WDR5 supports an N-Myc transcriptional complex that drives a protumorigenic gene expression signature in neuroblastoma

Y Sun, JL Bell, D Carter, S Gherardi, RC Poulos… - Cancer research, 2015 - AACR
Y Sun, JL Bell, D Carter, S Gherardi, RC Poulos, G Milazzo, JWH Wong, R Al-Awar, AE Tee…
Cancer research, 2015AACR
MYCN gene amplification in neuroblastoma drives a gene expression program that
correlates strongly with aggressive disease. Mechanistically, trimethylation of histone H3
lysine 4 (H3K4) at target gene promoters is a strict prerequisite for this transcriptional
program to be enacted. WDR5 is a histone H3K4 presenter that has been found to have an
essential role in H3K4 trimethylation. For this reason, in this study, we investigated the
relationship between WDR5-mediated H3K4 trimethylation and N-Myc transcriptional …
Abstract
MYCN gene amplification in neuroblastoma drives a gene expression program that correlates strongly with aggressive disease. Mechanistically, trimethylation of histone H3 lysine 4 (H3K4) at target gene promoters is a strict prerequisite for this transcriptional program to be enacted. WDR5 is a histone H3K4 presenter that has been found to have an essential role in H3K4 trimethylation. For this reason, in this study, we investigated the relationship between WDR5-mediated H3K4 trimethylation and N-Myc transcriptional programs in neuroblastoma cells. N-Myc upregulated WDR5 expression in neuroblastoma cells. Gene expression analysis revealed that WDR5 target genes included those with MYC-binding elements at promoters such as MDM2. We showed that WDR5 could form a protein complex at the MDM2 promoter with N-Myc, but not p53, leading to histone H3K4 trimethylation and activation of MDM2 transcription. RNAi-mediated attenuation of WDR5 upregulated expression of wild-type but not mutant p53, an effect associated with growth inhibition and apoptosis. Similarly, a small-molecule antagonist of WDR5 reduced N-Myc/WDR5 complex formation, N-Myc target gene expression, and cell growth in neuroblastoma cells. In MYCN-transgenic mice, WDR5 was overexpressed in precancerous ganglion and neuroblastoma cells compared with normal ganglion cells. Clinically, elevated levels of WDR5 in neuroblastoma specimens were an independent predictor of poor overall survival. Overall, our results identify WDR5 as a key cofactor for N-Myc–regulated transcriptional activation and tumorigenesis and as a novel therapeutic target for MYCN-amplified neuroblastomas. Cancer Res; 75(23); 5143–54. ©2015 AACR.
AACR