PUBLICATION

Swimming motility of a gut bacterial symbiont promotes resistance to intestinal expulsion and enhances inflammation

Authors
Wiles, T.J., Schlomann, B.H., Wall, E.S., Betancourt, R., Parthasarathy, R., Guillemin, K.
ID
ZDB-PUB-200403-137
Date
2020
Source
PLoS Biology   18: e3000661 (Journal)
Registered Authors
Guillemin, Karen
Keywords
none
MeSH Terms
  • Animals
  • Animals, Genetically Modified
  • Chemotaxis/genetics
  • Chemotaxis/physiology
  • Gastrointestinal Microbiome/physiology*
  • Gastrointestinal Motility/physiology*
  • Inflammation
  • Intestines/microbiology
  • Intestines/pathology*
  • Locomotion/genetics
  • Locomotion/physiology*
  • Macrophages/metabolism
  • Microbial Interactions
  • Mutation
  • Tumor Necrosis Factor-alpha/metabolism
  • Vibrio/genetics
  • Vibrio/physiology
  • Zebrafish/microbiology
  • Zebrafish/physiology
PubMed
32196484 Full text @ PLoS Biol.
Abstract
Some of the densest microbial ecosystems in nature thrive within the intestines of humans and other animals. To protect mucosal tissues and maintain immune tolerance, animal hosts actively sequester bacteria within the intestinal lumen. In response, numerous bacterial pathogens and pathobionts have evolved strategies to subvert spatial restrictions, thereby undermining immune homeostasis. However, in many cases, it is unclear how escaping host spatial control benefits gut bacteria and how changes in intestinal biogeography are connected to inflammation. A better understanding of these processes could uncover new targets for treating microbiome-mediated inflammatory diseases. To this end, we investigated the spatial organization and dynamics of bacterial populations within the intestine using larval zebrafish and live imaging. We discovered that a proinflammatory Vibrio symbiont native to zebrafish governs its own spatial organization using swimming motility and chemotaxis. Surprisingly, we found that Vibrio's motile behavior does not enhance its growth rate but rather promotes its persistence by enabling it to counter intestinal flow. In contrast, Vibrio mutants lacking motility traits surrender to host spatial control, becoming aggregated and entrapped within the lumen. Consequently, nonmotile and nonchemotactic mutants are susceptible to intestinal expulsion and experience large fluctuations in absolute abundance. Further, we found that motile Vibrio cells induce expression of the proinflammatory cytokine tumor necrosis factor alpha (TNFα) in gut-associated macrophages and the liver. Using inducible genetic switches, we demonstrate that swimming motility can be manipulated in situ to modulate the spatial organization, persistence, and inflammatory activity of gut bacterial populations. Together, our findings suggest that host spatial control over resident microbiota plays a broader role in regulating the abundance and persistence of gut bacteria than simply protecting mucosal tissues. Moreover, we show that intestinal flow and bacterial motility are potential targets for therapeutically managing bacterial spatial organization and inflammatory activity within the gut.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping