[HTML][HTML] Casein kinase 1α–dependent feedback loop controls autophagy in RAS-driven cancers

JK Cheong, F Zhang, PJ Chua, BH Bay… - The Journal of …, 2015 - Am Soc Clin Investig
JK Cheong, F Zhang, PJ Chua, BH Bay, A Thorburn, DM Virshup
The Journal of clinical investigation, 2015Am Soc Clin Investig
Activating mutations in the RAS oncogene are common in cancer but are difficult to
therapeutically target. RAS activation promotes autophagy, a highly regulated catabolic
process that metabolically buffers cells in response to diverse stresses. Here we report that
casein kinase 1α (CK1α), a ubiquitously expressed serine/threonine kinase, is a key
negative regulator of oncogenic RAS–induced autophagy. Depletion or pharmacologic
inhibition of CK1α enhanced autophagic flux in oncogenic RAS–driven human fibroblasts …
Activating mutations in the RAS oncogene are common in cancer but are difficult to therapeutically target. RAS activation promotes autophagy, a highly regulated catabolic process that metabolically buffers cells in response to diverse stresses. Here we report that casein kinase 1α (CK1α), a ubiquitously expressed serine/threonine kinase, is a key negative regulator of oncogenic RAS–induced autophagy. Depletion or pharmacologic inhibition of CK1α enhanced autophagic flux in oncogenic RAS–driven human fibroblasts and multiple cancer cell lines. FOXO3A, a master longevity mediator that transcriptionally regulates diverse autophagy genes, was a critical target of CK1α, as depletion of CK1α reduced levels of phosphorylated FOXO3A and increased expression of FOXO3A-responsive genes. Oncogenic RAS increased CK1α protein abundance via activation of the PI3K/AKT/mTOR pathway. In turn, elevated levels of CK1α increased phosphorylation of nuclear FOXO3A, thereby inhibiting transactivation of genes critical for RAS-induced autophagy. In both RAS-driven cancer cells and murine xenograft models, pharmacologic CK1α inactivation synergized with lysosomotropic agents to inhibit growth and promote tumor cell death. Together, our results identify a kinase feedback loop that influences RAS-dependent autophagy and suggest that targeting CK1α-regulated autophagy offers a potential therapeutic opportunity to treat oncogenic RAS–driven cancers.
The Journal of Clinical Investigation