Cargando…

FOXA1 promotes tumor cell proliferation through AR involving the Notch pathway in endometrial cancer

BACKGROUND: Increasing evidence suggests that forkhead box A1 (FOXA1) is frequently dysregulated in many types of human cancers. However, the exact function and mechanism of FOXA1 in human endometrial cancer (EC) remains unclear. METHODS: FOXA1 expression, androgen receptor (AR) expression, and the...

Descripción completa

Detalles Bibliográficos
Autores principales: Qiu, Meiting, Bao, Wei, Wang, Jingyun, Yang, Tingting, He, Xiaoying, Liao, Yun, Wan, Xiaoping
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3926330/
https://www.ncbi.nlm.nih.gov/pubmed/24512546
http://dx.doi.org/10.1186/1471-2407-14-78
_version_ 1782303960680366080
author Qiu, Meiting
Bao, Wei
Wang, Jingyun
Yang, Tingting
He, Xiaoying
Liao, Yun
Wan, Xiaoping
author_facet Qiu, Meiting
Bao, Wei
Wang, Jingyun
Yang, Tingting
He, Xiaoying
Liao, Yun
Wan, Xiaoping
author_sort Qiu, Meiting
collection PubMed
description BACKGROUND: Increasing evidence suggests that forkhead box A1 (FOXA1) is frequently dysregulated in many types of human cancers. However, the exact function and mechanism of FOXA1 in human endometrial cancer (EC) remains unclear. METHODS: FOXA1 expression, androgen receptor (AR) expression, and the relationships of these two markers with clinicopathological factors were determined by immunohistochemistry analysis. FOXA1 and AR were up-regulated by transient transfection with plasmids, and were down-regulated by transfection with siRNA or short hairpin RNA (shRNA). The effects of FOXA1 depletion and FOXA1 overexpression on AR-mediated transcription as well as Notch pathway and their impact on EC cell proliferation were examined by qRT-PCR, western blotting, co-immunoprecipitation, ChIP-PCR, MTT, colony-formation, and xenograft tumor–formation assays. RESULTS: We found that the expression of FOXA1 and AR in ECs was significantly higher than that in a typical hyperplasia and normal tissues. FOXA1 expression was significantly correlated with AR expression in clinical tissues. High FOXA1 levels positively correlated with pathological grade and depth of myometrial invasion in EC. High AR levels also positively correlated with pathological grade in EC. Moreover, the expression of XBP1, MYC, ZBTB16, and UHRF1, which are downstream targets of AR, was promoted by FOXA1 up-regulation or inhibited by FOXA1 down-regulation. Co-immunoprecipitation showed that FOXA1 interacted with AR in EC cells. ChIP-PCR assays showed that FOXA1 and AR could directly bind to the promoter and enhancer regions upstream of MYC. Mechanistic investigation revealed that over-expression of Notch1 and Hes1 proteins by FOXA1 could be reversed by AR depletion. In addition, we showed that down-regulation of AR attenuated FOXA1-up-regulated cell proliferation. However, AR didn’t influence the promotion effect of FOXA1 on cell migration and invasion. In vivo xenograft model, FOXA1 knockdown reduced the rate of tumor growth. CONCLUSIONS: These results suggest that FOXA1 promotes cell proliferation by AR and activates Notch pathway. It indicated that FOXA1 and AR may serve as potential gene therapy in EC.
format Online
Article
Text
id pubmed-3926330
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-39263302014-02-18 FOXA1 promotes tumor cell proliferation through AR involving the Notch pathway in endometrial cancer Qiu, Meiting Bao, Wei Wang, Jingyun Yang, Tingting He, Xiaoying Liao, Yun Wan, Xiaoping BMC Cancer Research Article BACKGROUND: Increasing evidence suggests that forkhead box A1 (FOXA1) is frequently dysregulated in many types of human cancers. However, the exact function and mechanism of FOXA1 in human endometrial cancer (EC) remains unclear. METHODS: FOXA1 expression, androgen receptor (AR) expression, and the relationships of these two markers with clinicopathological factors were determined by immunohistochemistry analysis. FOXA1 and AR were up-regulated by transient transfection with plasmids, and were down-regulated by transfection with siRNA or short hairpin RNA (shRNA). The effects of FOXA1 depletion and FOXA1 overexpression on AR-mediated transcription as well as Notch pathway and their impact on EC cell proliferation were examined by qRT-PCR, western blotting, co-immunoprecipitation, ChIP-PCR, MTT, colony-formation, and xenograft tumor–formation assays. RESULTS: We found that the expression of FOXA1 and AR in ECs was significantly higher than that in a typical hyperplasia and normal tissues. FOXA1 expression was significantly correlated with AR expression in clinical tissues. High FOXA1 levels positively correlated with pathological grade and depth of myometrial invasion in EC. High AR levels also positively correlated with pathological grade in EC. Moreover, the expression of XBP1, MYC, ZBTB16, and UHRF1, which are downstream targets of AR, was promoted by FOXA1 up-regulation or inhibited by FOXA1 down-regulation. Co-immunoprecipitation showed that FOXA1 interacted with AR in EC cells. ChIP-PCR assays showed that FOXA1 and AR could directly bind to the promoter and enhancer regions upstream of MYC. Mechanistic investigation revealed that over-expression of Notch1 and Hes1 proteins by FOXA1 could be reversed by AR depletion. In addition, we showed that down-regulation of AR attenuated FOXA1-up-regulated cell proliferation. However, AR didn’t influence the promotion effect of FOXA1 on cell migration and invasion. In vivo xenograft model, FOXA1 knockdown reduced the rate of tumor growth. CONCLUSIONS: These results suggest that FOXA1 promotes cell proliferation by AR and activates Notch pathway. It indicated that FOXA1 and AR may serve as potential gene therapy in EC. BioMed Central 2014-02-11 /pmc/articles/PMC3926330/ /pubmed/24512546 http://dx.doi.org/10.1186/1471-2407-14-78 Text en Copyright © 2014 Qiu et al.; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/2.0 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research Article
Qiu, Meiting
Bao, Wei
Wang, Jingyun
Yang, Tingting
He, Xiaoying
Liao, Yun
Wan, Xiaoping
FOXA1 promotes tumor cell proliferation through AR involving the Notch pathway in endometrial cancer
title FOXA1 promotes tumor cell proliferation through AR involving the Notch pathway in endometrial cancer
title_full FOXA1 promotes tumor cell proliferation through AR involving the Notch pathway in endometrial cancer
title_fullStr FOXA1 promotes tumor cell proliferation through AR involving the Notch pathway in endometrial cancer
title_full_unstemmed FOXA1 promotes tumor cell proliferation through AR involving the Notch pathway in endometrial cancer
title_short FOXA1 promotes tumor cell proliferation through AR involving the Notch pathway in endometrial cancer
title_sort foxa1 promotes tumor cell proliferation through ar involving the notch pathway in endometrial cancer
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3926330/
https://www.ncbi.nlm.nih.gov/pubmed/24512546
http://dx.doi.org/10.1186/1471-2407-14-78
work_keys_str_mv AT qiumeiting foxa1promotestumorcellproliferationthrougharinvolvingthenotchpathwayinendometrialcancer
AT baowei foxa1promotestumorcellproliferationthrougharinvolvingthenotchpathwayinendometrialcancer
AT wangjingyun foxa1promotestumorcellproliferationthrougharinvolvingthenotchpathwayinendometrialcancer
AT yangtingting foxa1promotestumorcellproliferationthrougharinvolvingthenotchpathwayinendometrialcancer
AT hexiaoying foxa1promotestumorcellproliferationthrougharinvolvingthenotchpathwayinendometrialcancer
AT liaoyun foxa1promotestumorcellproliferationthrougharinvolvingthenotchpathwayinendometrialcancer
AT wanxiaoping foxa1promotestumorcellproliferationthrougharinvolvingthenotchpathwayinendometrialcancer