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:

Inflammation-associated nitrate facilitates ectopic colonization of oral bacterium Veillonella parvula in the intestine

Abstract

Colonization of the intestine by oral microbes has been linked to multiple diseases such as inflammatory bowel disease and colon cancer, yet mechanisms allowing expansion in this niche remain largely unknown. Veillonella parvula, an asaccharolytic, anaerobic, oral microbe that derives energy from organic acids, increases in abundance in the intestine of patients with inflammatory bowel disease. Here we show that nitrate, a signature metabolite of inflammation, allows V. parvula to transition from fermentation to anaerobic respiration. Nitrate respiration, through the narGHJI operon, boosted Veillonella growth on organic acids and also modulated its metabolic repertoire, allowing it to use amino acids and peptides as carbon sources. This metabolic shift was accompanied by changes in carbon metabolism and ATP production pathways. Nitrate respiration was fundamental for ectopic colonization in a mouse model of colitis, because a V. parvula narG deletion mutant colonized significantly less than a wild-type strain during inflammation. These results suggest that V. parvula harness conditions present during inflammation to colonize in the intestine.

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 abundance of nitrate-respiring Veillonella spp. is increased during IBD.
Fig. 2: Nitrate respiration expands the metabolic repertoire of V. parvula.
Fig. 3: Metabolic changes associated with nitrate respiration.
Fig. 4: Nitrate respiration-mediated changes in membrane potential are harnessed for substrate transport.
Fig. 5: The conditional dispensability of ATP synthase unveils mechanistic aspects associated with fermentation and nitrate respiration.
Fig. 6: Nitrate reductase activity is required for efficient colonization in a mouse model of inflammation.

Similar content being viewed by others

Data availability

RNA-seq data are available on the Sequence Read Archive (SRA) under BioProject accession no. PRJNA861991. Human microbiome data were accessed through data deposited on the SRA under BioProject accession no. PRJNA436359. Source data are provided with this paper.

Code availability

No customized code was generated or used in the present study.

References

  1. Lamont, R. J., Koo, H. & Hajishengallis, G. The oral microbiota: dynamic communities and host interactions. Nat. Rev. Microbiol. 16, 745–759 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Dewhirst, F. E. et al. The human oral microbiome. J. Bacteriol. 192, 5002–5017 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Kitamoto, S. et al. The intermucosal connection between the mouth and gut in commensal pathobiont-driven colitis. Cell 182, 447–462.e414 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Atarashi, K. et al. Ectopic colonization of oral bacteria in the intestine drives TH1 cell induction and inflammation. Science 358, 359–365 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Gevers, D. et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe 15, 382–392 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Shaw, K. A. et al. Dysbiosis, inflammation, and response to treatment: a longitudinal study of pediatric subjects with newly diagnosed inflammatory bowel disease. Genome Med. 8, 75 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Ananthakrishnan, A. N. et al. Gut microbiome function predicts response to anti-integrin biologic therapy in inflammatory bowel diseases. Cell Host Microbe 21, 603–610 e603 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Santoru, M. L. et al. Cross sectional evaluation of the gut-microbiome metabolome axis in an Italian cohort of IBD patients. Sci. Rep. 7, 9523 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Geng, J. et al. Co-occurrence of driver and passenger bacteria in human colorectal cancer. Gut Pathogen. 6, 26 (2014).

    Article  Google Scholar 

  10. Kostic, A. D. et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 14, 207–215 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Flemer, B. et al. The oral microbiota in colorectal cancer is distinctive and predictive. Gut 67, 1454–1463 (2018).

    Article  PubMed  CAS  Google Scholar 

  12. Tunney, M. M. et al. Detection of anaerobic bacteria in high numbers in sputum from patients with cystic fibrosis. Am. J. Respir. Crit. Care Med 177, 995–1001 (2008).

    Article  PubMed  Google Scholar 

  13. Rogers, G. B. et al. characterization of bacterial community diversity in cystic fibrosis lung infections by use of 16s ribosomal DNA terminal restriction fragment length polymorphism profiling. J. Clin. Microbiol. 42, 5176–5183 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Rogers, G. B., Skelton, S., Serisier, D. J., van der Gast, C. J. & Bruce, K. D. Determining cystic fibrosis-affected lung microbiology: comparison of spontaneous and serially induced sputum samples by use of terminal restriction fragment length polymorphism profiling. J. Clin. Microbiol. 48, 78–86 (2010).

