Abstract

BackgroundLactobacillus reuteri strains are widely used as probiotics to prevent and treat inflammatory bowel disease by modulating the host’s immune system. However, the underlying mechanisms by which they communicate with the host have not been clearly understood. Bacterial extracellular vesicles (EVs) have been considered as important mediators of host-pathogen interactions, but their potential role in commensals-host crosstalk has not been widely studied. Here, we investigated the regulatory actions of EVs produced by L. reuteri BBC3, a gut-associated commensal bacterium of Black-Bone chicken, in the development of lipopolysaccharide (LPS)-induced intestinal inflammation in a chicken model using both in vivo and in vitro experiments.ResultsL. reuteri BBC3 produced nano-scale membrane vesicles with the size range of 60–250 nm. Biochemical and proteomic analyses showed that L. reuteri BBC3-derived EVs (LrEVs) carried DNA, RNA and several bioactive proteins previously described as mediators of other probiotics’ beneficial effects such as glucosyltransferase, serine protease and elongation factor Tu. In vivo broiler experiments showed that administration of LrEVs exerted similar effects as L. reuteri BBC3 in attenuating LPS-induced inflammation by improving growth performance, reducing mortality and decreasing intestinal injury. LrEVs suppressed the LPS-induced expression of pro-inflammatory genes (TNF-α, IL-1β, IL-6, IL-17 and IL-8), and improved the expression of anti-inflammatory genes (IL-10 and TGF-β) in the jejunum. LrEVs could be internalized by chicken macrophages. In vitro pretreatment with LrEVs reduced the gene expression of TNF-α, IL-1β and IL-6 by suppressing the NF-κB activity, and enhanced the gene expression of IL-10 and TGF-β in LPS-activated chicken macrophages. Additionally, LrEVs could inhibit Th1- and Th17-mediated inflammatory responses and enhance the immunoregulatory cells-mediated immunosuppression in splenic lymphocytes of LPS-challenged chickens through the activation of macrophages. Finally, we revealed that the reduced content of both vesicular proteins and nucleic acids attenuated the suppression of LrEVs on LPS-induced inflammatory responses in ex vivo experiments, suggesting that they are essential for the LrEVs-mediated immunoregulation.ConclusionsWe revealed that LrEVs participated in maintaining intestinal immune homeostasis against LPS-induced inflammatory responses in a chicken model. Our findings provide mechanistic insight into how commensal and probiotic Lactobacillus species modulate the host’s immune system in pathogens-induced inflammation.

Highlights

  • Lactobacillus reuteri strains are widely used as probiotics to prevent and treat inflammatory bowel disease by modulating the host’s immune system

  • We revealed that L. reuteri BBC3-derived EVs (LrEVs) participated in maintaining intestinal immune homeostasis against LPSinduced inflammatory responses in a chicken model

  • outer membrane vesicles (OMVs) can activate the innate and adaptive immune responses and induce the protective immunity similar to that induced by the intact bacteria in vivo, suggesting that these OMVs can be used for the development of novel vaccine candidates and adjuvants [5, 6]

Read more

Summary

Introduction

Lactobacillus reuteri strains are widely used as probiotics to prevent and treat inflammatory bowel disease by modulating the host’s immune system. Membrane vesicles (MVs) have been recognized as a form of cell-cell communication used by almost all domains of life: bacteria, archaea and eukaryotes [1]. Over the years, they have been largely ignored in the field of microbiology. Bacterial EVs contain various bioactive molecules of the parental bacteria, including proteins, lipids, nucleic acids and polysaccharides, which are involved in a large number of pathological and physiological functions in intercellular interactions such as nutrient acquisition, biofilm formation, stress response, delivery of toxins and virulence factors and invasion of host and immune regulation [2, 3]. OMVs can activate the innate and adaptive immune responses and induce the protective immunity similar to that induced by the intact bacteria in vivo, suggesting that these OMVs can be used for the development of novel vaccine candidates and adjuvants [5, 6]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call