiency promotes hepatocellular carcinoma proliferation via activation of the KEAP1/NRF2 pathway
title_sort gstz1 deficiency promotes hepatocellular carcinoma proliferation via activation of the keap1/nrf2 pathway
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6822483/
https://www.ncbi.nlm.nih.gov/pubmed/31666108
http://dx.doi.org/10.1186/s13046-019-1459-6
work_keys_str_mv AT lijingjing gstz1deficiencypromoteshepatocellularcarcinomaproliferationviaactivationofthekeap1nrf2pathway
AT wangqiujie gstz1deficiencypromoteshepatocellularcarcinomaproliferationviaactivationofthekeap1nrf2pathway
AT yangyi gstz1deficiencypromoteshepatocellularcarcinomaproliferationviaactivationofthekeap1nrf2pathway
AT leichong gstz1deficiencypromoteshepatocellularcarcinomaproliferationviaactivationofthekeap1nrf2pathway
AT yangfan gstz1deficiencypromoteshepatocellularcarcinomaproliferationviaactivationofthekeap1nrf2pathway
AT liangli gstz1deficiencypromoteshepatocellularcarcinomaproliferationviaactivationofthekeap1nrf2pathway
AT chenchang gstz1deficiencypromoteshepatocellularcarcinomaproliferationviaactivationofthekeap1nrf2pathway
AT xiajie gstz1deficiencypromoteshepatocellularcarcinomaproliferationviaactivationofthekeap1nrf2pathway
AT wangkai gstz1deficiencypromoteshepatocellularcarcinomaproliferationviaactivationofthekeap1nrf2pathway
AT tangni gstz1deficiencypromoteshepatocellularcarcinomaproliferationviaactivationofthekeap1nrf2pathway