Cargando…

BRAF-activated WT1 contributes to cancer growth and regulates autophagy and apoptosis in papillary thyroid carcinoma

BACKGROUND: Papillary thyroid carcinoma (PTC) is one of most prevalent malignant endocrine neoplasms, and it is associated with a high frequency of BRAF gene mutations, which lead to lymphatic metastasis and distant metastasis that promote tumor progression. The molecular mechanism of PTC and the ro...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Xing, Lin, Shan, Lin, Ying, Wu, Songsong, Zhuo, Minling, Zhang, Ailong, Zheng, Junjie, You, Zhenhui
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8818187/
https://www.ncbi.nlm.nih.gov/pubmed/35123502
http://dx.doi.org/10.1186/s12967-022-03260-7
_version_ 1784645779180224512
author Chen, Xing
Lin, Shan
Lin, Ying
Wu, Songsong
Zhuo, Minling
Zhang, Ailong
Zheng, Junjie
You, Zhenhui
author_facet Chen, Xing
Lin, Shan
Lin, Ying
Wu, Songsong
Zhuo, Minling
Zhang, Ailong
Zheng, Junjie
You, Zhenhui
author_sort Chen, Xing
collection PubMed
description BACKGROUND: Papillary thyroid carcinoma (PTC) is one of most prevalent malignant endocrine neoplasms, and it is associated with a high frequency of BRAF gene mutations, which lead to lymphatic metastasis and distant metastasis that promote tumor progression. The molecular mechanism of PTC and the role of BRAF mutation in PTC progression and development need to be further elucidated. METHODS: In this study, a comprehensive bioinformatics analysis was performed to identify the differentially expressed genes and signaling pathways in thyroid cancer patients carrying mutant BRAF. Then, we confirmed the prognostic role of WT1 in thyroid cancer patients. Immunohistochemistry was performed to measure the expression profile of WT1 in PTC tissue. Lentivirus shWT1 was transfected into BRAF(V600E) (mutant) PTC cells to stably inhibit WT1 expression. CCK-8, EdU, immunofluorescence, colony formation, cell migration, cell wound healing, apoptosis and autophagy assays were performed to assess the biological functions of WT1 in BRAF(V600E) PTC cells. RNA sequencing, immunohistochemistry and immunoblotting were performed to explore the molecular mechanism of WT1 in BRAF(V600E) PTC cells. RESULTS: The results confirmed that “epithelial cell proliferation”, “apoptosis” and “selective autophagy” were closely associated with this BRAF mutant in these thyroid cancer patients. Knocking down BRAF-activated WT1 effectively inhibited the proliferation and migration of BRAF(V600E) PTC cells. Silencing WT1 significantly inhibited autophagy and promoted the apoptosis of BRAF(V600E) PTC cells. Mechanistic investigations showed that silencing WT1 expression remarkably suppressed the AKT/mTOR and ERK/P65 signaling pathways in BRAF(V600E) PTC cells. CONCLUSION: All these results indicate that WT1 is a promising prognostic biomarker and facilitates PTC progression and development of cells carrying the BRAF(V600E) mutation. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-022-03260-7.
format Online
Article
Text
id pubmed-8818187
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-88181872022-02-07 BRAF-activated WT1 contributes to cancer growth and regulates autophagy and apoptosis in papillary thyroid carcinoma Chen, Xing Lin, Shan Lin, Ying Wu, Songsong Zhuo, Minling Zhang, Ailong Zheng, Junjie You, Zhenhui J Transl Med Research BACKGROUND: Papillary thyroid carcinoma (PTC) is one of most prevalent malignant endocrine neoplasms, and it is associated with a high frequency of BRAF gene mutations, which lead to lymphatic metastasis and distant metastasis that promote tumor progression. The molecular mechanism of PTC and the role of BRAF mutation in PTC progression and development need to be further elucidated. METHODS: In this study, a comprehensive bioinformatics analysis was performed to identify the differentially expressed genes and signaling pathways in thyroid cancer patients carrying mutant BRAF. Then, we confirmed the prognostic role of WT1 in thyroid cancer patients. Immunohistochemistry was performed to measure the expression profile of WT1 in PTC tissue. Lentivirus shWT1 was transfected into BRAF(V600E) (mutant) PTC cells to stably inhibit WT1 expression. CCK-8, EdU, immunofluorescence, colony formation, cell migration, cell wound healing, apoptosis and autophagy assays were performed to assess the biological functions of WT1 in BRAF(V600E) PTC cells. RNA sequencing, immunohistochemistry and immunoblotting were performed to explore the molecular mechanism of WT1 in BRAF(V600E) PTC cells. RESULTS: The results confirmed that “epithelial cell proliferation”, “apoptosis” and “selective autophagy” were closely associated with this BRAF mutant in these thyroid cancer patients. Knocking down BRAF-activated WT1 effectively inhibited the proliferation and migration of BRAF(V600E) PTC cells. Silencing WT1 significantly inhibited autophagy and promoted the apoptosis of BRAF(V600E) PTC cells. Mechanistic investigations showed that silencing WT1 expression remarkably suppressed the AKT/mTOR and ERK/P65 signaling pathways in BRAF(V600E) PTC cells. CONCLUSION: All these results indicate that WT1 is a promising prognostic biomarker and facilitates PTC progression and development of cells carrying the BRAF(V600E) mutation. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-022-03260-7. BioMed Central 2022-02-05 /pmc/articles/PMC8818187/ /pubmed/35123502 http://dx.doi.org/10.1186/s12967-022-03260-7 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis 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
Chen, Xing
Lin, Shan
Lin, Ying
Wu, Songsong
Zhuo, Minling
Zhang, Ailong
Zheng, Junjie
You, Zhenhui
BRAF-activated WT1 contributes to cancer growth and regulates autophagy and apoptosis in papillary thyroid carcinoma
title BRAF-activated WT1 contributes to cancer growth and regulates autophagy and apoptosis in papillary thyroid carcinoma
title_full BRAF-activated WT1 contributes to cancer growth and regulates autophagy and apoptosis in papillary thyroid carcinoma
title_fullStr BRAF-activated WT1 contributes to cancer growth and regulates autophagy and apoptosis in papillary thyroid carcinoma
title_full_unstemmed BRAF-activated WT1 contributes to cancer growth and regulates autophagy and apoptosis in papillary thyroid carcinoma
title_short BRAF-activated WT1 contributes to cancer growth and regulates autophagy and apoptosis in papillary thyroid carcinoma
title_sort braf-activated wt1 contributes to cancer growth and regulates autophagy and apoptosis in papillary thyroid carcinoma
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8818187/
https://www.ncbi.nlm.nih.gov/pubmed/35123502
http://dx.doi.org/10.1186/s12967-022-03260-7
work_keys_str_mv AT chenxing brafactivatedwt1contributestocancergrowthandregulatesautophagyandapoptosisinpapillarythyroidcarcinoma
AT linshan brafactivatedwt1contributestocancergrowthandregulatesautophagyandapoptosisinpapillarythyroidcarcinoma
AT linying brafactivatedwt1contributestocancergrowthandregulatesautophagyandapoptosisinpapillarythyroidcarcinoma
AT wusongsong brafactivatedwt1contributestocancergrowthandregulatesautophagyandapoptosisinpapillarythyroidcarcinoma
AT zhuominling brafactivatedwt1contributestocancergrowthandregulatesautophagyandapoptosisinpapillarythyroidcarcinoma
AT zhangailong brafactivatedwt1contributestocancergrowthandregulatesautophagyandapoptosisinpapillarythyroidcarcinoma
AT zhengjunjie brafactivatedwt1contributestocancergrowthandregulatesautophagyandapoptosisinpapillarythyroidcarcinoma
AT youzhenhui brafactivatedwt1contributestocancergrowthandregulatesautophagyandapoptosisinpapillarythyroidcarcinoma