Cargando…

Identification and validation of differentially expressed proteins in epithelial ovarian cancers using quantitative proteomics

Ovarian cancer is the most lethal gynecological malignant tumor because of its high recurrence rate. In the present work, in order to find new therapeutic targets, we identified 8480 proteins in thirteen pairs of ovarian cancer tissues and normal ovary tissues through quantitative proteomics. 498 pr...

Descripción completa

Detalles Bibliográficos
Autores principales: Qu, Hong, Chen, Yuling, Cao, Guangming, Liu, Chongdong, Xu, Jiatong, Deng, Haiteng, Zhang, Zhenyu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5347761/
https://www.ncbi.nlm.nih.gov/pubmed/27825122
http://dx.doi.org/10.18632/oncotarget.13077
_version_ 1782514104437571584
author Qu, Hong
Chen, Yuling
Cao, Guangming
Liu, Chongdong
Xu, Jiatong
Deng, Haiteng
Zhang, Zhenyu
author_facet Qu, Hong
Chen, Yuling
Cao, Guangming
Liu, Chongdong
Xu, Jiatong
Deng, Haiteng
Zhang, Zhenyu
author_sort Qu, Hong
collection PubMed
description Ovarian cancer is the most lethal gynecological malignant tumor because of its high recurrence rate. In the present work, in order to find new therapeutic targets, we identified 8480 proteins in thirteen pairs of ovarian cancer tissues and normal ovary tissues through quantitative proteomics. 498 proteins were found to be differentially expressed in ovarian cancer, which involved in various cellular processes, including metabolism, response to stimulus and biosynthetic process. The expression levels of chloride intracellular channel protein 1 (CLIC1) and lectin galactoside-binding soluble 3 binding protein (LGALS3BP) in epithelial ovarian cancer tissues were significantly higher than those in normal ovary tissues as confirmed by western blotting and immunohistochemistry. The knockdown of CLIC1 in A2780 cell line downregulated expression of CTPS1, leading to the decrease of CTP and an arrest of cell cycle G1 phase, which results into a slower proliferation. CLIC1-knockdown can also slow down the tumor growth in vivo. Besides, CLIC1-knockdown cells showed an increased sensitivity to hydrogen peroxide and cisplatin, suggesting that CLIC1 was involved in regulation of redox and drug resistance in ovarian cancer cells. These results indicate CLIC1 promotes tumorgenesis, and is a potential therapeutic target in epithelial ovarian cancer treatment.
format Online
Article
Text
id pubmed-5347761
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-53477612017-03-31 Identification and validation of differentially expressed proteins in epithelial ovarian cancers using quantitative proteomics Qu, Hong Chen, Yuling Cao, Guangming Liu, Chongdong Xu, Jiatong Deng, Haiteng Zhang, Zhenyu Oncotarget Research Paper Ovarian cancer is the most lethal gynecological malignant tumor because of its high recurrence rate. In the present work, in order to find new therapeutic targets, we identified 8480 proteins in thirteen pairs of ovarian cancer tissues and normal ovary tissues through quantitative proteomics. 498 proteins were found to be differentially expressed in ovarian cancer, which involved in various cellular processes, including metabolism, response to stimulus and biosynthetic process. The expression levels of chloride intracellular channel protein 1 (CLIC1) and lectin galactoside-binding soluble 3 binding protein (LGALS3BP) in epithelial ovarian cancer tissues were significantly higher than those in normal ovary tissues as confirmed by western blotting and immunohistochemistry. The knockdown of CLIC1 in A2780 cell line downregulated expression of CTPS1, leading to the decrease of CTP and an arrest of cell cycle G1 phase, which results into a slower proliferation. CLIC1-knockdown can also slow down the tumor growth in vivo. Besides, CLIC1-knockdown cells showed an increased sensitivity to hydrogen peroxide and cisplatin, suggesting that CLIC1 was involved in regulation of redox and drug resistance in ovarian cancer cells. These results indicate CLIC1 promotes tumorgenesis, and is a potential therapeutic target in epithelial ovarian cancer treatment. Impact Journals LLC 2016-11-04 /pmc/articles/PMC5347761/ /pubmed/27825122 http://dx.doi.org/10.18632/oncotarget.13077 Text en Copyright: © 2016 Qu et al. http://creativecommons.org/licenses/by/3.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Qu, Hong
Chen, Yuling
Cao, Guangming
Liu, Chongdong
Xu, Jiatong
Deng, Haiteng
Zhang, Zhenyu
Identification and validation of differentially expressed proteins in epithelial ovarian cancers using quantitative proteomics
title Identification and validation of differentially expressed proteins in epithelial ovarian cancers using quantitative proteomics
title_full Identification and validation of differentially expressed proteins in epithelial ovarian cancers using quantitative proteomics
title_fullStr Identification and validation of differentially expressed proteins in epithelial ovarian cancers using quantitative proteomics
title_full_unstemmed Identification and validation of differentially expressed proteins in epithelial ovarian cancers using quantitative proteomics
title_short Identification and validation of differentially expressed proteins in epithelial ovarian cancers using quantitative proteomics
title_sort identification and validation of differentially expressed proteins in epithelial ovarian cancers using quantitative proteomics
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5347761/
https://www.ncbi.nlm.nih.gov/pubmed/27825122
http://dx.doi.org/10.18632/oncotarget.13077
work_keys_str_mv AT quhong identificationandvalidationofdifferentiallyexpressedproteinsinepithelialovariancancersusingquantitativeproteomics
AT chenyuling identificationandvalidationofdifferentiallyexpressedproteinsinepithelialovariancancersusingquantitativeproteomics
AT caoguangming identificationandvalidationofdifferentiallyexpressedproteinsinepithelialovariancancersusingquantitativeproteomics
AT liuchongdong identificationandvalidationofdifferentiallyexpressedproteinsinepithelialovariancancersusingquantitativeproteomics
AT xujiatong identificationandvalidationofdifferentiallyexpressedproteinsinepithelialovariancancersusingquantitativeproteomics
AT denghaiteng identificationandvalidationofdifferentiallyexpressedproteinsinepithelialovariancancersusingquantitativeproteomics
AT zhangzhenyu identificationandvalidationofdifferentiallyexpressedproteinsinepithelialovariancancersusingquantitativeproteomics