Cargando…

Restoring the epigenetically silenced PCK2 suppresses renal cell carcinoma progression and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress

Rationale: Tumors have significant abnormalities in various biological properties. In renal cell carcinoma (RCC), metabolic abnormalities are characteristic biological dysfunction that cannot be ignored. Despite this, many aspects of this dysfunction have not been fully explained. The purpose of thi...

Descripción completa

Detalles Bibliográficos
Autores principales: Xiong, Zhiyong, Yuan, Changfei, shi, Jian, Xiong, Wei, Huang, Yu, Xiao, Wen, Yang, Hongmei, Chen, Ke, Zhang, Xiaoping
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7546001/
https://www.ncbi.nlm.nih.gov/pubmed/33052225
http://dx.doi.org/10.7150/thno.48469
_version_ 1783592146634276864
author Xiong, Zhiyong
Yuan, Changfei
shi, Jian
Xiong, Wei
Huang, Yu
Xiao, Wen
Yang, Hongmei
Chen, Ke
Zhang, Xiaoping
author_facet Xiong, Zhiyong
Yuan, Changfei
shi, Jian
Xiong, Wei
Huang, Yu
Xiao, Wen
Yang, Hongmei
Chen, Ke
Zhang, Xiaoping
author_sort Xiong, Zhiyong
collection PubMed
description Rationale: Tumors have significant abnormalities in various biological properties. In renal cell carcinoma (RCC), metabolic abnormalities are characteristic biological dysfunction that cannot be ignored. Despite this, many aspects of this dysfunction have not been fully explained. The purpose of this study was to reveal a new mechanism of metabolic and energy-related biological abnormalities in RCC. Methods: Molecular screening and bioinformatics analysis were performed in RCC based on data from The Cancer Genome Atlas (TCGA) database. Regulated pathways were investigated by qRT-PCR, immunoblot analysis and immunohistochemistry. A series of functional analyses was performed in cell lines and xenograft models. Results: By screening the biological abnormality core dataset-mitochondria-related dataset and the metabolic abnormality core dataset-energy metabolism-related dataset in public RCC databases, PCK2 was found to be differentially expressed in RCC compared with normal tissue. Further analysis by the TCGA database showed that PCK2 was significantly downregulated in RCC and predicted a poor prognosis. Through additional studies, it was found that a low expression of PCK2 in RCC was caused by methylation of its promoter region. Restoration of PCK2 expression in RCC cells repressed tumor progression and increased their sensitivity to sunitinib. Finally, mechanistic investigations indicated that PCK2 mediated the above processes by promoting endoplasmic reticulum stress. Conclusions: Collectively, our results identify a specific mechanism by which PCK2 suppresses the progression of renal cell carcinoma (RCC) and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress. This finding provides a new biomarker for RCC as well as novel targets and strategies for the treatment of RCC.
format Online
Article
Text
id pubmed-7546001
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-75460012020-10-12 Restoring the epigenetically silenced PCK2 suppresses renal cell carcinoma progression and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress Xiong, Zhiyong Yuan, Changfei shi, Jian Xiong, Wei Huang, Yu Xiao, Wen Yang, Hongmei Chen, Ke Zhang, Xiaoping Theranostics Research Paper Rationale: Tumors have significant abnormalities in various biological properties. In renal cell carcinoma (RCC), metabolic abnormalities are characteristic biological dysfunction that cannot be ignored. Despite this, many aspects of this dysfunction have not been fully explained. The purpose of this study was to reveal a new mechanism of metabolic and energy-related biological abnormalities in RCC. Methods: Molecular screening and bioinformatics analysis were performed in RCC based on data from The Cancer Genome Atlas (TCGA) database. Regulated pathways were investigated by qRT-PCR, immunoblot analysis and immunohistochemistry. A series of functional analyses was performed in cell lines and xenograft models. Results: By screening the biological abnormality core dataset-mitochondria-related dataset and the metabolic abnormality core dataset-energy metabolism-related dataset in public RCC databases, PCK2 was found to be differentially expressed in RCC compared with normal tissue. Further analysis by the TCGA database showed that PCK2 was significantly downregulated in RCC and predicted a poor prognosis. Through additional studies, it was found that a low expression of PCK2 in RCC was caused by methylation of its promoter region. Restoration of PCK2 expression in RCC cells repressed tumor progression and increased their sensitivity to sunitinib. Finally, mechanistic investigations indicated that PCK2 mediated the above processes by promoting endoplasmic reticulum stress. Conclusions: Collectively, our results identify a specific mechanism by which PCK2 suppresses the progression of renal cell carcinoma (RCC) and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress. This finding provides a new biomarker for RCC as well as novel targets and strategies for the treatment of RCC. Ivyspring International Publisher 2020-09-15 /pmc/articles/PMC7546001/ /pubmed/33052225 http://dx.doi.org/10.7150/thno.48469 Text en © The author(s) This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Xiong, Zhiyong
Yuan, Changfei
shi, Jian
Xiong, Wei
Huang, Yu
Xiao, Wen
Yang, Hongmei
Chen, Ke
Zhang, Xiaoping
Restoring the epigenetically silenced PCK2 suppresses renal cell carcinoma progression and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress
title Restoring the epigenetically silenced PCK2 suppresses renal cell carcinoma progression and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress
title_full Restoring the epigenetically silenced PCK2 suppresses renal cell carcinoma progression and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress
title_fullStr Restoring the epigenetically silenced PCK2 suppresses renal cell carcinoma progression and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress
title_full_unstemmed Restoring the epigenetically silenced PCK2 suppresses renal cell carcinoma progression and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress
title_short Restoring the epigenetically silenced PCK2 suppresses renal cell carcinoma progression and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress
title_sort restoring the epigenetically silenced pck2 suppresses renal cell carcinoma progression and increases sensitivity to sunitinib by promoting endoplasmic reticulum stress
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7546001/
https://www.ncbi.nlm.nih.gov/pubmed/33052225
http://dx.doi.org/10.7150/thno.48469
work_keys_str_mv AT xiongzhiyong restoringtheepigeneticallysilencedpck2suppressesrenalcellcarcinomaprogressionandincreasessensitivitytosunitinibbypromotingendoplasmicreticulumstress
AT yuanchangfei restoringtheepigeneticallysilencedpck2suppressesrenalcellcarcinomaprogressionandincreasessensitivitytosunitinibbypromotingendoplasmicreticulumstress
AT shijian restoringtheepigeneticallysilencedpck2suppressesrenalcellcarcinomaprogressionandincreasessensitivitytosunitinibbypromotingendoplasmicreticulumstress
AT xiongwei restoringtheepigeneticallysilencedpck2suppressesrenalcellcarcinomaprogressionandincreasessensitivitytosunitinibbypromotingendoplasmicreticulumstress
AT huangyu restoringtheepigeneticallysilencedpck2suppressesrenalcellcarcinomaprogressionandincreasessensitivitytosunitinibbypromotingendoplasmicreticulumstress
AT xiaowen restoringtheepigeneticallysilencedpck2suppressesrenalcellcarcinomaprogressionandincreasessensitivitytosunitinibbypromotingendoplasmicreticulumstress
AT yanghongmei restoringtheepigeneticallysilencedpck2suppressesrenalcellcarcinomaprogressionandincreasessensitivitytosunitinibbypromotingendoplasmicreticulumstress
AT chenke restoringtheepigeneticallysilencedpck2suppressesrenalcellcarcinomaprogressionandincreasessensitivitytosunitinibbypromotingendoplasmicreticulumstress
AT zhangxiaoping restoringtheepigeneticallysilencedpck2suppressesrenalcellcarcinomaprogressionandincreasessensitivitytosunitinibbypromotingendoplasmicreticulumstress