Molecular Oncology (May 2021)

Downregulation of Glutamine Synthetase, not glutaminolysis, is responsible for glutamine addiction in Notch1‐driven acute lymphoblastic leukemia

  • Tra Ly Nguyen,
  • Marie‐Julie Nokin,
  • Silvia Terés,
  • Mercedes Tomé,
  • Clément Bodineau,
  • Oriane Galmar,
  • Jean‐Max Pasquet,
  • Benoit Rousseau,
  • Sebastian vanLiempd,
  • Juan Manuel Falcon‐Perez,
  • Elodie Richard,
  • Elodie Muzotte,
  • Hamid‐Reza Rezvani,
  • Muriel Priault,
  • Marion Bouchecareilh,
  • Isabelle Redonnet‐Vernhet,
  • Julien Calvo,
  • Benjamin Uzan,
  • Françoise Pflumio,
  • Patricia Fuentes,
  • Maria L. Toribio,
  • Abdel‐Majid Khatib,
  • Pierre Soubeyran,
  • Piedad del Socorro Murdoch,
  • Raúl V. Durán

DOI
https://doi.org/10.1002/1878-0261.12877
Journal volume & issue
Vol. 15, no. 5
pp. 1412 – 1431

Abstract

Read online

The cellular receptor Notch1 is a central regulator of T‐cell development, and as a consequence, Notch1 pathway appears upregulated in > 65% of the cases of T‐cell acute lymphoblastic leukemia (T‐ALL). However, strategies targeting Notch1 signaling render only modest results in the clinic due to treatment resistance and severe side effects. While many investigations reported the different aspects of tumor cell growth and leukemia progression controlled by Notch1, less is known regarding the modifications of cellular metabolism induced by Notch1 upregulation in T‐ALL. Previously, glutaminolysis inhibition has been proposed to synergize with anti‐Notch therapies in T‐ALL models. In this work, we report that Notch1 upregulation in T‐ALL induced a change in the metabolism of the important amino acid glutamine, preventing glutamine synthesis through the downregulation of glutamine synthetase (GS). Downregulation of GS was responsible for glutamine addiction in Notch1‐driven T‐ALL both in vitro and in vivo. Our results also confirmed an increase in glutaminolysis mediated by Notch1. Increased glutaminolysis resulted in the activation of the mammalian target of rapamycin complex 1 (mTORC1) pathway, a central controller of cell growth. However, glutaminolysis did not play any role in Notch1‐induced glutamine addiction. Finally, the combined treatment targeting mTORC1 and limiting glutamine availability had a synergistic effect to induce apoptosis and to prevent Notch1‐driven leukemia progression. Our results placed glutamine limitation and mTORC1 inhibition as a potential therapy against Notch1‐driven leukemia.

Keywords