PELP1 Suppression Inhibits Gastric Cancer Through Downregulation of c-Src-PI3K-ERK Pathway

Background: Proline-, glutamic acid-, and leucine-rich protein 1 (PELP1), a co-activator of estrogen receptors alpha, was confirmed to be directly associated with the oncogenic process of multiple cancers, especially hormone-dependent cancers. The purpose of our research was to explore the biologica...

Full description

Bibliographic Details
Main Authors: Hongzhu Yan, Yanling Sun, Qian Wu, Zhe Wu, Meichun Hu, Yuanpeng Sun, Yusi Liu, Zi Ma, Shangqin Liu, Wuhan Xiao, Fuxing Liu, Zhifeng Ning
Format: Article
Language:English
Published: Frontiers Media S.A. 2020-02-01
Series:Frontiers in Oncology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fonc.2019.01423/full
id doaj-47d394fe678c439788e2e438f2707ac2
record_format Article
spelling doaj-47d394fe678c439788e2e438f2707ac22020-11-25T00:34:37ZengFrontiers Media S.A.Frontiers in Oncology2234-943X2020-02-01910.3389/fonc.2019.01423461305PELP1 Suppression Inhibits Gastric Cancer Through Downregulation of c-Src-PI3K-ERK PathwayHongzhu Yan0Hongzhu Yan1Yanling Sun2Qian Wu3Zhe Wu4Meichun Hu5Yuanpeng Sun6Yusi Liu7Zi Ma8Shangqin Liu9Wuhan Xiao10Fuxing Liu11Zhifeng Ning12Basic Medical School, Hubei University of Science and Technology, Xianning, ChinaInstitute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, ChinaBasic Medical School, Hubei University of Science and Technology, Xianning, ChinaBasic Medical School, Hubei University of Science and Technology, Xianning, ChinaBasic Medical School, Hubei University of Science and Technology, Xianning, ChinaBasic Medical School, Hubei University of Science and Technology, Xianning, ChinaBasic Medical School, Hubei University of Science and Technology, Xianning, ChinaBasic Medical School, Hubei University of Science and Technology, Xianning, ChinaWuhan University Zhongnan Hospital, Wuhan, ChinaWuhan University Zhongnan Hospital, Wuhan, ChinaThe Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, ChinaBasic Medical School, Hubei University of Science and Technology, Xianning, ChinaBasic Medical School, Hubei University of Science and Technology, Xianning, ChinaBackground: Proline-, glutamic acid-, and leucine-rich protein 1 (PELP1), a co-activator of estrogen receptors alpha, was confirmed to be directly associated with the oncogenic process of multiple cancers, especially hormone-dependent cancers. The purpose of our research was to explore the biological function, clinical significance, and therapeutic targeted value of PELP1 in gastric cancer (GC).Methods: The expression status of PELP1 in GC cell lines or tissues was analyzed through bioinformatics data mining. Thirty-six GC tissue chip was applied to demonstrate the results of bioinformatics data mining assayed by immunohistochemical method. The expression status of PELP1 in GC cell lines was also analyzed using western blot. Correlation analysis between PELP1 expression and clinicopathological parameter was performed. Kaplan-Meier survival analysis was applied to analyze the relationship between PELP1 expression and total survival time. Three pairs of siRNA were designed to silence the expression of PELP1 in GC. After PELP1 was silenced by siRNA or activated by saRNA, the growth, plate colony formation, migration and invasion ability of the GC cell or normal gastric epithelium cell line was tested in vitro. Cell cycle was tested by flow cytometry. Nude mice xenograft experiment was performed after PELP1 was silenced. The downstream molecular pathway regulated by PELP1 was explored. Molecular docking tool was applied to combine chlorpromazine with PELP1. The inhibitory effect of chlorpromazine in GC was assayed, then it was tested whether PELP1 was a therapeutic target of chlorpromazine in GC.Results: PELP1 expression was elevated in GC cell lines and clinical GC tissue samples. PELP1 silence by siRNA compromised the malignant traits of GC. PELP1 expression positively correlated with tumor invasion depth, lymph node metastasis, tissue grade, TNM stage, but had no correlation with patient age, sex, tumor size, and tumor numbers. Kaplan-Meier survival analysis revealed high PELP1 expression had a shorter survival period in GC patients after follow-up. Q-PCR and western blot revealed PELP1 suppression in GC decreased expression of the c-Src-PI3K-ERK pathway. It was also implied that chlorpromazine (CPZ) can inhibit the malignant traits of GC and downregulate the expression of PELP1.Conclusions: In a word, PELP1 is an oncogene in gastric cancer and c-Src-PI3K-ERK pathway activation may be responsible for its tumorigenesis, PELP1 may be a potential therapeutic target of chlorpromazine in GC.https://www.frontiersin.org/article/10.3389/fonc.2019.01423/fullPELP1oncogenemaster genegastric cancerc-Src-PI3K-ERK pathwaychlorpromazine
collection DOAJ
language English
format Article
sources DOAJ
author Hongzhu Yan
Hongzhu Yan
Yanling Sun
Qian Wu
Zhe Wu
