Histone H2A variants alpha1-extension helix directs RNF168-mediated ubiquitination

Nat Commun. 2020 May 18;11(1):2462. doi: 10.1038/s41467-020-16307-4.

Abstract

Histone ubiquitination plays an important role in the DNA damage response (DDR) pathway. RNF168 catalyzes H2A and H2AX ubiquitination on lysine 13/15 (K13/K15) upon DNA damage and promotes the accrual of downstream repair factors at damaged chromatin. Here, we report that RNF168 ubiquitinates the non-canonical H2A variants H2AZ and macroH2A1/2 at the divergent N-terminal tail lysine residue. In addition to their evolutionarily conserved nucleosome acidic patch, we identify the positively charged alpha1-extension helix as essential for RNF168-mediated ubiquitination of H2A variants. Moreover, mutation of the RNF168 UMI (UIM- and MIU-related UBD) hydrophilic acidic residues abolishes RNF168-mediated ubiquitination as well as 53BP1 and BRCA1 ionizing radiation-induced foci formation. Our results reveal a juxtaposed bipartite electrostatic interaction utilized by the nucleosome to direct RNF168 orientation towards the target lysine residues in proximity to the H2A alpha1-extension helix, which plays an important role in the DDR pathway.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Amino Acid Sequence
  • Evolution, Molecular
  • HEK293 Cells
  • Histones / chemistry*
  • Histones / genetics
  • Histones / metabolism*
  • Humans
  • Lysine / metabolism
  • Protein Structure, Secondary
  • Substrate Specificity
  • Tumor Suppressor p53-Binding Protein 1 / metabolism
  • Ubiquitin-Protein Ligases / metabolism*
  • Ubiquitination*

Substances

  • Histones
  • TP53BP1 protein, human
  • Tumor Suppressor p53-Binding Protein 1
  • macroH2A histone
  • RNF168 protein, human
  • Ubiquitin-Protein Ligases
  • Lysine