Cell Death Discovery (Aug 2024)

HDAC3 genetic and pharmacologic inhibition radiosensitizes fusion positive rhabdomyosarcoma by promoting DNA double-strand breaks

  • Matteo Cassandri,
  • Antonella Porrazzo,
  • Silvia Pomella,
  • Beatrice Noce,
  • Clemens Zwergel,
  • Francesca Antonella Aiello,
  • Francesca Vulcano,
  • Luisa Milazzo,
  • Simona Camero,
  • Deborah Pajalunga,
  • Massimo Spada,
  • Valeria Manzi,
  • Giovanni Luca Gravina,
  • Silvia Codenotti,
  • Michela Piccione,
  • Miriam Tomaciello,
  • Michele Signore,
  • Giovanni Barillari,
  • Cinzia Marchese,
  • Alessandro Fanzani,
  • Biagio De Angelis,
  • Concetta Quintarelli,
  • Christopher R. Vakoc,
  • Eleanor Y. Chen,
  • Francesca Megiorni,
  • Franco Locatelli,
  • Sergio Valente,
  • Antonello Mai,
  • Rossella Rota,
  • Francesco Marampon

DOI
https://doi.org/10.1038/s41420-024-02115-y
Journal volume & issue
Vol. 10, no. 1
pp. 1 – 20

Abstract

Read online

Abstract Radiotherapy (RT) plays a critical role in the management of rhabdomyosarcoma (RMS), the prevalent soft tissue sarcoma in childhood. The high risk PAX3-FOXO1 fusion-positive subtype (FP-RMS) is often resistant to RT. We have recently demonstrated that inhibition of class-I histone deacetylases (HDACs) radiosensitizes FP-RMS both in vitro and in vivo. However, HDAC inhibitors exhibited limited success on solid tumors in human clinical trials, at least in part due to the presence of off-target effects. Hence, identifying specific HDAC isoforms that can be targeted to radiosensitize FP-RMS is imperative. We, here, found that only HDAC3 silencing, among all class-I HDACs screened by siRNA, radiosensitizes FP-RMS cells by inhibiting colony formation. Thus, we dissected the effects of HDAC3 depletion using CRISPR/Cas9-dependent HDAC3 knock-out (KO) in FP-RMS cells, which resulted in Endoplasmatic Reticulum Stress activation, ERK inactivation, PARP1- and caspase-dependent apoptosis and reduced stemness when combined with irradiation compared to single treatments. HDAC3 loss-of-function increased DNA damage in irradiated cells augmenting H2AX phosphorylation and DNA double-strand breaks (DSBs) and counteracting irradiation-dependent activation of ATM and DNA-Pkcs as well as Rad51 protein induction. Moreover, HDAC3 depletion hampers FP-RMS tumor growth in vivo and maximally inhibits the growth of irradiated tumors compared to single approaches. We, then, developed a new HDAC3 inhibitor, MC4448, which showed specific cell anti-tumor effects and mirrors the radiosensitizing effects of HDAC3 depletion in vitro synergizing with ERKs inhibition. Overall, our findings dissect the pro-survival role of HDAC3 in FP-RMS and suggest HDAC3 genetic or pharmacologic inhibition as a new promising strategy to overcome radioresistance in this tumor.