Correlation Engine 2.0
Clear Search sequence regions


Sizes of these terms reflect their relevance to your search.

We previously identified the mechanistic target of rapamycin complex 2 (mTORC2) as an effector of Ras for the control of directed cell migration in Dictyostelium. Recently, the Ras-mediated regulation of mTORC2 was found to be conserved in mammalian cells, and mTORC2 was shown to be an effector of oncogenic Ras. Interestingly, mTORC2 has been linked to cancer cell migration, and particularly in breast cancer. Here, we investigated the role of Ras in promoting the migration and invasion of breast cancer cells through mTORC2. We observed that both Ras and mTORC2 promote the migration of different breast cancer cells and breast cancer cell models. Using HER2 and oncogenic Ras-transformed breast epithelial MCF10A cells, we found that both wild-type Ras and oncogenic Ras promote mTORC2 activation and an mTORC2-dependent migration and invasion in these breast cancer models. We further observed that, whereas oncogenic Ras-transformed MCF10A cells display uncontrolled cell proliferation and invasion, disruption of mTORC2 leads to loss of invasiveness only. Together, our findings suggest that, whereas the Ras-mediated activation of mTORC2 is expected to play a minor role in breast tumor formation, the Ras-mTORC2 pathway plays an important role in promoting the migration and invasion of breast cancer cells.

Citation

Shannon E Collins, Mollie E Wiegand, Alyssa N Werner, Isabella N Brown, Mary I Mundo, Douglas J Swango, Ghassan Mouneimne, Pascale G Charest. Ras-mediated activation of mTORC2 promotes breast epithelial cell migration and invasion. Molecular biology of the cell. 2023 Feb 01;34(2):ar9

Expand section icon Mesh Tags

Expand section icon Substances


PMID: 36542482

View Full Text