Small RNA SmsR1 modulates acidogenicity and cariogenic virulence by affecting protein acetylation in Streptococcus mutans

PLoS Pathog. 2024 Apr 15;20(4):e1012147. doi: 10.1371/journal.ppat.1012147. eCollection 2024 Apr.

Abstract

Post-transcriptional regulation by small RNAs and post-translational modifications (PTM) such as lysine acetylation play fundamental roles in physiological circuits, offering rapid responses to environmental signals with low energy consumption. Yet, the interplay between these regulatory systems remains underexplored. Here, we unveil the cross-talk between sRNAs and lysine acetylation in Streptococcus mutans, a primary cariogenic pathogen known for its potent acidogenic virulence. Through systematic overexpression of sRNAs in S. mutans, we identified sRNA SmsR1 as a critical player in modulating acidogenicity, a key cariogenic virulence feature in S. mutans. Furthermore, combined with the analysis of predicted target mRNA and transcriptome results, potential target genes were identified and experimentally verified. A direct interaction between SmsR1 and 5'-UTR region of pdhC gene was determined by in vitro binding assays. Importantly, we found that overexpression of SmsR1 reduced the expression of pdhC mRNA and increased the intracellular concentration of acetyl-CoA, resulting in global changes in protein acetylation levels. This was verified by acetyl-proteomics in S. mutans, along with an increase in acetylation level and decreased activity of LDH. Our study unravels a novel regulatory paradigm where sRNA bridges post-transcriptional regulation with post-translational modification, underscoring bacterial adeptness in fine-tuning responses to environmental stress.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Acetylation
  • Animals
  • Bacterial Proteins* / genetics
  • Bacterial Proteins* / metabolism
  • Dental Caries / metabolism
  • Dental Caries / microbiology
  • Female
  • Gene Expression Regulation, Bacterial*
  • Protein Processing, Post-Translational*
  • RNA, Bacterial / genetics
  • RNA, Bacterial / metabolism
  • RNA, Small Untranslated / genetics
  • RNA, Small Untranslated / metabolism
  • Rats
  • Streptococcus mutans* / genetics
  • Streptococcus mutans* / metabolism
  • Streptococcus mutans* / pathogenicity
  • Virulence

Substances

  • Bacterial Proteins
  • RNA, Bacterial
  • RNA, Small Untranslated

Grants and funding

This work was supported by the National Natural Science Foundation of China (82170947 to J.Z., 32170046 to Y.L.), the Sichuan Science and Technology Program (2022YFH0048 to Y.L., 2023NSFSC1509 to Q.Z.), the Key Research and Development Projects of Science and Technology Department of Sichuan Province (23ZDYF2052 to J.Z.), the State Key Laboratory of Oral Diseases (SKLOD2022OF03 to Y.L.), the Fundamental Research Funds for the Central Universities, Research and Develop Program, West China Hospital of Stomatology Sichuan University (RD-02-202402 to Q.M.), and the Research Funding from West China School/Hospital of Stomatology Sichuan University (RCDWJS2023-10 to Q.M.). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.