Cargando…

Genetic ablation of TAZ induces HepG2 liver cancer cell apoptosis through activating the CaMKII/MIEF1 signaling pathway

BACKGROUND AND OBJECTIVE: Transcriptional coactivator with PDZ-binding motif (TAZ) has been found to be associated with tumor progression. Mitochondrial homeostasis regulates cancer cell viability and metastasis. However, the roles of TAZ and mitochondrial homeostasis in liver cancer viability have...

Descripción completa

Detalles Bibliográficos
Autores principales: Hou, Yi, Lan, Chunna, Kong, Ying, Zhu, Chunjiao, Peng, Wenna, Huang, Zhichao, Zhang, Changjie
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Dove Medical Press 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6402445/
https://www.ncbi.nlm.nih.gov/pubmed/30881030
http://dx.doi.org/10.2147/OTT.S196142
_version_ 1783400400683008000
author Hou, Yi
Lan, Chunna
Kong, Ying
Zhu, Chunjiao
Peng, Wenna
Huang, Zhichao
Zhang, Changjie
author_facet Hou, Yi
Lan, Chunna
Kong, Ying
Zhu, Chunjiao
Peng, Wenna
Huang, Zhichao
Zhang, Changjie
author_sort Hou, Yi
collection PubMed
description BACKGROUND AND OBJECTIVE: Transcriptional coactivator with PDZ-binding motif (TAZ) has been found to be associated with tumor progression. Mitochondrial homeostasis regulates cancer cell viability and metastasis. However, the roles of TAZ and mitochondrial homeostasis in liver cancer viability have not been explored. The aim of our study was to investigate the influence of TAZ on HepG2 liver cancer cell apoptosis. MATERIALS AND METHODS: HepG2 liver cancer cell was used in the present study, and shRNA against TAZ was transfected into HepG2 cell to knockdown TAZ expression. Mitochondrial function was analyzed using Western blotting, immunofluorescence assay, and flow cytometry. Pathway blocker was used to confirm the role of CaMKII pathway in TAZ-mediated cancer cell death. RESULTS: Our results indicated that TAZ deletion induced death in HepG2 cell via apoptosis. Biological analysis demonstrated that mitochondrial stress, including mitochondrial bioenergetics disorder, mitochondrial oxidative stress, and mitochondrial apoptosis, were activated by TAZ deletion. Furthermore, we found that TAZ affected mitochondrial stress by triggering mitochondrial elongation factor 1 (MIEF1)-related mitochondrial dysfunction. The loss of MIEF1 sustained mitochondrial function and promoted cancer cell survival. Molecular investigation illustrated that TAZ regulated MIEF1 expression via the CaMKII signaling pathway. Blockade of the CaMKII pathway prevented TAZ-mediated MIEF1 upregulation and improved cancer cell survival. CONCLUSION: Taken together, our results highlight the key role of TAZ as a master regulator of HepG2 liver cancer cell viability via the modulation of MIEF1-related mitochondrial stress and the CaMKII signaling pathway. These findings define TAZ and MIEF1-related mitochondrial dysfunction as tumor suppressors that act by promoting cancer apoptosis via the CaMKII signaling pathway, with potential implications for new approaches to liver cancer therapy.
format Online
Article
Text
id pubmed-6402445
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Dove Medical Press
record_format MEDLINE/PubMed
spelling pubmed-64024452019-03-16 Genetic ablation of TAZ induces HepG2 liver cancer cell apoptosis through activating the CaMKII/MIEF1 signaling pathway Hou, Yi Lan, Chunna Kong, Ying Zhu, Chunjiao Peng, Wenna Huang, Zhichao Zhang, Changjie Onco Targets Ther Original Research BACKGROUND AND OBJECTIVE: Transcriptional coactivator with PDZ-binding motif (TAZ) has been found to be associated with tumor progression. Mitochondrial homeostasis regulates cancer cell viability and metastasis. However, the roles of TAZ and mitochondrial homeostasis in liver cancer viability have not been explored. The aim of our study was to investigate the influence of TAZ on