Interplay of m6A and H3K27 trimethylation restrains inflammation during bacterial infection

Sci Adv. 2020 Aug 19;6(34):eaba0647. doi: 10.1126/sciadv.aba0647. eCollection 2020 Aug.

Abstract

While N 6-methyladenosine (m6A) is the most prevalent modification of eukaryotic messenger RNA (mRNA) involved in various cellular responses, its role in modulating bacteria-induced inflammatory response remains elusive. Here, we showed that loss of the m6A reader YTH-domain family 2 (YTHDF2) promoted demethylation of histone H3 lysine-27 trimethylation (H3K27me3), which led to enhanced production of proinflammatory cytokines and facilitated the deposition of m6A cotranscriptionally. Mechanistically, the mRNA of lysine demethylase 6B (KDM6B) was m6A-modified and its decay mediated by YTHDF2. YTHDF2 deficiency stabilized KDM6B to promote H3K27me3 demethylation of multiple proinflammatory cytokines and subsequently enhanced their transcription. Furthermore, we identified H3K27me3 as a barrier for m6A modification during transcription. KDM6B recruits the m6A methyltransferase complex to facilitate the methylation of m6A in transcribing mRNA by removing adjacent H3K27me3 barriers. These results revealed cross-talk between m6A and H3K27me3 during bacterial infection, which has broader implications for deciphering epitranscriptomics in immune homeostasis.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Bacterial Infections* / genetics
  • Cytokines
  • Histones* / genetics
  • Humans
  • Inflammation / genetics
  • Jumonji Domain-Containing Histone Demethylases / genetics
  • Lysine
  • RNA, Messenger / genetics

Substances

  • Cytokines
  • Histones
  • RNA, Messenger
  • Jumonji Domain-Containing Histone Demethylases
  • KDM6B protein, human
  • Lysine