Poster Presentation & Flash Talk Presentation 28th Lorne Cancer Conference 2016

A genome-wide RNAi screen for bypass of AKT-induced senescence (#127)

Keefe T Chan 1 , Jeannine Diesch 2 , Katherine M Hannan 3 , Kaylene J Simpson 1 , Ross D Hannan 3 , Rick B Pearson 1
  1. Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
  2. Josep Carreras Leukaemia Research Institute, Badalona, Barcelona, Spain
  3. Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia

Hyperactivation of the PI3K/AKT/mTORC1 signalling pathway is a hallmark of many sporadic human cancers. However, we and others have demonstrated that chronic activation of this pathway in normal cells induces senescence, which effectively acts as a “brake” on the progression to malignancy. We hypothesise that specific genetic changes overcome this brake and permit the increased cell proliferation and transformation required for cancer development. Understanding the basis of oncogene-induced senescence in normal cells and how this is subverted in cancer cells will provide insight into the mechanism of cancer development and how it can be targeted.

Our previous work showed that AKT-induced senescence in normal human cells occurs via a p53 and mTORC1-dependent mechanism (Astle et al, Oncogene 2011). To further understand the critical mechanisms underlying AKT-induced senescence, we performed transcriptome profiling by RNA sequencing. Gene ontology analysis of differentially regulated genes between senescent and normal fibroblasts demonstrated significant upregulation of NF-kb target genes associated with a senescence-associated secretory phenotype (SASP), which is consistent with recent studies of mTORC1 signalling in SASP regulation (Laberge et al, Nat Cell Biol 2015; Herranz et al., Nat Cell Biol 2015). To determine the genetic changes required to overcome AKT-induced senescence, we have completed a genome-wide RNAi gain-of-function screen using multi-parametric readouts including cell number, proliferation, and senescence-associated beta-galactosidase staining. We identified 98 candidate gene targets, several of which are known and potential tumour suppressors. We are investigating how AKT governs these pathways and how they can be exploited in cancer.

  1. Astle MV, Hannan KM, Ng PY, Lee RS, George AJ, Hsu AK, Haupt Y, Hannan RD, Pearson RB. AKT induces senescence in human cells via mTORC1 and p53 in the absence of DNA damage: implications for targeting mTOR during malignancy. Oncogene. 2012 Apr 12;31(15):1949-62.
  2. Laberge RM, Sun Y, Orjalo AV, Patil CK, Freund A, Zhou L, Curran SC, Davalos AR, Wilson-Edell KA, Liu S, Limbad C, Demaria M, Li P, Hubbard GB, Ikeno Y, Javors M, Desprez PY, Benz CC, Kapahi P, Nelson PS, Campisi J. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat Cell Biol. 2015 Aug;17(8):1049-61.
  3. Herranz N, Gallage S, Mellone M, Wuestefeld T, Klotz S, Hanley CJ, Raguz S, Acosta JC, Innes AJ, Banito A, Georgilis A, Montoya A, Wolter K, Dharmalingam G, Faull P, Carroll T, Martínez-Barbera JP, Cutillas P, Reisinger F, Heikenwalder M, Miller RA, Withers D, Zender L, Thomas GJ, Gil J. mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat Cell Biol. 2015 Sep;17(9):1205-17.