American Association for Cancer Research
Browse
00085472can150879-sup-147559_3_supp_0_s46pvb.docx (53.64 kB)

Supplementary Methods from MYC-Driven Neuroblastomas Are Addicted to a Telomerase-Independent Function of Dyskerin

Download (53.64 kB)
journal contribution
posted on 2023-03-30, 23:53 authored by Rosemary O'Brien, Sieu L. Tran, Michelle F. Maritz, Bing Liu, Cheng Fei Kong, Stefania Purgato, Chen Yang, Jayne Murray, Amanda J. Russell, Claudia L. Flemming, Georg von Jonquieres, Hilda A. Pickett, Wendy B. London, Michelle Haber, Preethi H. Gunaratne, Murray D. Norris, Giovanni Perini, Jamie I. Fletcher, Karen L. MacKenzie

Text file containing detailed methods.

Funding

Cancer Council New South Wales

National Health and Medical Research Council

Cancer Institute New South Wales

Italian Association for Cancer Research (AIRC)

History

ARTICLE ABSTRACT

The RNA-binding protein dyskerin, encoded by the DKC1 gene, functions as a core component of the telomerase holoenzyme as well as ribonuclear protein complexes involved in RNA processing and ribosome biogenesis. The diverse roles of dyskerin across many facets of RNA biology implicate its potential contribution to malignancy. In this study, we examined the expression and function of dyskerin in neuroblastoma. We show that DKC1 mRNA levels were elevated relative to normal cells across a panel of 15 neuroblastoma cell lines, where both N-Myc and c-Myc directly targeted the DKC1 promoter. Upregulation of MYCN was shown to dramatically increase DKC1 expression. In two independent neuroblastoma patient cohorts, high DKC1 expression correlated strongly with poor event-free and overall survival (P < 0.0001), independently of established prognostic factors. RNAi-mediated depletion of dyskerin inhibited neuroblastoma cell proliferation, including cells immortalized via the telomerase-independent ALT mechanism. Furthermore, dyskerin attenuation impaired anchorage-independent proliferation and tumor growth. Overexpression of the telomerase RNA component, hTR, demonstrated that this proliferative impairment was not a consequence of telomerase suppression. Instead, ribosomal stress, evidenced by depletion of small nucleolar RNAs and nuclear dispersal of ribosomal proteins, was the likely cause of the proliferative impairment in dyskerin-depleted cells. Accordingly, dyskerin suppression caused p53-dependent G1 cell-cycle arrest in p53 wild-type cells, and a p53-independent pathway impaired proliferation in cells with p53 dysfunction. Together, our findings highlight dyskerin as a new therapeutic target in neuroblastoma with crucial telomerase-independent functions and broader implications for the spectrum of malignancies driven by MYC family oncogenes. Cancer Res; 76(12); 3604–17. ©2016 AACR.