Nature Communications (Feb 2024)

Targeting DNA2 overcomes metabolic reprogramming in multiple myeloma

  • Natthakan Thongon,
  • Feiyang Ma,
  • Natalia Baran,
  • Pamela Lockyer,
  • Jintan Liu,
  • Christopher Jackson,
  • Ashley Rose,
  • Ken Furudate,
  • Bethany Wildeman,
  • Matteo Marchesini,
  • Valentina Marchica,
  • Paola Storti,
  • Giannalisa Todaro,
  • Irene Ganan-Gomez,
  • Vera Adema,
  • Juan Jose Rodriguez-Sevilla,
  • Yun Qing,
  • Min Jin Ha,
  • Rodrigo Fonseca,
  • Caleb Stein,
  • Caleb Class,
  • Lin Tan,
  • Sergio Attanasio,
  • Guillermo Garcia-Manero,
  • Nicola Giuliani,
  • David Berrios Nolasco,
  • Andrea Santoni,
  • Claudio Cerchione,
  • Carlos Bueso-Ramos,
  • Marina Konopleva,
  • Philip Lorenzi,
  • Koichi Takahashi,
  • Elisabet Manasanch,
  • Gabriella Sammarelli,
  • Rashmi Kanagal-Shamanna,
  • Andrea Viale,
  • Marta Chesi,
  • Simona Colla

DOI
https://doi.org/10.1038/s41467-024-45350-8
Journal volume & issue
Vol. 15, no. 1
pp. 1 – 13

Abstract

Read online

Abstract DNA damage resistance is a major barrier to effective DNA-damaging therapy in multiple myeloma (MM). To discover mechanisms through which MM cells overcome DNA damage, we investigate how MM cells become resistant to antisense oligonucleotide (ASO) therapy targeting Interleukin enhancer binding factor 2 (ILF2), a DNA damage regulator that is overexpressed in 70% of MM patients whose disease has progressed after standard therapies have failed. Here, we show that MM cells undergo adaptive metabolic rewiring to restore energy balance and promote survival in response to DNA damage activation. Using a CRISPR/Cas9 screening strategy, we identify the mitochondrial DNA repair protein DNA2, whose loss of function suppresses MM cells’ ability to overcome ILF2 ASO−induced DNA damage, as being essential to counteracting oxidative DNA damage. Our study reveals a mechanism of vulnerability of MM cells that have an increased demand for mitochondrial metabolism upon DNA damage activation.