Oncotarget

Research Papers:

RALB provides critical survival signals downstream of Ras in acute myeloid leukemia

Craig E. Eckfeldt _, Emily J. Pomeroy, Robin D.W. Lee, Katherine S. Hazen, Lindsey A. Lee, Branden S. Moriarity and David A. Largaespada

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:65147-65156. https://doi.org/10.18632/oncotarget.11431

Metrics: PDF 1894 views  |   HTML 2346 views  |   ?  


Abstract

Craig E. Eckfeldt1,2, Emily J. Pomeroy1, Robin D.W. Lee1, Katherine S. Hazen1, Lindsey A. Lee1, Branden S. Moriarity2,3, David A. Largaespada2,3,4

1Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA

2Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA

3Department of Pediatrics, Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN 55455, USA

4Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA

Correspondence to:

Craig E. Eckfeldt, email: [email protected]

Keywords: RAS signaling, RALB, TBK1, BCL2, acute myeloid leukemia (AML)

Received: April 20, 2016     Accepted: August 11, 2016     Published: August 20, 2016

ABSTRACT

Mutations that activate RAS proto-oncogenes and their effectors are common in acute myeloid leukemia (AML); however, efforts to therapeutically target Ras or its effectors have been unsuccessful, and have been hampered by an incomplete understanding of which effectors are required for AML proliferation and survival. We investigated the role of Ras effector pathways in AML using murine and human AML models. Whereas genetic disruption of NRAS(V12) expression in an NRAS(V12) and Mll-AF9-driven murine AML induced apoptosis of leukemic cells, inhibition of phosphatidylinositol-3-kinase (PI3K) and/or mitogen-activated protein kinase (MAPK) signaling did not reproduce this effect. Conversely, genetic disruption of RALB signaling induced AML cell death and phenocopied the effects of suppressing oncogenic Ras directly – uncovering a novel role for RALB signaling in AML survival. Knockdown of RALB led to decreased phosphorylation of TBK1 and reduced BCL2 expression, providing mechanistic insight into RALB survival signaling in AML. Notably, we found that patient-derived AML blasts have higher levels of RALB-TBK1 signaling compared to normal blood leukocytes, supporting a pathophysiologic role for RALB signaling for AML patients. Overall, our work provides new insight into the specific roles of Ras effector pathways in AML and has identified RALB signaling as a key survival pathway.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 11431