Using co-immunoprecipitation (Co-IP) coupled mass spectrometry (MS), we found that the NHEJ-related protein Ku80 directly interacts with EZH2 and regulates its methyltransferase activity

Using co-immunoprecipitation (Co-IP) coupled mass spectrometry (MS), we found that the NHEJ-related protein Ku80 directly interacts with EZH2 and regulates its methyltransferase activity. suggesting DNA-PK-mediated EZH2 phosphorylation impairs EZH2 histone methyltransferase activity. On the other hand, EZH2 inhibition increased the DNA damage level at the late phase of T-cell activation, suggesting EZH2 involved in genomic integrity maintenance. In conclusion, our study is the first to demonstrate that EZH2 is phosphorylated by the DNA damage responsive complex DNA-PK and regulates DNA damage-mediated T-cell apoptosis, which reveals a novel functional crosstalk between epigenetic regulation and genomic integrity. The elimination of expanded T cells and the regulation of T-cell apoptosis in the late phase of the immune response are crucial for maintaining immune homeostasis.1 In recent years, OGT2115 an understanding of how the DNA damage response contributes to the regulation of T-cell fate in the immune response has emerged. In response to DNA damage occurring during the inflammatory response, OGT2115 cells initiate DNA repair pathways that are required for host cell survival. If the damage is too severe, cell cycle arrest/apoptosis is initiated.2 Lymphocytes are particularly susceptible to DNA damage-induced apoptosis; it has been suggested that this sensitivity serves as a fail-safe mechanism to counter these cells’ intrinsic high potential for mutation and clonal expansion. However, the regulatory network of DNA damage-induced apoptosis is not yet completely understood. Polycomb repressive complex 2 (PRC2) mediates gene silencing by catalyzing the tri-methylation of lysine 27 on histone H3 (H3K27me3) within the gene promoter region. PRC2 controls normal stem cell differentiation and is associated with many malignant tumors.3 EZH2, the catalytic subunit of PRC2, is an essential epigenetic regulator of multiple cellular events. Interestingly, PRC2 components have recently been reported to be recruited to DNA damage sites, thus suggesting that EZH2 may be involved in DNA damage response mechanisms.4, 5, 6, 7 The roles of EZH2 in governing T-cell survival have been noted by several groups. EZH2 has been shown to have a non-redundant role in T helper (Th)-cell lineage survival, and EZH2 deficiency accelerates effector Th-cell death via death receptor-mediated extrinsic and intrinsic apoptotic pathways.8 We have also identified a defect in Bim expression that rescues EZH2-mediated cell death in a graft-versus-host disease mouse model, thus providing a different mechanism.9 Furthermore, a recent study has revealed a non-redundant and cell-intrinsic requirement for EZH2 in both regulatory T-cell differentiation and effector T-cell expansion.10 Given OGT2115 the diversity of mechanisms by which EZH2 regulates T-cell apoptosis, further exploration is needed. During DNA repair, a protein kinase, DNA-dependent protein kinase (DNA-PK), functions as a OGT2115 sensor of DNA double-strand breaks (DSBs) and is involved in the non-homologous end-joining (NHEJ) DNA repair pathway.11 Once DNA damage is present, the DNA-PK catalytic subunit (DNA-PKcs) is recruited to DNA Rabbit Polyclonal to DUSP6 lesion sites and promotes DNA repair by mediating the phosphorylation of downstream proteins.12, 13 The regulatory subunit of DNA-PK, Ku80, together with Ku70, functions as a bridge between the kinase and its substrates and mediates the phosphorylation of many proteins, such as p53, HSP90, TFIID, and c-Jun.12, 14, 15 Accumulating evidence indicates that the activity and stability of EZH2 are regulated by posttranslational modifications that are critical for the biological function of PRC2, especially phosphorylation.16 However, whether the exact mechanism and function of PRC2 at sites of DSBs correlate with the phosphorylase kinase DNA-PK is still unknown. We have previously shown that EZH2 has critical roles in regulating the T-cell response in several immune diseases.9, 17, 18 Given that EZH2’s function and target genes largely depend on its interacting proteins, we sought to reveal a new EZH2 regulatory pathway by identifying new EZH2-interacting proteins in T cells, in hopes of facilitating the OGT2115 development of new drug targets for treating immune diseases. We.