Cargando…

Epigenomic characterization of a p53-regulated 3p22.2 tumor suppressor that inhibits STAT3 phosphorylation via protein docking and is frequently methylated in esophageal and other carcinomas

Rationale: Oncogenic STAT3 signaling activation and 3p22-21.3 locus alteration are common in multiple tumors, especially carcinomas of the nasopharynx, esophagus and lung. Whether these two events are linked remains unclear. Our CpG methylome analysis identified a 3p22.2 gene, DLEC1, as a methylated...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Lili, Xu, Juan, Qiu, Guohua, Ying, Jianming, Du, Zhenfang, Xiang, Tingxiu, Wong, Kai Yau, Srivastava, Gopesh, Zhu, Xiao-Feng, Mok, Tony S, Chan, Anthony TC, Chan, Francis KL, Ambinder, Richard F, Tao, Qian
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5743460/
https://www.ncbi.nlm.nih.gov/pubmed/29290793
http://dx.doi.org/10.7150/thno.20893
_version_ 1783288569119375360
author Li, Lili
Xu, Juan
Qiu, Guohua
Ying, Jianming
Du, Zhenfang
Xiang, Tingxiu
Wong, Kai Yau
Srivastava, Gopesh
Zhu, Xiao-Feng
Mok, Tony S
Chan, Anthony TC
Chan, Francis KL
Ambinder, Richard F
Tao, Qian
author_facet Li, Lili
Xu, Juan
Qiu, Guohua
Ying, Jianming
Du, Zhenfang
Xiang, Tingxiu
Wong, Kai Yau
Srivastava, Gopesh
Zhu, Xiao-Feng
Mok, Tony S
Chan, Anthony TC
Chan, Francis KL
Ambinder, Richard F
Tao, Qian
author_sort Li, Lili
collection PubMed
description Rationale: Oncogenic STAT3 signaling activation and 3p22-21.3 locus alteration are common in multiple tumors, especially carcinomas of the nasopharynx, esophagus and lung. Whether these two events are linked remains unclear. Our CpG methylome analysis identified a 3p22.2 gene, DLEC1, as a methylated target in esophageal squamous cell (ESCC), nasopharyngeal (NPC) and lung carcinomas. Thus, we further characterized its epigenetic abnormalities and functions. Methods: CpG methylomes were established by methylated DNA immunoprecipitation. Promoter methylation was analyzed by methylation-specific PCR and bisulfite genomic sequencing. DLEC1 expression and clinical significance were analyzed using TCGA database. DLEC1 functions were analyzed by transfections followed by various cell biology assays. Protein-protein interaction was assessed by docking, Western blot and immunoprecipitation analyses. Results: We defined the DLEC1 promoter within a CpG island and p53-regulated. DLEC1 was frequently downregulated in ESCC, lung and NPC cell lines and primary tumors, but was readily expressed in normal tissues and immortalized normal epithelial cells, with mutations rarely detected. DLEC1 methylation was frequently detected in ESCC tumors and correlated with lymph node metastasis, tumor recurrence and progression, with DLEC1 as the most frequently methylated among the established 3p22.2 tumor suppressors (RASSF1A, PLCD1 and ZMYND10/BLU). DLEC1 inhibits carcinoma cell growth through inducing cell cycle arrest and apoptosis, and also suppresses cell metastasis by reversing epithelial-mesenchymal transition (EMT) and cell stemness. Moreover, DLEC1 represses oncogenic signaling including JAK/STAT3, MAPK/ERK, Wnt/β-catenin and AKT pathways in multiple carcinoma types. Particularly, DLEC1 inhibits IL-6-induced STAT3 phosphorylation in a dose-dependent manner. DLEC1 contains three YXXQ motifs and forms a protein complex with STAT3 via protein docking, which blocks STAT3-JAK2 interaction and STAT3 phosphorylation. IL-6 stimulation enhances the binding of DLEC1 with STAT3, which diminishes their interaction with JAK2 and further leads to decreased STAT3 phosphorylation. The YXXQ motifs of DLEC1 are crucial for its inhibition of STAT3 phosphorylation, and disruption of these motifs restores STAT3 phosphorylation through abolishing DLEC1 binding to STAT3. Conclusions: Our study demonstrates, for the first time, predominant epigenetic silencing of DLEC1 in ESCC, and a novel mechanistic link of epigenetic DLEC1 disruption with oncogenic STAT3 signaling in multiple carcinomas.
