Citation

  • Authors: Bonagas N. et al.
  • Year: 2022
  • Journal: Nat Cancer 3 156-172
  • Applications: in vitro / DNA, siRNA / INTERFERin, jetPEI
  • Cell type: U-2 OS
    Description: Human bone osteosarcoma
    Known as: U2OS

Method

Transient siRNA knockdown of MTHFD2 Depletion of endogenous MTHFD2 was achieved using three different siRNA oligonucleotides targeting MTHFD2 (Dharmacon). Nontargeting siRNA was used as a control (Dharmacon). Transfection was performed on log(phase) cells using 10 nM siRNA and INTERFERin transfection reagent (Polyplus-Transfection), following the manufacturer’s protocol. Briefly, cells were seeded 1 d before transfection and allowed to attach overnight. Control and targeting siRNAs were mixed with INTERFERin in serum-free medium, vortexed for 10 s and incubated at room temperature for 15 min to allow transfection complex formation. Cells were washed and changed to fresh complete medium before adding the transfection mix. The cells were incubated with the siRNAs at 37 °C with 5% CO2 in a humidified incubator until harvesting. Transfection was performed on log(phase) U2OS cells using 4 µg of plasmid DNA and jetPEI transfection reagent (Polyplus-Transfection), according to the manufacturer’s protocol. Successful and stable plasmid integration was maintained by culturing the cells in complete medium supplemented with 250 μg ml−1 of G418 disulfate (Sigma-Aldrich).

Abstract

The folate metabolism enzyme MTHFD2 (methylenetetrahydrofolate dehydrogenase/cyclohydrolase) is consistently overexpressed in cancer but its roles are not fully characterized, and current candidate inhibitors have limited potency for clinical development. In the present study, we demonstrate a role for MTHFD2 in DNA replication and genomic stability in cancer cells, and perform a drug screen to identify potent and selective nanomolar MTHFD2 inhibitors; protein cocrystal structures demonstrated binding to the active site of MTHFD2 and target engagement. MTHFD2 inhibitors reduced replication fork speed and induced replication stress followed by S-phase arrest and apoptosis of acute myeloid leukemia cells in vitro and in vivo, with a therapeutic window spanning four orders of magnitude compared with nontumorigenic cells. Mechanistically, MTHFD2 inhibitors prevented thymidine production leading to misincorporation of uracil into DNA and replication stress. Overall, these results demonstrate a functional link between MTHFD2-dependent cancer metabolism and replication stress that can be exploited therapeutically with this new class of inhibitors.

Go to