    Article  PubMed  CAS  Google Scholar 

  15. Seedorf, H. et al. Bacteria from diverse habitats colonize and compete in the mouse gut. Cell 159, 253–266 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Hove, H. & Mortensen, P. B. Influence of intestinal inflammation (IBD) and small and large bowel length on fecal short-chain fatty acids and lactate. Dig. Dis. Sci. 40, 1372–1380 (1995).

    Article  PubMed  CAS  Google Scholar 

  17. Franzosa, E. A. et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat. Microbiol. 4, 293–305 (2019).

    Article  PubMed  CAS  Google Scholar 

  18. Schicho, R. et al. Quantitative metabolomic profiling of serum, plasma, and urine by 1H NMR spectroscopy discriminates between patients with inflammatory bowel disease and healthy individuals. J. Proteome Res. 11, 3344–3357 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Winter, S. E. et al. Host-derived nitrate boosts growth of E. coli in the inflamed gut. Science 339, 708–711 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Hughes, E. R. et al. Microbial respiration and formate oxidation as metabolic signatures of inflammation-associated dysbiosis. Cell Host Microbe 21, 208–219 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Gillis, C. C. et al. Dysbiosis-associated change in host metabolism generates lactate to support Salmonella growth. Cell Host Microbe 23, 54–64 e56 (2018).

    Article  PubMed  CAS  Google Scholar 

  23. 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  PubMed  PubMed Central  CAS  Google Scholar 

  24. Lopez, C. A., Rivera-Chavez, F., Byndloss, M. X. & Baumler, A. J. The periplasmic nitrate reductase NapABC supports luminal growth of Salmonella enterica serovar Typhimurium during colitis. Infect. Immun. 83, 3470–3478 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Lopez, C. A. et al. Phage-mediated acquisition of a type III secreted effector protein boosts growth of Salmonella by nitrate respiration. mBio. 3, e00143-12 (2012).

  26. Hughes, C. V., Kolenbrander, P. E., Andersen, R. N. & Moore, L. V. Coaggregation properties of human oral Veillonella spp.: relationship to colonization site and oral ecology. Appl. Environ. Microbiol. 54, 1957–1963 (1988).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Mashima, I. & Nakazawa, F. The interaction between Streptococcus spp. and Veillonella tobetsuensis in the early stages of oral biofilm formation. J. Bacteriol. 197, 2104–2111 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. van der Hoeven, J. S., Toorop, A. I. & Mikx, R. H. Symbiotic relationship of Veillonella alcalescens and Streptococcus mutans in dental plaque in gnotobiotic rats. Caries Res 12, 142–147 (1978).

    Article  PubMed  Google Scholar 

  29. Mikx, F. H. & Van der Hoeven, J. S. Symbiosis of Streptococcus mutans and Veillonella alcalescens in mixed continuous cultures. Arch. Oral. Biol. 20, 407–410 (1975).

    Article  PubMed  CAS  Google Scholar 

  30. Periasamy, S. & Kolenbrander, P. E. Central role of the early colonizer Veillonella sp. in establishing multispecies biofilm communities with initial, middle, and late colonizers of enamel. J. Bacteriol. 192, 2965–2972 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Delwiche, E. A., Pestka, J. J. & Tortorello, M. L. The veillonellae: Gram-negative cocci with a unique physiology. Annu. Rev. Microbiol. 39, 175–193 (1985).

    Article  PubMed  CAS  Google Scholar 

  32. Schirmer, M. et al. Compositional and temporal changes in the gut microbiome of pediatric ulcerative colitis patients are linked to disease course. Cell Host Microbe 24, 600–610.e604 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Kugathasan, S. et al. Prediction of complicated disease course for children newly diagnosed with Crohn’s disease: a multicentre inception cohort study. Lancet 389, 1710–1718 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Rogosa, M. The genus Veillonella. I. General cultural, ecological, and biochemical considerations. J. Bacteriol. 87, 162–170 (1964).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Doel, J. J., Benjamin, N., Hector, M. P., Rogers, M. & Allaker, R. P. Evaluation of bacterial nitrate reduction in the human oral cavity. Eur. J. Oral Sci. 113, 14–19 (2005).