format Online
Article
Text
id pubmed-5743460
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-57434602018-01-01 Epigenomic characterization of a p53-regulated 3p22.2 tumor suppressor that inhibits STAT3 phosphorylation via protein docking and is frequently methylated in esophageal and other carcinomas Li, Lili Xu, Juan Qiu, Guohua Ying, Jianming Du, Zhenfang Xiang, Tingxiu Wong, Kai Yau Srivastava, Gopesh Zhu, Xiao-Feng Mok, Tony S Chan, Anthony TC Chan, Francis KL Ambinder, Richard F Tao, Qian Theranostics Research Paper Rationale: Oncogenic STAT3 signaling activation and 3p22-21.3 locus alteration are common in multiple tumors, especially carcinomas of the nasopharynx, esophagus and lung. Whether these two events are linked remains unclear. Our CpG methylome analysis identified a 3p22.2 gene, DLEC1, as a methylated target in esophageal squamous cell (ESCC), nasopharyngeal (NPC) and lung carcinomas. Thus, we further characterized its epigenetic abnormalities and functions. Methods: CpG methylomes were established by methylated DNA immunoprecipitation. Promoter methylation was analyzed by methylation-specific PCR and bisulfite genomic sequencing. DLEC1 expression and clinical significance were analyzed using TCGA database. DLEC1 functions were analyzed by transfections followed by various cell biology assays. Protein-protein interaction was assessed by docking, Western blot and immunoprecipitation analyses. Results: We defined the DLEC1 promoter within a CpG island and p53-regulated. DLEC1 was frequently downregulated in ESCC, lung and NPC cell lines and primary tumors, but was readily expressed in normal tissues and immortalized normal epithelial cells, with mutations rarely detected. DLEC1 methylation was frequently detected in ESCC tumors and correlated with lymph node metastasis, tumor recurrence and progression, with DLEC1 as the most frequently methylated among the established 3p22.2 tumor suppressors (RASSF1A, PLCD1 and ZMYND10/BLU). DLEC1 inhibits carcinoma cell growth through inducing cell cycle arrest and apoptosis, and also suppresses cell metastasis by reversing epithelial-mesenchymal transition (EMT) and cell stemness. Moreover, DLEC1 represses oncogenic signaling including JAK/STAT3, MAPK/ERK, Wnt/β-catenin and AKT pathways in multiple carcinoma types. Particularly, DLEC1 inhibits IL-6-induced STAT3 phosphorylation in a dose-dependent manner. DLEC1 contains three YXXQ motifs and forms a protein complex with STAT3 via protein docking, which blocks STAT3-JAK2 interaction and STAT3 phosphorylation. IL-6 stimulation enhances the binding of DLEC1 with STAT3, which diminishes their interaction with JAK2 and further leads to decreased STAT3 phosphorylation. The YXXQ motifs of DLEC1 are crucial for its inhibition of STAT3 phosphorylation, and disruption of these motifs restores STAT3 phosphorylation through abolishing DLEC1 binding to STAT3. Conclusions: Our study demonstrates, for the first time, predominant epigenetic silencing of DLEC1 in ESCC, and a novel mechanistic link of epigenetic DLEC1 disruption with oncogenic STAT3 signaling in multiple carcinomas. Ivyspring International Publisher 2018-01-01 /pmc/articles/PMC5743460/ /pubmed/29290793 http://dx.doi.org/10.7150/thno.20893 Text en © Ivyspring International Publisher This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Li, Lili
Xu, Juan
Qiu, Guohua
Ying, Jianming
Du, Zhenfang
Xiang, Tingxiu
Wong, Kai Yau
Srivastava, Gopesh
Zhu, Xiao-Feng
Mok, Tony S
Chan, Anthony TC
Chan, Francis KL
Ambinder, Richard F
Tao, Qian
Epigenomic characterization of a p53-regulated 3p22.2 tumor suppressor that inhibits STAT3 phosphorylation via protein docking and is frequently methylated in esophageal and other carcinomas
title Epigenomic characterization of a p53-regulated 3p22.2 tumor suppressor that inhibits STAT3 phosphorylation via protein docking and is frequently methylated in esophageal and other carcinomas
title_full Epigenomic characterization of a p53-regulated 3p22.2 tumor suppressor that inhibits STAT3 phosphorylation via protein docking and is frequently methylated in esophageal and other carcinomas
title_fullStr Epigenomic characterization of a p53-regulated 3p22.2 tumor suppressor that inhibits STAT3 phosphorylation via protein docking and is frequently methylated in esophageal and other carcinomas
title_full_unstemmed Epigenomic characterization of a p53-regulated 3p22.2 tumor suppressor that inhibits STAT3 phosphorylation via protein docking and is frequently methylated in esophageal and other carcinomas
title_short Epigenomic characterization of a p53-regulated 3p22.2 tumor suppressor that inhibits STAT3 phosphorylation via protein docking and is frequently methylated in esophageal and other carcinomas
title_sort epigenomic characterization of a p53-regulated 3p22.2 tumor suppressor that inhibits stat3 phosphorylation via protein docking and is frequently methylated in esophageal and other carcinomas
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5743460/
https://www.ncbi.nlm.nih.gov/pubmed/29290793
http://dx.doi.org/10.7150/thno.20893
work_keys_str_mv AT lilili epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT xujuan epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT qiuguohua epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT yingjianming epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT duzhenfang epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT xiangtingxiu epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT wongkaiyau epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT srivastavagopesh epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT zhuxiaofeng epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT moktonys epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT chananthonytc epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT chanfranciskl epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT ambinderrichardf epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas
AT taoqian epigenomiccharacterizationofap53regulated3p222tumorsuppressorthatinhibitsstat3phosphorylationviaproteindockingandisfrequentlymethylatedinesophagealandothercarcinomas