Phosphorylation of MLL by ATR is required for execution of mammalian S-phase checkpoint

H Liu, S Takeda, R Kumar, TD Westergard, EJ Brown… - Nature, 2010 - nature.com
H Liu, S Takeda, R Kumar, TD Westergard, EJ Brown, TK Pandita, EHY Cheng, JJD Hsieh
Nature, 2010nature.com
Cell cycle checkpoints are implemented to safeguard the genome, avoiding the
accumulation of genetic errors,. Checkpoint loss results in genomic instability and
contributes to the evolution of cancer. Among G1-, S-, G2-and M-phase checkpoints, genetic
studies indicate the role of an intact S-phase checkpoint in maintaining genome integrity,.
Although the basic framework of the S-phase checkpoint in multicellular organisms has
been outlined, the mechanistic details remain to be elucidated. Human chromosome-11 …
Abstract
Cell cycle checkpoints are implemented to safeguard the genome, avoiding the accumulation of genetic errors,. Checkpoint loss results in genomic instability and contributes to the evolution of cancer. Among G1-, S-, G2- and M-phase checkpoints, genetic studies indicate the role of an intact S-phase checkpoint in maintaining genome integrity,. Although the basic framework of the S-phase checkpoint in multicellular organisms has been outlined, the mechanistic details remain to be elucidated. Human chromosome-11 band-q23 translocations disrupting the MLL gene lead to poor prognostic leukaemias,,,,. Here we assign MLL as a novel effector in the mammalian S-phase checkpoint network and identify checkpoint dysfunction as an underlying mechanism of MLL leukaemias. MLL is phosphorylated at serine 516 by ATR in response to genotoxic stress in the S phase, which disrupts its interaction with, and hence its degradation by, the SCFSkp2 E3 ligase, leading to its accumulation. Stabilized MLL protein accumulates on chromatin, methylates histone H3 lysine 4 at late replication origins and inhibits the loading of CDC45 to delay DNA replication. Cells deficient in MLL showed radioresistant DNA synthesis and chromatid-type genomic abnormalities, indicative of S-phase checkpoint dysfunction. Reconstitution of Mll−/− (Mll also known as Mll1) mouse embryonic fibroblasts with wild-type but not S516A or ΔSET mutant MLL rescues the S-phase checkpoint defects. Moreover, murine myeloid progenitor cells carrying an Mll–CBP knock-in allele that mimics human t(11;16) leukaemia show a severe radioresistant DNA synthesis phenotype. MLL fusions function as dominant negative mutants that abrogate the ATR-mediated phosphorylation/stabilization of wild-type MLL on damage to DNA, and thus compromise the S-phase checkpoint. Together, our results identify MLL as a key constituent of the mammalian DNA damage response pathway and show that deregulation of the S-phase checkpoint incurred by MLL translocations probably contributes to the pathogenesis of human MLL leukaemias.
nature.com