    Article  PubMed  CAS  Google Scholar 

  36. Inderlied, C. B. & Delwiche, E. A. Nitrate reduction and the growth of Veillonella alcalescens. J. Bacteriol. 114, 1206–1212 (1973).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Mitsui, T., Saito, M. & Harasawa, R. Salivary nitrate-nitrite conversion capacity after nitrate ingestion and incidence of Veillonella spp. in elderly individuals. J. Oral Sci. 60, 405–410 (2018).

    Article  PubMed  Google Scholar 

  38. Wicaksono, D. P., Washio, J., Abiko, Y., Domon, H. & Takahashi, N. Nitrite production from nitrate and its link with lactate metabolism in oral Veillonella spp. Appl. Environ. Microbiol. https://doi.org/10.1128/AEM.01255-20 (2020).

  39. Moreno-Vivian, C., Cabello, P., Martinez-Luque, M., Blasco, R. & Castillo, F. Prokaryotic nitrate reduction: molecular properties and functional distinction among bacterial nitrate reductases. J. Bacteriol. 181, 6573–6584 (1999).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Knapp, S. et al. Natural competence is common among clinical Isolates of Veillonella parvula and is useful for genetic manipulation of this key member of the oral microbiome. Front. Cell Infect. Microbiol 7, 139 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Reuss, O. & Morschhauser, J. A family of oligopeptide transporters is required for growth of Candida albicans on proteins. Mol. Microbiol. 60, 795–812 (2006).

    Article  PubMed  CAS  Google Scholar 

  42. Berntsson, R. P. et al. The structural basis for peptide selection by the transport receptor OppA. EMBO J. 28, 1332–1340 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. de Vries, W., Rietveld-Struijk, R. M. & Stouthamer, A. H. ATP formation associated with fumarate and nitrate reduction in growing cultures of Veillonella alcalescens. Antonie Van Leeuwenhoek 43, 153–167 (1977).

    Article  PubMed  Google Scholar 

  44. Galivan, J. H. & Allen, S. H. Methylmalonyl coenzyme A decarboxylase. Its role in succinate decarboxylation by Micrococcus lactilyticus. J. Biol. Chem. 243, 1253–1261 (1968).

    Article  PubMed  CAS  Google Scholar 

  45. de Vries, W., van Wijck-Kapteyn, W. M. & Oosterhuis, S. K. The presence and function of cytochromes in Selenomonas ruminantium, Anaerovibrio lipolytica and Veillonella alcalescens. J. Gen. Microbiol. 81, 69–78 (1974).

    PubMed  Google Scholar 

  46. McCormick, N. G., Ordal, E. J. & Whiteley, H. R. Degradation of pyruvate Micrococcus lactilyticus. I. General properties of the formate-exchange reaction. J. Bacteriol. 83, 887–898 (1962).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Griffith, M. J. & Nishimura, J. S. Acetate kinase from Veillonella alcalescens. Purification and physical properties. J. Biol. Chem. 254, 442–446 (1979).

    Article  PubMed  CAS  Google Scholar 

  48. Pelroy, R. A. & Whiteley, H. R. Regulatory properties of acetokinase from Veillonella alcalescens. J. Bacteriol. 105, 259–267 (1971).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Bowman, C. M., Valdez, R. O. & Nishimura, J. S. Acetate kinase from Veillonella alcalescens. Regulation of enzyme activity by succinate and substrates. J. Biol. Chem. 251, 3117–3121 (1976).

    Article  PubMed  CAS  Google Scholar 

  50. Kolios, G., Valatas, V. & Ward, S. G. Nitric oxide in inflammatory bowel disease: a universal messenger in an unsolved puzzle. Immunology 113, 427–437 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Anderson, C. J. et al. Microbes exploit death-induced nutrient release by gut epithelial cells. Nature 596, 262–267 (2021).

