Cargando…

UBE2E2 enhances Snail-mediated epithelial-mesenchymal transition and Nrf2-mediated antioxidant activity in ovarian cancer

Dissemination of ovarian cancer (OvCa) cells can lead to inoperable metastatic lesions in the bowel and omentum, which have a poor prognosis despite surgical and chemotherapeutical options. A better understanding of the mechanisms underlying metastasis is urgently needed. In this study, bioinformati...

Descripción completa

Detalles Bibliográficos
Autores principales: Hong, Xiaoling, Ma, Ning, Li, Danjie, Zhang, Mengwen, Dong, Wenqiuzi, Huang, Jie, Ci, Xinxin, Zhang, Songling
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9918489/
https://www.ncbi.nlm.nih.gov/pubmed/36765041
http://dx.doi.org/10.1038/s41419-023-05636-z
_version_ 1784886620434989056
author Hong, Xiaoling
Ma, Ning
Li, Danjie
Zhang, Mengwen
Dong, Wenqiuzi
Huang, Jie
Ci, Xinxin
Zhang, Songling
author_facet Hong, Xiaoling
Ma, Ning
Li, Danjie
Zhang, Mengwen
Dong, Wenqiuzi
Huang, Jie
Ci, Xinxin
Zhang, Songling
author_sort Hong, Xiaoling
collection PubMed
description Dissemination of ovarian cancer (OvCa) cells can lead to inoperable metastatic lesions in the bowel and omentum, which have a poor prognosis despite surgical and chemotherapeutical options. A better understanding of the mechanisms underlying metastasis is urgently needed. In this study, bioinformatics analyses revealed that UBE2E2, a less-studied ubiquitin (Ub)-conjugating enzyme (E2), was upregulated in OvCa and was associated with poor prognosis. Subsequently, we performed western blot analysis and IHC staining with 88 OvCa and 26 normal ovarian tissue samples, which further confirmed that UBE2E2 protein is highly expressed in OvCa tissue but weakly expressed in normal tissue. Furthermore, the silencing of UBE2E2 blocked OvCa cell migration, epithelial-mesenchymal transition (EMT) and metastasis in vitro, whereas UBE2E2 overexpression exerted the opposite effects. Mechanistically, UBE2E2 promoted p62 accumulation and increased the activity of the Nrf2-antioxidant response element (ARE) system, which ultimately activated the Snail signaling pathway by inhibiting the ubiquitin-mediated degradation of Snail. Additionally, co-IP and immunofluorescence demonstrated that a direct interaction exists between UBE2E2 and Nrf2, and the N-terminal of UBE2E2 (residues 1-52) is required and sufficient for its interaction with Nrf2 protein. Mutations in the active site cysteine (Cys139) impaired both the function and cellular distribution of UBE2E2. More importantly, the deletion of UBE2E2 reduced tumorigenicity and metastasis in xenograft OvCa mouse models. Taken together, our findings reveal the role of the UBE2E2-Nrf2-p62-Snail signaling axis in OvCa and thus provides novel therapeutic targets for the prevention of OvCa metastasis.
format Online
Article
Text
id pubmed-9918489
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-99184892023-02-12 UBE2E2 enhances Snail-mediated epithelial-mesenchymal transition and Nrf2-mediated antioxidant activity in ovarian cancer Hong, Xiaoling Ma, Ning Li, Danjie Zhang, Mengwen Dong, Wenqiuzi Huang, Jie Ci, Xinxin Zhang, Songling Cell Death Dis Article Dissemination of ovarian cancer (OvCa) cells can lead to inoperable metastatic lesions in the bowel and omentum, which have a poor prognosis despite surgical and chemotherapeutical options. A better understanding of the mechanisms underlying metastasis is urgently needed. In this study, bioinformatics analyses revealed that UBE2E2, a less-studied ubiquitin (Ub)-conjugating enzyme (E2), was upregulated in OvCa and was associated with poor prognosis. Subsequently, we performed western blot analysis and IHC staining with 88 OvCa and 26 normal ovarian tissue samples, which further confirmed that UBE2E2 protein is highly expressed in OvCa tissue but weakly expressed in normal tissue. Furthermore, the silencing of UBE2E2 blocked OvCa cell migration, epithelial-mesenchymal transition (EMT) and metastasis in vitro, whereas UBE2E2 overexpression exerted the opposite effects. Mechanistically, UBE2E2 promoted p62 accumulation and increased the activity of the Nrf2-antioxidant response element (ARE) system, which ultimately activated the Snail signaling pathway by inhibiting the ubiquitin-mediated degradation of Snail. Additionally, co-IP and immunofluorescence demonstrated that a direct interaction exists between UBE2E2 and Nrf2, and the N-terminal of UBE2E2 (residues 1-52) is required and sufficient for its interaction with Nrf2 protein. Mutations in the active site cysteine (Cys139) impaired both the function and cellular distribution of UBE2E2. More importantly, the deletion of UBE2E2 reduced tumorigenicity and metastasis in xenograft OvCa mouse models. Taken together, our findings reveal the role of the UBE2E2-Nrf2-p62-Snail signaling axis in OvCa and thus provides novel therapeutic targets for the prevention of OvCa metastasis. Nature Publishing Group UK 2023-02-10 /pmc/articles/PMC9918489/ /pubmed/36765041 http://dx.doi.org/10.1038/s41419-023-05636-z 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 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/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Hong, Xiaoling
Ma, Ning
Li, Danjie
Zhang, Mengwen
Dong, Wenqiuzi
Huang, Jie
Ci, Xinxin
Zhang, Songling
UBE2E2 enhances Snail-mediated epithelial-mesenchymal transition and Nrf2-mediated antioxidant activity in ovarian cancer
title UBE2E2 enhances Snail-mediated epithelial-mesenchymal transition and Nrf2-mediated antioxidant activity in ovarian cancer
title_full UBE2E2 enhances Snail-mediated epithelial-mesenchymal transition and Nrf2-mediated antioxidant activity in ovarian cancer
title_fullStr UBE2E2 enhances Snail-mediated epithelial-mesenchymal transition and Nrf2-mediated antioxidant activity in ovarian cancer
title_full_unstemmed UBE2E2 enhances Snail-mediated epithelial-mesenchymal transition and Nrf2-mediated antioxidant activity in ovarian cancer
title_short UBE2E2 enhances Snail-mediated epithelial-mesenchymal transition and Nrf2-mediated antioxidant activity in ovarian cancer
title_sort ube2e2 enhances snail-mediated epithelial-mesenchymal transition and nrf2-mediated antioxidant activity in ovarian cancer
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9918489/
https://www.ncbi.nlm.nih.gov/pubmed/36765041
http://dx.doi.org/10.1038/s41419-023-05636-z
work_keys_str_mv AT hongxiaoling ube2e2enhancessnailmediatedepithelialmesenchymaltransitionandnrf2mediatedantioxidantactivityinovariancancer
AT maning ube2e2enhancessnailmediatedepithelialmesenchymaltransitionandnrf2mediatedantioxidantactivityinovariancancer
AT lidanjie ube2e2enhancessnailmediatedepithelialmesenchymaltransitionandnrf2mediatedantioxidantactivityinovariancancer
AT zhangmengwen ube2e2enhancessnailmediatedepithelialmesenchymaltransitionandnrf2mediatedantioxidantactivityinovariancancer
AT dongwenqiuzi ube2e2enhancessnailmediatedepithelialmesenchymaltransitionandnrf2mediatedantioxidantactivityinovariancancer
AT huangjie ube2e2enhancessnailmediatedepithelialmesenchymaltransitionandnrf2mediatedantioxidantactivityinovariancancer
AT cixinxin ube2e2enhancessnailmediatedepithelialmesenchymaltransitionandnrf2mediatedantioxidantactivityinovariancancer
AT zhangsongling ube2e2enhancessnailmediatedepithelialmesenchymaltransitionandnrf2mediatedantioxidantactivityinovariancancer