Lactobacillus reuteri attenuated allergic inflammation induced by HDM in the mouse and modulated gut microbes

PLoS One. 2020 Apr 21;15(4):e0231865. doi: 10.1371/journal.pone.0231865. eCollection 2020.

Abstract

Gut microbiome plays an essential role in asthma development, and probiotic-based manipulation of the gut microbiome has been proposed to prevent asthma. Although the preventive effect of Lactobacillus supplementation against allergies has been reported, the precise Lactobacillus species beneficial for effective prevention of asthma remain unidentified and the underlying mechanisms remain unclear. Therefore, we aimed to investigate the efficacy of oral administration of six Lactobacillus species and the mechanism underlying asthma prevention via gut microbiome modulation. We investigated the effects of oral administration of L. rhamnosus, L. fermentum, L. casei, L. gasseri, L. salivarius, and L. reuteri (five strains of each species) on asthma and gut microbiome of house dust mite (HDM)-treated murine models of asthma. Of these, L. reuteri administration was the most effective: it alleviated airway inflammation, decreased total IgE and HDM-IgG1, and reduced Th2-associated pro-inflammatory cytokines. Moreover, modulation of specific microbial genera by L. reuteri was more effective in asthma prevention than the modulation of the overall microbiota composition. Lactobacillus and Enterococcus were enriched after L. reuteri supplementation and were closely associated with total IgE and IL-13 production. Furthermore, L. reuteri specifically altered the gut microbial function toward butyrate generation. Thus, L. reuteri may reduce the risk of asthma development by modulating specific gut microbiota to improve the lung immune environment. Our study suggests a novel option for gut microbiome manipulation via L. reuteri supplementation for suppression of asthma and other allergic diseases.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Administration, Oral
  • Animals
  • Asthma / immunology
  • Asthma / pathology*
  • Butyrates / metabolism
  • Cecum / metabolism
  • Disease Models, Animal
  • Gastrointestinal Microbiome*
  • Immunoglobulin E / blood
  • Interleukin-13 / metabolism
  • Limosilactobacillus reuteri / physiology*
  • Lung / immunology
  • Lung / metabolism
  • Lung / pathology
  • Mice
  • Pyroglyphidae / immunology*
  • Th2 Cells / cytology
  • Th2 Cells / immunology
  • Th2 Cells / metabolism

Substances

  • Butyrates
  • Interleukin-13
  • Immunoglobulin E

Grants and funding

This work was supported by the National Natural Science Foundation of China (Grant No. 31820103010, 31771953, 31871774), the Fundamental Research Funds for the Central Universities (JUSRP51903B), the National first class discipline program of Food Science and Technology (JUFSTR20180102) The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.