EEF1D overexpression promotes osteosarcoma cell proliferation by facilitating Akt-mTOR and Akt-bad signaling

Abstract Background Dysregulation of eukaryotic translation elongation factor 1 delta (EEF1D) in cancers has been reported; however, the role and mechanisms of EEF1D in osteosarcoma remain poorly understood. The aim of this study is to investigate the expression and role of EEF1D in osteosarcoma and...

Full description

Bibliographic Details
Main Authors: Dong-dong Cheng, Shi-jie Li, Bin Zhu, Shu-min Zhou, Qing-cheng Yang
Format: Article
Language:English
Published: BMC 2018-03-01
Series:Journal of Experimental & Clinical Cancer Research
Subjects:
Online Access:http://link.springer.com/article/10.1186/s13046-018-0715-5
id doaj-ee9485b70efe4f4d843c3532543a6fd3
record_format Article
spelling doaj-ee9485b70efe4f4d843c3532543a6fd32020-11-25T00:24:50ZengBMCJournal of Experimental & Clinical Cancer Research1756-99662018-03-0137111010.1186/s13046-018-0715-5EEF1D overexpression promotes osteosarcoma cell proliferation by facilitating Akt-mTOR and Akt-bad signalingDong-dong Cheng0Shi-jie Li1Bin Zhu2Shu-min Zhou3Qing-cheng Yang4Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalDepartment of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalDepartment of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalInstitution of microsurgery for limbs, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalDepartment of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalAbstract Background Dysregulation of eukaryotic translation elongation factor 1 delta (EEF1D) in cancers has been reported; however, the role and mechanisms of EEF1D in osteosarcoma remain poorly understood. The aim of this study is to investigate the expression and role of EEF1D in osteosarcoma and to elucidate its underlying mechanisms. Methods The expression of EEF1D in osteosarcomas and cell lines was evaluated by qRT-PCR, Western blotting and immunohistochemistry. EEF1D knockdown using small interfering RNA (siRNA) was employed to analyze the role of EEF1D in osteosarcoma cell proliferation and cell cycle progression. The host signaling pathways affected by EEF1D knockdown were detected using PathScan® intracellular signaling array kit. Results The expression of EEF1D was found to be up-regulated in human osteosarcoma tissues and cell lines. Its expression was positively correlated with Enneking stage and the tumor recurrence. EEF1D knockdown inhibited osteosarcoma cell proliferation, colony-forming ability, and cell cycle G2/M transition in vitro. In addition, EEF1D knockdown decreased the levels of phospho-Akt, phospho-mTOR, and phospho-Bad proteins. Conclusions EEF1D is upregulated in osteosarcoma and plays a tumor promoting role by facilitating Akt-mTOR and Akt-Bad signaling pathways. Accordingly, EEF1D is a potential target for cancer therapy.http://link.springer.com/article/10.1186/s13046-018-0715-5EEF1DProliferationAkt-mTOR signaling pathwayAkt-bad signaling pathwayOsteosarcoma
collection DOAJ
language English
format Article
sources DOAJ
author Dong-dong Cheng
Shi-jie Li
Bin Zhu
Shu-min Zhou
Qing-cheng Yang
spellingShingle Dong-dong Cheng
Shi-jie Li
Bin Zhu
Shu-min Zhou
Qing-cheng Yang
EEF1D overexpression promotes osteosarcoma cell proliferation by facilitating Akt-mTOR and Akt-bad signaling
Journal of Experimental & Clinical Cancer Research
EEF1D
Proliferation
Akt-mTOR signaling pathway
Akt-bad signaling pathway
Osteosarcoma
author_facet Dong-dong Cheng
Shi-jie Li
Bin Zhu
Shu-min Zhou
Qing-cheng Yang
author_sort Dong-dong Cheng
title EEF1D overexpression promotes osteosarcoma cell proliferation by facilitating Akt-mTOR and Akt-bad signaling
title_short EEF1D overexpression promotes osteosarcoma cell proliferation by facilitating Akt-mTOR and Akt-bad signaling
title_full EEF1D overexpression promotes osteosarcoma cell proliferation by facilitating Akt-mTOR and Akt-bad signaling
title_fullStr EEF1D overexpression promotes osteosarcoma cell proliferation by facilitating Akt-mTOR and Akt-bad signaling
title_full_unstemmed EEF1D overexpression promotes osteosarcoma cell proliferation by facilitating Akt-mTOR and Akt-bad signaling
title_sort eef1d overexpression promotes osteosarcoma cell proliferation by facilitating akt-mtor and akt-bad signaling
publisher BMC
series Journal of Experimental & Clinical Cancer Research
issn 1756-9966
publishDate 2018-03-01
description Abstract Background Dysregulation of eukaryotic translation elongation factor 1 delta (EEF1D) in cancers has been reported; however, the role and mechanisms of EEF1D in osteosarcoma remain poorly understood. The aim of this study is to investigate the expression and role of EEF1D in osteosarcoma and to elucidate its underlying mechanisms. Methods The expression of EEF1D in osteosarcomas and cell lines was evaluated by qRT-PCR, Western blotting and immunohistochemistry. EEF1D knockdown using small interfering RNA (siRNA) was employed to analyze the role of EEF1D in osteosarcoma cell proliferation and cell cycle progression. The host signaling pathways affected by EEF1D knockdown were detected using PathScan® intracellular signaling array kit. Results The expression of EEF1D was found to be up-regulated in human osteosarcoma tissues and cell lines. Its expression was positively correlated with Enneking stage and the tumor recurrence. EEF1D knockdown inhibited osteosarcoma cell proliferation, colony-forming ability, and cell cycle G2/M transition in vitro. In addition, EEF1D knockdown decreased the levels of phospho-Akt, phospho-mTOR, and phospho-Bad proteins. Conclusions EEF1D is upregulated in osteosarcoma and plays a tumor promoting role by facilitating Akt-mTOR and Akt-Bad signaling pathways. Accordingly, EEF1D is a potential target for cancer therapy.
topic EEF1D
Proliferation
Akt-mTOR signaling pathway
Akt-bad signaling pathway
Osteosarcoma
url http://link.springer.com/article/10.1186/s13046-018-0715-5
work_keys_str_mv AT dongdongcheng eef1doverexpressionpromotesosteosarcomacellproliferationbyfacilitatingaktmtorandaktbadsignaling
AT shijieli eef1doverexpressionpromotesosteosarcomacellproliferationbyfacilitatingaktmtorandaktbadsignaling
AT binzhu eef1doverexpressionpromotesosteosarcomacellproliferationbyfacilitatingaktmtorandaktbadsignaling
AT shuminzhou eef1doverexpressionpromotesosteosarcomacellproliferationbyfacilitatingaktmtorandaktbadsignaling
AT qingchengyang eef1doverexpressionpromotesosteosarcomacellproliferationbyfacilitatingaktmtorandaktbadsignaling
_version_ 1725351236922769408