Meichun Hu
Yuanpeng Sun
Yusi Liu
Zi Ma
Shangqin Liu
Wuhan Xiao
Fuxing Liu
Zhifeng Ning
spellingShingle Hongzhu Yan
Hongzhu Yan
Yanling Sun
Qian Wu
Zhe Wu
Meichun Hu
Yuanpeng Sun
Yusi Liu
Zi Ma
Shangqin Liu
Wuhan Xiao
Fuxing Liu
Zhifeng Ning
PELP1 Suppression Inhibits Gastric Cancer Through Downregulation of c-Src-PI3K-ERK Pathway
Frontiers in Oncology
PELP1
oncogene
master gene
gastric cancer
c-Src-PI3K-ERK pathway
chlorpromazine
author_facet Hongzhu Yan
Hongzhu Yan
Yanling Sun
Qian Wu
Zhe Wu
Meichun Hu
Yuanpeng Sun
Yusi Liu
Zi Ma
Shangqin Liu
Wuhan Xiao
Fuxing Liu
Zhifeng Ning
author_sort Hongzhu Yan
title PELP1 Suppression Inhibits Gastric Cancer Through Downregulation of c-Src-PI3K-ERK Pathway
title_short PELP1 Suppression Inhibits Gastric Cancer Through Downregulation of c-Src-PI3K-ERK Pathway
title_full PELP1 Suppression Inhibits Gastric Cancer Through Downregulation of c-Src-PI3K-ERK Pathway
title_fullStr PELP1 Suppression Inhibits Gastric Cancer Through Downregulation of c-Src-PI3K-ERK Pathway
title_full_unstemmed PELP1 Suppression Inhibits Gastric Cancer Through Downregulation of c-Src-PI3K-ERK Pathway
title_sort pelp1 suppression inhibits gastric cancer through downregulation of c-src-pi3k-erk pathway
publisher Frontiers Media S.A.
series Frontiers in Oncology
issn 2234-943X
publishDate 2020-02-01
description Background: Proline-, glutamic acid-, and leucine-rich protein 1 (PELP1), a co-activator of estrogen receptors alpha, was confirmed to be directly associated with the oncogenic process of multiple cancers, especially hormone-dependent cancers. The purpose of our research was to explore the biological function, clinical significance, and therapeutic targeted value of PELP1 in gastric cancer (GC).Methods: The expression status of PELP1 in GC cell lines or tissues was analyzed through bioinformatics data mining. Thirty-six GC tissue chip was applied to demonstrate the results of bioinformatics data mining assayed by immunohistochemical method. The expression status of PELP1 in GC cell lines was also analyzed using western blot. Correlation analysis between PELP1 expression and clinicopathological parameter was performed. Kaplan-Meier survival analysis was applied to analyze the relationship between PELP1 expression and total survival time. Three pairs of siRNA were designed to silence the expression of PELP1 in GC. After PELP1 was silenced by siRNA or activated by saRNA, the growth, plate colony formation, migration and invasion ability of the GC cell or normal gastric epithelium cell line was tested in vitro. Cell cycle was tested by flow cytometry. Nude mice xenograft experiment was performed after PELP1 was silenced. The downstream molecular pathway regulated by PELP1 was explored. Molecular docking tool was applied to combine chlorpromazine with PELP1. The inhibitory effect of chlorpromazine in GC was assayed, then it was tested whether PELP1 was a therapeutic target of chlorpromazine in GC.Results: PELP1 expression was elevated in GC cell lines and clinical GC tissue samples. PELP1 silence by siRNA compromised the malignant traits of GC. PELP1 expression positively correlated with tumor invasion depth, lymph node metastasis, tissue grade, TNM stage, but had no correlation with patient age, sex, tumor size, and tumor numbers. Kaplan-Meier survival analysis revealed high PELP1 expression had a shorter survival period in GC patients after follow-up. Q-PCR and western blot revealed PELP1 suppression in GC decreased expression of the c-Src-PI3K-ERK pathway. It was also implied that chlorpromazine (CPZ) can inhibit the malignant traits of GC and downregulate the expression of PELP1.Conclusions: In a word, PELP1 is an oncogene in gastric cancer and c-Src-PI3K-ERK pathway activation may be responsible for its tumorigenesis, PELP1 may be a potential therapeutic target of chlorpromazine in GC.
topic PELP1
oncogene
master gene
gastric cancer
c-Src-PI3K-ERK pathway
chlorpromazine
url https://www.frontiersin.org/article/10.3389/fonc.2019.01423/full
work_keys_str_mv AT hongzhuyan pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
AT hongzhuyan pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
AT yanlingsun pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
AT qianwu pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
AT zhewu pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
AT meichunhu pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
AT yuanpengsun pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
AT yusiliu pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
AT zima pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
AT shangqinliu pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
AT wuhanxiao pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
AT fuxingliu pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
AT zhifengning pelp1suppressioninhibitsgastriccancerthroughdownregulationofcsrcpi3kerkpathway
_version_ 1725312534201761792