Cargando…

ATP8B1 Knockdown Activated the Choline Metabolism Pathway and Induced High-Level Intracellular REDOX Homeostasis in Lung Squamous Cell Carcinoma

SIMPLE SUMMARY: We found that low expression of ATP8B1 was associated with poor prognosis, and involved in the dysregulation of glutathione (GSH) synthesis and choline metabolism in lung squamous cell carcinoma (LUSC) samples of The Cancer Genome Atlas (TCGA) and Tianjin Medical University Cancer In...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Xiao, Zhang, Rui, Liu, Pengpeng, Zhang, Runjiao, Ning, Junya, Ye, Yingnan, Yu, Wenwen, Yu, Jinpu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8834475/
https://www.ncbi.nlm.nih.gov/pubmed/35159102
http://dx.doi.org/10.3390/cancers14030835
_version_ 1784649196009160704
author Zhang, Xiao
Zhang, Rui
Liu, Pengpeng
Zhang, Runjiao
Ning, Junya
Ye, Yingnan
Yu, Wenwen
Yu, Jinpu
author_facet Zhang, Xiao
Zhang, Rui
Liu, Pengpeng
Zhang, Runjiao
Ning, Junya
Ye, Yingnan
Yu, Wenwen
Yu, Jinpu
author_sort Zhang, Xiao
collection PubMed
description SIMPLE SUMMARY: We found that low expression of ATP8B1 was associated with poor prognosis, and involved in the dysregulation of glutathione (GSH) synthesis and choline metabolism in lung squamous cell carcinoma (LUSC) samples of The Cancer Genome Atlas (TCGA) and Tianjin Medical University Cancer Institute and Hospital (TJMUCH) cohort. We further constructed ATP8B1 knockdown of LUSC cell lines H520(SH)(-ATP8B1) and SK-MES-1(SH)(-ATP8B1) to investigate how ATP8B1 knockdown promoted cell proliferation, migration, and invasion in vitro and in vivo via upregulation of the CHKA-dependent choline metabolism pathway. We identified that ATP8B1 knockdown and CHKA upregulation can lead to mitochondrial dysfunction and high reduction-oxidation (REDOX) homeostasis, which may be involved in the roles of cardiolipin in maintaining mitochondrial dynamics and phospholipid homeostasis. Therefore, we proposed ATP8B1 as a novel predictive biomarker in LUSC and targeting ATP8B1-driven specific metabolic disorder might be a promising therapeutic strategy for LUSC. ABSTRACT: The flippase ATPase class I type 8b member 1 (ATP8B1) is essential for maintaining the stability and polarity of the epithelial membrane and can translocate specific phospholipids from the outer membrane to the inner membrane of the cell. Although ATP8B1 plays important roles in cell functions, ATP8B1 has been poorly studied in tumors and its prognostic value in patients with lung squamous cell carcinoma (LUSC) remains unclear. By investigating the whole genomic expression profiles of LUSC samples from The Cancer Genome Atlas (TCGA) database and Tianjin Medical University Cancer Institute and Hospital (TJMUCH) cohort, we found that low expression of ATP8B1 was associated with poor prognosis of LUSC patients. The results from cellular experiments and a xenograft-bearing mice model indicated that ATP8B1 knockdown firstly induced mitochondrial dysfunction and promoted ROS production. Secondly, ATP8B1 knockdown promoted glutathione synthesis via upregulation of the CHKA-dependent choline metabolism pathway, therefore producing and maintaining high-level intracellular REDOX homeostasis to aggravate carcinogenesis and progression of LUSC. In summary, we proposed ATP8B1 as a novel predictive biomarker in LUSC and targeting ATP8B1-driven specific metabolic disorder might be a promising therapeutic strategy for LUSC.
format Online
Article
Text
id pubmed-8834475
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-88344752022-02-12 ATP8B1 Knockdown Activated the Choline Metabolism Pathway and Induced High-Level Intracellular REDOX Homeostasis in Lung Squamous Cell Carcinoma Zhang, Xiao Zhang, Rui Liu, Pengpeng Zhang, Runjiao Ning, Junya Ye, Yingnan Yu, Wenwen Yu, Jinpu Cancers (Basel) Article SIMPLE SUMMARY: We found that low expression of ATP8B1 was associated with poor prognosis, and involved in the dysregulation of glutathione (GSH) synthesis and choline metabolism in lung squamous cell carcinoma (LUSC) samples of The Cancer Genome Atlas (TCGA) and Tianjin Medical University Cancer Institute and Hospital (TJMUCH) cohort. We further constructed ATP8B1 knockdown of LUSC cell lines H520(SH)(-ATP8B1) and SK-MES-1(SH)(-ATP8B1) to investigate how ATP8B1 knockdown promoted cell proliferation, migration, and invasion in vitro and in vivo via upregulation of the CHKA-dependent choline metabolism pathway. We identified that ATP8B1 knockdown and CHKA upregulation can lead to mitochondrial dysfunction and high reduction-oxidation (REDOX) homeostasis, which may be involved in the roles of cardiolipin in maintaining mitochondrial dynamics and phospholipid homeostasis. Therefore, we proposed ATP8B1 as a novel predictive biomarker in LUSC and targeting ATP8B1-driven specific metabolic disorder might be a promising therapeutic strategy for LUSC. ABSTRACT: The flippase ATPase class I type 8b member 1 (ATP8B1) is essential for maintaining the stability and polarity of the epithelial membrane and can translocate specific phospholipids from the outer membrane to the inner membrane of the cell. Although ATP8B1 plays important roles in cell functions, ATP8B1 has been poorly studied in tumors and its prognostic value in patients with lung squamous cell carcinoma (LUSC) remains unclear. By investigating the whole genomic expression profiles of LUSC samples from The Cancer Genome Atlas (TCGA) database and Tianjin Medical University Cancer Institute and Hospital (TJMUCH) cohort, we found that low expression of ATP8B1 was associated with poor prognosis of LUSC patients. The results from cellular experiments and a xenograft-bearing mice model indicated that ATP8B1 knockdown firstly induced mitochondrial dysfunction and promoted ROS production. Secondly, ATP8B1 knockdown promoted glutathione synthesis via upregulation of the CHKA-dependent choline metabolism pathway, therefore producing and maintaining high-level intracellular REDOX homeostasis to aggravate carcinogenesis and progression of LUSC. In summary, we proposed ATP8B1 as a novel predictive biomarker in LUSC and targeting ATP8B1-driven specific metabolic disorder might be a promising therapeutic strategy for LUSC. MDPI 2022-02-07 /pmc/articles/PMC8834475/ /pubmed/35159102 http://dx.doi.org/10.3390/cancers14030835 Text en © 2022 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Zhang, Xiao
Zhang, Rui
Liu, Pengpeng
Zhang, Runjiao
Ning, Junya
Ye, Yingnan
Yu, Wenwen
Yu, Jinpu
ATP8B1 Knockdown Activated the Choline Metabolism Pathway and Induced High-Level Intracellular REDOX Homeostasis in Lung Squamous Cell Carcinoma
title ATP8B1 Knockdown Activated the Choline Metabolism Pathway and Induced High-Level Intracellular REDOX Homeostasis in Lung Squamous Cell Carcinoma
title_full ATP8B1 Knockdown Activated the Choline Metabolism Pathway and Induced High-Level Intracellular REDOX Homeostasis in Lung Squamous Cell Carcinoma
title_fullStr ATP8B1 Knockdown Activated the Choline Metabolism Pathway and Induced High-Level Intracellular REDOX Homeostasis in Lung Squamous Cell Carcinoma
title_full_unstemmed ATP8B1 Knockdown Activated the Choline Metabolism Pathway and Induced High-Level Intracellular REDOX Homeostasis in Lung Squamous Cell Carcinoma
title_short ATP8B1 Knockdown Activated the Choline Metabolism Pathway and Induced High-Level Intracellular REDOX Homeostasis in Lung Squamous Cell Carcinoma
title_sort atp8b1 knockdown activated the choline metabolism pathway and induced high-level intracellular redox homeostasis in lung squamous cell carcinoma
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8834475/
https://www.ncbi.nlm.nih.gov/pubmed/35159102
http://dx.doi.org/10.3390/cancers14030835
work_keys_str_mv AT zhangxiao atp8b1knockdownactivatedthecholinemetabolismpathwayandinducedhighlevelintracellularredoxhomeostasisinlungsquamouscellcarcinoma
AT zhangrui atp8b1knockdownactivatedthecholinemetabolismpathwayandinducedhighlevelintracellularredoxhomeostasisinlungsquamouscellcarcinoma
AT liupengpeng atp8b1knockdownactivatedthecholinemetabolismpathwayandinducedhighlevelintracellularredoxhomeostasisinlungsquamouscellcarcinoma
AT zhangrunjiao atp8b1knockdownactivatedthecholinemetabolismpathwayandinducedhighlevelintracellularredoxhomeostasisinlungsquamouscellcarcinoma
AT ningjunya atp8b1knockdownactivatedthecholinemetabolismpathwayandinducedhighlevelintracellularredoxhomeostasisinlungsquamouscellcarcinoma
AT yeyingnan atp8b1knockdownactivatedthecholinemetabolismpathwayandinducedhighlevelintracellularredoxhomeostasisinlungsquamouscellcarcinoma
AT yuwenwen atp8b1knockdownactivatedthecholinemetabolismpathwayandinducedhighlevelintracellularredoxhomeostasisinlungsquamouscellcarcinoma
AT yujinpu atp8b1knockdownactivatedthecholinemetabolismpathwayandinducedhighlevelintracellularredoxhomeostasisinlungsquamouscellcarcinoma