The University of Southampton
University of Southampton Institutional Repository

A Kinome-Wide RNAi Screen in Drosophila Glia Reveals That the RIO Kinases Mediate Cell Proliferation and Survival through TORC2-Akt Signaling in Glioblastoma

A Kinome-Wide RNAi Screen in Drosophila Glia Reveals That the RIO Kinases Mediate Cell Proliferation and Survival through TORC2-Akt Signaling in Glioblastoma
A Kinome-Wide RNAi Screen in Drosophila Glia Reveals That the RIO Kinases Mediate Cell Proliferation and Survival through TORC2-Akt Signaling in Glioblastoma
Glioblastoma, the most common primary malignant brain tumor, is incurable with current therapies. Genetic and molecular analyses demonstrate that glioblastomas frequently display mutations that activate receptor tyrosine kinase (RTK) and Pi-3 kinase (PI3K) signaling pathways. In Drosophila melanogaster, activation of RTK and PI3K pathways in glial progenitor cells creates malignant neoplastic glial tumors that display many features of human glioblastoma. In both human and Drosophila, activation of the RTK and PI3K pathways stimulates Akt signaling along with other as-yet-unknown changes that drive oncogenesis. We used this Drosophila glioblastoma model to perform a kinome-wide genetic screen for new genes required for RTK- and PI3K-dependent neoplastic transformation. Human orthologs of novel kinases uncovered by these screens were functionally assessed in mammalian glioblastoma models and human tumors. Our results revealed that the atypical kinases RIOK1 and RIOK2 are overexpressed in glioblastoma cells in an Akt-dependent manner. Moreover, we found that overexpressed RIOK2 formed a complex with RIOK1, mTor, and mTor-complex-2 components, and that overexpressed RIOK2 upregulated Akt signaling and promoted tumorigenesis in murine astrocytes. Conversely, reduced expression of RIOK1 or RIOK2 disrupted Akt signaling and caused cell cycle exit, apoptosis, and chemosensitivity in glioblastoma cells by inducing p53 activity through the RpL11-dependent ribosomal stress checkpoint. These results imply that, in glioblastoma cells, constitutive Akt signaling drives RIO kinase overexpression, which creates a feedforward loop that promotes and maintains oncogenic Akt activity through stimulation of mTor signaling. Further study of the RIO kinases as well as other kinases identified in our Drosophila screen may reveal new insights into defects underlying glioblastoma and related cancers and may reveal new therapeutic opportunities for these cancers.
1553-7390
Read, RD
b5caca7b-0063-438a-b703-7ecbb6fc2b51
Fenton, TR
087260ba-f6a1-405a-85df-099d05810a84
Gomez, GG
0dd77897-fe6e-4ca4-808f-4be30b708991
Wykosky, J
411e9851-1e7e-4548-b23a-d43f56437455
Vandenberg, SR
ea04cace-bb69-4ee2-81db-90039d834ce2
Babic, I
dea1f220-998e-4c01-b5a3-086fb48a4b4c
Yang, H
0f90e934-3d0c-4be7-9b3a-b676373d15e3
Iwanami, A
56823670-3090-4bd7-b51a-8a7d777aac74
Cavenee, WK
e92aa948-8cc9-419f-a596-b36ed9ceeb09
Mischel, PS
03cba385-ecb8-4c4a-883a-db8303888c36
Furnari, FB
eb84de55-9ec1-480b-8424-11703ab8db3d
Thomas, JB
7b5051a9-1e81-40cd-bd71-c0f40f8110ab
Read, RD
b5caca7b-0063-438a-b703-7ecbb6fc2b51
Fenton, TR
087260ba-f6a1-405a-85df-099d05810a84
Gomez, GG
0dd77897-fe6e-4ca4-808f-4be30b708991
Wykosky, J
411e9851-1e7e-4548-b23a-d43f56437455
Vandenberg, SR
ea04cace-bb69-4ee2-81db-90039d834ce2
Babic, I
dea1f220-998e-4c01-b5a3-086fb48a4b4c
Yang, H
0f90e934-3d0c-4be7-9b3a-b676373d15e3
Iwanami, A
56823670-3090-4bd7-b51a-8a7d777aac74
Cavenee, WK
e92aa948-8cc9-419f-a596-b36ed9ceeb09
Mischel, PS
03cba385-ecb8-4c4a-883a-db8303888c36
Furnari, FB
eb84de55-9ec1-480b-8424-11703ab8db3d
Thomas, JB
7b5051a9-1e81-40cd-bd71-c0f40f8110ab

