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:

Inflammatory monocytes promote granuloma control of Yersinia infection

Abstract

Granulomas are organized immune cell aggregates formed in response to chronic infection or antigen persistence. The bacterial pathogen Yersiniapseudotuberculosis (Yp) blocks innate inflammatory signalling and immune defence, inducing neutrophil-rich pyogranulomas (PGs) within lymphoid tissues. Here we uncover that Yp also triggers PG formation within the murine intestinal mucosa. Mice lacking circulating monocytes fail to form defined PGs, have defects in neutrophil activation and succumb to Yp infection. Yersinia lacking virulence factors that target actin polymerization to block phagocytosis and reactive oxygen burst do not induce PGs, indicating that intestinal PGs form in response to Yp disruption of cytoskeletal dynamics. Notably, mutation of the virulence factor YopH restores PG formation and control of Yp in mice lacking circulating monocytes, demonstrating that monocytes override YopH-dependent blockade of innate immune defence. This work reveals an unappreciated site of Yersinia intestinal invasion and defines host and pathogen drivers of intestinal granuloma formation.

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: Intestinal PGs form upon oral Yersinia infection.
Fig. 2: Intestinal inflammation is spatially restricted to PGs.
Fig. 3: Inflammatory monocytes maintain PGs to restrict infection.
Fig. 4: Inflammatory monocytes control systemic Yersinia.
Fig. 5: Yersinia virulence factors induce intestinal PGs.
Fig. 6: Neutrophils control YopH-deficient Yersinia in absence of monocytes.

Similar content being viewed by others

Data availability

Raw RNA sequencing data are available on the Gene Expression Omnibus (accession no. GSE194334). All other raw data are available upon request to the corresponding author.

Code availability

Code for RNA sequencing analysis is available in the Supplementary Code File.

References

  1. Pagán, A. J. & Ramakrishnan, L. The formation and function of granulomas. Annu. Rev. Immunol. 36, 639–665 (2018).

    Article  PubMed  Google Scholar 

  2. Petersen, H. J. & Smith, A. M. The role of the innate immune system in granulomatous disorders. Front. Immunol. 4, 1–11 (2013).

    Article  CAS  Google Scholar 

  3. Casson, C. N. et al. Neutrophils and Ly6Chi monocytes collaborate in generating an optimal cytokine response that protects against pulmonary Legionella pneumophila infection. PLoS Pathog. 13, e1006309 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Dunay, I. R. et al. Gr1+ inflammatory monocytes are required for mucosal resistance to the pathogen Toxoplasma gondii. Immunity 29, 306–317 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Espinosa, V. et al. Inflammatory monocytes orchestrate innate antifungal immunity in the lung. PLoS Pathog. 10, e1003940 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Seo, S.-U. et al. Intestinal macrophages arising from CCR2+ monocytes control pathogen infection by activating innate lymphoid cells. Nat. Commun. 6, 8010 (2015).

    Article  CAS  PubMed  Google Scholar 

  7. Antonelli, L. R. et al. Intranasal Poly-IC treatment exacerbates tuberculosis in mice through the pulmonary recruitment of a pathogen-permissive monocyte/macrophage population. J. Clin. Invest. 120, 1674–1682 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. St. John, A. L. et al. S1P-dependent trafficking of intracellular Yersinia pestis through lymph nodes establishes buboes and systemic infection. Immunity 41, 440–450 (2014).

    Article  Google Scholar 

  9. Eisele, N. A. et al. Salmonella require the fatty acid regulator PPARδ for the establishment of a metabolic environment essential for long term persistence. Cell Host Microbe 14, 171–182 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Paff, J. R., Triplett, D. A. & Saari, T. N. Clinical and laboratory aspects of Yersinia pseudotuberculosis infections, with a report of two cases. Am. J. Clin. Pathol. 66, 101–110 (1976).

    Article  CAS  PubMed  Google Scholar 

  11. El-Maraghi, N. R. H. & Mair, N. S. The histopathology of enteric infection with Yersinia pseudotuberculosis. Am. J. Clin. Pathol. 71, 631–639 (1979).

    Article  CAS  PubMed  Google Scholar 

  12. Barnes, P. D., Bergman, M. A., Mecsas, J. C. & Isberg, R. R. Yersinia pseudotuberculosis disseminates directly from a replicating bacterial pool in the intestine. J. Exp. Med. 203, 1591–1601 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lamps, L. W. et al. The role of Yersinia enterocolitica and Yersinia pseudotuberculosis in granulomatous appendicitis: a histologic and molecular study. Am. J. Surg. Pathol. 25, 508–515 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Zhang, Y., Tam, J. W., Mena, P., van der Velden, A. W. M. & Bliska, J. B. CCR2+ inflammatory dendritic cells and translocation of antigen by type III secretion are required for the exceptionally large CD8+ T cell response to the protective YopE69-77 epitope during Yersinia infection. PLoS Pathog. 11, e1005167 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Zhang, Y., Khairallah, C., Sheridan, B. S., van der Velden, A. W. M. & Bliska, J. B. CCR2+ inflammatory monocytes are recruited to Yersinia pseudotuberculosis pyogranulomas and dictate adaptive responses at the expense of innate immunity during oral infection. Infect. Immun. 86, e00782–17 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kuziel, W. A. et al. Severe reduction in leukocyte adhesion and monocyte extravasation in mice deficient in CC chemokine receptor 2. Proc. Natl Acad. Sci. USA 94, 12053–12058 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Serbina, N. V. & Pamer, E. G. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat. Immunol. 7, 311–317 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Peterson, L. W. et al. RIPK1-dependent apoptosis bypasses pathogen blockade of innate signaling to promote immune defense. J. Exp. Med. 214, 3171–3182 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Tsutomu, U. Studies on the pathogenicity of Yersinia enterocolitica III. Comparative studies between Y. enterocolitica and Y. pseudotuberculosis. Microbiol. Immunol. 21, 505–516 (1977).

    Article  Google Scholar 

  20. Bliska, J. B., Brodsky, I. E. & Mecsas, J. Role of the Yersinia pseudotuberculosis virulence plasmid in pathogen-phagocyte interactions in mesenteric lymph nodes. EcoSal 9, eESP-0014–eESP-2021 (2021).

    Google Scholar 

  21. Davis, K. M. All Yersinia are not created equal: phenotypic adaptation to distinct niches within mammalian tissues. Front. Cell. Infect. Microbiol. 8, 1–8 (2018).

    Article  Google Scholar 

  22. Davis, K. M., Mohammadi, S. & Isberg, R. R. Community behavior and spatial regulation within a bacterial microcolony in deep tissue sites serves to protect against host attack. Cell Host Microbe 17, 21–31 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Clark, M. A., Hirst, B. H. & Jepson, M. A. M-cell surface β1 integrin expression and invasin-mediated targeting of Yersinia pseudotuberculosis to mouse Peyer’s patch M cells. Infect. Immun. 66, 1237–1243 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Fasciano, A. C. et al. Yersinia pseudotuberculosis YopE prevents uptake by M cells and instigates M cell extrusion in human ileal enteroid-derived monolayers. Gut Microbes 13, 1988390 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Nuss, A. M. et al. Tissue dual RNA-seq allows fast discovery of infection-specific functions and riboregulators shaping host-pathogen transcriptomes. Proc. Natl Acad. Sci. USA 114, E791–E800 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Serbina, N. V., Salazar-Mather, T. P., Biron, C. A., Kuziel, W. A. & Pamer, E. G. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity 19, 59–70 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. Zigmond, E. et al. Ly6C hi monocytes in the inflamed colon give rise to proinflammatory effector cells and migratory antigen-presenting cells. Immunity 37, 1076–1090 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Grainger, J. R. et al. Inflammatory monocytes regulate pathologic responses to commensals during acute gastrointestinal infection. Nat. Med. 19, 713–721 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Davis, J. M. & Ramakrishnan, L. The role of the granuloma in expansion and dissemination of early tuberculous infection. Cell 136, 37–49 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Satpathy, A. T. et al. Notch2-dependent classical dendritic cells orchestrate intestinal immunity to attaching-and-effacing bacterial pathogens. Nat. Immunol. 14, 937–948 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Boring, L. et al. Impaired monocyte migration and reduced type 1 (Th1) cytokine responses in C-C chemokine receptor 2 knockout mice. J. Clin. Invest. 100, 2552–2561 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Borjesson, D. L., Simon, S. I., Hodzic, E., Ballantyne, C. M. & Barthold, S. W. Kinetics of CD11b/CD18 up-regulation during infection with the agent of human granulocytic ehrlichiosis in mice. Lab. Investig. 82, 303–311 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Mann, B. S. & Chung, K. F. Blood neutrophil activation markers in severe asthma: lack of inhibition by prednisolone therapy. Respir. Res. 7, 1–10 (2006).

    Article  Google Scholar 

  34. Yoon, J. W., Pahl, M. V. & Vaziri, N. D. Spontaneous leukocyte activation and oxygen-free radical generation in end-stage renal disease. Kidney Int. 71, 167–172 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Othman, A., Sekheri, M. & Filep, J. G. Roles of neutrophil granule proteins in orchestrating inflammation and immunity. FEBS J. 289, 3932–3953 (2022).

    Article  CAS  PubMed  Google Scholar 

  36. Rijneveld, A. W., De Vos, A. F., Florquin, S., Verbeek, J. S. & van der Poll, T. CD11b limits bacterial outgrowth and dissemination during murine pneumococcal pneumonia. J. Infect. Dis. 191, 1755–1760 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Latger-Cannard, V., Besson, I., Doco-Lecompte, T. & Lecompte, T. A standardized procedure for quantitation of CD11b on polymorphonuclear neutrophil by flow cytometry: potential application in infectious diseases. Clin. Lab. Haematol. 26, 177–186 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Portnoy, D. A., Wolf-Watz, H., Bolin, I., Beeder, A. B. & Falkow, S. Characterization of common virulence plasmids in yersinia species and their role in the expression of outer membrane proteins. Infect. Immun. 43, 108–114 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Balada-Llasat, J. M. & Mecsas, J. C. Yersinia has a tropism for B and T cell zones of lymph nodes that is independent of the type III secretion system. PLoS Pathog. 2, e86 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Mukherjee, S. et al. Yersinia YopJ acetylates and inhibits kinase activation by blocking phosphorylation. Science 312, 1211–1214 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Brodsky, I. E. & Medzhitov, R. Reduced secretion of YopJ by Yersinia limits in vivo cell death but enhances bacterial virulence. PLoS Pathog. 4, e1000067 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Chung, L. K. et al. The Yersinia virulence factor yopm hijacks host kinases to inhibit type III effector-triggered activation of the pyrin inflammasome. Cell Host Microbe 20, 296–306 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Rolán, H. G., Durand, E. A. & Mecsas, J. Identifying Yersinia YopH-targeted signal transduction pathways that impair neutrophil responses during in vivo murine infection. Cell Host Microbe 14, 306–317 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Green, S. P., Hartland, E. L., Robins-Browne, R. M. & Phillips, W. A. Role of YopH in the suppression of tyrosine phosphorylation and respiratory burst activity in murine macrophages infected with Yersinia enterocolitica. J. Leukoc. Biol. 57, 972–977 (1995).

    Article  CAS  PubMed  Google Scholar 

  45. Shaban, L. et al. Yersinia pseudotuberculosis YopH targets SKAP2-dependent and independent signaling pathways to block neutrophil antimicrobial mechanisms during infection. PLoS Pathog. 16, e1008576 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Mecsas, J., Raupach, B. & Falkow, S. The Yersinia Yops inhibit invasion of Listeria, Shigella and Edwardsiella but not Salmonella into epithelial cells. Mol. Microbiol. 28, 1269–1281 (1998).

    Article  CAS  PubMed  Google Scholar 

  47. Logsdon, L. K. & Mecsas, J. C. Requirement of the Yersinia pseudotuberculosis effectors YopH and YopE in colonization and persistence in intestinal and lymph tissues. Infect. Immun. 71, 4595–4607 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Fisher, M. L., Castillo, C. & Mecsas, J. Intranasal inoculation of mice with Yersinia pseudotuberculosis causes a lethal lung infection that is dependent on Yersinia Outer Proteins and PhoP. Infect. Immun. 75, 429–442 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Westermark, L., Fahlgren, A. & Fällman, M. Yersinia pseudotuberculosis efficiently escapes polymorphonuclear neutrophils during early infection. Infect. Immun. 82, 1181–1191 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Stackowicz, J., Jönsson, F. & Reber, L. L. Mouse models and tools for the in vivo study of neutrophils. Front. Immunol. 10, 3130 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Wojtasiak, M. et al. Depletion of Gr-1+, but not Ly6G+, immune cells exacerbates virus replication and disease in an intranasal model of herpes simplex virus type 1 infection. J. Gen. Virol. 91, 2158–2166 (2010).

    Article  CAS  PubMed  Google Scholar 

  52. Bozue, J. et al. A Yersinia pestis tat mutant is attenuated in bubonic and small-aerosol pneumonic challenge models of infection but not as attenuated by intranasal challenge. PLoS ONE 9, e104524 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Durand, E. A., Maldonado-Arocho, F. J., Castillo, C., Walsh, R. L. & Mecsas, J. C. The presence of professional phagocytes dictates the number of host cells targeted for Yop translocation during infection. Cell. Microbiol. 12, 1064–1082 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Taheri, N., Fahlgren, A. & Fällman, M. Yersinia pseudotuberculosis blocks neutrophil degranulation. Infect. Immun. 84, 3369–3378 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Murphy, A. J. et al. Neutrophil activation is attenuated by high-density lipoprotein and apolipoprotein A-I in in vitro and in vivo models of inflammation. Arterioscler. Thromb. Vasc. Biol. 31, 1333–1341 (2011).

    Article  CAS  PubMed  Google Scholar 

  56. Simonet, M. & Falkow, S. Invasin expression in Yersinia pseudotuberculosis. Infect. Immun. 60, 4414–4417 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Portnoy, D. A., Moseley, S. L. & Falkow, S. Characterization of plasmids and plasmid-associated determinants of Yersinia enterocolitica pathogenesis. Infect. Immun. 31, 775–782 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Zhang, Y., Murtha, J., Roberts, M. A., Siegel, R. M. & Bliska, J. B. Type III secretion decreases bacterial and host survival following phagocytosis of Yersinia pseudotuberculosis by macrophages. Infect. Immun. 76, 4299–4310 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Mack, M. et al. Expression and characterization of the chemokine receptors CCR2 and CCR5 in mice. J. Immunol. 166, 4697–4704 (2001).

    Article  CAS  PubMed  Google Scholar 

  60. Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    Article  CAS  PubMed  Google Scholar 

  61. Huber, W. et al. Orchestrating high-throughput genomic analysis with Bioconductor. Nat. Methods 12, 115–121 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Soneson, C., Love, M. I. & Robinson, M. D. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Research 4, 1521 (2016).

    Article  PubMed Central  Google Scholar 

  63. 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  CAS  PubMed  Google Scholar 

  64. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Bliska for generously providing plasmids for Yop mutant Yp strains, as well as K. Davis for generously providing the mCherry+ Yp plasmid. We thank the staff at the PennVet Comparative Pathology Core for their help in preparing the histological samples. We thank D. Christian and A. Stout for key advice on confocal microscopy methods, and S. Shin for constructive editorial comments and scientific discussion. This work was supported by NIH Awards R01AI128530 (I.E.B.), R0AI1139102A1 (I.E.B.), R01DK123528 (I.E.B.) and a BWF Investigator in the Pathogenesis of Infectious Disease Award (I.E.B.); the Foundation Blanceflor Postdoctoral Scholarship (D.S.), the Swedish Society for Medical Research postdoctoral fellowship (D.S.) and the Sweden-America Foundation J. Sigfrid Edström award (D.S.); NIH NRSA F31AI160741-01 (R.M.); NIH T32 AI141393-2 in Microbial Pathogenesis and Genomics (R.M.); Mark Foundation Grant 19-011MIA (I.E.B.), F32 AI164655 (J.P.G.); and NSF GRFP Award (S.P.). We thank members of the Brodsky laboratory for scientific discussion and D. Grubaugh for comments on the manuscript. We thank R. Kratofil for scientific discussion and advice on assays to measure neutrophil activation.

Author information

Authors and Affiliations

Authors

Contributions

D.S. established the initial findings of intestinal granulomas. D.S., R.M. and I.E.B. conceptualized the study and devised experiments. D.S. and R.M. devised the methodology and performed experiments. S.T.P., J.P.G. and I.R. performed experiments. C.-A.A., E.R. and M.L. performed the histology and histopathological scoring. E.K. prepared the RNA sequencing libraries. M.M. provided the anti-CCR2 antibody. R.M. and D.B. analysed the RNA sequencing data. I.E.B. acquired the funding and supervised the study. D.S., R.M. and I.E.B. wrote the original draft. All authors reviewed and edited the manuscript.

Corresponding author

Correspondence to Igor E. Brodsky.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Microbiology thanks the anonymous reviewers 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 Intestinal pyogranulomas form upon oral Yersinia infection.

(a) Flow cytometry plots displaying the gating strategy employed to identify eosinophils, dendritic cells, B cells and T cells in small-intestinal tissue at day 5 post-Yp infection. Representative of four independent experiments. (b) Frequency and total number of eosinophils, dendritic cells, B cells, CD4+ T cells and CD8+ T cells in small-intestinal tissue at day 5 post Yp-infection. Each data point represents one mouse (n = 15-20). Lines represent median. Pooled from four independent experiments. Wilcoxon test (two-tailed) was performed for paired analyses (PG- vs PG + ). Mann-Whitney U test (two-tailed) was performed for all remaining statistical analyses. * (p < 0.05), ** (p < 0.01), *** (p < 0.001), **** (p < 0.0001), NS (not significant, p > 0.05).

Extended Data Fig. 2 Non-pyogranuloma tissue does not undergo inflammation.

(a) Heat map of top 30 significantly upregulated genes in PG- compared to uninfected samples in descending order by fold change. False discovery rate < 0.05 using Benjamini–Hochberg procedure. (b) Gene ontology analysis of top 30 upregulated genes by fold change only in PG- compared to uninfected samples. Dotted line denotes p = 0.05.

Extended Data Fig. 3 CCR2-deficient mice cannot control intestinal Yersinia.

(a) Quantification of total number of intestinal lesions at day 3 post infection. Each data point indicates one mouse (n = 9-11). Line represents median. Pooled from two independent experiments. (b) H&E-stained paraffin-embedded small-intestinal sections containing Peyer’s patches from WT and Ccr2gfp/gfp mice at day 5 post infection. Dashed circle denotes area containing pyogranuloma or necrosuppurative lesion. Scale bars = 500 μm. Representative images from two independent experiments. (c) Bacterial burdens in small intestinal PG- and PG + tissue at day 3 post infection. Each data point represents mean CFU of 3-5 pooled punch biopsies from one mouse (n = 15-25). Lines represent geometric mean. Pooled data from four independent experiments. (d) Frequency of monocytes in blood at day 5 post infection. Each data point represents one mouse (n = 29-30). Lines indicate median. Pooled data from four independent experiments. (e) Bacterial burdens in small intestinal PG- and PG + tissue at day 5 post infection. Each data point represents the mean CFU of 3-5 pooled punch biopsies from one mouse (n = 21-30). Lines indicate geometric mean. Data pooled from four independent experiments. (f) Bacterial burdens in small intestinal PG- and PG + tissue at day 3 post infection. Each data point represents the mean CFU of 3-5 pooled punch biopsies from one mouse (n = 10-13). Lines indicate geometric mean. Pooled data from two independent experiments. (g) H&E-stained paraffin-embedded sections containing pyogranulomas from WT and Ccr2gfp/gfp mice at day 3 post-infection. Dashed circle denotes area containing pyogranuloma. Scale bars = 100 μm. Representative images of one experiment. All statistical analyses by Mann-Whitney U test (two-tailed). * (p < 0.05), ** (p < 0.01), *** (p < 0.001), **** (p < 0.0001), NS (not significant, p > 0.05).

Extended Data Fig. 4 Effects of monocyte deficiency on other cell types.

(a) Total numbers and frequencies of CD4+ T cells, CD8+ T cells, B cells, and DCs in small intestinal PG + tissue. Each data point indicates the mean of 3-10 pooled punch biopsies from one mouse (n = 20-22). Lines represent median. Pooled data from three to five independent experiments. (b) Frequency of neutrophils in MLN and spleen at day 5 post infection. Each data point represents one mouse (n = 8-12). Lines represent median. Pooled data from two independent experiments. (c) Neutrophil surface CD11b expression in MLN at day 5 post-infection was measured by flow cytometry. Each data point represents one mouse (n = 6). Lines represent median. Representative of two independent experiments. (d) Intracellular levels of CD11b in neutrophils in small intestinal PG + tissue at day 5 post-infection were measured by flow cytometry. Each data point represents the mean of 3-10 pooled punch biopsies from one mouse (n = 6-7). Lines represent median. Representative of four independent experiments. (e) Cytokine levels in homogenates of tissue punch biopsies at day 5 post infection were measured by cytometric bead array. Each data point represents the mean of 3-10 pooled punch biopsies from one mouse (n = 18-22). Lines represent median. Pooled data from three independent experiments. (f) Intracellular levels of cytokines and lipocalin in neutrophils in small intestinal PG + tissue at day 5 post-infection were measured by flow cytometry. Each data point represents the mean of 3-10 pooled punch biopsies from one mouse (n = 18-19). Lines represent median. Pooled data from three independent experiments. All statistical analyses by Mann-Whitney U test (two-tailed). * (p < 0.05), ** (p < 0.01), *** (p < 0.001), **** (p < 0.0001), NS (not significant, p > 0.05).

Extended Data Fig. 5 CCR2-deficient mice cannot control systemic Yersinia.

(a) Bacterial burdens in indicated organs at day 3 post infection. Each data point represents one mouse (n = 10-14 for PP, MLN, lung; 21-25 for spleen, liver). Lines represent geometric mean. Pooled data from two to four independent experiments. (b) Bacterial burdens in indicated organs at day 5 post infection. Each data point represents one mouse (n = 29-30). Lines represent geometric mean. Pooled data from four independent experiments. (c) Bacterial burdens in indicated organs at day 3 post infection. Each data point represents one mouse (n = 10-13). Lines represent geometric mean. Pooled data from two independent experiments. (d) Survival of infected mice. Pooled data from two independent experiments. Statistical analyses by (a-c) Mann-Whitney U test (two-tailed) or (d) Mantel-Cox test. * (p < 0.05), ** (p < 0.01), *** (p < 0.001), **** (p < 0.0001), NS (not significant, p > 0.05).

Extended Data Fig. 6 Yersinia virulence factors induce intestinal pyogranulomas.

(a) Cumulative bacterial burdens in PG- tissue at day 5 post infection. Each symbol represents one mouse (n = 9-11). Lines represent geometric mean. Dotted line represents limit of detection. Pooled from two independent experiments. (b) Frequency of monocytes in small-intestinal tissue and MLN. Each symbol represents one mouse (n = 3-9). Lines represent median. Pooled and representative data from two independent experiments. (c) Frequency of neutrophils in small-intestinal tissue and MLN. Each symbol represents one mouse (n = 3-9). Lines represent median. Pooled and representative data from two independent experiments. (d) Bacterial burdens in indicated organs at day 5 post-infection. Each symbol represents one mouse (n = 10-43). Lines represent geometric mean. Pooled from 2-6 independent experiments. (e) Total number of intestinal lesions at day 5 post infection. Each symbol represents one mouse (n = 8-10). Lines represent median. Pooled from two independent experiments. (f) Total number of intestinal lesions at day 5 post infection. Each symbol represents one mouse (n = 11). Lines represent median. Pooled from three independent experiments. (g) Frequency and total number of monocytes in small-intestinal PG + tissue at day 5 post WT or YopHR409A Yp infection. Each data point represents one mouse (n = 8-10). Lines represent median. Pooled from two independent experiments. (h) Bacterial burdens in indicated organs at day 3 post WT or YopHR409A Yp infection. Each symbol represents one mouse (n = 10-13). Lines represent geometric mean. Pooled from two independent experiments. Statistical analyses by (a, d, e) Kruskal–Wallis test with Dunn’s post-test and (b, c, f, g, h) Mann-Whitney U test (two-tailed). * (p < 0.05), ** (p < 0.01), *** (p < 0.001), **** (p < 0.0001), NS (not significant, p > 0.05).

Extended Data Fig. 7 Anti-Gr-1 effectively depletes neutrophils during infection.

(a) Flow cytometry plots displaying the gating strategy employed to identify neutrophils and monocytes upon anti-Gr-1 administration. Due to masking of Ly-6G and Ly-6C epitopes by anti-Gr-1, monocytes were identified as CCR2-GFP+ cells (green box) and neutrophils were identified as SSC high cells (pink boxes). Representative images of three independent experiments. (b) Frequency of monocytes in blood at day 5 post infection was determined by flow cytometry. Each symbol represents one mouse (n = 10-11). Lines represent median. Data from three independent experiments. Statistical analyses by Kruskal-Wallis test with Dunn’s post-test. * (p < 0.05), ** (p < 0.01), *** (p < 0.001), **** (p < 0.0001), NS (not significant, p > 0.05).

Supplementary information

Reporting Summary

Supplementary Tables 1 and 2

Table 1. Gene Ontology analysis of PG+ versus PG− samples. Table 2. Gene Ontology analysis of PG− versus uninfected samples.

Supplementary Code File 1

Code for RNA sequencing analysis.

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

Sorobetea, D., Matsuda, R., Peterson, S.T. et al. Inflammatory monocytes promote granuloma control of Yersinia infection. Nat Microbiol 8, 666–678 (2023). https://doi.org/10.1038/s41564-023-01338-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-023-01338-6

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