HepG2 liver cancer cell apoptosis. MATERIALS AND METHODS: HepG2 liver cancer cell was used in the present study, and shRNA against TAZ was transfected into HepG2 cell to knockdown TAZ expression. Mitochondrial function was analyzed using Western blotting, immunofluorescence assay, and flow cytometry. Pathway blocker was used to confirm the role of CaMKII pathway in TAZ-mediated cancer cell death. RESULTS: Our results indicated that TAZ deletion induced death in HepG2 cell via apoptosis. Biological analysis demonstrated that mitochondrial stress, including mitochondrial bioenergetics disorder, mitochondrial oxidative stress, and mitochondrial apoptosis, were activated by TAZ deletion. Furthermore, we found that TAZ affected mitochondrial stress by triggering mitochondrial elongation factor 1 (MIEF1)-related mitochondrial dysfunction. The loss of MIEF1 sustained mitochondrial function and promoted cancer cell survival. Molecular investigation illustrated that TAZ regulated MIEF1 expression via the CaMKII signaling pathway. Blockade of the CaMKII pathway prevented TAZ-mediated MIEF1 upregulation and improved cancer cell survival. CONCLUSION: Taken together, our results highlight the key role of TAZ as a master regulator of HepG2 liver cancer cell viability via the modulation of MIEF1-related mitochondrial stress and the CaMKII signaling pathway. These findings define TAZ and MIEF1-related mitochondrial dysfunction as tumor suppressors that act by promoting cancer apoptosis via the CaMKII signaling pathway, with potential implications for new approaches to liver cancer therapy. Dove Medical Press 2019-03-01 /pmc/articles/PMC6402445/ /pubmed/30881030 http://dx.doi.org/10.2147/OTT.S196142 Text en © 2019 Hou et al. This work is published and licensed by Dove Medical Press Limited The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed.
spellingShingle Original Research
Hou, Yi
Lan, Chunna
Kong, Ying
Zhu, Chunjiao
Peng, Wenna
Huang, Zhichao
Zhang, Changjie
Genetic ablation of TAZ induces HepG2 liver cancer cell apoptosis through activating the CaMKII/MIEF1 signaling pathway
title Genetic ablation of TAZ induces HepG2 liver cancer cell apoptosis through activating the CaMKII/MIEF1 signaling pathway
title_full Genetic ablation of TAZ induces HepG2 liver cancer cell apoptosis through activating the CaMKII/MIEF1 signaling pathway
title_fullStr Genetic ablation of TAZ induces HepG2 liver cancer cell apoptosis through activating the CaMKII/MIEF1 signaling pathway
title_full_unstemmed Genetic ablation of TAZ induces HepG2 liver cancer cell apoptosis through activating the CaMKII/MIEF1 signaling pathway
title_short Genetic ablation of TAZ induces HepG2 liver cancer cell apoptosis through activating the CaMKII/MIEF1 signaling pathway
title_sort genetic ablation of taz induces hepg2 liver cancer cell apoptosis through activating the camkii/mief1 signaling pathway
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6402445/
https://www.ncbi.nlm.nih.gov/pubmed/30881030
http://dx.doi.org/10.2147/OTT.S196142
work_keys_str_mv AT houyi geneticablationoftazinduceshepg2livercancercellapoptosisthroughactivatingthecamkiimief1signalingpathway
AT lanchunna geneticablationoftazinduceshepg2livercancercellapoptosisthroughactivatingthecamkiimief1signalingpathway
AT kongying geneticablationoftazinduceshepg2livercancercellapoptosisthroughactivatingthecamkiimief1signalingpathway
AT zhuchunjiao geneticablationoftazinduceshepg2livercancercellapoptosisthroughactivatingthecamkiimief1signalingpathway
AT pengwenna geneticablationoftazinduceshepg2livercancercellapoptosisthroughactivatingthecamkiimief1signalingpathway
AT huangzhichao geneticablationoftazinduceshepg2livercancercellapoptosisthroughactivatingthecamkiimief1signalingpathway
AT zhangchangjie geneticablationoftazinduceshepg2livercancercellapoptosisthroughactivatingthecamkiimief1signalingpathway