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.

  • Article
  • Published:

Mycobiota and diet-derived fungal xenosiderophores promote Salmonella gastrointestinal colonization

Abstract

The fungal gut microbiota (mycobiota) has been implicated in diseases that disturb gut homeostasis, such as inflammatory bowel disease. However, little is known about functional relationships between bacteria and fungi in the gut during infectious colitis. Here we investigated the role of fungal metabolites during infection with the intestinal pathogen Salmonella enterica serovar Typhimurium, a major cause of gastroenteritis worldwide. We found that, in the gut lumen, both the mycobiota and fungi present in the diet can be a source of siderophores, small molecules that scavenge iron from the host. The ability to use fungal siderophores, such as ferrichrome and coprogen, conferred a competitive growth advantage to Salmonella strains expressing the fungal siderophore receptors FhuA or FhuE in vitro and in a mouse model. Our study highlights the role of inter-kingdom cross-feeding between fungi and Salmonella and elucidates an additional function of the gut mycobiota, revealing the importance of these understudied members of the gut ecosystem during bacterial infection.

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: Salmonella is able to utilize purified fungal siderophores.
Fig. 2: Intestinal fungi can be a source of fungal siderophores.
Fig. 3: Dietary changes profoundly alter gut mycobiome composition.
Fig. 4: Mouse and human diets can contain fungal siderophores that promote Salmonella growth.
Fig. 5: Fungal siderophores confer a competitive advantage to Salmonella in vivo.

Similar content being viewed by others

Data availability

Raw sequence reads have been deposited at NCBI Sequence Read Archive under project PRJNA769709, and the UNITE (v.8.2) database78 was used for all microbiome analysis. Data are additionally available at https://github.com/BehnsenLab/Siderophores. Data used to generate figures are available as supplementary tables and as source data files published alongside this manuscript. Source data are provided with this paper.

Code availability

Original code used has been deposited at https://github.com/BehnsenLab/Siderophores.

References

  1. Scallan, E. et al. Foodborne illness acquired in the United States—major pathogens. Emerg. Infect. Dis. 17, 7–15 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Hohmann, E. L. Nontyphoidal salmonellosis. Clin. Infect. Dis. 32, 263–269 (2001).

    Article  CAS  PubMed  Google Scholar 

  3. Parry, C. M. et al. A retrospective study of secondary bacteraemia in hospitalised adults with community acquired non-typhoidal Salmonella gastroenteritis. BMC Infect. Dis. 13, 107 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Ng, K. M. et al. Microbiota-liberated host sugars facilitate post-antibiotic expansion of enteric pathogens. Nature 502, 96–99 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Faber, F. et al. Respiration of microbiota-derived 1,2-propanediol drives salmonella expansion during colitis. PLoS Pathog. 13, e1006129 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Winter, S. E. et al. Gut inflammation provides a respiratory electron acceptor for Salmonella. Nature 467, 426–429 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Thiennimitr, P. et al. Intestinal inflammation allows Salmonella to use ethanolamine to compete with the microbiota. Proc. Natl Acad. Sci. USA 108, 17480–17485 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ott, S. J. et al. Fungi and inflammatory bowel diseases: alterations of composition and diversity. Scand. J. Gastroenterol. 43, 831–841 (2008).

