The aberrant activation of oncogene MYC (also termed c-MYC) is one of the most common features in human cancer. The unleashed MYC oncogene frequently produces abundant MYC protein, which mediates a transcriptional response involved in a variety of biological processes, contributing to almost every aspect of tumorigenesis. The significance of MYC deregulation has been recognized in T-cell acute lymphoblastic leukemia (T-ALL), a life-threatening hematological malignancy with dismal outcome due to disease relapse and drug resistance. Therapeutic efforts aimed at inhibiting MYC expression or activity should have an important clinical relevance. However, attempts to directly disrupt MYC function have met with limited success, in part due to its "undruggable" protein structure.

Aurora kinases, a multi-genic family of serine/threonine kinases, consist Aurora A (AURKA), Aurora B (AURKB) and Aurora C (AURKC). They are known to play an integral role in the regulation of cell division and chromosomal segregation. Amplification or overexpression of Aurora kinases is frequently found in human cancers with clear evidence of oncogenic potential, implicating Aurora kinases as rational anti-tumor targets. AURKB is the catalytic subunit of the chromosomal passenger protein complex (CPPC) which regulates multiple facets of cell division. Overexpression of AURKB has been reported in a variety of cancers and predicts poor overall survival. AZD1152 is a highly potent and selective inhibitor of Aurora B; preclinical evidence of anti-tumor efficacy with AZD1152 has extended to the clinical setting with tolerable toxicity. Despite considerable study, it remains largely unclear how expression of AURKB is elevated and, in particular, how elevated levels of AURKB reprogram cells to promote the cancer progression.

We identified AURKB as a novel MYC binding partner using liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis. To assess the potential role of AURKB in regulating MYC, we inhibited AURKB in multiple T-ALL cell lines and found that MYC protein expression was diminished significantly. Notably, depletion of AURKB failed to downregulate MYC mRNA, suggesting a post-transcriptional regulation of the MYC protein. We then assessed MYC protein decay in the presence or absence of AURKB expression. Time course experiments revealed that knockdown of AURKB significantly shortened the half-life of endogenous MYC in T-ALL cells. Consistently, AURKB depletion resulted in significant downregulation of MYC-induced gene expression programs, corroborating an essential role of AURKB in sustaining MYC transcriptional activities.

Regulation of MYC degradation by ubiquitin-proteasome system is dependent on MYC phosphorylation. Initiation of MYC turnover requires GSK3b-mediated phosphorylation of threonine 58 (p-T58). Time-course analysis showed that treatment with AZD1152 in T-ALL cells resulted in a notable increase in MYC p-T58. We performed in vitro kinase assays and found that MYC is phosphorylated directly by AURKB. This phosphorylation counteracted GSK3b-directed Thr58 phosphorylation and subsequent FBW7-mediated proteasomal degradation and enhanced the MYC protein stability.

The AURKB mRNA and protein are generally more abundant in human T-ALL than normal thymocytes. We demonstrated that MYC, in concert with TAL1, directly binds to the AURKB promoter and activates its transcription, thus constituting a reciprocal regulation between MYC and AURKB.

In order to clarify the biological significance of the AURKB-MYC circuit, we co-injected the one-cell-stage zebrafish embryos with the rag2:EGFP-Myc construct alone or together with rag2:AURKB followed by fluorescent microscopy analysis. Our results demonstrate that AURKB phosphorylation of MYC is functionally important for T cell leukemogenesis in vivo. Moreover, inhibition of AURKB elicits dramatic MYC degradation in association with robust apoptosis in wild type-FBW7 T-ALL cells, suggesting that FBW7 mutational status may serve as a genetic predictor of sensitivity to AURKB inhibitors.

In this study, we unravel a non-mitotic role of AURKB in promoting MYC stabilization via direct MYC phosphorylation at an unconventional site. Our findings decipher a previously unsuspected mechanism involved in MYC deregulation and highlight disruption of the AURKB-MYC signaling circuit as a promising T-ALL therapeutic strategy.

Disclosures

No relevant conflicts of interest to declare.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution