Hunk negatively regulates c-myc to promote Akt-mediated cell survival and mammary tumorigenesis induced by loss of Pten

ES Yeh, GK Belka, AE Vernon… - Proceedings of the …, 2013 - National Acad Sciences
ES Yeh, GK Belka, AE Vernon, CC Chen, JJ Jung, LA Chodosh
Proceedings of the National Academy of Sciences, 2013National Acad Sciences
The protooncogenes Akt and c-myc each positively regulate cell growth and proliferation,
but have opposing effects on cell survival. These oncogenes cooperate to promote
tumorigenesis, in part because the prosurvival effects of Akt offset the proapoptotic effects of
c-myc. Akt's ability to counterbalance c-myc's proapoptotic effects has primarily been
attributed to Akt-induced stimulation of prosurvival pathways that indirectly antagonize the
effects of c-myc. We report a more direct mechanism by which Akt modulates the …
The protooncogenes Akt and c-myc each positively regulate cell growth and proliferation, but have opposing effects on cell survival. These oncogenes cooperate to promote tumorigenesis, in part because the prosurvival effects of Akt offset the proapoptotic effects of c-myc. Akt’s ability to counterbalance c-myc’s proapoptotic effects has primarily been attributed to Akt-induced stimulation of prosurvival pathways that indirectly antagonize the effects of c-myc. We report a more direct mechanism by which Akt modulates the proapoptotic effects of c-myc. Specifically, we demonstrate that Akt up-regulates the adenosine monophosphate-associated kinase (AMPK)-related protein kinase, Hormonally up-regulated neu-associated kinase (Hunk), which serves as an effector of Akt prosurvival signaling by suppressing c-myc expression in a kinase-dependent manner to levels that are compatible with cell survival. Consequently, Akt pathway activation in the mammary glands of Hunk−/− mice results in induction of c-myc expression to levels that induce apoptosis. c-myc knockdown rescues the increase in apoptosis induced by Hunk deletion in cells in which Akt has been activated, indicating that repression of c-myc is a principal mechanism by which Hunk mediates the prosurvival effects of Akt. Consistent with this mechanism of action, we find that Hunk is required for c-myc suppression and mammary tumorigenesis induced by phosphatase and tensin homolog (Pten) deletion in mice. Together, our findings establish a prosurvival function for Hunk in tumorigenesis, define an essential mechanism by which Akt suppresses c-myc–induced apoptosis, and identify Hunk as a previously unrecognized link between the Akt and c-myc oncogenic pathways.
National Acad Sciences