NXN suppresses metastasis of hepatocellular carcinoma by promoting degradation of Snail through binding to DUB3

Abstract The poor prognosis of hepatocellular carcinoma (HCC) could be attributed to its high metastasis rate. Here, we report the role of nucleoredoxin (NXN), a multifunctional redox-active protein, in HCC metastasis. The expression of NXN in HCC tissues was measured by immunohistochemistry. The ro...

Full description

Bibliographic Details
Main Authors: Yuanping Zhang, Dinglan Zuo, Jiliang Qiu, Kai Li, Yi Niu, Yichuan Yuan, Yuxiong Qiu, Liang Qiao, Wei He, Chenwei Wang, Yunfei Yuan, Binkui Li
Format: Article
Language:English
Published: Nature Publishing Group 2022-08-01
Series:Cell Death and Disease
Online Access:https://doi.org/10.1038/s41419-022-05135-7
_version_ 1811342772438827008
author Yuanping Zhang
Dinglan Zuo
Jiliang Qiu
Kai Li
Yi Niu
Yichuan Yuan
Yuxiong Qiu
Liang Qiao
Wei He
Chenwei Wang
Yunfei Yuan
Binkui Li
author_facet Yuanping Zhang
Dinglan Zuo
Jiliang Qiu
Kai Li
Yi Niu
Yichuan Yuan
Yuxiong Qiu
Liang Qiao
Wei He
Chenwei Wang
Yunfei Yuan
Binkui Li
author_sort Yuanping Zhang
collection DOAJ
description Abstract The poor prognosis of hepatocellular carcinoma (HCC) could be attributed to its high metastasis rate. Here, we report the role of nucleoredoxin (NXN), a multifunctional redox-active protein, in HCC metastasis. The expression of NXN in HCC tissues was measured by immunohistochemistry. The role of NXN on HCC proliferation was determined by CCK-8, EdU and colony formation assays in vitro and subcutaneous tumor formation model in vivo. Transwell and wound healing assays and tail vein injection model were performed to assess the function of NXN on HCC metastasis. Co-immunoprecipitation assay was performed to examine the interaction among NXN, Snail and DUB3. Our results showed that NXN was downregulated in HCC tissues compared to adjacent liver tissues. Patients with low NXN expression had shorter overall survival (OS) time (P < 0.001) than those with high NXN expression. Biologically, ectopic expression of NXN significantly inhibited the proliferation and metastasis of HCC cells both in vitro and in vivo by suppressing epithelial-mesenchymal transition (EMT). Mechanistically, NXN promoted the ubiquitin-proteasome-mediated degradation of Snail through interaction with DUB3. Further, depletion of Snail abolished NXN-inhibited cell proliferation and metastasis. In summary, NXN suppressed the proliferation and metastasis of HCC by inhibiting DUB3-mediated deubiquitylation of Snail protein. Our study demonstrates that NXN, DUB3 and Snail complex functioned as an important regulatory mechanism of HCC progression and indicates a potential therapeutic approach for the treatment of HCC metastasis.
first_indexed 2024-04-13T19:16:49Z
format Article
id doaj.art-6fefaeace3bb4aac970fa686c3df9cde
institution Directory Open Access Journal
issn 2041-4889
language English
last_indexed 2024-04-13T19:16:49Z
publishDate 2022-08-01
publisher Nature Publishing Group
record_format Article
series Cell Death and Disease
spelling doaj.art-6fefaeace3bb4aac970fa686c3df9cde2022-12-22T02:33:39ZengNature Publishing GroupCell Death and Disease2041-48892022-08-0113811110.1038/s41419-022-05135-7NXN suppresses metastasis of hepatocellular carcinoma by promoting degradation of Snail through binding to DUB3Yuanping Zhang0Dinglan Zuo1Jiliang Qiu2Kai Li3Yi Niu4Yichuan Yuan5Yuxiong Qiu6Liang Qiao7Wei He8Chenwei Wang9Yunfei Yuan10Binkui Li11State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer CenterDepartment of Liver Surgery, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer CenterDepartment of Liver Surgery, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer CenterAbstract The poor prognosis of hepatocellular carcinoma (HCC) could be attributed to its high metastasis rate. Here, we report the role of nucleoredoxin (NXN), a multifunctional