A novel phosphatidylinositol 3-kinase (PI3K) inhibitor directs a potent FOXO-dependent, p53-independent cell cycle arrest phenotype characterized by the differential induction of a subset of FOXO-regulated genes. | - CCMAR -

Journal Article

TítuloA novel phosphatidylinositol 3-kinase (PI3K) inhibitor directs a potent FOXO-dependent, p53-independent cell cycle arrest phenotype characterized by the differential induction of a subset of FOXO-regulated genes.
Publication TypeJournal Article
AuthorsHill, R, Kalathur, RKiran Redd, Callejas, S, Colaço, L, Brandão, R, Serelde, B, Cebriá, A, Blanco-Aparicio, C, Pastor, J, Futschik, ME, Dopazo, A, Link, W
Year of Publication2014
JournalBreast Cancer Res
Volume16
Questão6
Date Published2014 Dec 09
Pagination482
ISSN1465-542X
Palavras-chaveAdenocarcinoma, Breast Neoplasms, Cell Cycle Checkpoints, Cell Line, Tumor, Cell Proliferation, Computer Simulation, Female, Forkhead Transcription Factors, HCT116 Cells, Humans, In Vitro Techniques, MCF-7 Cells, Phosphatidylinositol 3-Kinase, Proto-Oncogene Proteins c-akt, Pyrazoles, Pyrimidines, Real-Time Polymerase Chain Reaction, Signal Transduction
Abstract

INTRODUCTION: The activation of the phosphoinositide 3-kinase (PI3K)/AKT signalling pathway is one the most frequent genetic events in breast cancer, consequently the development of PI3K inhibitors has attracted much attention. Here we evaluate the effect of PI3K inhibition on global gene expression in breast cancer cells.

METHODS: We used a range of methodologies that include in silico compound analysis, in vitro kinase assays, cell invasion assays, proliferation assays, genome-wide transcription studies (Agilent Technologies full genome arrays), gene set enrichment analysis, quantitative real-time PCR, immunoblotting in addition to chromatin immunoprecipitation.

RESULTS: We defined the physico-chemical and the biological properties of ETP-45658, a novel potent PI3K inhibitor. We demonstrated that ETP-45658 potently inhibited cell proliferation within a broad range of human cancer cells, most potently suppressing the growth of breast cancer cells via inhibiting cell cycle. We show that this response is Forkhead box O (FOXO) protein dependent and p53 independent. Our genome-wide microarray analysis revealed that the cell cycle was the most affected biological process after exposure to ETP-45658 (or our control PI3K inhibitor PI-103), that despite the multiple transcription factors that are regulated by the PI3K/AKT signalling cascade, only the binding sites for FOXO transcription factors were significantly enriched and only a subset of all FOXO-dependent genes were induced. This disparity in gene transcription was not due to differential FOXO promoter recruitment.

CONCLUSIONS: The constitutive activation of PI3Ks and thus the exclusion of FOXO transcription factors from the nucleus is a key feature of breast cancer. Our results presented here highlight that PI3K inhibition activates specific FOXO-dependent genes that mediate cell cycle arrest in breast cancer cells.

DOI10.1186/s13058-014-0482-y
Sapientia

http://www.ncbi.nlm.nih.gov/pubmed/25488803?dopt=Abstract

Alternate JournalBreast Cancer Res.
PubMed ID25488803
PubMed Central IDPMC4303209
CCMAR Authors