    Article  PubMed  CAS  Google Scholar 

  52. Madej, M. et al. Structural and functional insights into oligopeptide acquisition by the RagAB transporter from Porphyromonas gingivalis. Nat. Microbiol. 5, 1016–1025 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Pucino, V. et al. Lactate buildup at the site of chronic inflammation promotes disease by inducing CD4+ T. Cell Metab. Rewiring Cell Metab. 30, 1055–1074.e1058 (2019).

    PubMed  CAS  Google Scholar 

  54. Dalgaard, P. & Koutsoumanis, K. Comparison of maximum specific growth rates and lag times estimated from absorbance and viable count data by different mathematical models. J. Microbiol. Methods 43, 183–196 (2001).

    Article  PubMed  CAS  Google Scholar 

  55. Pasolli, E. et al. Extensive unexplored human microbiome diversity revealed by over 150,000 genomes from metagenomes spanning age, geography, and lifestyle. Cell 176, 649–662 e620 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Edgar, R. C. Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26, 2460–2461 (2010).

    Article  PubMed  CAS  Google Scholar 

  57. Segata, N., Bornigen, D., Morgan, X. C. & Huttenhower, C. PhyloPhlAn is a new method for improved phylogenetic and taxonomic placement of microbes. Nat. Commun. 4, 2304 (2013).

    Article  PubMed  Google Scholar 

  58. Shishkin, A. A. et al. Simultaneous generation of many RNA-seq libraries in a single reaction. Nat. Methods 12, 323–325 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Zhu, Y. Y., Machleder, E. M., Chenchik, A., Li, R. & Siebert, P. D. Reverse transcriptase template switching: a SMART approach for full-length cDNA library construction. Biotechniques 30, 892–897 (2001).

    Article  PubMed  CAS  Google Scholar 

  60. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  PubMed  CAS  Google Scholar 

  62. Abeel, T., Van Parys, T., Saeys, Y., Galagan, J. & Van de Peer, Y. GenomeView: a next-generation genome browser. Nucleic Acids Res. 40, e12 (2012).

    Article  PubMed  CAS  Google Scholar 

  63. Mascanfroni, I. D. et al. Metabolic control of type 1 regulatory T cell differentiation by AHR and HIF1-alpha. Nat. Med. 21, 638–646 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Ng, S. K. & Hamilton, I. R. Lactate metabolism by Veillonella parvula. J. Bacteriol. 105, 999–1005 (1971).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. L. Merritt at Oregon Health & Science University for the SKV38 strain of V. parvula and the pBSLJ1 plasmid, as well as J. Vargas Asencio at MIT for helpful discussions and T. Reimels for editorial assistance. RNA-seq libraries were constructed and sequenced at the Broad Institute of MIT and Harvard by the Microbial ’Omics Core and Genomics Platform, respectively. The Microbial ’Omics Core also provided preliminary analysis for all RNA-seq data. We thank the gnotobiotic animal facility at the Broad Institute, specifically C. Umana and A. Discua, for helping with experiments and T. Caron for providing support for running the facility. This work was supported by grants from the National Institutes of Health (grant no. P30 DK043351), Center for Microbiome Informatics and Therapeutics, and Crohn’s and Colitis Foundation to R.J.X and from the Deutsche Forschungsgemeinschaft (German Research Foundation, grant no. 426120468) to M.S.

Author information

Authors and Affiliations

Authors

Contributions

D.F.R.-T., H.V. and R.J.X. conceived the study. D.F.R.-T., E.M.B., E.R.T., K.A.P. and J.A.-P. provided the methodology. D.F.R.-T., M.S. and R.L.W. did the computational analyses. D.F.R.-T., E.M.B., E.R.T., M.A.O., A.M.T.M., K.D., T.M. and K.A.P. carried out the investigations. D.F.R.-T., E.M.B., H.V. and R.J.X. wrote the manuscript. All authors edited the manuscript. C.B.C., H.V. and R.J.X. supervised the study.

Corresponding author

Correspondence to Ramnik J. Xavier.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Microbiology thanks Andrés Vázquez-Torres 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 Phylogenetic analysis of nitrate reductases.

