The gut microbiota mediates protective immunity against tuberculosis via modulation of lncRNA

Gut Microbes. 2022 Jan-Dec;14(1):2029997. doi: 10.1080/19490976.2022.2029997.

Abstract

The gut-lung axis has been implicated as a potential therapeutic target in lung disorders. While increasing evidence suggests that gut microbiota plays a critical role in regulating host immunity and contributing to tuberculosis (TB) development and progression, the underlying mechanisms whereby gut microbiota may impact TB outcomes are not fully understood. Here, we found that broad-spectrum antibiotics treatment increased susceptibility to Mycobacterium tuberculosis (M. tuberculosis) infection and modulated pulmonary inflammatory responses in mouse M. tuberculosis infection model. We then identified a commensal gut bacteria-regulated lncRNA, termed lncRNA-CGB, which was down-regulated by dysbiosis of gut microbiota during TB infection. Furthermore, we found that Bacteroides fragilis (B. fragilis) was a direct regulator of lncRNA-CGB, and oral administration of B. fragilis enhanced expression of lncRNA-CGB and promoted anti-TB immunity. Genomic knock-out of lncRNA-CGB led to reduced IFN-γ expression and impaired anti-TB immunity, therefore leading to detrimental effects on M. tuberculosis infection. Mechanistically, lncRNA-CGB interacted with EZH2 and negatively regulated H3K27 tri-methylation (H3K27Me3) epigenetic programming, leading to enhanced IFN-γ expression. Thus, this work not only uncovered previously unrecognized importance of gut bacteria-lncRNA-EZH2-H3K27Me3 axis in conferring immune protection against TB but also identified a potential new paradigm to develop a microbiota-based treatment against TB and potentially other diseases.

Keywords: Tuberculosis; commensal gut bacteria; gut-lung axis.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Dysbiosis / microbiology
  • Gastrointestinal Microbiome*
  • Mice
  • Mycobacterium tuberculosis* / genetics
  • RNA, Long Noncoding* / genetics
  • Tuberculosis* / drug therapy
  • Tuberculosis* / microbiology

Substances

  • RNA, Long Noncoding

Grants and funding

This work was supported by NSFC Grants 82072250 (To G.Z.), National Science and Technology Major Project “Construction of Demonstration Zone for Integrated Prevention and Control of Infectious Diseases” 2018ZX10715004-002-012 (To F.Y.), China Postdoctoral Science Foundation 2017M612811 (To F.Y.). National Key Research and Development Plan 2020YFA0907200 and 2019YFC0840602 (To G.L.Z.), the Guangdong Foundation for Basic and Applied Basic Research 2019B1515120041 (To G.L.Z.), the Guangdong Scientific and Technological Foundation 2020B1111170014 (To G.L.Z.), and the Shenzhen Scientific and Technological Foundation KCXFZ202002011007083 (To G.L.Z.). Shanghai Science and Technology Committee Basic Research Grant 20JC1417800 (To H.S.).