    Article  CAS  PubMed  Google Scholar 

  9. Dollive, S. et al. Fungi of the murine gut: episodic variation and proliferation during antibiotic treatment. PLoS ONE 8, e71806 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Eriksson, M. et al. The C-type lectin receptor SIGNR3 binds to fungi present in commensal microbiota and influences immune regulation in experimental colitis. Front. Immunol. 4, 196 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Foster, M. L., Dowd, S. E., Stephenson, C., Steiner, J. M. & Suchodolski, J. S. Characterization of the fungal microbiome (mycobiome) in fecal samples from dogs. Vet. Med. Int. 2013, 658373 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Hoffmann, C. et al. Archaea and fungi of the human gut microbiome: correlations with diet and bacterial residents. PLoS ONE 8, e66019 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Frykman, P. K. et al. Characterization of bacterial and fungal microbiome in children with hirschsprung disease with and without a history of enterocolitis: a multicenter study. PLoS ONE 10, e0124172 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Donovan, P. D., Gonzalez, G., Higgins, D. G., Butler, G. & Ito, K. Identification of fungi in shotgun metagenomics datasets. PLoS ONE 13, e0192898 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Sokol, H. et al. Fungal microbiota dysbiosis in IBD. Gut 66, 1039–1048 (2017).

    Article  CAS  PubMed  Google Scholar 

  16. Wheeler, M. L. et al. Immunological consequences of intestinal fungal dysbiosis. Cell Host Microbe 19, 865–873 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Skalski, J. H. et al. Expansion of commensal fungus Wallemia mellicola in the gastrointestinal mycobiota enhances the severity of allergic airway disease in mice. PLoS Pathog. 14, e1007260 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Yang, A. M. et al. Intestinal fungi contribute to development of alcoholic liver disease. J. Clin. Investig. 127, 2829–2841 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Bacher, P. et al. Human anti-fungal Th17 immunity and pathology rely on cross-reactivity against Candida albicans. Cell 176, 1340–1355.e15 (2019).

    Article  CAS  PubMed  Google Scholar 

  20. Zhu, F. et al. Autoreactive T cells and chronic fungal infection drive esophageal carcinogenesis. Cell Host Microbe 21, 478–493.e7 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Aykut, B. et al. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature 574, 264–267 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhai, B. et al. High-resolution mycobiota analysis reveals dynamic intestinal translocation preceding invasive candidiasis. Nat. Med. 26, 59–64 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Rolling, T. et al. Haematopoietic cell transplantation outcomes are linked to intestinal mycobiota dynamics and an expansion of Candida parapsilosis complex species. Nat. Microbiol. 6, 1505–1515 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Deriu, E. et al. Probiotic bacteria reduce salmonella typhimurium intestinal colonization by competing for iron. Cell Host Microbe 14, 26–37 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Andrews, S. C., Robinson, A. K. & Rodríguez-Quiñones, F. Bacterial iron homeostasis. FEMS Microbiol. Rev. 27, 215–237 (2003).

    Article  CAS  PubMed  Google Scholar 

  26. Holden, V. I. & Bachman, M. A. Diverging roles of bacterial siderophores during infection. Metallomics 7, 986–995 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Flo, T. H. et al. Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron. Nature 432, 917–921 (2004).

    Article  CAS  PubMed  Google Scholar 

  28. Berger, T. et al. Lipocalin 2-deficient mice exhibit increased sensitivity to Escherichia coli infection but not to ischemia–reperfusion injury. Proc. Natl Acad. Sci. USA 103, 1834–1839 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Crosa, J. H. & Walsh, C. T. Genetics and assembly line enzymology of siderophore biosynthesis in bacteria. Microbiol. Mol. Biol. Rev. 66, 223–249 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bachman, M. A., Miller, V. L. & Weiser, J. N. Mucosal lipocalin 2 has pro-inflammatory and iron-sequestering effects in response to bacterial enterobactin. PLoS Pathog. 5, e1000622 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Raffatellu, M. et al. Lipocalin-2 resistance confers an advantage to Salmonella enterica serotype Typhimurium for growth and survival in the inflamed intestine. Cell Host Microbe 5, 476–486 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Holmes, M. A., Paulsene, W., Jide, X., Ratledge, C. & Strong, R. K. Siderocalin (Lcn 2) also binds carboxymycobactins, potentially defending against mycobacterial infections through iron sequestration. Structure 13, 29–41 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Hantke, K. Identification of an iron uptake system specific for coprogen and rhodotorulic acid in Escherichia coli K12. Mol. Genet. Genom. 191, 301–306 (1983).