Analysis of the Nar, Nap, and Nas nitrate reductase families in human (a) gut and (b) oral microbes, using the NarG and NapA protein sequences of E. coli, and NasA of K. pneumoniae as templates. The sequences were searched in the assembled gene catalog from Pasolli et al. (2019)55. (c) Phylogenetic tree of NarG in gut microbes reveals that Veillonella is one of the few genera within the Firmicutes with a putative nitrate reductase. Genes with at least 45% amino acid similarity were used for analysis.

Extended Data Fig. 2 Nitrate-stimulated growth of various Veillonella strains, and narI::chl complementation experiments.

(a) The growth of V. parvula SKV38 was assessed in different complex media supplemented or not with 40 mM nitrate (n = 4 biological replicates). (b) Growth curves of WT and narI::chl strains in SK, SKL, SKN, and SKLN media. Cells lacking the NarI protein are unable to grow in SKN, and nitrate does not stimulate its growth when lactate is present (n = 3 biological replicates). (c) The growth of various Veillonella strains was assessed in SK, SKL, SKN, and SKLN media (n = 3 biological replicates). Growth rates (µmax) were calculated for each growth curve. Data (a-c) represent mean ± SEM, analyzed by one-way ANOVA and Tukey’s HSD test. NS, not significant.

Source data

Extended Data Fig. 3 Nitrate metabolism in Veillonella.

(a) Levels of nitrate and nitrite were measured at the mid-exponential phase in cells growing on SKN medium supplemented or not with lactate as the carbon source (n = 3 biological replicates). The measured concentrations of nitrate and nitrite in the uninoculated SK medium were 43.161 ± 0.410 mM and <0.050 mM, respectively (n = 3 biological replicates). (b) Ammonium (n = 6 biological replicates) or (c) nitrite (n = 4 biological replicates) were used to determine if the positive effect of nitrate on growth of V. parvula was due to nitrate being used as a source of nitrogen or an electron acceptor, respectively. Growth rates (µmax) were calculated for each growth curve. Data represent mean ± SEM, analyzed by one-way ANOVA and Tukey’s HSD test (a, b) or Dunnett’s test (c). NS, not significant.

Source data

Extended Data Fig. 4 Amino acids and peptides stimulate the growth of different Veillonella strains.

(a) The growth of various Veillonella strains was studied in SKbN with 0.5% yeast extract, and with and without 1% casitone, 1% casamino acids, or 50 mM L-aspartic acid (n = 3 biological replicates). (b) The addition of lactate or malate stimulates growth of V. parvula in SKb medium (n = 4 biological replicates). Growth rates (µmax) were calculated for each growth curve. Data represent mean ± SEM, analyzed by one-way ANOVA and Tukey’s HSD test (a) or Dunnett’s test (b). NS, not significant.

Source data

Extended Data Fig. 5 LC-MS analysis of amino acids in unspent and spent media.

LC-MS analysis of the unspent (n = 6 biological replicates) and spent SKL, SKN, and SKLN media (n = 3 biological replicates) was used to study amino acid consumption. Supernatants were collected at the (a) mid-exponential and (b) late exponential phases. Data represent mean ± SEM, analyzed by one-way ANOVA and Tukey’s HSD test. NS, not significant.

Source data

Extended Data Fig. 6 Role of the PMF in the production of ATP and transport, and phenotype of the L-lactate permease (lldP::chl) mutant.

(a) Measurement of the proton motive force (PMF) in WT and narG::tet cells growing on SK with and without lactate or malate, and with or without CCCP. The PMF was measured before and 40 min after addition of 40 mM nitrate. CCCP was added to confirm that the observed changes were associated with the proton gradient (n = 3 biological replicates). (b) Lactate (n = 3 biological replicates) and aspartate (n = 2 biological replicates) consumption were measured in resting WT cells resuspended in resting cell (RC) buffer with or without nitrate. (c) Lactate consumption (n = 4 biological replicates) was measured in growing atp::tet cells with or without nitrate. Both metabolites were measured in the supernatant using colorimetric techniques. (d) Growth experiments with WT Veillonella and a strain lacking LldP (lldP::chl) in SK, SKL, SKN, and SKLN media (n = 4 biological replicates). Cells lacking the LldP transporter are nearly unable to grow with lactate as the carbon source. Growth rates (µmax) were calculated for each growth curve. Data (ad) represent mean ± SEM, analyzed by two-sided t-test (a, b, and c), or one-way ANOVA and Tukey’s HSD test (growth rates in (d). NS, not significant.

