Abstract

BackgroundRASSF1A, a tumor suppressor gene, is frequently inactivated in lung cancer leading to a YAP-dependent epithelial-mesenchymal transition (EMT). Such effects are partly due to the inactivation of the anti-migratory RhoB GTPase via the inhibitory phosphorylation of GEF-H1, the GDP/GTP exchange factor for RhoB. However, the kinase responsible for RhoB/GEF-H1 inactivation in RASSF1A-depleted cells remained unknown.MethodsNDR1/2 inactivation by siRNA or shRNA effects on epithelial-mesenchymal transition, invasion, xenograft formation and growth in SCID−/− Beige mice, apoptosis, proliferation, cytokinesis, YAP/TAZ activation were investigated upon RASSF1A loss in human bronchial epithelial cells (HBEC).ResultsWe demonstrate here that depletion of the YAP-kinases NDR1/2 reverts migration and metastatic properties upon RASSF1A loss in HBEC. We show that NDR2 interacts directly with GEF-H1 (which contains the NDR phosphorylation consensus motif HXRXXS/T), leading to GEF-H1 phosphorylation. We further report that the RASSF1A/NDR2/GEF-H1/RhoB/YAP axis is involved in proper cytokinesis in human bronchial cells, since chromosome proper segregation are NDR-dependent upon RASSF1A or GEF-H1 loss in HBEC.ConclusionTo summarize, our data support a model in which, upon RASSF1A silencing, NDR2 gets activated, phosphorylates and inactivates GEF-H1, leading to RhoB inactivation. This cascade induced by RASSF1A loss in bronchial cells is responsible for metastasis properties, YAP activation and cytokinesis defects.

Highlights

  • RASSF1A, a tumor suppressor gene, is frequently inactivated in lung cancer leading to a YAP-dependent epithelial-mesenchymal transition (EMT)

  • Depletion of the NDR1/2 kinases reverts the migratory and metastatic phenotypes induced by RASSF1A loss in human bronchial epithelial cells (HBEC) Forty-eight hours after silencing RASSF1A or NDR1/2 in HBEC cells using siRNAs (Additional file 1: Table S2), we performed wound healing (Fig. 1a) and invasion (Fig. 1b) assays in the presence of mitomycin C, to inhibit the contribution of cell division in wound repair

  • The increase of migration and invasion induced by RASSF1A loss in HBEC-3 cells ([3], Fig. 1a and b) was inhibited by NDR1/2 depletion (Fig. 1a and b) without leading to cell death

Read more

Summary

Introduction

RASSF1A, a tumor suppressor gene, is frequently inactivated in lung cancer leading to a YAP-dependent epithelial-mesenchymal transition (EMT) Such effects are partly due to the inactivation of the anti-migratory RhoB GTPase via the inhibitory phosphorylation of GEF-H1, the GDP/GTP exchange factor for RhoB. RASSF1A [Ras association (RalGDS/AF-6) domain family member 1], a tumor and metastatic suppressor gene, is frequently inactivated and an independent predictor of poor prognosis in resected early-stage non–small cell lung cancer (NSCLC) [1, 2]. RhoB inactivation leads to the cancer-associated phenotypes observed in RASSF1A-depleted HBEC

Methods
Results
Discussion
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.