redox-active protein, in HCC metastasis. The expression of NXN in HCC tissues was measured by immunohistochemistry. The role of NXN on HCC proliferation was determined by CCK-8, EdU and colony formation assays in vitro and subcutaneous tumor formation model in vivo. Transwell and wound healing assays and tail vein injection model were performed to assess the function of NXN on HCC metastasis. Co-immunoprecipitation assay was performed to examine the interaction among NXN, Snail and DUB3. Our results showed that NXN was downregulated in HCC tissues compared to adjacent liver tissues. Patients with low NXN expression had shorter overall survival (OS) time (P < 0.001) than those with high NXN expression. Biologically, ectopic expression of NXN significantly inhibited the proliferation and metastasis of HCC cells both in vitro and in vivo by suppressing epithelial-mesenchymal transition (EMT). Mechanistically, NXN promoted the ubiquitin-proteasome-mediated degradation of Snail through interaction with DUB3. Further, depletion of Snail abolished NXN-inhibited cell proliferation and metastasis. In summary, NXN suppressed the proliferation and metastasis of HCC by inhibiting DUB3-mediated deubiquitylation of Snail protein. Our study demonstrates that NXN, DUB3 and Snail complex functioned as an important regulatory mechanism of HCC progression and indicates a potential therapeutic approach for the treatment of HCC metastasis.https://doi.org/10.1038/s41419-022-05135-7
spellingShingle Yuanping Zhang
Dinglan Zuo
Jiliang Qiu
Kai Li
Yi Niu
Yichuan Yuan
Yuxiong Qiu
Liang Qiao
Wei He
Chenwei Wang
Yunfei Yuan
Binkui Li
NXN suppresses metastasis of hepatocellular carcinoma by promoting degradation of Snail through binding to DUB3
Cell Death and Disease
title NXN suppresses metastasis of hepatocellular carcinoma by promoting degradation of Snail through binding to DUB3
title_full NXN suppresses metastasis of hepatocellular carcinoma by promoting degradation of Snail through binding to DUB3
title_fullStr NXN suppresses metastasis of hepatocellular carcinoma by promoting degradation of Snail through binding to DUB3
title_full_unstemmed NXN suppresses metastasis of hepatocellular carcinoma by promoting degradation of Snail through binding to DUB3
title_short NXN suppresses metastasis of hepatocellular carcinoma by promoting degradation of Snail through binding to DUB3
title_sort nxn suppresses metastasis of hepatocellular carcinoma by promoting degradation of snail through binding to dub3
url https://doi.org/10.1038/s41419-022-05135-7
work_keys_str_mv AT yuanpingzhang nxnsuppressesmetastasisofhepatocellularcarcinomabypromotingdegradationofsnailthroughbindingtodub3
AT dinglanzuo nxnsuppressesmetastasisofhepatocellularcarcinomabypromotingdegradationofsnailthroughbindingtodub3
AT jiliangqiu nxnsuppressesmetastasisofhepatocellularcarcinomabypromotingdegradationofsnailthroughbindingtodub3
AT kaili nxnsuppressesmetastasisofhepatocellularcarcinomabypromotingdegradationofsnailthroughbindingtodub3
AT yiniu nxnsuppressesmetastasisofhepatocellularcarcinomabypromotingdegradationofsnailthroughbindingtodub3
AT yichuanyuan nxnsuppressesmetastasisofhepatocellularcarcinomabypromotingdegradationofsnailthroughbindingtodub3
AT yuxiongqiu nxnsuppressesmetastasisofhepatocellularcarcinomabypromotingdegradationofsnailthroughbindingtodub3
AT liangqiao nxnsuppressesmetastasisofhepatocellularcarcinomabypromotingdegradationofsnailthroughbindingtodub3
AT weihe nxnsuppressesmetastasisofhepatocellularcarcinomabypromotingdegradationofsnailthroughbindingtodub3
AT chenweiwang nxnsuppressesmetastasisofhepatocellularcarcinomabypromotingdegradationofsnailthroughbindingtodub3
AT yunfeiyuan nxnsuppressesmetastasisofhepatocellularcarcinomabypromotingdegradationofsnailthroughbindingtodub3
AT binkuili nxnsuppressesmetastasisofhepatocellularcarcinomabypromotingdegradationofsnailthroughbindingtodub3