Cargando…

Intercellular adhesion molecule 2 as a novel prospective tumor suppressor induced by ERG promotes ubiquitination-mediated radixin degradation to inhibit gastric cancer tumorigenicity and metastasis

BACKGROUND: Gastric cancer (GC) is a fatal cancer with unclear pathogenesis. In this study, we explored the function and potential mechanisms of intercellular adhesion molecule 2 (ICAM2) in the development and advancement of GC. METHODS: Quantitative real-time polymerase chain reaction (qRT-PCR) and...

Descripción completa

Detalles Bibliográficos
Autores principales: Tang, Xiaocheng, Huang, Jintuan, Jiang, Yingming, Qiu, Jun, Li, Tuoyang, Li, Weiyao, Chen, Zijian, Huang, Zhenze, Yu, Xihu, Yang, Tao, Ji, Xiang, Tan, Rongchang, lv, Li, Yang, Zuli, Chen, Hao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10536727/
https://www.ncbi.nlm.nih.gov/pubmed/37759298
http://dx.doi.org/10.1186/s12967-023-04536-2
_version_ 1785112939101945856
author Tang, Xiaocheng
Huang, Jintuan
Jiang, Yingming
Qiu, Jun
Li, Tuoyang
Li, Weiyao
Chen, Zijian
Huang, Zhenze
Yu, Xihu
Yang, Tao
Ji, Xiang
Tan, Rongchang
lv, Li
Yang, Zuli
Chen, Hao
author_facet Tang, Xiaocheng
Huang, Jintuan
Jiang, Yingming
Qiu, Jun
Li, Tuoyang
Li, Weiyao
Chen, Zijian
Huang, Zhenze
Yu, Xihu
Yang, Tao
Ji, Xiang
Tan, Rongchang
lv, Li
Yang, Zuli
Chen, Hao
author_sort Tang, Xiaocheng
collection PubMed
description BACKGROUND: Gastric cancer (GC) is a fatal cancer with unclear pathogenesis. In this study, we explored the function and potential mechanisms of intercellular adhesion molecule 2 (ICAM2) in the development and advancement of GC. METHODS: Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were performed to quantify ICAM2 expression in harvested GC tissues and cultured cell lines. Immunohistochemical analyses were conducted on a GC tissue microarray to quantify ICAM2 expression and explore its implication on the prognosis of GC patients. In vitro experiments were carried out to reveal the biological functions of ICAM2 in GC cell lines. Further, in vivo experiments were conducted using xenograft models to assess the impact of ICAM2 on GC development and metastasis. Western blot, immunofluorescence, immunoprecipitation, luciferase assay, chromatin immunoprecipitation, and ubiquitination analysis were employed to investigate the underlying mechanisms. RESULTS: ICAM2 expression was downregulated in GC, positively correlating with advanced T stage, distant metastasis, advanced clinical stage, vessel invasion, and shorter patient survival time. ICAM2 overexpression suppressed the proliferation, migration, invasion, metastasis of GC cells as well as their ability to form tumors, whereas ICAM2 knockdown yielded opposite results. Erythroblast transformation-specific-related gene (ERG) as a transcription factor promoted the transcription of ICAM2 by binding to the crucial response element localized within its promoter region. Further analysis revealed that ICAM2 reduced radixin (RDX) protein stability and expression. In these cells, ICAM2 bound to the RDX protein to promote the ubiquitination and degradation of RDX via NEDD4 Like E3 Ubiquitin Protein Ligase (NEDD4L), and this post-translational modification resulted in the inhibition of GC. CONCLUSIONS: In summary, this study demonstrates that ICAM2, which is induced by ERG, suppresses GC progression by enhancing the ubiquitination and degradation of RDX in a NEDD4L-dependent manner. Therefore, these results suggest that ICAM2 is a potential prognostic marker and a therapeutic target for GC. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-023-04536-2.
format Online
Article
Text
id pubmed-10536727
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-105367272023-09-29 Intercellular adhesion molecule 2 as a novel prospective tumor suppressor induced by ERG promotes ubiquitination-mediated radixin degradation to inhibit gastric cancer tumorigenicity and metastasis Tang, Xiaocheng Huang, Jintuan Jiang, Yingming Qiu, Jun Li, Tuoyang Li, Weiyao Chen, Zijian Huang, Zhenze Yu, Xihu Yang, Tao Ji, Xiang Tan, Rongchang lv, Li Yang, Zuli Chen, Hao J Transl Med Research BACKGROUND: Gastric cancer (GC) is a fatal cancer with unclear pathogenesis. In this study, we explored the function and potential mechanisms of intercellular adhesion molecule 2 (ICAM2) in the development and advancement of GC. METHODS: Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were performed to quantify ICAM2 expression in harvested GC tissues and cultured cell lines. Immunohistochemical analyses were conducted on a GC tissue microarray to quantify ICAM2 expression and explore its implication on the prognosis of GC patients. In vitro experiments were carried out to reveal the biological functions of ICAM2 in GC cell lines. Further, in vivo experiments were conducted using xenograft models to assess