Abstract

PRL-3, a metastasis-associated phosphatase, is known to exert its oncogenic functions through activation of PI3K/Akt, which is a key regulator of the rapamycin-sensitive mTOR complex 1 (mTORC1), but a coherent link between PRL-3 and activation of mTOR has not yet been formally demonstrated. We report a positive correlation between PRL-3 expression and mTOR phospho-activation in clinical tumour samples and mouse models of cancer and demonstrate that PRL-3 increased downstream signalling to the mTOR substrates, p70S6K and 4E-BP1, by increasing PI3K/Akt-mediated activation of Rheb-GTP via TSC2 suppression. We also show that PRL-3 increases mTOR translocation to lysosomes via increased mTOR binding affinity to Rag GTPases in an Akt-independent manner, demonstrating a previously undescribed mechanism of action for PRL-3. PRL-3 also enhanced matrix metalloproteinase-2 secretion and cellular invasiveness via activation of mTOR, attributes which were sensitive to rapamycin treatment. The downstream effects of PRL-3 were maintained even under conditions of environmental stress, suggesting that PRL-3 provides a strategic survival advantage to tumour cells via its effects on mTOR.

Highlights

  • We further investigated the above finding in the spontaneous mouse mammary tumour virus (MMTV) transgenic model, which harbours the polyomavirus middle T oncoprotein (PyMT) under transcriptional control of the MMTV promoter-enhancer, resulting in the formation of palpable mammary tumours in mice as early as 6 weeks of age[26]

  • We found that PRL-3 overexpression induces an aberrant activation of mechanistic target of rapamycin (mTOR) kinase in cancer cells, as reflected by hyperphosphorylation of the direct substrates of mTOR complex 1 (mTORC1), 4E (eIF4E)-binding protein 1 (4E-BP1) and p70 S6 kinase (p70S6K)

  • We formally demonstrate the signalling pathway by which PRL-3 induces mTORC1 activation as via the Akt-TSC2-Rheb signalling pathway

Read more

Summary

Introduction

Full mTORC1 activation is a two-pronged process, requiring growth factor signalling (via PI3K/Akt) to activate Rheb, and mTORC1 translocation to Rheb-resident endomembranes, lysosomes[11]. Through the activation of upstream RTKs, PRL-3 enhances cell growth and survival through multiple oncogenic effector pathways, including PI3K/Akt, Ras/MAPK, and SRC17–19. Given that cells overexpressing PRL-3 exhibit shared characteristics with cells possessing hyperactive PI3K/Akt/mTOR signalling, including enhanced cell proliferation, survival, and motility, we hypothesized that PRL-3 might potentially play a role in mTOR regulation, as well in cancer progression. PRL-3 was shown previously to promote autophagy in ovarian cancers[23], a phenomenon typically inhibited by mTOR activity[24] This observation was later shown to be rapamycin-insensitive, discounting a role for mTORC1 as a proxy for PRL-3-driven autophagy. Our results uncover a role for oncogenic PRL-3 signalling via mTORC1 both in vivo and in vitro

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.