Source data

Extended Data Fig. 7 Phenotype of cells lacking components of the ATP synthase, and expression analysis of the atp operon.

(a) Growth experiments with WT Veillonella and the AtpC (atpC::chl) and AtpD-AtpC (atpDC::chl) deletion strains in SK, SKL, SKN, and SKLN media (n = 4 biological replicates). These deletion strains exhibited a phenotype similar to the one seen in the atp::tet strain. Growth rates (µmax) were calculated for each growth curve. (b) Transcriptional analysis of the atpA gene in cells growing in SKL, SKN, and SKLN media (n = 3 biological replicates). Data represent mean ± SEM, analyzed by one-way ANOVA and Tukey’s HSD test. NS, not significant.

Source data

Extended Data Fig. 8 Validation of the DSS-treated mouse model.

(a) Measurements of IFN-γ, TNF, IL-6, IL-17A, and IL-10 in the mesenteric lymph nodes of WT-colonized ASF mice without (n = 4) and with (n = 5) DSS treatment. (b) Lamina propria cells were isolated from colonic tissue and analyzed via flow cytometry. From left to right: Cells were first gated for positive expression of the pan immune cell marker CD45 while excluding dead cells staining for the viability dye Zombie UV. Next, gating was further cleaned by gating on FSC-A to exclude debris and FSC-A versus FSC-H to exclude doublets. Finally neutrophils were gated based on double positive expression of the two surface protein markers Ly6G and Ly6C. c) Left: representative contour plots showing the proportion of neutrophils based on positive expression of Ly6C and Ly6G on live CD45+ lymphocytes isolated from mouse colonic lamina propria. Right: Frequency of neutrophils among total CD45+ colonic lamina propria lymphocytes. (d) Total number of neutrophils isolated from mouse colons based on gating in c. The data in a, c, and d are representative of two independent experiments. For c and d, boxplots display the first and third quartiles with a thick line representing the median; n = 4 mice without DSS, and n = 5 mice with DSS treatment. A Student’s t-test was used in a, c, and d for statistical analyses.

Source data

Extended Data Fig. 9 Colonization of Veillonella in a mouse model of colitis.

(a) Raw Ct values of bacterial 16 S rRNA gene measured by qPCR in the stool of WT-colonized ASF mice with and without DSS treatment (n = 8–11 mice). (b) Absolute abundance of V. parvula in the stool of ASF mice as measured by CFU per mg of stool on selective SKLN media (n = 3 mice). (c) Absolute abundance of V. parvula in the colonic tissue of ASF mice as measured by CFU per mg of tissue on selective SKLN media (n = 4 mice). (d) Concentration of lactate in the gut of control- and DSS-treated ASF mice colonized with V. parvula (n = 8 mice). (e) Relative expression of the IIdD, lldP, hisA and argB genes in WT Veillonella colonizing the stool of mice with and without DSS treatment (n = 3–4). The V. parvula gyrA gene was used as an internal control. (f) Colon lengths of WT and iNOS KO SPF mice (n = 4–7). Data represent two independent experiments and show mean (a, df) ± SEM (df) or median (b, c) values. Data were analyzed by one-way ANOVA and Tukey’s HSD test or Mann-Whitney U-test.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Table 1.

Reporting summary

Supplementary Tables

Supplementary Table 2 Resistance genes used for V. parvula mutants. Supplementary Table 3 Primers for mutants. Supplementary Table 4 Primers for qPCR.

Supplementary Data File 1

LC–MS data.

Supplementary Data File 2

LC–MS analysis of unspent and spent SKL, SKN and SKLN media at the late exponential phase used to study dipeptide consumption.

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 Fig. 6

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.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

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

Rojas-Tapias, D.F., Brown, E.M., Temple, E.R. et al. Inflammation-associated nitrate facilitates ectopic colonization of oral bacterium Veillonella parvula in the intestine. Nat Microbiol 7, 1673–1685 (2022). https://doi.org/10.1038/s41564-022-01224-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-022-01224-7

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