the impact of ICAM2 on GC development and metastasis. Western blot, immunofluorescence, immunoprecipitation, luciferase assay, chromatin immunoprecipitation, and ubiquitination analysis were employed to investigate the underlying mechanisms. RESULTS: ICAM2 expression was downregulated in GC, positively correlating with advanced T stage, distant metastasis, advanced clinical stage, vessel invasion, and shorter patient survival time. ICAM2 overexpression suppressed the proliferation, migration, invasion, metastasis of GC cells as well as their ability to form tumors, whereas ICAM2 knockdown yielded opposite results. Erythroblast transformation-specific-related gene (ERG) as a transcription factor promoted the transcription of ICAM2 by binding to the crucial response element localized within its promoter region. Further analysis revealed that ICAM2 reduced radixin (RDX) protein stability and expression. In these cells, ICAM2 bound to the RDX protein to promote the ubiquitination and degradation of RDX via NEDD4 Like E3 Ubiquitin Protein Ligase (NEDD4L), and this post-translational modification resulted in the inhibition of GC. CONCLUSIONS: In summary, this study demonstrates that ICAM2, which is induced by ERG, suppresses GC progression by enhancing the ubiquitination and degradation of RDX in a NEDD4L-dependent manner. Therefore, these results suggest that ICAM2 is a potential prognostic marker and a therapeutic target for GC. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-023-04536-2. BioMed Central 2023-09-27 /pmc/articles/PMC10536727/ /pubmed/37759298 http://dx.doi.org/10.1186/s12967-023-04536-2 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/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 licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence 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 licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Tang, Xiaocheng
Huang, Jintuan
Jiang, Yingming
Qiu, Jun
Li, Tuoyang
Li, Weiyao
Chen, Zijian
Huang, Zhenze
Yu, Xihu
Yang, Tao
Ji, Xiang
Tan, Rongchang
lv, Li
Yang, Zuli
Chen, Hao
Intercellular adhesion molecule 2 as a novel prospective tumor suppressor induced by ERG promotes ubiquitination-mediated radixin degradation to inhibit gastric cancer tumorigenicity and metastasis
title Intercellular adhesion molecule 2 as a novel prospective tumor suppressor induced by ERG promotes ubiquitination-mediated radixin degradation to inhibit gastric cancer tumorigenicity and metastasis
title_full Intercellular adhesion molecule 2 as a novel prospective tumor suppressor induced by ERG promotes ubiquitination-mediated radixin degradation to inhibit gastric cancer tumorigenicity and metastasis
title_fullStr Intercellular adhesion molecule 2 as a novel prospective tumor suppressor induced by ERG promotes ubiquitination-mediated radixin degradation to inhibit gastric cancer tumorigenicity and metastasis
title_full_unstemmed Intercellular adhesion molecule 2 as a novel prospective tumor suppressor induced by ERG promotes ubiquitination-mediated radixin degradation to inhibit gastric cancer tumorigenicity and metastasis
title_short Intercellular adhesion molecule 2 as a novel prospective tumor suppressor induced by ERG promotes ubiquitination-mediated radixin degradation to inhibit gastric cancer tumorigenicity and metastasis
title_sort intercellular adhesion molecule 2 as a novel prospective tumor suppressor induced by erg promotes ubiquitination-mediated radixin degradation to inhibit gastric cancer tumorigenicity and metastasis
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10536727/
https://www.ncbi.nlm.nih.gov/pubmed/37759298
http://dx.doi.org/10.1186/s12967-023-04536-2
work_keys_str_mv AT tangxiaocheng intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT huangjintuan intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT jiangyingming intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT qiujun intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT lituoyang intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT liweiyao intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT chenzijian intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT huangzhenze intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT yuxihu intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT yangtao intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT jixiang intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT tanrongchang intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT lvli intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT yangzuli intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis
AT chenhao intercellularadhesionmolecule2asanovelprospectivetumorsuppressorinducedbyergpromotesubiquitinationmediatedradixindegradationtoinhibitgastriccancertumorigenicityandmetastasis