Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

The intestinal regionalization of acute norovirus infection is regulated by the microbiota via bile acid-mediated priming of type III interferon

Abstract

Evidence has accumulated to demonstrate that the intestinal microbiota enhances mammalian enteric virus infections1. For example, we and others previously reported that commensal bacteria stimulate acute and persistent murine norovirus infections2,3,4. However, in apparent contradiction of these results, the virulence of murine norovirus infection was unaffected by antibiotic treatment. This prompted us to perform a detailed investigation of murine norovirus infection in microbially deplete mice, revealing a more complex picture in which commensal bacteria inhibit viral infection of the proximal small intestine while simultaneously stimulating the infection of distal regions of the gut. Thus, commensal bacteria can regulate viral regionalization along the intestinal tract. We further show that the mechanism underlying bacteria-dependent inhibition of norovirus infection in the proximal gut involves bile acid priming of type III interferon. Finally, the regional effects of the microbiota on norovirus infection may result from distinct regional expression profiles of key bile acid receptors that regulate the type III interferon response. Overall, these findings reveal that the biotransformation of host metabolites by the intestinal microbiota directly and regionally impacts infection by a pathogenic enteric virus.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The intestinal microbiota plays opposing roles in the regulation of proximal gut versus distal gut MNV infection.
Fig. 2: Bacteria-dependent type III IFN signalling in the proximal gut protects intestinal immune cells from MNV-1 infection.
Fig. 3: Bile acids biotransformed by the intestinal microbiota prime type III IFN induction to suppress MNV infection of the proximal gut.
Fig. 4: Bile acids prime IFN-λ induction in vitro.

Similar content being viewed by others

Data availability

All of the data that support the findings of this study are available from the corresponding authors on request. The source data for bile acid analyses performed in this study are included in Supplementary Table 2.

References

  1. Karst, S. M. The influence of commensal bacteria on infection with enteric viruses. Nat. Rev. Microbiol. 14, 197–204 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Pfeiffer, J. K. & Virgin, H. W. Transkingdom control of viral infection and immunity in the mammalian intestine. Science 351, aad5872 (2016).

    Article  PubMed  CAS  Google Scholar 

  3. Kuss, S. K. et al. Intestinal microbiota promote enteric virus replication and systemic pathogenesis. Science 334, 249–252 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kane, M. et al. Successful transmission of a retrovirus depends on the commensal microbiota. Science 334, 245–249 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kernbauer, E., Ding, Y. & Cadwell, K. An enteric virus can replace the beneficial function of commensal bacteria. Nature 516, 94–98 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Jones, M. K. et al. Enteric bacteria promote human and murine norovirus infection of B cells. Science 346, 755–759 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Baldridge, M. T. et al. Commensal microbes and interferon-λ determine persistence of enteric murine norovirus infection. Science 347, 266–269 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Uchiyama, R., Chassaing, B., Zhang, B. & Gewirtz, A. T. Antibiotic treatment suppresses rotavirus infection and enhances specific humoral immunity. J. Infect. Dis. 210, 171–182 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Robinson, C. M., Jesudhasan, P. R. & Pfeiffer, J. K. Bacterial lipopolysaccharide binding enhances virion stability and promotes environmental fitness of an enteric virus. Cell Host Microbe 15, 36–46 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wilks, J. et al. Mammalian lipopolysaccharide receptors incorporated into the retroviral envelope augment virus transmission. Cell Host Microbe 18, 456–462 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sansone, C. L. et al. Microbiota-dependent priming of antiviral intestinal immunity in Drosophila. Cell Host Microbe 18, 571–581 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ramirez, J. L. et al. Reciprocal tripartite interactions between the Aedes aegypti midgut microbiota, innate immune system and dengue virus influences vector competence. PLoS Negl. Trop. Dis. 6, e1561 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Wu, P. et al. A gut commensal bacterium promotes mosquito permissiveness to arboviruses. Cell Host Microbe 25, 101–112 (2019).

    Article  CAS  PubMed  Google Scholar 

  14. Ichinohe, T. et al. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc. Natl Acad. Sci. USA 108, 5354–5359 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Abt, M. C. et al. Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity 37, 158–170 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. McFarlane, A. J. et al. Enteric helminth-induced type I interferon signaling protects against pulmonary virus infection through interaction with the microbiota. J. Allergy Clin. Immunol. 140, 1068–1078 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Thackray, L. B. et al. Oral antibiotic treatment of mice exacerbates the disease severity of multiple flavivirus infections. Cell Rep. 22, 3440–3453 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Steed, A. L. et al. The microbial metabolite desaminotyrosine protects from influenza through type I interferon. Science 357, 498–502 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhu, S. et al. Regulation of norovirus virulence by the VP1 protruding domain correlates with B cell infection efficiency. J. Virol. 90, 2858–2867 (2015).

    Article  PubMed  CAS  Google Scholar 

  20. Zhu, S. et al. Identification of immune and viral correlates of norovirus protective immunity through comparative study of intra-cluster norovirus strains. PLoS Pathog. 9, e1003592 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Thackray, L. B. et al. Murine noroviruses comprising a single genogroup exhibit biological diversity despite limited sequence divergence. J. Virol. 81, 10460–10473 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hsu, C. C., Riley, L. K., Wills, H. M. & Livingston, R. S. Persistent infection with and serologic crossreactivity of three novel murine noroviruses. Comp. Med. 56, 247–251 (2006).

    CAS  PubMed  Google Scholar 

  23. Arias, A., Bailey, D., Chaudhry, Y. & Goodfellow, I. G. Development of a reverse genetics system for murine norovirus 3; long-term persistence occurs in the caecum and colon. J. Gen. Virol. 93, 1432–1441 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Grau, K. R. et al. The major targets of acute norovirus infection are immune cells in the gut-associated lymphoid tissue. Nat. Microbiol. 2, 1586–1591 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sommereyns, C., Paul, S., Staeheli, P. & Michiels, T. IFN-lambda (IFN-λ) is expressed in a tissue-dependent fashion and primarily acts on epithelial cells in vivo. PLoS Pathog. 4, e1000017 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Wilen, C. B. et al. Tropism for tuft cells determines immune promotion of norovirus pathogenesis. Science 360, 204–208 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lazear, H. M., Nice, T. J. & Diamond, M. S. Interferon-λ: immune functions at barrier surfaces and beyond. Immunity 43, 15–28 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Thomas, C., Pellicciari, R., Pruzanski, M., Auwerx, J. & Schoonjans, K. Targeting bile-acid signalling for metabolic diseases. Nat. Rev. Drug Discov. 7, 678–693 (2008).

    Article  CAS  PubMed  Google Scholar 

  29. Joyce, S. A. & Gahan, C. G. M. The gut microbiota and the metabolic health of the host. Curr. Opin. Gastroenterol. 30, 120–127 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Fiorucci, S. & Distrutti, E. Bile acid-activated receptors, intestinal microbiota, and the treatment of metabolic disorders. Trends Mol. Med. 21, 702–714 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Vavassori, P., Mencarelli, A., Renga, B., Distrutti, E. & Fiorucci, S. The bile acid receptor FXR is a modulator of intestinal innate immunity. J. Immunol. 183, 6251–6261 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Schupp, A.-K. et al. Bile acids act as soluble host restriction factors limiting cytomegalovirus replication in hepatocytes. J. Virol. 90, 6686–6698 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kim, Y. & Chang, K.-O. Inhibitory effects of bile acids and synthetic farnesoid X receptor agonists on rotavirus replication. J. Virol. 85, 12570–12577 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chang, K.-O. et al. Bile acids are essential for porcine enteric calicivirus replication in association with down-regulation of signal transducer and activator of transcription 1. Proc. Natl Acad. Sci. USA 101, 8733–8738 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ettayebi, K. et al. Replication of human noroviruses in stem cell–derived human enteroids. Science 353, 1387–1393 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Buffie, C. G. et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature 517, 205–208 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Sun, X. et al. Microbiota-derived metabolic factors reduce campylobacteriosis in mice. Gastroenterology 154, 1751–1763 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Wells, J. E. & Hylemon, P. B. Identification and characterization of a bile acid 7α-dehydroxylation operon in Clostridium sp. strain TO-931, a highly active 7α-dehydroxylating strain isolated from human feces. Appl. Environ. Microbiol. 66, 1107–1113 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nelson, C. A. et al. Structural basis for murine norovirus engagement of bile acids and the CD300lf receptor. Proc. Natl Acad. Sci. USA 115, E9201–E9210 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Lee, S. et al. Norovirus cell tropism is determined by combinatorial action of a viral non-structural protein and host cytokine. Cell Host Microbe 22, 449–459 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhang, Y., Kast-Woelbern, H. R. & Edwards, P. A. Natural structural variants of the nuclear receptor farnesoid X receptor affect transcriptional activation. J. Biol. Chem. 278, 104–110 (2003).

    Article  CAS  PubMed  Google Scholar 

  42. Bookout, A. L. et al. Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network. Cell 126, 789–799 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Higashiyama, H., Kinoshita, M. & Asano, S. Immunolocalization of farnesoid X receptor (FXR) in mouse tissues using tissue microarray. Acta Histochem. 110, 86–93 (2008).