Read, RD, Fenton, TR, Gomez, GG, Wykosky, J, Vandenberg, SR, Babic, I, Yang, H, Iwanami, A, Cavenee, WK, Mischel, PS, Furnari, FB and Thomas, JB (2013) A Kinome-Wide RNAi Screen in Drosophila Glia Reveals That the RIO Kinases Mediate Cell Proliferation and Survival through TORC2-Akt Signaling in Glioblastoma. PLoS Genetics, 9 (2). (doi:10.1249/MSS.0b013e31822cb0d2).

Record type: Article

Abstract

Glioblastoma, the most common primary malignant brain tumor, is incurable with current therapies. Genetic and molecular analyses demonstrate that glioblastomas frequently display mutations that activate receptor tyrosine kinase (RTK) and Pi-3 kinase (PI3K) signaling pathways. In Drosophila melanogaster, activation of RTK and PI3K pathways in glial progenitor cells creates malignant neoplastic glial tumors that display many features of human glioblastoma. In both human and Drosophila, activation of the RTK and PI3K pathways stimulates Akt signaling along with other as-yet-unknown changes that drive oncogenesis. We used this Drosophila glioblastoma model to perform a kinome-wide genetic screen for new genes required for RTK- and PI3K-dependent neoplastic transformation. Human orthologs of novel kinases uncovered by these screens were functionally assessed in mammalian glioblastoma models and human tumors. Our results revealed that the atypical kinases RIOK1 and RIOK2 are overexpressed in glioblastoma cells in an Akt-dependent manner. Moreover, we found that overexpressed RIOK2 formed a complex with RIOK1, mTor, and mTor-complex-2 components, and that overexpressed RIOK2 upregulated Akt signaling and promoted tumorigenesis in murine astrocytes. Conversely, reduced expression of RIOK1 or RIOK2 disrupted Akt signaling and caused cell cycle exit, apoptosis, and chemosensitivity in glioblastoma cells by inducing p53 activity through the RpL11-dependent ribosomal stress checkpoint. These results imply that, in glioblastoma cells, constitutive Akt signaling drives RIO kinase overexpression, which creates a feedforward loop that promotes and maintains oncogenic Akt activity through stimulation of mTor signaling. Further study of the RIO kinases as well as other kinases identified in our Drosophila screen may reveal new insights into defects underlying glioblastoma and related cancers and may reveal new therapeutic opportunities for these cancers.

Text
Reliability_of_Cycling_Gross_Efficiency_Using_the.15 - Version of Record
Available under License Creative Commons Attribution.
Download (190kB)

More information

Published date: 1 February 2013
Additional Information: copyright 2013 Read et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Identifiers

Local EPrints ID: 453909
URI: http://eprints.soton.ac.uk/id/eprint/453909
ISSN: 1553-7390
PURE UUID: 48a0f58e-1659-4ac6-94a7-1f72de100b18
ORCID for RD Read: ORCID iD orcid.org/0000-0002-4297-6728
ORCID for TR Fenton: ORCID iD orcid.org/0000-0002-4737-8233

Catalogue record

Date deposited: 25 Jan 2022 17:52
Last modified: 17 Mar 2024 04:11

Export record

Altmetrics

Contributors

Author: RD Read ORCID iD
Author: TR Fenton ORCID iD
Author: GG Gomez
Author: J Wykosky
Author: SR Vandenberg
Author: I Babic
Author: H Yang
Author: A Iwanami
Author: WK Cavenee
Author: PS Mischel
Author: FB Furnari
Author: JB Thomas

Download statistics

Downloads from ePrints over the past year. Other digital versions may also be available to download e.g. from the publisher's website.

View more statistics

Atom RSS 1.0 RSS 2.0

Contact ePrints Soton: eprints@soton.ac.uk

ePrints Soton supports OAI 2.0 with a base URL of http://eprints.soton.ac.uk/cgi/oai2

This repository has been built using EPrints software, developed at the University of Southampton, but available to everyone to use.

We use cookies to ensure that we give you the best experience on our website. If you continue without changing your settings, we will assume that you are happy to receive cookies on the University of Southampton website.

×