Human Gut Symbiont Roseburia hominis Promotes and Regulates Innate Immunity

Angela M Patterson, Imke E Mulder, Anthony J Travis, Annaig Lan, Nadine Cerf-Bensussan, Valerie Gaboriau-Routhiau, Karen Garden, Elizabeth Logan, Margaret I Delday, Alistair G P Coutts, Edouard Monnais, Vanessa C Ferraria, Ryo Inoue, George Grant, Rustam I Aminov

Research output: Contribution to journalArticlepeer-review

110 Citations (Scopus)
10 Downloads (Pure)

Abstract

OBJECTIVE: Roseburia hominis is a flagellated gut anaerobic bacterium belonging to the Lachnospiraceae family within the Firmicutes phylum. A significant decrease of R. hominis colonization in the gut of ulcerative colitis patients has recently been demonstrated. In this work, we have investigated the mechanisms of R. hominis-host cross talk using both murine and in vitro models.

DESIGN: The complete genome sequence of R. hominis A2-183 was determined. C3H/HeN germ-free mice were mono-colonized with R. hominis, and the host-microbe interaction was studied using histology, transcriptome analyses and FACS. Further investigations were performed in vitro and using the TLR5KO and DSS-colitis murine models.

RESULTS: In the bacterium, R. hominis, host gut colonization upregulated genes involved in conjugation/mobilization, metabolism, motility, and chemotaxis. In the host cells, bacterial colonization upregulated genes related to antimicrobial peptides, gut barrier function, toll-like receptors (TLR) signaling, and T cell biology. CD4(+)CD25(+)FoxP3(+) T cell numbers increased in the lamina propria of both mono-associated and conventional mice treated with R. hominis. Treatment with the R. hominis bacterium provided protection against DSS-induced colitis. The role of flagellin in host-bacterium interaction was also investigated.

CONCLUSION: Mono-association of mice with R. hominis bacteria results in specific bidirectional gene expression patterns. A set of genes thought to be important for host colonization are induced in R. hominis, while the host cells respond by strengthening gut barrier function and enhancing Treg population expansion, possibly via TLR5-flagellin signaling. Our data reveal the immunomodulatory properties of R. hominis that could be useful for the control and treatment of gut inflammation.

Original languageEnglish
Article number1166
Pages (from-to)1-14
Number of pages14
JournalFrontiers in Immunology
Volume8
DOIs
Publication statusPublished - 26 Sept 2017

Bibliographical note

Acknowledgments
The authors would like to thank Andrew Gewirtz (Georgia State University, Atlanta, GA, USA) and Adam Cunningham (University of Birmingham, Edgbaston, Birmingham, UK) for generously providing the germ-free and conventional TLR5KO mice, respectively. The authors also wish to acknowledge a significant contribution to the inception of the work, experimental work, interpretation of results, and writing of the manuscript by Denise Kelly, as well as the support from RESAS (Rural and Environmental Science and Analytical Services) of the Scottish Government. EM and VF were supported by the Marie Curie Initial training network Fellowships funded by the EU (grant #215532). The authors acknowledge a generous support of Prof. Harry Flint and Dr. Sylvia Duncan in the microbiological part of this work. The skilled technical support of Gillian Campbell and Pauline Young at RINH Genomics is also gratefully acknowledged.

Keywords

  • Journal Article
  • Roseburia
  • T lymphocytes
  • immune tolerance
  • inflammatory bowel disease
  • flagellin
  • TLR5

Fingerprint

Dive into the research topics of 'Human Gut Symbiont Roseburia hominis Promotes and Regulates Innate Immunity'. Together they form a unique fingerprint.

Cite this