    Article  CAS  Google Scholar 

  34. Rabsch, W. Characterization of the catecholate indicator strain S. typhimurium TA2700 as an ent fhuC double mutant. FEMS Microbiol. Lett. 163, 79–84 (1998).

    Article  CAS  PubMed  Google Scholar 

  35. Kingsley, R. A. et al. Ferrioxamine-mediated iron(III) utilization by Salmonella enterica. Appl. Environ. Microbiol. 65, 1610–1618 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Luckey, M., Pollack, J. R., Wayne, R., Ames, B. N. & Neilands, J. B. Iron uptake in Salmonella typhimurium: utilization of exogenous siderochromes as iron carriers. J. Bacteriol. 111, 731–738 (1972).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Heithoff, D. M., Conner, C. P. & Mahan, M. J. Dissecting the biology of a pathogen during infection. Trends Microbiol. 5, 509–513 (1997).

    Article  CAS  PubMed  Google Scholar 

  38. Rollenhagen, C. & Bumann, D. Salmonella enterica highly expressed genes are disease specific. Infect. Immun. 74, 1649–1660 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nuccio, S. P. & Bäumler, A. J. Comparative analysis of Salmonella genomes identifies a metabolic network for escalating growth in the inflamed gut. MBio 5, e00929–14 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  40. de Jong, H. K., Parry, C. M., van der Poll, T. & Wiersinga, W. J. Host–pathogen interaction in invasive salmonellosis. PLoS Pathog. 8, e1002933 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Galán, J. E. Typhoid toxin provides a window into typhoid fever and the biology of Salmonella typhi. Proc. Natl Acad. Sci. USA 113, 6338–6344 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Galet, J. et al. Pseudomonas fluorescens pirates both ferrioxamine and ferricoelichelin siderophores from Streptomyces ambofaciens. Appl. Environ. Microbiol. 81, 3132–3141 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Palyada, K., Threadgill, D. & Stintzi, A. Iron acquisition and regulation in Campylobacter jejuni. J. Bacteriol. 186, 4714–4729 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhu, W. et al. Xenosiderophore utilization promotes Bacteroides thetaiotaomicron resilience during colitis. Cell Host Microbe 27, 376–388.e8 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pierce, E. C. et al. Bacterial-fungal interactions revealed by genome-wide analysis of bacterial mutant fitness. Nat. Microbiol 6, 87–102 (2021).

    Article  CAS  PubMed  Google Scholar 

  46. Sassone-Corsi, M. et al. Microcins mediate competition among Enterobacteriaceae in the inflamed gut. Nature 540, 280–283 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Barthel, M. et al. Pretreatment of mice with streptomycin provides a Salmonella enterica serovar Typhimurium colitis model that allows analysis of both pathogen and host. Infect. Immun. 71, 2839–2858 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Behnsen, J. et al. The cytokine IL-22 promotes pathogen colonization by suppressing related commensal bacteria. Immunity 40, 262–273 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Charlang, G., Ng, B., Horowitz, N. H. & Horowitz, R. M. Cellular and extracellular siderophores of Aspergillus nidulans and Penicillium chrysogenum. Mol. Cell. Biol. 1, 94–100 (1981).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Haas, H., Eisendle, M. & Turgeon, B. G. Siderophores in fungal physiology and virulence. Annu. Rev. Phytopathol. 46, 149–187 (2008).

    Article  CAS  PubMed  Google Scholar 

  51. Osman, Y., Gebreil, A., Mowafy, A. M., Anan, T. I. & Hamed, S. M. Characterization of Aspergillus niger siderophore that mediates bioleaching of rare earth elements from phosphorites. World J. Microbiol. Biotechnol. 35, 93 (2019).

    Article  PubMed  Google Scholar 

  52. Santus, W., Devlin, J. R. & Behnsen, J. Crossing kingdoms: how the mycobiota and fungal-bacterial interactions impact host health and disease. Infect. Immun. 89, e00648-20 (2021).

  53. Kim, Y. & Mylonakis, E. Killing of Candida albicans filaments by Salmonella enterica serovar Typhimurium is mediated by sopB effectors, parts of a type III secretion system. Eukaryot. Cell 10, 782–790 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Nemeth, E. et al. IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J. Clin. Investig. 113, 1271–1276 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Institute of Medicine et al. Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc (National Academies Press, 2002).

  56. Scupham, A. J. et al. Abundant and diverse fungal microbiota in the murine intestine. Appl. Environ. Microbiol. 72, 793–801 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Mims, T. S. et al. The gut mycobiome of healthy mice is shaped by the environment and correlates with metabolic outcomes in response to diet. Commun. Biol. 4, 281 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. David, L. A. et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559–563 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Suhr, M. J. & Hallen-Adams, H. E. The human gut mycobiome: pitfalls and potentials—a mycologist’s perspective. Mycologia 107, 1057–1073 (2015).

    Article  CAS  PubMed  Google Scholar 

  60. Leonardi, I. et al. Mucosal fungi promote gut barrier function and social behavior via type 17 immunity. Cell 185, 831–846.e14 (2022).

    Article  CAS  PubMed  Google Scholar 

  61. Schütze, E. et al. Siderophore production by streptomycetes—stability and alteration of ferrihydroxamates in heavy metal-contaminated soil. Environ. Sci. Pollut. Res. 22, 19376–19383 (2015).

    Article  Google Scholar 

  62. Shields-Cutler, R. R. et al. Human metabolome-derived cofactors are required for the antibacterial activity of siderocalin in urine. J. Biol. Chem. 291, 25901–25910 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Watts, R. E. et al. Contribution of siderophore systems to growth and urinary tract colonization of asymptomatic bacteriuria Escherichia coli. Infect. Immun. 80, 333–344 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Oide, S., Berthiller, F., Wiesenberger, G., Adam, G. & Turgeon, B. G. Individual and combined roles of malonichrome, ferricrocin, and TAFC siderophores in Fusarium graminearum pathogenic and sexual development. Front. Microbiol. 5, 759 (2014).

    PubMed  Google Scholar 

  65. Indriati Hood, M. & Skaar, E. P. Nutritional immunity: transition metals at the pathogen–host interface. Nat. Rev. Microbiol. 10, 525–537 (2012).

    Article  PubMed  Google Scholar 

  66. Heymann, P., Ernst, J. F. & Winkelmann, G. A gene of the major facilitator superfamily encodes a transporter for enterobactin (Enb1p) in Saccharomyces cerevisiae. Biometals 13, 65–72 (2000).

    Article  CAS  PubMed  Google Scholar 

  67. Haas, H. et al. Characterization of the Aspergillus nidulans transporters for the siderophores enterobactin and triacetylfusarinine C. Biochem. J. 371, 505–513 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Yeung, F. et al. Altered immunity of laboratory mice in the natural environment is associated with fungal colonization. Cell Host Microbe 27, 809–822.e6 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Auchtung, T. A. et al. Investigating colonization of the healthy adult gastrointestinal tract by fungi. mSphere 3, e00092–18 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. European Food Safety Authority & European Centre for Disease Prevention and Control. The European Union one health 2020 zoonoses report. EFSA J. 19, e06406 (2021).

  71. Sarma-Rupavtarm, R. B., Ge, Z., Schauer, D. B., Fox, J. G. & Polz, M. F. Spatial distribution and stability of the eight microbial species of the altered schaedler flora in the mouse gastrointestinal tract. Appl. Environ. Microbiol. 70, 2791–2800 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Metzenberg, R. L. Vogel’s Medium N salts: avoiding the need for ammonium nitrate. Fungal Genet. Rep. 50, 14–14 (2003).

    Article  Google Scholar 

  73. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 10–12 (2011).

    Article  Google Scholar 

  74. Bolyen, E. et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 37, 852–857 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Callahan, B. J. et al. DADA2: high-resolution sample inference from Illumina amplicon data. Nat. Methods 13, 581–583 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Quast, C. et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 41, D590–D596 (2013).

    Article  CAS  PubMed  Google Scholar 

  77. Rivers, A. R., Weber, K. C., Gardner, T. G., Liu, S. & Armstrong, S. D. ITSxpress: software to rapidly trim internally transcribed spacer sequences with quality scores for marker gene analysis. F1000Res. 7, 1418 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Kõljalg, U. et al. The taxon hypothesis paradigm—on the unambiguous detection and communication of taxa. Microorganisms 8, 1910 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  79. R Core Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, 2020).

  80. Wickham, H. ggplot2: Elegant Graphics for Data Analysis (Springer International Publishing, 2016).

  81. Wickham, H., François, R., Henry, L. & Müller, K. dplyr: A Grammar of Data Manipulation (R Foundation for Statistical Computing, 2020).

  82. Paradis, E. & Schliep, K. ape 5.0: an environment for modern phylogenetics and evolutionary analyses in R. Bioinformatics 35, 526–528 (2019).

    Article  CAS  PubMed  Google Scholar 

  83. Neuwirth, E. RColorBrewer: ColorBrewer Palettes (R Foundation for Statistical Computing, 2014).

  84. Lahti, L. et al. Introduction to the Microbiome R Package http://bioconductor.statistik.tu-dortmund.de/packages/3.6/bioc/vignettes/microbiome/inst/doc/vignette.html (TU Dortmund, 2018).

  85. McMurdie, P. J. & Holmes, S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE 8, e61217 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Liu, C. M. et al. FungiQuant: a broad-coverage fungal quantitative real-time PCR assay. BMC Microbiol. 12, 255 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all the members of the Behnsen lab for technical assistance, helpful discussion and critical reading of the manuscript. D. Kiani, K. Perfecto, K. Jaswal and O. A. Todd assisted with the husbandry of germ-free mice. This work was supported by UIC Institutional Startup Funds to J.B. and grants from the National Institute of Health (R01DK093426 to D.M.U. and R01AI143641 to J.B.). Illustrations were created at Biorender.com.

Author information

Authors and Affiliations

Authors

Contributions

J.B. conceived and administrated the study and was the primary supervisor. W.S. additionally supervised parts of the study and was responsible for execution and analysis of the majority of the experimental work, including all mouse experiments, and most of the in vitro experiments. J.B., J.R.D., K.A.K., C.C.J. and J.T. performed select bacterial growth curves, and J.B., K.A.K., J.R.D. and J.T. generated select Salmonella strains. W.S., A.P.R., K.A.K., J.R.D. and J.B. were involved in the husbandry of germ-free mice. D.M.U. was responsible for microbiome sequencing, and D.M.U. and A.P.R. analysed microbiome data and generated custom scripts. W.S. wrote the original draft of the manuscript. J.B., W.S. and D.M.U. revised the draft with input from all authors.

Corresponding author

Correspondence to Judith Behnsen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Microbiology thanks Ilse Jacobsen, Tobias Hohl and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Purified fungal siderophores promote Salmonella growth.

(a) Growth of Salmonella strains WT (n = 5), iroN (DMEM n = 5, DMEM + Lcn2 n = 6), iroN fhuA (DMEM n = 3, DMEM + Lcn2 n = 4), iroN fhuE (n = 3) at different time points (2, 5, 8 h) in DMEM/F12 + 10% FBS or DMEM/F12 + 10% FBS + 100 ng/ml lipocalin-2. (b) Growth of Salmonella strains WT, iroN, iroN fhuA or iroN fhuE in DMEM/F12 + 10% FBS + 100 nM coprogen (n = 3) or DMEM/F12 + 10% FBS + 100 nM ferrichrome (n = 3). (c) Growth of Salmonella strains WT, iroN fepA and iroN fepA fhuA fhuE in LB. Time points were taken at 2, 5, 8 and 24 h (n = 3). (d) Growth of Salmonella strains WT and fhuA fhuE at 1 h, 3 h, 5 h, 7 h post inoculation in co-culture in minimal medium (n = 3), minimal medium + 100 nM ferrichrome (n = 3), minimal medium + 100 ng/ml deferoxamine (n = 3). Statistics: 2way ANOVA with Tukey’s multiple comparisons test. Data represent geometric mean + /- geometric SD. Numerical p values are indicated within the figure.

Source data

Extended Data Fig. 2 Fungal siderophores from gut fungi promote Salmonella growth.

(a) Viability of fungi at 48 h in minimal medium measured as dry weight, mg/tube for A. niger and mouse isolate or CFU/mg for S. cerevisiae and M. furfur (n = 3). (b-d) Growth of Salmonella strains iroN fepA and iroN fepA fhuA fhuE at 8 h post inoculation in co-culture in minimal media with or without the addition of (b) live fungi (n = 4), (c) fungal supernatant (n = 3) or (d) fungal homogenate (n = 3). (e) Growth of Salmonella WT and Malassezia furfur in co-culture or alone in LB supplemented with 1% Tween40 and 0.2% oleic acid (n = 4). Data represent geometric mean and +/− geometric SD. Statistics: one-way ANOVA with Tukey’s multiple comparisons test. Numerical p values are indicated within the figure.

Source data

Extended Data Fig. 3 Diet composition affects composition of the bacterial microbiota.

(a) Principal component analysis of 16 S, left panel, and ITS, right panel, sequencing data of diet switch from chow LM485 to purified 50ppm diet (n = 5). (b) Relative abundance of selected bacterial genera prior and after diet change (n = 5) (c) Relative abundance of selected fungal genera prior and after diet change (n = 5). (b, c) Data represent mean +/− SD. Statistics: unpaired t test. Numerical p values are indicated within the figure.

Source data

Extended Data Fig. 4 Diet composition affects composition of the fungal microbiota.

(a) Principal component analysis of 16 S, left panel, and ITS, right panel, sequencing data of diet switch from chow LM485 to chow 2914 (n = 5). (b) Relative abundance of selected bacterial genera prior and after diet change (n = 5) (c) Relative abundance of selected fungal genera prior and after diet change (n = 5). (b, c) Data represent mean +/− SD. Statistics: unpaired t test. Numerical p values are indicated within the figure.

Source data

Extended Data Fig. 5 Fungal siderophores from mouse and human food promote Salmonella growth.

(a) Growth of Salmonella strains iroN fepA and iroN fepA fhuA fhuE at 8 h post inoculation in minimal medium with the addition of chow suspensions from chow 5066 (n = 7), chow LM485 (n = 7), purified 200ppm (n = 7) and purified 50ppm (n = 3) in aerobic shaking liquid culture. (b) Growth of Salmonella strains iroN fepA and iroN fepA fhuA fhuE at 8 h post inoculation in minimal medium with the addition of chow suspensions and ferrichrome (5 nM, 10 nM, 50 nM) (n = 3) (c, d) Growth of Salmonella strains iroN fepA and iroN fepA fhuA fhuE at 8 h post inoculation in minimal medium with the addition of chow suspensions in (c) aerobic non-shaking liquid culture (n = 3) and (d) anaerobic non-shaking liquid culture (n = 3). (e) Growth of Salmonella strains iroN fepA and iroN fepA fhuA fhuE at 8 h post inoculation in minimal medium with the addition of human food suspensions (minimal medium, Agaricus bisporus mushrooms, Quorn™, sourdough n = 3; beer, red wine, white bread, chicken, flour, lettuce, banana n = 4; blue cheese n = 5). Data are depicted as geometric mean +/− geometric SD. Statistics: Ordinary one-way ANOVA with Tukey’s multiple comparisons test. Numerical p values are indicated within the figure.

Source data

Extended Data Fig. 6 Siderophores from diet and mycobiota promote Salmonella gut colonization.

(a) mRNA expression of innate immune genes extracted from cecal tissue 96 h post infection with WT Salmonella or iroN fepA Salmonella. Expression is normalized to housekeeping gene Actb (Tnf, Ifng, Il17a, Il22 n = 8; Cxcl1 WT n = 7, iroN fepA n = 8). (b) Colonization measured as CFU/mg of Salmonella strains iroN fepA and iroN fepA fhuA in fecal samples of SPF mice collected at different time points post infection [24 h (n = 8), 48 h (n = 8), 72 h (n = 5), 96 h (n = 8)]. (c) Dissemination measured as CFU/mg of Salmonella strains iroN fepA and iroN fepA fhuA in peripheral organs of SPF mice collected at 96 h post infection [liver, spleen (n = 7), Peyer’s patches (PP) and mesenteric lymph nodes (MLN) (n = 8)]. (d) Competitive index of Salmonella strain iroN fepA over iroN fepA fhuA in organ homogenate liver, spleen (n = 7), Peyer’s patches (PP) and mesenteric lymph nodes (MLN) (n = 8) at 96 h. (e) Colonization measured as CFU/mg of Salmonella strains iroN fepA and iroN fepA fhuA fhuE in fecal samples of ASF mice collected at different time points post infection [24 h (chow n = 19, purified n = 20), 48 h (chow n = 19, purified n = 20), 72 h (chow = 15, purified n = 16), 96 h (chow n = 14, purified n = 16)]. (f) Competitive index (CI) of Salmonella strain iroN fepA over iroN fepA fhuA fhuE in fecal samples of ASF mice at 24 h, 48 h, 72 h, and 96 h. Each line represents CI development in one mouse (shown are mice with samples at more than 2 time points; chow n = 15, purified n = 16). (g) Slope of CI development in (f) (mice with samples at all time points; chow n = 14, purified n = 16). (h) CI of Salmonella strain iroN fepA over iroN fepA fhuA fhuE in fecal samples of ASF mice at 96 h compared to 24 h (mice with samples at both time points; chow n = 14, purified n = 16). (i) Colonization measured as CFU/mg of Salmonella strains iroN fepA and iroN fepA fhuA fhuE in fecal samples of ASF mice collected at 96 h post infection (chow n = 5, purified n = 6, purified + fungi n = 7). Data are depicted as geometric mean +/− geometric SD (a-c, e, i), box plot from 25th to 75th percentile, median, minimum and maximum (d), or median with 95% confidence interval (g, h). Statistics: For panel (a), (d), (g), and (h) one sample t test compared to no competitive advantage (CI = 1) was used. For panel (g) and (h) unpaired Mann-Whitney test was used. Numerical p values are indicated within the figure.

Source data

Extended Data Fig. 7 Proposed model.

Early during Salmonella infection, before the onset of inflammation, Salmonella can acquire iron in the gut environment via secretion of siderophores enterobactin and salmochelin. Other sources of iron are siderophores present in the mouse diet and siderophores produced by the mycobiota. At later stages of infection, after the onset of inflammation, host protein lipocalin-2 binds enterobactin and prevents Salmonella to acquire iron via this siderophore, rendering fungal siderophores present in the diet and produced by the mycobiota more important for Salmonella growth in the murine gut.

Supplementary information

Reporting Summary

Supplementary Table

Relative abundances 16S and ITS, diet information and resources table.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Santus, W., Rana, A.P., Devlin, J.R. et al. Mycobiota and diet-derived fungal xenosiderophores promote Salmonella gastrointestinal colonization. Nat Microbiol 7, 2025–2038 (2022). https://doi.org/10.1038/s41564-022-01267-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-022-01267-w

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

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