4.7 Article

microRNA regulation of mammalian target of rapamycin expression and activity controls estrogen receptor function and RADOO1 sensitivity

Journal

MOLECULAR CANCER
Volume 13, Issue -, Pages -

Publisher

BIOMED CENTRAL LTD
DOI: 10.1186/1476-4598-13-229

Keywords

miR-155; mTOR; breast cancer; miRNA; Estrogen receptor

Funding

  1. National Institutes of Health [CA125806, R01CA138268, R01AI106676, R01AI101046]
  2. Office of Naval Research [N00014-16-1-1136]
  3. National Center for Research Resources [P20RR020152]
  4. Department of Defense Breast Cancer Research Program [BC085426]
  5. Epigenetic Changes in Cancer Genomes (The Integrative Cancer Biology Program (ICBP): Centers for Cancer Systems Biology (CCSB) (KP Nephew) [NCI-U54 CA113001-07]

Ask authors/readers for more resources

Background: The AKT/mammalian target of rapamycin (mTOR) signaling pathway is regulated by 17a-estradiol (E2) signaling and mediates E2-induced proliferation and progesterone receptor (PgR) expression in breast cancer. Methods and results: Here we use deep sequencing analysis of previously published data from The Cancer Genome Atlas to demonstrate that expression of a key component of mTOR signaling, rapamycin-insensitive companion of mTOR (Rictor), positively correlated with an estrogen receptor-a positive (ERa+) breast tumor signature. Through increased microRNA-155 (miR-155) expression in the ERa+ breast cancer cells we demonstrate repression of Rictor enhanced activation of mTOR complex 1 (mTORC1) signaling with both qPCR and western blot. miR-155-mediated mTOR signaling resulted in deregulated ERa signaling both in cultured cells in vitro and in xenografts in vivo in addition to repressed PgR expression and activity. Furthermore we observed that miR-155 enhanced mTORC1 signaling (observed through western blot for increased phosphorylation on mTOR S2448) and induced inhibition of mTORC2 signaling (evident through repressed Rictor and tuberous sclerosis 1 (TSC1) gene expression). mTORC1 induced deregulation of E2 signaling was confirmed using qPCR and the mTORC1-specific inhibitor RAD001. Co-treatment of MCF7 breast cancer cells stably overexpressing miR-155 with RAD001 and E2 restored E2-induced PgR gene expression. RAD001 treatment of SCID/CB17 mice inhibited E2-induced tumorigenesis of the MCF7 miR-155 overexpressing cell line. Finally we demonstrated a strong positive correlation between Rictor and PgR expression and a negative correlation with Raptor expression in Luminal B breast cancer samples, a breast cancer histological subtype known for having an altered ERa-signaling pathway. Conclusions: miRNA mediated alterations in mTOR and ERa signaling establishes a new mechanism for altered estrogen responses independent of growth factor stimulation.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available