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

Wiles, Travis J. and Schlomann, Brandon H. and Wall, Elena S. and Betancourt, Reina and Parthasarathy, Raghuveer and Guillemin, Karen and Ayres, Janelle S. (2020) Swimming motility of a gut bacterial symbiont promotes resistance to intestinal expulsion and enhances inflammation. PLOS Biology, 18 (3). e3000661. ISSN 1545-7885

[thumbnail of file_id=10.1371%2Fjournal.pbio.3000661&type=printable] Text
file_id=10.1371%2Fjournal.pbio.3000661&type=printable - Published Version

Download (4MB)

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.

Item Type: Article
Subjects: Science Repository > Biological Science
Depositing User: Managing Editor
Date Deposited: 27 Jan 2023 05:10
Last Modified: 22 Jun 2024 07:56
URI: http://research.manuscritpub.com/id/eprint/1346

Actions (login required)

View Item
View Item