Loss of phosphatase CTDNEP1 potentiates aggressive medulloblastoma by triggering MYC amplification and genomic instability

MYC-driven medulloblastomas are highly aggressive childhood brain tumors, however, the molecular and genetic events triggering MYC amplification and malignant transformation remain elusive. Here we report that mutations in CTDNEP1, a CTD nuclear-envelope-phosphatase, are the most significantly enric...

Full description

Saved in:
Bibliographic Details
Published inNature communications Vol. 14; no. 1; p. 762
Main Authors Luo, Zaili, Xin, Dazhuan, Liao, Yunfei, Berry, Kalen, Ogurek, Sean, Zhang, Feng, Zhang, Liguo, Zhao, Chuntao, Rao, Rohit, Dong, Xinran, Li, Hao, Yu, Jianzhong, Lin, Yifeng, Huang, Guoying, Xu, Lingli, Xin, Mei, Nishinakamura, Ryuichi, Yu, Jiyang, Kool, Marcel, Pfister, Stefan M, Roussel, Martine F, Zhou, Wenhao, Weiss, William A, Andreassen, Paul, Lu, Q Richard
Format Journal Article
LanguageEnglish
Published England Nature Publishing Group 10.02.2023
Nature Publishing Group UK
Nature Portfolio
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:MYC-driven medulloblastomas are highly aggressive childhood brain tumors, however, the molecular and genetic events triggering MYC amplification and malignant transformation remain elusive. Here we report that mutations in CTDNEP1, a CTD nuclear-envelope-phosphatase, are the most significantly enriched recurrent alterations in MYC-driven medulloblastomas, and define high-risk subsets with poorer prognosis. Ctdnep1 ablation promotes the transformation of murine cerebellar progenitors into Myc-amplified medulloblastomas, resembling their human counterparts. CTDNEP1 deficiency stabilizes and activates MYC activity by elevating MYC serine-62 phosphorylation, and triggers chromosomal instability to induce p53 loss and Myc amplifications. Further, phosphoproteomics reveals that CTDNEP1 post-translationally modulates the activities of key regulators for chromosome segregation and mitotic checkpoint regulators including topoisomerase TOP2A and checkpoint kinase CHEK1. Co-targeting MYC and CHEK1 activities synergistically inhibits CTDNEP1-deficient MYC-amplified tumor growth and prolongs animal survival. Together, our studies demonstrate that CTDNEP1 is a tumor suppressor in highly aggressive MYC-driven medulloblastomas by controlling MYC activity and mitotic fidelity, pointing to a CTDNEP1-dependent targetable therapeutic vulnerability.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:2041-1723
2041-1723
DOI:10.1038/s41467-023-36400-8