eLife (Jun 2023)

Defining function of wild-type and three patient-specific TP53 mutations in a zebrafish model of embryonal rhabdomyosarcoma

  • Jiangfei Chen,
  • Kunal Baxi,
  • Amanda E Lipsitt,
  • Nicole Rae Hensch,
  • Long Wang,
  • Prethish Sreenivas,
  • Paulomi Modi,
  • Xiang Ru Zhao,
  • Antoine Baudin,
  • Daniel G Robledo,
  • Abhik Bandyopadhyay,
  • Aaron Sugalski,
  • Anil K Challa,
  • Dias Kurmashev,
  • Andrea R Gilbert,
  • Gail E Tomlinson,
  • Peter Houghton,
  • Yidong Chen,
  • Madeline N Hayes,
  • Eleanor Y Chen,
  • David S Libich,
  • Myron S Ignatius

DOI
https://doi.org/10.7554/eLife.68221
Journal volume & issue
Vol. 12

Abstract

Read online

In embryonal rhabdomyosarcoma (ERMS) and generally in sarcomas, the role of wild-type and loss- or gain-of-function TP53 mutations remains largely undefined. Eliminating mutant or restoring wild-type p53 is challenging; nevertheless, understanding p53 variant effects on tumorigenesis remains central to realizing better treatment outcomes. In ERMS, >70% of patients retain wild-type TP53, yet mutations when present are associated with worse prognosis. Employing a kRASG12D-driven ERMS tumor model and tp53 null (tp53-/-) zebrafish, we define wild-type and patient-specific TP53 mutant effects on tumorigenesis. We demonstrate that tp53 is a major suppressor of tumorigenesis, where tp53 loss expands tumor initiation from <35% to >97% of animals. Characterizing three patient-specific alleles reveals that TP53C176F partially retains wild-type p53 apoptotic activity that can be exploited, whereas TP53P153Δ and TP53Y220C encode two structurally related proteins with gain-of-function effects that predispose to head musculature ERMS. TP53P153Δ unexpectedly also predisposes to hedgehog-expressing medulloblastomas in the kRASG12D-driven ERMS-model.

Keywords