mTOR dependent transformed human cells have a distinct set of essential genes from bcr-abl transformed cells

Constitutively active intracellular signaling drives and sustains cancer growth. The mTOR kinase integrates multiple inputs sensing nutrient, energy and growth factor levels to promote protein synthesis and anabolic metabolism, and is hyperactivated in a broad range of cancers. The bcr-abl kinase is a fusion protein generated by chromosomal translocation and gives rise to chronic myeloid leukemia and a small sub-set of leukemias. Using an in vitro transformed murine cell model, we performed shRNA knockdown of 25 genes previously shown to be essential for mTOR dependent oncogenic growth and survival. None of the genes were essential in the bcr-abl transformed line. Interrogation of this gene set in human cancer cell lines revealed that many of these genes were essential in cells dependent on mTOR signaling (defined by sensitivity to pharmacological mTOR inhibition). However, none of the genes were essential in bcr-abl transformed K562 cells that are insensitive to mTOR inhibition. Thus there is a clear divide between cells transformed via bcr-abl directed oncogenesis and other modes of transformation where mTOR, due to its central role in regulating cell growth and metabolism, is recruited as part of an oncogenic program. These validated hits represent a set of genes essential for executing critical functions downstream of mTOR and may be novel therapeutic targets for cancer.

[1]  K. Guan,et al.  mTOR as a central hub of nutrient signalling and cell growth , 2019, Nature Cell Biology.

[2]  J. Murray,et al.  Oncogenic Signalling through Mechanistic Target of Rapamycin (mTOR): A Driver of Metabolic Transformation and Cancer Progression , 2018, Cancers.

[3]  HIF prolyl hydroxylase PHD3 regulates translational machinery and glucose metabolism in clear cell renal cell carcinoma , 2017, Cancer & metabolism.

[4]  D. Sabatini,et al.  mTOR Signaling in Growth, Metabolism, and Disease , 2017, Cell.

[5]  M. Colombi,et al.  An Isogenic Cell Panel Identifies Compounds That Inhibit Proliferation of mTOR-Pathway Addicted Cells by Different Mechanisms , 2014, Journal of biomolecular screening.

[6]  M. Climent,et al.  Prospective study assessing hypoxia-related proteins as markers for the outcome of treatment with sunitinib in advanced clear-cell renal cell carcinoma. , 2013, Annals of oncology : official journal of the European Society for Medical Oncology.

[7]  Qi-Lian Liang,et al.  Function and expression of prolyl hydroxylase 3 in cancers , 2013, Archives of medical science : AMS.

[8]  M. Hall,et al.  HIF-independent role of prolyl hydroxylases in the cellular response to amino acids , 2012, Oncogene.

[9]  P. Wysocki mTOR inhibitors in renal cell carcinoma , 2011 .

[10]  D. Cho,et al.  mTOR inhibitors in renal cell carcinoma. , 2011, Therapy.

[11]  C. Schaefer,et al.  Genome-wide shRNA screen reveals increased mitochondrial dependence upon mTORC2 addiction , 2011, Oncogene.

[12]  D. Sabatini,et al.  An ATP-competitive Mammalian Target of Rapamycin Inhibitor Reveals Rapamycin-resistant Functions of mTORC1* , 2009, Journal of Biological Chemistry.

[13]  Sang Gyun Kim,et al.  Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation , 2008, Proceedings of the National Academy of Sciences.

[14]  M. Colombi,et al.  Isolation and characterization of dominant and recessive IL-3-independent hematopoietic transformants , 2006, Oncogene.

[15]  Razelle Kurzrock,et al.  Philadelphia ChromosomePositive Leukemias: From Basic Mechanisms to Molecular Therapeutics , 2003, Annals of Internal Medicine.

[16]  C. Moroni,et al.  Reversible abrogation of IL‐3 dependence by an inducible H‐ras oncogene. , 1989, The EMBO journal.

[17]  S. Collins,et al.  Replacement of serum by insulin and transferrin supports growth and differentiation of the human promyelocytic cell line, HL-60. , 1980, Experimental cell research.