Cargando…

Upregulation of miR-181s reverses mesenchymal transition by targeting KPNA4 in glioblastoma

The goal of this work was to explore the most effective miRNAs affecting glioblastoma multiforme (GBM) phenotype transition and malignant progression. We annotated 491 TCGA samples’ miRNA expression profiles according to their mRNA-based subtypes and found that the mesenchymal tumors had significant...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Hongjun, Tao, Tao, Yan, Wei, Feng, Yan, Wang, Yongzhi, Cai, Jinquan, You, Yongping, Jiang, Tao, Jiang, Chuanlu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4539550/
https://www.ncbi.nlm.nih.gov/pubmed/26283154
http://dx.doi.org/10.1038/srep13072
_version_ 1782386127707045888
author Wang, Hongjun
Tao, Tao
Yan, Wei
Feng, Yan
Wang, Yongzhi
Cai, Jinquan
You, Yongping
Jiang, Tao
Jiang, Chuanlu
author_facet Wang, Hongjun
Tao, Tao
Yan, Wei
Feng, Yan
Wang, Yongzhi
Cai, Jinquan
You, Yongping
Jiang, Tao
Jiang, Chuanlu
author_sort Wang, Hongjun
collection PubMed
description The goal of this work was to explore the most effective miRNAs affecting glioblastoma multiforme (GBM) phenotype transition and malignant progression. We annotated 491 TCGA samples’ miRNA expression profiles according to their mRNA-based subtypes and found that the mesenchymal tumors had significantly decreased miR-181 family expression compared with the other three subtypes while the proneural subtype harbored extremely high miR-181 family expression. Patients with high miR-181 family expression had longer overall survival (p = 0.0031). We also confirmed that NF-κB-targeting genes and the EMT (epithelial-mesenchymal transition) pathway were inversely correlated with miR-181 family expression and that the entire miR-181 family inhibited glioma cell invasion and proliferation; of these, miR-181b was the most effective suppressor. Furthermore, miR-181b was validated to suppress EMT by targeting KPNA4 and was associated with survival outcome in the TCGA and CGGA datasets and in another independent cohort. The EMT-inhibitory effect of miR-181b was lost after KPNA4 expression was restored. We also identified the antitumorigenic activity of miR-181b in vitro and in vivo. Our results showed that miR-181 family expression was closely correlated with TCGA subtypes and patients’ overall survival, indicating that miR-181b, a tumor-suppressive miRNA, could be a novel therapeutic candidate for treating gliomas.
format Online
Article
Text
id pubmed-4539550
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-45395502015-08-26 Upregulation of miR-181s reverses mesenchymal transition by targeting KPNA4 in glioblastoma Wang, Hongjun Tao, Tao Yan, Wei Feng, Yan Wang, Yongzhi Cai, Jinquan You, Yongping Jiang, Tao Jiang, Chuanlu Sci Rep Article The goal of this work was to explore the most effective miRNAs affecting glioblastoma multiforme (GBM) phenotype transition and malignant progression. We annotated 491 TCGA samples’ miRNA expression profiles according to their mRNA-based subtypes and found that the mesenchymal tumors had significantly decreased miR-181 family expression compared with the other three subtypes while the proneural subtype harbored extremely high miR-181 family expression. Patients with high miR-181 family expression had longer overall survival (p = 0.0031). We also confirmed that NF-κB-targeting genes and the EMT (epithelial-mesenchymal transition) pathway were inversely correlated with miR-181 family expression and that the entire miR-181 family inhibited glioma cell invasion and proliferation; of these, miR-181b was the most effective suppressor. Furthermore, miR-181b was validated to suppress EMT by targeting KPNA4 and was associated with survival outcome in the TCGA and CGGA datasets and in another independent cohort. The EMT-inhibitory effect of miR-181b was lost after KPNA4 expression was restored. We also identified the antitumorigenic activity of miR-181b in vitro and in vivo. Our results showed that miR-181 family expression was closely correlated with TCGA subtypes and patients’ overall survival, indicating that miR-181b, a tumor-suppressive miRNA, could be a novel therapeutic candidate for treating gliomas. Nature Publishing Group 2015-08-18 /pmc/articles/PMC4539550/ /pubmed/26283154 http://dx.doi.org/10.1038/srep13072 Text en Copyright © 2015, Macmillan Publishers Limited http://creativecommons.org/licenses/by/4.0/ This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/
spellingShingle Article
Wang, Hongjun
Tao, Tao
Yan, Wei
Feng, Yan
Wang, Yongzhi
Cai, Jinquan
You, Yongping
Jiang, Tao
Jiang, Chuanlu
Upregulation of miR-181s reverses mesenchymal transition by targeting KPNA4 in glioblastoma
title Upregulation of miR-181s reverses mesenchymal transition by targeting KPNA4 in glioblastoma
title_full Upregulation of miR-181s reverses mesenchymal transition by targeting KPNA4 in glioblastoma
title_fullStr Upregulation of miR-181s reverses mesenchymal transition by targeting KPNA4 in glioblastoma
title_full_unstemmed Upregulation of miR-181s reverses mesenchymal transition by targeting KPNA4 in glioblastoma
title_short Upregulation of miR-181s reverses mesenchymal transition by targeting KPNA4 in glioblastoma
title_sort upregulation of mir-181s reverses mesenchymal transition by targeting kpna4 in glioblastoma
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4539550/
https://www.ncbi.nlm.nih.gov/pubmed/26283154
http://dx.doi.org/10.1038/srep13072
work_keys_str_mv AT wanghongjun upregulationofmir181sreversesmesenchymaltransitionbytargetingkpna4inglioblastoma
AT taotao upregulationofmir181sreversesmesenchymaltransitionbytargetingkpna4inglioblastoma
AT yanwei upregulationofmir181sreversesmesenchymaltransitionbytargetingkpna4inglioblastoma
AT fengyan upregulationofmir181sreversesmesenchymaltransitionbytargetingkpna4inglioblastoma
AT wangyongzhi upregulationofmir181sreversesmesenchymaltransitionbytargetingkpna4inglioblastoma
AT caijinquan upregulationofmir181sreversesmesenchymaltransitionbytargetingkpna4inglioblastoma
AT youyongping upregulationofmir181sreversesmesenchymaltransitionbytargetingkpna4inglioblastoma
AT jiangtao upregulationofmir181sreversesmesenchymaltransitionbytargetingkpna4inglioblastoma
AT jiangchuanlu upregulationofmir181sreversesmesenchymaltransitionbytargetingkpna4inglioblastoma