Cargando…

MYBBP1A suppresses breast cancer tumorigenesis by enhancing the p53 dependent anoikis

BACKGROUND: Tumor suppressor p53 is mutated in a wide variety of human cancers and plays a critical role in anoikis, which is essential for preventing tumorigenesis. Recently, we found that a nucleolar protein, Myb-binding protein 1a (MYBBP1A), was involved in p53 activation. However, the function o...

Descripción completa

Detalles Bibliográficos
Autores principales: Akaogi, Kensuke, Ono, Wakana, Hayashi, Yuki, Kishimoto, Hiroyuki, Yanagisawa, Junn
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2013
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3575238/
https://www.ncbi.nlm.nih.gov/pubmed/23388179
http://dx.doi.org/10.1186/1471-2407-13-65
_version_ 1782259679270797312
author Akaogi, Kensuke
Ono, Wakana
Hayashi, Yuki
Kishimoto, Hiroyuki
Yanagisawa, Junn
author_facet Akaogi, Kensuke
Ono, Wakana
Hayashi, Yuki
Kishimoto, Hiroyuki
Yanagisawa, Junn
author_sort Akaogi, Kensuke
collection PubMed
description BACKGROUND: Tumor suppressor p53 is mutated in a wide variety of human cancers and plays a critical role in anoikis, which is essential for preventing tumorigenesis. Recently, we found that a nucleolar protein, Myb-binding protein 1a (MYBBP1A), was involved in p53 activation. However, the function of MYBBP1A in cancer prevention has not been elucidated. METHODS: Relationships between MYBBP1A expression levels and breast cancer progression were examined using patient microarray databases and tissue microarrays. Colony formation, xenograft, and anoikis assays were conducted using cells in which MYBBP1A was either knocked down or overexpressed. p53 activation and interactions between p53 and MYBBP1A were assessed by immunoprecipitation and western blot. RESULTS: MYBBP1A expression was negatively correlated with breast cancer tumorigenesis. In vivo and in vitro experiments using the breast cancer cell lines MCF-7 and ZR-75-1, which expresses wild type p53, showed that tumorigenesis, colony formation, and anoikis resistance were significantly enhanced by MYBBP1A knockdown. We also found that MYBBP1A binds to p53 and enhances p53 target gene transcription under anoikis conditions. CONCLUSIONS: These results suggest that MYBBP1A is required for p53 activation during anoikis; therefore, it is involved in suppressing colony formation and the tumorigenesis of breast cancer cells. Collectively, our results suggest that MYBBP1A plays a role in tumor prevention in the context of p53 activation.
format Online
Article
Text
id pubmed-3575238
institution National Center for Biotechnology Information
language English
publishDate 2013
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-35752382013-02-19 MYBBP1A suppresses breast cancer tumorigenesis by enhancing the p53 dependent anoikis Akaogi, Kensuke Ono, Wakana Hayashi, Yuki Kishimoto, Hiroyuki Yanagisawa, Junn BMC Cancer Research Article BACKGROUND: Tumor suppressor p53 is mutated in a wide variety of human cancers and plays a critical role in anoikis, which is essential for preventing tumorigenesis. Recently, we found that a nucleolar protein, Myb-binding protein 1a (MYBBP1A), was involved in p53 activation. However, the function of MYBBP1A in cancer prevention has not been elucidated. METHODS: Relationships between MYBBP1A expression levels and breast cancer progression were examined using patient microarray databases and tissue microarrays. Colony formation, xenograft, and anoikis assays were conducted using cells in which MYBBP1A was either knocked down or overexpressed. p53 activation and interactions between p53 and MYBBP1A were assessed by immunoprecipitation and western blot. RESULTS: MYBBP1A expression was negatively correlated with breast cancer tumorigenesis. In vivo and in vitro experiments using the breast cancer cell lines MCF-7 and ZR-75-1, which expresses wild type p53, showed that tumorigenesis, colony formation, and anoikis resistance were significantly enhanced by MYBBP1A knockdown. We also found that MYBBP1A binds to p53 and enhances p53 target gene transcription under anoikis conditions. CONCLUSIONS: These results suggest that MYBBP1A is required for p53 activation during anoikis; therefore, it is involved in suppressing colony formation and the tumorigenesis of breast cancer cells. Collectively, our results suggest that MYBBP1A plays a role in tumor prevention in the context of p53 activation. BioMed Central 2013-02-07 /pmc/articles/PMC3575238/ /pubmed/23388179 http://dx.doi.org/10.1186/1471-2407-13-65 Text en Copyright ©2013 Akaogi 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.
spellingShingle Research Article
Akaogi, Kensuke
Ono, Wakana
Hayashi, Yuki
Kishimoto, Hiroyuki
Yanagisawa, Junn
MYBBP1A suppresses breast cancer tumorigenesis by enhancing the p53 dependent anoikis
title MYBBP1A suppresses breast cancer tumorigenesis by enhancing the p53 dependent anoikis
title_full MYBBP1A suppresses breast cancer tumorigenesis by enhancing the p53 dependent anoikis
title_fullStr MYBBP1A suppresses breast cancer tumorigenesis by enhancing the p53 dependent anoikis
title_full_unstemmed MYBBP1A suppresses breast cancer tumorigenesis by enhancing the p53 dependent anoikis
title_short MYBBP1A suppresses breast cancer tumorigenesis by enhancing the p53 dependent anoikis
title_sort mybbp1a suppresses breast cancer tumorigenesis by enhancing the p53 dependent anoikis
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3575238/
https://www.ncbi.nlm.nih.gov/pubmed/23388179
http://dx.doi.org/10.1186/1471-2407-13-65
work_keys_str_mv AT akaogikensuke mybbp1asuppressesbreastcancertumorigenesisbyenhancingthep53dependentanoikis
AT onowakana mybbp1asuppressesbreastcancertumorigenesisbyenhancingthep53dependentanoikis
AT hayashiyuki mybbp1asuppressesbreastcancertumorigenesisbyenhancingthep53dependentanoikis
AT kishimotohiroyuki mybbp1asuppressesbreastcancertumorigenesisbyenhancingthep53dependentanoikis
AT yanagisawajunn mybbp1asuppressesbreastcancertumorigenesisbyenhancingthep53dependentanoikis