Cargando…

Insulin-like growth factor 1-induced enolase 2 deacetylation by HDAC3 promotes metastasis of pancreatic cancer

Enolase 2 (ENO2) is a key glycolytic enzyme in the metabolic process of glycolysis, but its potential function in pancreatic ductal adenocarcinoma (PDAC) is unclear. In this study, we observed a significant overexpression of ENO2 in PDAC tissues, and its expression was correlated with metastasis and...

Descripción completa

Detalles Bibliográficos
Autores principales: Zheng, Yan, Wu, Chao, Yang, Jimeng, Zhao, Yue, Jia, Huliang, Xue, Min, Xu, Da, Yang, Feng, Fu, Deliang, Wang, Chaoqun, Hu, Beiyuan, Zhang, Ze, Li, Tianen, Yan, Shican, Wang, Xuan, Nelson, Peter J., Bruns, Christiane, Qin, Lunxiu, Dong, Qiongzhu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7217878/
https://www.ncbi.nlm.nih.gov/pubmed/32398667
http://dx.doi.org/10.1038/s41392-020-0146-6
_version_ 1783532679037190144
author Zheng, Yan
Wu, Chao
Yang, Jimeng
Zhao, Yue
Jia, Huliang
Xue, Min
Xu, Da
Yang, Feng
Fu, Deliang
Wang, Chaoqun
Hu, Beiyuan
Zhang, Ze
Li, Tianen
Yan, Shican
Wang, Xuan
Nelson, Peter J.
Bruns, Christiane
Qin, Lunxiu
Dong, Qiongzhu
author_facet Zheng, Yan
Wu, Chao
Yang, Jimeng
Zhao, Yue
Jia, Huliang
Xue, Min
Xu, Da
Yang, Feng
Fu, Deliang
Wang, Chaoqun
Hu, Beiyuan
Zhang, Ze
Li, Tianen
Yan, Shican
Wang, Xuan
Nelson, Peter J.
Bruns, Christiane
Qin, Lunxiu
Dong, Qiongzhu
author_sort Zheng, Yan
collection PubMed
description Enolase 2 (ENO2) is a key glycolytic enzyme in the metabolic process of glycolysis, but its potential function in pancreatic ductal adenocarcinoma (PDAC) is unclear. In this study, we observed a significant overexpression of ENO2 in PDAC tissues, and its expression was correlated with metastasis and poor prognosis in PDAC patients. K394 was identified as a major acetylation site in ENO2 that regulates its enzymatic activity, cell metabolism and PDAC progression. Knockdown of ENO2 suppressed tumor growth and liver metastasis in PDAC. Re-expression of wild-type (WT) ENO2, but not the K394 acetylation mimetic mutant, could reverse the decreased tumor malignancy. We further characterized histone deacetylase 3 (HDAC3) and P300/CBP-associated factor (PCAF) as the potential deacetylase and acetyltransferase for ENO2, respectively. HDAC3-mediated deacetylation was shown to lead to ENO2 activation and enhancement of glycolysis. Importantly, insulin-like growth factor-1 (IGF-1) was found to decrease K394 acetylation and stimulate ENO2 activity in a dose- and time-dependent manner. The PI3K/AKT/mTOR pathway facilitated the phosphorylation of HDAC3 on S424, which promoted K394 deacetylation and activation of ENO2. Linsitinib, an oral small-molecule inhibitor of IGF-1R, could inhibit IGF-1-induced ENO2 deacetylation by HDAC3 and the PI3K/AKT/mTOR pathway. Furthermore, linsitinib showed a different effect on the growth and metastasis of PDAC depending on the overexpression of WT versus K394-mutant ENO2. Our results reveal a novel mechanism by which acetylation negatively regulates ENO2 activity in the metastasis of PDAC by modulating glycolysis. Blockade of IGF-1-induced ENO2 deacetylation represents a promising strategy to prevent the development of PDAC.
format Online
Article
Text
id pubmed-7217878
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-72178782020-05-14 Insulin-like growth factor 1-induced enolase 2 deacetylation by HDAC3 promotes metastasis of pancreatic cancer Zheng, Yan Wu, Chao Yang, Jimeng Zhao, Yue Jia, Huliang Xue, Min Xu, Da Yang, Feng Fu, Deliang Wang, Chaoqun Hu, Beiyuan Zhang, Ze Li, Tianen Yan, Shican Wang, Xuan Nelson, Peter J. Bruns, Christiane Qin, Lunxiu Dong, Qiongzhu Signal Transduct Target Ther Article Enolase 2 (ENO2) is a key glycolytic enzyme in the metabolic process of glycolysis, but its potential function in pancreatic ductal adenocarcinoma (PDAC) is unclear. In this study, we observed a significant overexpression of ENO2 in PDAC tissues, and its expression was correlated with metastasis and poor prognosis in PDAC patients. K394 was identified as a major acetylation site in ENO2 that regulates its enzymatic activity, cell