International Journal of Molecular Sciences (Oct 2020)

Deferasirox-Dependent Iron Chelation Enhances Mitochondrial Dysfunction and Restores p53 Signaling by Stabilization of p53 Family Members in Leukemic Cells

  • Chiara Calabrese,
  • Cristina Panuzzo,
  • Serena Stanga,
  • Giacomo Andreani,
  • Silvia Ravera,
  • Alessandro Maglione,
  • Lucrezia Pironi,
  • Jessica Petiti,
  • Muhammad Shahzad Ali,
  • Patrizia Scaravaglio,
  • Francesca Napoli,
  • Carmen Fava,
  • Marco De Gobbi,
  • Francesco Frassoni,
  • Giuseppe Saglio,
  • Enrico Bracco,
  • Barbara Pergolizzi,
  • Daniela Cilloni

DOI
https://doi.org/10.3390/ijms21207674
Journal volume & issue
Vol. 21, no. 20
p. 7674

Abstract

Read online

Iron is crucial to satisfy several mitochondrial functions including energy metabolism and oxidative phosphorylation. Patients affected by Myelodysplastic Syndromes (MDS) and acute myeloid leukemia (AML) are frequently characterized by iron overload (IOL), due to continuous red blood cell (RBC) transfusions. This event impacts the overall survival (OS) and it is associated with increased mortality in lower-risk MDS patients. Accordingly, the oral iron chelator Deferasirox (DFX) has been reported to improve the OS and delay leukemic transformation. However, the molecular players and the biological mechanisms laying behind remain currently mostly undefined. The aim of this study has been to investigate the potential anti-leukemic effect of DFX, by functionally and molecularly analyzing its effects in three different leukemia cell lines, harboring or not p53 mutations, and in human primary cells derived from 15 MDS/AML patients. Our findings indicated that DFX can lead to apoptosis, impairment of cell growth only in a context of IOL, and can induce a significant alteration of mitochondria network, with a sharp reduction in mitochondrial activity. Moreover, through a remarkable reduction of Murine Double Minute 2 (MDM2), known to regulate the stability of p53 and p73 proteins, we observed an enhancement of p53 transcriptional activity after DFX. Interestingly, this iron depletion-triggered signaling is enabled by p73, in the absence of p53, or in the presence of a p53 mutant form. In conclusion, we propose a mechanism by which the increased p53 family transcriptional activity and protein stability could explain the potential benefits of iron chelation therapy in terms of improving OS and delaying leukemic transformation.

Keywords