Abstract

HIC1 (hypermethylated in cancer 1) is a tumor suppressor gene frequently epigenetically silenced in human cancers. HIC1 encodes a transcriptional repressor involved in the regulation of growth control and DNA damage response. We previously demonstrated that HIC1 can be either acetylated or SUMOylated on lysine 314. This deacetylation/SUMOylation switch is governed by an unusual complex made up of SIRT1 and HDAC4 which deacetylates and thereby favors SUMOylation of HIC1 by a mechanism not yet fully deciphered. This switch regulates the interaction of HIC1 with MTA1, a component of the NuRD complex and potentiates the repressor activity of HIC1. Here, we show that HIC1 silencing in human fibroblasts impacts the repair of DNA double-strand breaks whereas ectopic expression of wild-type HIC1, but not of nonsumoylatable mutants, leads to a reduced number of γH2AX foci induced by etoposide treatment. In this way, we demonstrate that DNA damage leads to (i) an enhanced HDAC4/Ubc9 interaction, (ii) the activation of SIRT1 by SUMOylation (Lys-734), and (iii) the SUMO-dependent recruitment of HDAC4 by SIRT1 which permits the deacetylation/SUMOylation switch of HIC1. Finally, we show that this increase of HIC1 SUMOylation favors the HIC1/MTA1 interaction, thus demonstrating that HIC1 regulates DNA repair in a SUMO-dependent way. Therefore, epigenetic HIC1 inactivation, which is an early step in tumorigenesis, could contribute to the accumulation of DNA mutations through impaired DNA repair and thus favor tumorigenesis.

Highlights

  • The tumor suppressor gene Hypermethylated in Cancer 1 (HIC1) encodes a transcriptional repressor SUMOylated at Lys-314

  • MEF2 transcription factors [11, 12], we previously showed that the deacetylation/SUMOylation switch of HIC1 is orchestrated by a complex containing two deacetylases belonging to different functional classes: SIRT1 that deacetylates HIC1 and HDAC4 that favors its SUMOylation, probably via an E3 ligase activity [7]

  • Our results show that DNA double-strand breaks (DSBs) induce the formation of a SIRT1-SUMO1/HDAC4/Ubc9 complex that spawns the deacetylation/SUMOylation switch of HIC1, reinforcing the hypothesis that HDAC4 can play the role of SUMO E3 ligase

Read more

Summary

Background

The tumor suppressor gene HIC1 (hypermethylated in cancer 1) encodes a transcriptional repressor SUMOylated at Lys-314. We show that HIC1 silencing in human fibroblasts impacts the repair of DNA double-strand breaks whereas ectopic expression of wild-type HIC1, but not of nonsumoylatable mutants, leads to a reduced number of ␥H2AX foci induced by etoposide treatment In this way, we demonstrate that DNA damage leads to (i) an enhanced HDAC4/Ubc interaction, (ii) the activation of SIRT1 by SUMOylation (Lys-734), and (iii) the SUMO-dependent recruitment of HDAC4 by SIRT1 which permits the deacetylation/SUMOylation switch of HIC1. Ectopic expression of wild-type HIC1 but not of nonSUMOylatable mutants leads to a reduced number of ␥H2AX foci supporting a role of HIC1 in the regulation of DNA repair in a SUMO-dependent manner In accordance with this latter observation, we demonstrate that etoposide, bleomycin, or UVinduced DNA double-strand breaks (DSBs) lead to an increase of HIC1 modification by SUMO2 in an ATM (ataxia telangectasia mutated)-dependent way. Our results show that DNA DSBs induce the formation of a SIRT1-SUMO1/HDAC4/Ubc complex that spawns the deacetylation/SUMOylation switch of HIC1, reinforcing the hypothesis that HDAC4 can play the role of SUMO E3 ligase

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call