metabolism and PDAC progression. Knockdown of ENO2 suppressed tumor growth and liver metastasis in PDAC. Re-expression of wild-type (WT) ENO2, but not the K394 acetylation mimetic mutant, could reverse the decreased tumor malignancy. We further characterized histone deacetylase 3 (HDAC3) and P300/CBP-associated factor (PCAF) as the potential deacetylase and acetyltransferase for ENO2, respectively. HDAC3-mediated deacetylation was shown to lead to ENO2 activation and enhancement of glycolysis. Importantly, insulin-like growth factor-1 (IGF-1) was found to decrease K394 acetylation and stimulate ENO2 activity in a dose- and time-dependent manner. The PI3K/AKT/mTOR pathway facilitated the phosphorylation of HDAC3 on S424, which promoted K394 deacetylation and activation of ENO2. Linsitinib, an oral small-molecule inhibitor of IGF-1R, could inhibit IGF-1-induced ENO2 deacetylation by HDAC3 and the PI3K/AKT/mTOR pathway. Furthermore, linsitinib showed a different effect on the growth and metastasis of PDAC depending on the overexpression of WT versus K394-mutant ENO2. Our results reveal a novel mechanism by which acetylation negatively regulates ENO2 activity in the metastasis of PDAC by modulating glycolysis. Blockade of IGF-1-induced ENO2 deacetylation represents a promising strategy to prevent the development of PDAC. Nature Publishing Group UK 2020-05-13 /pmc/articles/PMC7217878/ /pubmed/32398667 http://dx.doi.org/10.1038/s41392-020-0146-6 Text en © The Author(s) 2020 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
spellingShingle Article
Zheng, Yan
Wu, Chao
Yang, Jimeng
Zhao, Yue
Jia, Huliang
Xue, Min
Xu, Da
Yang, Feng
Fu, Deliang
Wang, Chaoqun
Hu, Beiyuan
Zhang, Ze
Li, Tianen
Yan, Shican
Wang, Xuan
Nelson, Peter J.
Bruns, Christiane
Qin, Lunxiu
Dong, Qiongzhu
Insulin-like growth factor 1-induced enolase 2 deacetylation by HDAC3 promotes metastasis of pancreatic cancer
title Insulin-like growth factor 1-induced enolase 2 deacetylation by HDAC3 promotes metastasis of pancreatic cancer
title_full Insulin-like growth factor 1-induced enolase 2 deacetylation by HDAC3 promotes metastasis of pancreatic cancer
title_fullStr Insulin-like growth factor 1-induced enolase 2 deacetylation by HDAC3 promotes metastasis of pancreatic cancer
title_full_unstemmed Insulin-like growth factor 1-induced enolase 2 deacetylation by HDAC3 promotes metastasis of pancreatic cancer
title_short Insulin-like growth factor 1-induced enolase 2 deacetylation by HDAC3 promotes metastasis of pancreatic cancer
title_sort insulin-like growth factor 1-induced enolase 2 deacetylation by hdac3 promotes metastasis of pancreatic cancer
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7217878/
https://www.ncbi.nlm.nih.gov/pubmed/32398667
http://dx.doi.org/10.1038/s41392-020-0146-6
work_keys_str_mv AT zhengyan insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT wuchao insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT yangjimeng insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT zhaoyue insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT jiahuliang insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT xuemin insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT xuda insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT yangfeng insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT fudeliang insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT wangchaoqun insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT hubeiyuan insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT zhangze insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT litianen insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT yanshican insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT wangxuan insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT nelsonpeterj insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT brunschristiane insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT qinlunxiu insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer
AT dongqiongzhu insulinlikegrowthfactor1inducedenolase2deacetylationbyhdac3promotesmetastasisofpancreaticcancer