    Article  PubMed  Google Scholar 

  44. Inagaki, T. et al. Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proc. Natl Acad. Sci. USA 103, 3920–3925 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chang, K.-O. & George, D. W. Bile acids promote the expression of hepatitis C virus in replicon-harboring cells. J. Virol. 81, 9633–9640 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Xiong, Q. et al. Metabolite-sensing G protein coupled receptor TGR5 protects host from viral infection through amplifying type I interferon responses. Front. Immunol. 9, 2289 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Mowat, A. M. & Agace, W. W. Regional specialization within the intestinal immune system. Nat. Rev. Immunol. 14, 667–685 (2014).

    Article  CAS  PubMed  Google Scholar 

  48. Padilla-Nash, H. M. et al. Spontaneous transformation of murine epithelial cells requires the early acquisition of specific chromosomal aneuploidies and genomic imbalances. Genes Chromosomes Cancer 51, 353–374 (2012).

    Article  CAS  PubMed  Google Scholar 

  49. González-Hernández, M. B., Perry, J. W. & Wobus, C. E. Neutral red assay for murine norovirus replication and detection in a mouse. Bio Protoc. 3, e415 (2013).

    Article  PubMed  Google Scholar 

  50. Kahan, S. M. et al. Comparative murine norovirus studies reveal a lack of correlation between intestinal virus titers and enteric pathology. Virology 421, 202–210 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Gonzalez-Hernandez, M. B. et al. Efficient norovirus and reovirus replication in the mouse intestine requires microfold (M) cells. J. Virol. 88, 6934–6943 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Bagyánszki, M. et al. Chronic alcohol consumption affects gastrointestinal motility and reduces the proportion of neuronal NOS‐immunoreactive myenteric neurons in the murine jejunum. Anat. Rec. 293, 1536–1542 (2010).

    Article  Google Scholar 

Download references

Acknowledgements

We acknowledge the Washington University Genome Engineering and iPSC Center, M. White and D. Kreamalmeyer. S.M.K. was funded by grant nos. NIH R01AI116892, NIH R01AI081921 and NIH R01AI141478. M.T.B. was supported by grant nos. NIH R01AI141478, NIH K22 AI127846-01, DDRCC P30 DK052574 and the Global Probiotics Council’s Young Investigator Grant for Probiotics Research. C.E.W. was funded in part by NIH R21 AI103961 and the University of Michigan Host–Microbiome Initiative. E.W.H. and M.P. were supported by grant no. T90DE021990. H.T. was supported by grant no. T32DK094775. C.B.W. was supported by grant no. NIH K08 AI28043 and the Burroughs Wellcome Fund.

Author information

Authors and Affiliations

Authors

Contributions

K.R.G., S.Z., S.T.P., E.W.H., D.P., M.P., A.H., H.T., P.F., V.R.G. and M.T.B. performed the experiments. K.R.G., S.Z., E.W.H., D.P., M.P., H.T., C.B.W., C.E.W., M.T.B. and S.M.K. analysed the results. M.T.B. and S.M.K. designed the project. K.R.G., M.T.B. and S.M.K. wrote the manuscript. All of the authors read and edited the manuscript.

Corresponding authors

Correspondence to Megan T. Baldridge or Stephanie M. Karst.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Cumulative intestinal titers.

Intestinal titers from SI-1, SI-2, SI-3 cecum and colon were combined for each mouse, and data for all mice per group averaged. Data for individual intestinal regions for Ifnar-/- mice are shown in Fig. 1c, for B6 mice are shown in Fig. 1d, and for germ-free mice are shown in Fig. 1e.

Extended Data Fig. 2 Tuft cells are not required for MNV-1 infection.

Tuft cell-deficient Pou2f3-/- or wild-type littermates (n = 9 mice per group) were challenged with 106 plaque-forming units (PFU) MNV-1 and viral genomes were quantified at 7 dpi. Pou2f3-/- (KO) and WT mice had statistically similar viral genomes in ileum, colon. MLNs, and spleen. Data is pooled from three independent experiments. Error bars indicate the mean of all data points. LOD, limit of detection.

Extended Data Fig. 3 Measurements of individual bile acids.

Lumenal contents were collected from SI-1 (a) and SI-3 (b) of groups of B6 mice (n = 5) treated with PBS, Abx, clindamycin, or nalidixic acid. The indicated bile acids were measured at the University of Pennsylvania Microbial Culture and Metabolomics Core using a Waters Acquity vPLC System. Source data are provided in Supplementary Table 2.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Grau, K.R., Zhu, S., Peterson, S.T. et al. The intestinal regionalization of acute norovirus infection is regulated by the microbiota via bile acid-mediated priming of type III interferon. Nat Microbiol 5, 84–92 (2020). https://doi.org/10.1038/s41564-019-0602-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-019-0602-7

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing