Abstract
Granulomas are organized immune cell aggregates formed in response to chronic infection or antigen persistence. The bacterial pathogen Yersinia pseudotuberculosis (Yp) blocks innate inflammatory signaling and immune defense, inducing neutrophil-rich pyogranulomas within lymphoid tissues. Here, we uncover that Yp also triggers pyogranuloma formation within the murine intestinal mucosa. Mice lacking circulating monocytes fail to form defined pyogranulomas, 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 pyogranulomas, indicating that intestinal pyogranulomas form in response to Yp disruption of cytoskeletal dynamics. Notably, mutation of the virulence factor YopH restores pyogranuloma formation and control of Yp in mice lacking circulating monocytes, demonstrating that monocytes override YopH-dependent blockade of innate immune defense. This work reveals an unappreciated site of Yersinia intestinal invasion and defines host and pathogen drivers of intestinal granuloma formation.
Microbial pathogens utilize diverse mechanisms to subvert host immunity in order to replicate and spread to new hosts. While acute infections can be cleared rapidly by the immune system, some pathogens evade immune defenses to cause chronic disease. Chronic infections often result in formation of structures termed granulomas that limit pathogen dissemination and tissue damage1. Granulomas are characterized by the presence of activated phagocytes, notably monocytes and macrophages, and form in response to a wide variety of infections2. Monocytes are rapidly recruited to infected tissues where they produce inflammatory cytokines and anti-microbial effector molecules, contributing to defense against multiple pathogens3–6. Some pathogens, however, exploit monocytes as a means of dissemination, including Salmonella enterica, Yersinia pestis, and Mycobacterium species7–9. The pathogen-specific signals that induce granuloma formation remain poorly defined.
Enteropathogenic Yersinia, including Y. pseudotuberculosis (Yp) and Y. enterocolitica (Ye), cause self-limiting gastroenteritis and mesenteric lymphadenopathy following enteric infection10,11. In immune-compromised patients, however, bacteria disseminate and cause a systemic plague-like disease, indicating that the intestinal immune system is critical for control of acute infection. Indeed, the intestine constitutes a bottleneck against Yersinia dissemination, as bacteria in systemic organs are thought to originate predominantly from the intestinal lumen rather than gut-associated lymphoid tissues12. A hallmark of Yersinia infections is the presence of chronic pyogranulomas (PG) in lymphoid tissue, characterized by nodular infiltrates of activated monocytes and macrophages surrounding a core of activated neutrophils13. Notably, the contribution of monocytes to pyogranuloma formation and their role in Yersinia restriction are unclear14,15.
Here we report that pyogranulomas form acutely in the murine intestinal mucosa during enteric Yersinia infection. PG are enriched in neutrophils and inflammatory monocytes, and contain live bacteria at levels comparable to Peyer’s patches (PP). Notably, CCR2-deficient mice, which lack circulating inflammatory monocytes16,17, form disorganized necrosuppurative lesions rather than defined PG, are unable to contain bacteria within the lesions, and succumb rapidly to infection. Moreover, mice lacking circulating monocytes exhibit reduced levels of IL-1 cytokines and surface expression of the neutrophil activation marker CD11b within intestinal pyogranulomas. Yp lacking either the virulence plasmid encoding the type III secreted Yersinia Outer Proteins (Yops), or lacking Yops that block phagocytosis and the reactive oxygen burst, do not induce detectable pyogranulomas, indicating that pyogranulomas are induced in response to Yersinia blockade of innate immune defense. Notably, CCR2-deficient mice infected with bacteria lacking the virulence factor YopH, which blocks actin cytoskeleton dynamics, were able to restrict bacterial burdens and form defined granulomatous lesions, accompanied by restored neutrophil CD11b surface expression. Neutrophil depletion in CCR2-deficient animals abrogates control of YopH-mutant Yersinia, demonstrating that inflammatory monocytes overcome YopH-mediated disruption of neutrophil function. Altogether, our study identifies an unappreciated site of Yersinia colonization within the murine intestinal mucosa, and reveals an essential function for inflammatory monocytes in maintainance of PG architecture during Yp infection.
Results
Intestinal pyogranulomas form upon oral Yersinia infection
Yersinia pseudotuberculosis colonizes gut-associated lymphoid tissues, resulting in acute pyogranuloma (PG) formation following oral infection18. Interactions between Yersinia and immune cells within systemic tissues have been extensively documented19–21. In our efforts to dissect intestinal immune responses to Yp, we observed numerous macroscopically visible nodular lesions in the gastrointestinal tract five days post-infection, which appeared as punctate areas of increased opacity (Fig. 1a). Lesions were most prevalent in the jejunum and ileum, ranged in number from two to over forty in individual mice, and also formed in response to Yersinia enterocolitica infection (Fig. 1b, c). Histology of Yp-infected intestines revealed focal inflammation characterized by crypt hyperplasia, edema, and submucosal to transmural cellular infiltration, whereas non-lesional areas of infected intestines appeared largely unaffected (Fig. 1d). The lesions contained infiltrates of macrophages and neutrophils surrounding colonies of coccobacilli, similar to structures that we and others observed in lymphoid tissues and have termed pyogranulomas (Fig. 1e)15,18,22. Consistently, flow-cytometric analysis of intestinal punch biopsies containing pyogranulomas (PG+), adjacent non-granulomatous tissue (PG−), and uninfected control tissue (uninf) revealed that neutrophils were the most enriched cell type within PG+ tissue, followed by macrophages and inflammatory monocytes (Fig. 1f, g). We also observed increases in eosinophils, dendritic cells, and CD4+ T cells in PG+ tissue, although the relative frequencies of these populations were decreased, due to even larger increases in neutrophils, monocytes, and macrophages (Extended Data Fig. 1a, b).
Figure 1. Intestinal pyogranulomas form upon oral Yersinia infection.
(a) Small-intestinal segments from uninfected and Yp-infected mice with arrows depicting lesions, and a magnified lesion with dotted circle depicting size of punch biopsies. Scale bars = 3 mm (left) and 0.5 mm (right). Representative of >3 independent experiments.
(b) Frequency distribution of lesions along the intestine, with graphical key of anatomical segments. Each colored bar represents the mean frequency of lesions in a given segment (n = 20 mice). Only mice with >9 total lesions (>80% of mice) were included. Pooled from three independent experiments.
(c) Quantification of total intestinal lesions at day 5 post-infection with Yp or Ye. Each circle represents one mouse (n = 16-17). Lines represent median. Pooled from three independent experiments.
(d) H&E-stained paraffin-embedded longitudinal small-intestinal sections from uninf and Yp-infected mice. Scale bars = 200 μm. a = lesion, b = crypt hyperplasia, c = submucosal inflammation, d = edema. Representative of three independent experiments.
(e) H&E-stained paraffin-embedded small-intestinal pyogranuloma depicting an encircled bacterial colony. Scale bar = 10 μm. Representative of three independent experiments.
(f) Flow-cytometry plots identifying CD11b+Ly-6G+ neutrophils, CD11b+CD64+Ly-6C+ monocytes, and CD11b+CD64+Ly-6C−MHC-II+ macrophages in small-intestinal tissue. Representative of four independent experiments.
(g) Frequency and total number of neutrophils, monocytes and macrophages in small-intestinal tissue. Each circle represents one mouse (n = 15-19). Lines represent median. Pooled from four independent experiments.
(h) Fluorescently labeled small-intestinal pyogranuloma from a Ccr2gfp/+ mouse. White (Yp-mCherry), magenta (Ly-6G-AF647), green (CCR2-GFP). Scale bars = 100 μm. Representative of two independent experiments.
(i) Bacterial burdens in small-intestinal tissue. Each circle represents one mouse (n = 24-25). Lines represent geometric mean. Dotted line represents detection limit. Pooled from four independent experiments.
(j) Cumulative bacterial burdens in PG+ tissue and Peyer’s patches. Each circle represents one mouse (n = 29-30). Lines represent geometric mean. Pooled from five independent experiments.
Wilcoxon test (two-tailed) was performed for paired analyses (PG− vs PG+). Mann-Whitney U test (two-tailed) was performed for remaining statistical analyses. * (p < 0.05), ** (p < 0.01), **** (p < 0.0001), NS (not significant, p > 0.05), ND (not detected).
In lymphoid tissues Yp pyogranulomas consist of a central bacterial colony, surrounded by neutrophils, which are bordered in turn by monocytes and macrophages15,22. Confocal microscopy demonstrated that intestinal PG also contained a central Yp colony surrounded by a dense population of Ly-6G+ neutrophils (Fig. 1h). Interestingly, in contrast to lymphoid tissues, CCR2+ monocytes and macrophages formed a mesh-like network of cells that both overlapped with and bordered the neutrophils (Fig. 1h). Consistent with the presence of bacterial colonies detected by histology and fluorescence microscopy, PG+ tissue harbored high numbers of viable bacteria comparable to that found in the Peyer’s patches (Fig. 1i, j). Since PP are a major entry point and replicative niche for enteropathogenic Yersinia following oral innoculation23,24, altogether, these findings reveal intestinal pyogranulomas as a previously unappreciated location of Yersinia invasion within the intestinal mucosa and a potential site of bacterial restriction or dissemination.
Intestinal inflammation is spatially restricted to PGs
To test whether PGs exhibit location-specific inflammatory responses, we next performed RNA sequencing of pyogranulomas, adjacent non-pyogranuloma tissue, and uninfected tissue. Principal component analysis showed distinct clustering by sample type (Fig. 2a), and comparison of PG+ and PG− samples revealed 355 upregulated and 363 downregulated genes (Fig. 2b). Top upregulated genes included granulocyte- and monocyte-recruiting chemokines (Cxcl1, Cxcl2, Cxcl3, Cxcl5), pro-inflammatory cytokines (Il1b, Il22), metal-sequestration proteins (S100a8, S100a9), and matrix metalloproteases (Mmp3) (Fig. 2c). Gene-ontology analysis indicated that chemotaxis of myeloid cells and defense against bacterial pathogens predominated the top upregulated responses within PG+ biopsies (Fig. 2d, Extended Data Table 1). These responses were strikingly similar to previously reported Yp-infected PP25. Consistently, gene set-enrichment analysis indicated that a set of 50 upregulated genes previously reported in Yp-infected PP were also enriched in PG+ samples (Fig. 2e). Furthermore, protein levels of the pro-inflammatory cytokines IL-1α, IL-1β, IL-6, TNF, and CCL2 were significantly elevated within PG+ biopsies (Fig. 2f). Consistent with histology and microscopy, the inflammatory transcriptional response was localized to PG+ tissue, as PG− samples did not exhibit enrichment of genes or ontology terms related to myeloid cell migration or innate immune-cell activation (Extended Data Fig. 2, Extended Data Table 2). Likewise, production of pro-inflammatory cytokines was not detected in PG- tissue (Fig. 2e). Altogether, these data indicate that the pro-inflammatory response to Yp infection in the gut mucosa is spatially restricted to PG.
Figure 2. Intestinal inflammation is spatially restricted to PGs.
(a) Principal component analysis of PG+ (pink), PG− (green), and uninf (gray) samples at day 5 post-infection. Five biopsies were pooled per mouse.
(b) Heatmap of all differentially expressed genes in PG+ compared to PG− samples. False discovery rate < 0.05 using Benjamini-Hochberg procedure. Pink and orange bars denote two clusters grouped based on Pearson correlation.
(c) Heatmap of top 30 significantly upregulated genes in PG+ compared to PG− samples in descending order by fold change. False discovery rate < 0.05 using Benjamini-Hochberg procedure.
(d) Gene ontology analysis of top 30 upregulated genes by fold change only in PG+ compared to PG− samples.
(e) Gene set enrichment analysis (GSEA) of top 50 upregulated genes in Yp-infected Peyer’s patches. NES = normalized enrichment score; FDR = false discovery rate.
(f) Cytokine levels in homogenates of tissue punch biopsies at day 5 post-infection. Lines represent group mean. Each circle represents one mouse (n = 6-9). Statistical analysis 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). Data from one or two pooled independent experiments.
Inflammatory monocytes maintain PGs to restrict infection
Inflammatory monocytes promote host defense by differentiating into phagocytes and antigen presenting cells4,6,26,27, producing pro-inflammatory mediators3, and modulating other immune cell functions5,28. Monocyte-derived cells can also promote pathogen replication or dissemination to new sites7,29. Mice lacking the chemokine receptor CCR2 have a ten-fold reduction in circulating monocytes due to defective bone marrow-egress16,17. Monocytes are rapidly recruited to intestinal PGs (Fig. 1) and sites of systemic Yp infection15,18,22, raising the question of their role in Yp infection. CCR2-deficiency has been associated with more rapid bacterial clearance from the MLN following enteric infection15, but increased susceptibility to intravenous Yp infection14. Interestingly, we found that while Ccr2gfp/gfp mice, which lack circulating mononcytes due to lack of CCR2 expression30,31, had similar overall numbers of macroscopic intestinal lesions as WT mice (Fig. 3a, Extended Data Fig. 3a), their lesions had a disorganized appearance, and exhibited central caseation with tissue necrosis (Fig. 3b). In contrast to WT PG which exhibited robust inflammatory infiltrates and a defined cellular organization encapsulating central bacterial colonies, Ccr2gfp/gfp intestinal lesions contained expanded coalescing bacterial colonies with limited immune cell recruitment (Fig. 3b, c). PP of Ccr2gfp/gfp mice had similarly disorganized lesions with central tissue necrosis, suggesting that monocytes are required to establish or maintain organized PGs during Yp infection (Extended Data Fig. 3b). Two independent CCR2-deficient mouse lines showed significantly higher bacterial burdens in PG+ and PG− tissues compared to WT mice (Fig. 3d, Extended Data Fig. 3c). Moreover, acute depletion of monocytes in WT mice with anti-CCR2 specific antibodies resulted in increased PG− and PG+ bacterial burden (Extended Data Fig. 3d, e), demonstrating that the requirement for monocytes in control of Yp is not due to developmental defects in CCR2-deficient mice. Interestingly, at day 3 post-infection, Ccr2gfp/gfp mice had elevated bacterial burdens in PG− tissue but not PG+ tissue and did not exhibit overt signs of tissue necrosis in PGs (Extended Data Fig. 3f, g), indicating that the defect in bacterial control and PG architecture develops between day 3 and 5. Together, these data suggest that monocytes enable maintenance of PG architecture and restrict Yp within intestinal PG.
Figure 3. Inflammatory monocytes maintain PGs to restrict infection.
(a) Quantification of total intestinal lesions at day 5 post-infection. Each circle represents one mouse (n = 20). Lines represent median. Pooled data from three independent experiments.
(b) H&E-stained small intestinal sections from Yp-infected mice at day 5 post-infection. 1 denotes the Yersinia microcolony, 2 denotes necrotic tissue. Scale bars = 250 μm (top) and 50 μm (bottom). Representative images of two independent experiments.
(c) Histopathological scores of small intestinal tissue at day 5 post-infection. Each mouse was given a score between 0-4 (healthy-severe) for presence of bacterial colonies free from immune cell infiltrate. Each circle represents one mouse (n = 7-9 for uninfected, 9-16 for Yp). Lines represent median. Pooled data from two independent experiments.
(d) Bacterial burdens in PG− and PG+ tissue at day 5 post-infection. Each circle represents the mean Yp-CFU of 3-5 pooled punch biopsies from one mouse (n = 31-37). Lines represent geometric mean. Pooled data from six independent experiments.
(e) Total numbers and frequencies of indicated cells in small intestinal uninf, PG−, and PG+ tissue at day 5 post-infection. Each circle represents the mean of 3-10 pooled punch biopsies from one mouse (n = 24). Lines represent median. Pooled data from five independent experiments.
(f) Fluorescently labeled PG+ tissue from Yp-infected Ccr2gfp/+ (top) and Ccr2gfp/gfp (bottom) mice at day 5 post-infection. Scale bars = 100 μm. Representative images of two independent experiments.
(g) PG+ neutrophil surface CD11b expression at day 5 post-infection. Each circle represents the mean of 3-10 pooled punch biopsies from one mouse (n = 6). Lines represent median. Representative of four independent experiments.
(h) Cytokine levels in homogenates of tissue punch biopsies were measured by cytometric bead array at day 5 post-infection. Each circle represents the mean of 3-10 pooled punch biopsies from one mouse (n = 18-21). Lines represent median. Pooled data from three independent experiments.
Mann-Whitney U test (two-tailed) was performed for all statistical analyses. * (p < 0.05), ** (p < 0.01), *** (p < 0.001), **** (p < 0.0001), NS (not significant, p > 0.05)
Consistent with their reduced overall cellularity, Ccr2gfp/gfp lesions exhibited decreased frequency and numbers of viable CD45+ hematopoietic cells compared to WT PG (Fig. 3e). Consistent with the important role of CCR2 in promoting monocyte egress from bone marrow16,17, Ccr2gfp/gfp intestinal lesions contained significantly lower numbers of monocytes, macrophages, and dendritic cells compared with PG from WT mice (Fig. 3e, Extended Data Fig. 4a). Notably, CCR2 deficiency did not impact T or B cell numbers in intestinal PGs, indicating that the defect in enteric control of Yp in CCR2-deficient mice was independent of adaptive immune cells (Extended Data Fig. 4a). Ccr2gfp/gfp intestinal lesions also showed a trend toward reduced neutrophil numbers (Fig. 3e), suggesting that monocytes promote recruitment, retention, or survival of neutrophils within intestinal PG during Yp infection. This defect was specific to the intestinal lesions, as we observed similar frequencies of neutrophils in the MLN and spleen (Extended Data Fig. 4b). Immunofluorescence microscopy of the lesions indicated that neutrophils were unable to effectively contain Yp in the absence of monocytes, as the bacterial colony expanded outside the range of the neutrophil marker Ly-6G. In contrast, Yp was fully encapsulated by neutrophils and CCR2+ cells within WT PGs (Fig. 3f).
Intriguingly, surface expression of the integrin CD11b, a well-established marker of neutrophil activation32–34, was significantly reduced in both PGs and MLN of Ccr2gfp/gfp mice compared to WT counterparts (Fig. 3g, Extended Data Fig. 4c), suggesting a defect in neutrophil activation in the absence of monocytes. CD11b is present on the neutrophil cell surface and membranes of intracellular granules, and increased CD11b surface expression occurs in numerous inflammatory settings35–37. Notably, PG neutrophils in CCR2-deficient mice exhibited increased intracellular CD11b levels, suggesting that translocation of CD11b from intracellular granules to the cell surface is defective in the absence of monocytes (Extended Data Fig. 4d). Interestingly, total IL-1α and IL-1β levels were significantly reduced in Ccr2gfp/gfp PGs, whereas other pro-inflammatory cytokines were unaffected (Fig. 3h, Extended Data Fig. 4e), indicating that monocytes or monocyte-derived cells specifically produce IL-1, or specifically promote IL-1 production by other cells within intestinal PGs. Notably, intracellular levels of IL-1 cytokines, TNF, and lipocalin were unaffected in PG neutrophils in Ccr2gfp/gfp mice (Extended Data Fig. 4f), suggesting that inflammatory monocytes do not regulate neutrophil-intrinsic inflammatory cytokine and antimicrobial protein production. Altogether, these results demonstrate that inflammatory monocytes or monocyte-derived cells promote maintenance of functional granulomas that limit intestinal bacterial replication and dissemination.
Inflammatory monocytes control systemic Yersinia
Following systemic dissemination, Yp colonizes and induces PGs in lymphoid tissues15,18,22. Critically, both CCR2-deficient and anti-CCR2 depleted mice had significantly higher Yp burdens in the MLN and systemic organs (Fig. 4a, Extended Data Fig. 5a, 5b). Similar to intestinal tissue, systemic bacterial burdens in Ccr2gfp/gfp mice were unaffected at day 3 post-infection, indicating that this defect in control develops between day 3 and 5 (Extended Data Fig. 5c). Consistent with our findings that monocytes were required for maintenance of intestinal pyogranuloma architecture, infected Ccr2gfp/gfp spleens exhibited widespread tissue necrosis, free bacterial colonies, and sparse immune cell recruitment, in contrast to WT spleens where neutrophils and monocytes effectively encapsulated Yp microcolonies within organized PG (Fig. 4b, c). Notably, mice lacking CCR2 succumbed rapidly to acute infection (Fig. 4d, Extended Data Fig. 5d). Importantly, co-housed littermate Ccr2+/+ and Ccr2gfp/+ mice were equally resistant to Yp infection while Ccr2gfp/gfp littermates succumbed (Fig. 4e), indicating that increased susceptibility of Ccr2gfp/gfp to Yp infection is not due to differences in composition of a vertically-transmitted intestinal microbiota. Collectively, our findings demonstrate that inflammatory monocytes maintain organized pyogranulomas in host tissues, thereby limiting tissue necrosis and systemic bacterial dissemination, ultimately enabling bacterial control and host survival following oral Yp infection.
Figure 4. Inflammatory monocytes control systemic Yersinia.
(a) Bacterial burdens in indicated organs at day 5 post-infection. Each circle represents one mouse (n = 20-25). Lines represent geometric mean. Pooled data from four independent experiments.
(b) H&E-stained paraffin-embedded longitudinal spleen sections from WT and Ccr2gfp/gfp mice at day 5 post-infection. Dashed circle denotes area with bacterial microcolonies and neutrophils. Scale bars = 500 μm (top) and 50 μm (top). Representative images of two independent experiments.
(c) Histopathological scores of spleens from uninfected and Yp-infected mice at day 5 post-infection. Each mouse was given a score between 0-4 (healthy-severe) for presence of bacterial colonies free from immune cell infiltrate. Each circle represents one mouse (n = 9 for uninfected, 16 for Yp). Lines represent median. Pooled data from two independent experiments.
(d) Survival of infected WT (n=16) and Ccr2gfp/gfp (n=13) mice. Pooled data from two independent experiments.
(e) Survival of infected littermate wild-type Ccr2+/+ (n=20), heterozygous Ccr2gfp/+ (n=19), and homozygous Ccr2gfp/gfp (n=15) mice. Pooled data from three independent experiments.
Statistical analyses by (a, c) Mann-Whitney U test (two-tailed) and (d, e) Mantel-Cox test. * (p < 0.05), ** (p < 0.01), *** (p < 0.001), **** (p < 0.0001), NS (not significant, p > 0.05).
Yersinia virulence factors induce intestinal pyogranulomas
Yersinia inject Yersinia Outer Proteins (Yops), which are encoded on a virulence plasmid (pYV), into target cells38 to block phagocytosis, production of reactive oxygen species (ROS), and degranulation20. Interestingly, Yp lacking the virulence plasmid (pYV-) still colonize the intestine during the acute phase of infection without causing disease39. Since granulomas form in response to pathogens that thwart immune defenses, we hypothesized that intestinal pyogranulomas may be triggered by the activity of Yp effector proteins. Indeed, even at a ten-fold higher infectious dose, we did not observe intestinal lesions at day 5 post-infection with pYV- bacteria (Fig. 5a), despite detectable (although reduced) intestinal colonization (Extended Data Fig. 6a).
Figure 5. Yersinia virulence factors induce intestinal pyogranulomas.
(a) Quantification of total intestinal lesions at day 5 post-Yp infection. Each symbol represents one mouse (n = 10-43). Lines represent median. Pooled from 2-6 independent experiments.
(b) H&E-stained paraffin-embedded transverse small intestinal sections from WT and Ccr2gfp/gfp mice infected with WT or YopHR409A Yp at day 5 post infection depicting encircled bacterial colonies. Scale bars = 100 μm. Representative of two independent experiments.
(c) Frequency and total number of neutrophils in small-intestinal PG+ tissue at day 5 post WT or YopHR409A Yp infection. Each circle represents one mouse (n = 12-14). Lines represent median. Pooled from three independent experiments.
(d) Mean fluorescent intensity (MFI) of surface CD11b expression on neutrophils in PG+ tissue at day 5 post WT or YopHR409A Yp infection. Each symbol represents one mouse (n = 7-10). Lines represent median. Pooled from two independent experiments.
(e) Bacterial burdens in small-intestinal PG− and PG+ tissue at day 5 post WT or YopHR409A Yp infection. Each symbol represents one mouse (n = 25-26). Lines represent geometric mean. Pooled from four independent experiments.
(f) Bacterial burdens in indicated organs at day 5 post WT or YopHR409A Yp infection. Each symbol represents one mouse (n = 15-22). Lines represent geometric mean. Pooled from four independent experiments.
(g) Survival of WT mice infected with WT (n = 20) or YopHR409A (n = 20) Yp and Ccr2−/− mice infected with WT (n = 10) or YopHR409A (n = 17) Yp Pooled from two independent experiments.
Statistical analyses by (a, c, d, e, f) Kruskal-Wallis test with Dunn’s post-test, and (g) Mantel Cox test. * (p < 0.05), ** (p < 0.01), *** (p < 0.001), **** (p < 0.0001), NS (not significant, p > 0.05).
pYV- bacteria still induced monocyte recruitment to both the intestinal mucosa and MLN, indicating that lack of detectable PG was not due to an absence of immune infiltration (Extended Data Fig. 6b). However, neutrophil accumulation was absent upon pYV- infection, demonstrating that neutrophil recruitment occurs in response to Yp virulence (Extended Data Fig. 6c). Although comparable in the intestinal mucosa, pYV- bacterial burdens were reduced in other gut-associated and systemic lymphoid tissues (Extended Data Fig. 6d), illustrating that intestinal PG formation is dispensable for control of pYV- Yersinia.
Intestinal lesions formed at WT levels in mice infected with Yp individually deficient in either YopM or YopJ enzymatic activity, which block pyrin inflammasome assembly or NF-κB and MAPK signaling40–42, respectively, suggesting that neither YopM nor YopJ are singly required for pyogranuloma formation (Extended Data Fig. 6e). Several Yops function together to disrupt the actin cytoskeleton, thereby blocking phagocytosis and the reactive oxygen burst20. These Yops (E, H, and T), have partially overlapping functions and can compensate for one another in certain settings20. Notably, Yp with combined point mutations in each of the catalytic residues of these Yops (YopER144A, YopTC139A, and YopHR409A) did not induce intestinal lesions and had burdens similar to pYV- infection (Fig. 5a and Extended Data Fig. 6a, d). In contrast, bacteria lacking YopE and YopT, or YopE alone, induced WT numbers of intestinal lesions (Fig. 5a) and were only attenuated in systemic organs (Extended Data Fig. 6d), indicating that YopH is sufficient, in the absence of YopE and YopT, to induce pyogranuloma formation. Interestingly, YopHR409A mutants that lack YopH tyrosine phosphatase activity induced fewer pyogranulomas than wild-type bacteria (Fig. 5a), suggesting that YopH is sufficient and partially responsible for pyogranuloma formation. In addition, MLN colonization was abrogated in the absence of YopH activity, whereas YopE was dispensable for MLN colonization (Extended Data Fig. 6d). Intriguingly, bacteria lacking both YopE and YopH (yopEH) induced no detectable lesions (Fig. 5a) and showed similar levels of colonization to yopETH bacteria (Extended Data Fig. 6a, d), suggesting that the host response to YopE and YopH drives pyogranuloma formation.
YopH blocks innate cell phagocytosis and ROS production20,43–46. YopH-deficient Yp are therefore more susceptible to neutrophil killing in vitro and are attenuated in vivo47–49. Consequently, neutrophil depletion increases susceptibility to Yp and restores virulence to YopH-mutant bacteria45,46, indicating that neutrophil defenses are an important target of YopH in vivo. However, whether monocytes combat YopH-dependent blockade of neutrophil function is unknown. As in WT mice, we found that YopH-mutant bacteria induced significantly fewer intestinal lesions in Ccr2gfp/gfp mice than WT bacteria (Extended Data Fig. 6f). Strikingly, despite the lack of monocytes, YopHR409A infection of CCR2-deficient mice restored neutrophil recruitment and induced neutrophil-rich lesions without necrosis (Fig. 5b, c, Extended Data Fig. 6g). Intriguingly, CD11b surface expression was restored in PG+ tissue of CCR2-deficient animals infected with YopHR409A bacteria, indicating that YopH limits neutrophil activation in the absence of monocytes (Fig. 5d). Furthermore, CCR2-deficient mice effectively controlled bacterial burdens in intestinal and systemic tissues following infection with YopHR409A bacteria (Fig. 5e, f and Extended Data Fig. 6h). Consistently, while CCR2-deficient mice rapidly succumbed to WT Yp, they survived infection with YopHR409A bacteria at levels similarl to WT mice infected with WT Yp, suggesting that monocytes control Yersinia infection by overcoming the virulence of YopH (Fig. 5g). Altogether, these data indicate that intestinal pyogranulomas form in response to Yersinia disruption of innate immune defense, and that monocytes counteract YopH activity in vivo.
Neutrophils control YopH-deficient Yersinia in absence of monocytes
While monocytes are dispensable for control of YopH-deficient Yp, the cell type that mediates this control in the absence of monocytes is unknown. Neutrophil recruitment to PGs was restored during infection of Ccr2gfp/gfp mice with YopH-deficient Yp, raising the possibility that neutrophils mediate control of Yersinia in the absence of monocytes. Since Ccr2gfp/gfp mice largely lack circulating monocytes, anti-Gr-1, which depletes both neutrophils and monocytes and is highly efficient at depleting neutrophils in infectious or inflammatory settings50,51, allowed us to interrogate the role of neutrophils in control of YopH-mutant Yp (Fig. 6a, Extended Data Fig. 7a). Strikingly, Ccr2gfp/gfp mice injected with anti-Gr-1 had elevated burdens of YopH-mutant Yp in PG− tissue (Fig. 6b). Anti-Gr-1 did not further reduce blood monocyte frequencies in Ccr2gfp/gfp mice (Extended Data Fig. 7b), indicating that increased susceptibility of these mice was not attributable to depletion of remaining Ly-6C+ monocytes. CCR2-deficient mice infected with YopHR409A bacteria had very few detectable PG (Fig. 6c), making it difficult to robustly analyze bacterial burdens in this tissue. Intriguingly, Ccr2gfp/gfp mice injected with anti-Gr-1 had elevated numbers of macroscopic intestinal lesions during infection with YopHR409A bacteria (Fig. 6c). Moreover, in peripheral organs, Ccr2gfp/gfp mice injected with anti-Gr-1 had elevated bacterial burdens following YopH-mutant infection (Fig. 6d), demonstrating that in the absence of monocytes, neutrophils play a key role in control of YopH-deficient bacteria. Notably, mice infected with WT bacteria did not exhibit increased systemic bacterial burdens upon neutrophil depletion (Fig. 6d), consistent with YopH blockade of neutrophil functions. Collectively, these data indicate that neutrophils control YopH-deficient Yp in the absence of monocytes.
Figure 6. Neutrophils control YopH-deficient Yersinia in absence of monocytes.
(a) Frequency of neutrophils in blood at day 5 post infection was determined by flow cytometry. Each symbol represents one mouse (n = 10-11). Lines represent median. Pooled data from three independent experiments.
(b) Bacterial burdens in small-intestinal PG− tissue at day 5 post WT or YopHR409A Yp infection. Each symbol represents one mouse (n = 10-12). Lines represent geometric mean. Pooled data from three independent experiments.
(c) Quantification of total number of intestinal lesions at day 5 post infection. Each symbol represents one mouse (n = 10-12). Lines represent median. Pooled data from three independent experiments.
(d) Bacterial burdens in indicated organs at day 5 post WT or YopHR409A Yp infection. Each symbol represents one mouse (n = 10-12). Lines represent geometric mean. Pooled data from three independent experiments.
All 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).
Discussion
Granulomas are a conserved response to persistent or long-term infectious and non-infectious stimuli1,2. The natural rodent and human pathogen Yersinia pseudotuberculosis induces formation of neutrophil-rich pyogranulomas (PG) in infected lymphoid tissues11,13. Here, we report that PG form throughout the intestine during murine enteropathogenic Yersinia infection. Peyer’s patches are considered the primary site of Yersinia intestinal infection. However, intestinal pyogranulomas harbored a similar total bacterial burden as the PPs, suggesting that PGs are a previously unrecognized niche for enteropathogenic Yersinia intestinal colonization. Notably, wild-type mice almost entirely restricted intestinal Yp and inflammation to these granulomatous foci. The transcriptional profile of PGs was similar to Yp-infected Peyer’s patches25, indicating a shared response to intestinal Yp infection driven by recruitment and activation of innate immune cells, which may serve to limit bacterial spread and tissue damage within the gut mucosa.
While initial formation of PGs was similar in CCR2-deficient and WT mice, monocytes were critical for maintaining architectural integrity of PGs and bacterial control in the intestine and deeper tissues during Yersinia infection, as monocyte deficiency was associated with widespread tissue necrosis and elevated bacterial burdens in intestinal and systemic tissues that occurred between day 3 and day 5. Interestingly, Y. pestis, which causes more severe disease than Y. pseudotuberculosis, is commonly associated with necrotic and necrosuppurative lesions52, suggesting that Y. pestis has evolved to overcome monocyte-mediated host defense and pyogranuloma formation in order to enhance systemic dissemination and transmission.
Monocytes were previously reported to be dispensable for acute control of Yp following oral inoculation15. These studies employed a Ccr2−/− mouse line17,27 originally generated on the 129 mouse background and backcrossed to C57BL/616, raising the possibility that distinct polymorphisms in this mouse line might account for this difference. Ccr2gfp/gfp mice were generated directly on the C57BL/6J background30, making it unlikely that our findings are due to immunologically impactful polymorphisms that co-segregate with the Ccr2 locus. Infection of co-housed littermates also demonstrated that susceptibility of CCR2-deficient mice is not due to differences in maternally-transmitted or environmentally-acquired microbiota. Additionally, acute depletion of monocytes with anti-CCR2 antibodies phenocopied CCR2-deficient mice in abrogating control of intestinal and systemic bacterial burdens, demonstrating that monocytes are critical for acute control of enteric Yersinia infection.
Lack of both YopE and YopH abrogated PG formation, whereas either effector alone was sufficient to induce PG formation, implying a key role for actin cytoskeleton disruption in PG formation. Notably, ablation of YopH activity alone significantly reduced numbers of intestinal PGs, indicating that YopH was predominantly responsible for PG formation. Precisely how YopH and YopE lead to pyogranuloma formation remains to be determined.
Monocytes and neutrophils comprise a large proportion of Yop-injected cells in vivo53. YopH blocks neutrophil degranulation, ROS production, phagocytosis, and release of neutrophil extracellular traps20,45,46,49,54. The virulence of YopH-deficient bacteria is restored upon neutrophil depletion or ROS-deficiency45,46, suggesting that YopH potently targets neutrophil function in tissues during infection. In line with these observations, PGs of CCR2-deficient mice exhibited decreased numbers of neutrophils and reduced surface expression of the activation marker CD11b, which were restored in the setting of YopH deficiency. CD11b is expressed basally on the plasma membrane and in a subset of neutrophil granules, which are recruited to the surface upon neutrophil activation35–37,55. Our findings imply that monocytes enhance neutrophil degranulation to overcome YopH-dependent blockade of their antimicrobial functions. Consistently, while monocytes were required for host survival in response to WT bacteria, they were dispensable for controlling YopH mutant Yp. Furthermore, depletion of neutrophils in YopHR409A-infected CCR2-deficient mice restored bacterial virulence. Overall, our findings imply that monocytes play an important role in host defense by promoting maintenance of PG architecture and neutrophil function in the face of YopH-dependent blockade. Our study reveals a previously unappreciated site of Yersinia colonization within the intestine and provides insight into granuloma formation and function during Yersinia infection.
Methods
Animals
C57BL/6 wild-type and Ccr2gfp/gfp mice30 (in which insertion of EGFP into the translation initiation site of Ccr2 disrupts its expression) were acquired from the Jackson Laboratory and bred at the University of Pennsylvania. Ccr2r−/− mice31 were provided by Dr. Sunny Shin (University of Pennsylvania). Unless specifically noted, all animals were bred by homozygous mating and housed separately by genotype. Mice of either sex between 8-12 weeks of age were used for all experiments. All animal studies were performed in accordance with University of Pennsylvania IACUC-approved protocols (protocol #804523).
Bacteria
Wild-type Yp (clinical isolate strain 32777, serogroup O1)56 and isogenic mutants were provided by Dr. James Bliska (Dartmouth College). Ye (strain 8081, serogroup O8)57 was provided by Dr. Stanley Falkow (Stanford University). Additional mutants lacking YopE (ΔyopE), enzymatic activity of YopH (YopHR409A), both (denoted yopEH) or YopE/YopT/YopH (YopER144ATC139AHR409A, denoted yopETH) were generated by two-step allelic recombination as previously described58 with plasmids provided by Dr. James Bliska. Fluorescent Yp (mCherry+) was generated from plasmids provided by Dr. Kimberly Davis (Johns Hopkins University).
Infections
Yp and Ye were cultured to stationary phase at 28°C and 250 rpm shaking for 16 hours in 2xYT broth supplemented with 2 μg/ml triclosan (Millipore Sigma). Mice were fasted for 16 hours and subsequently inoculated by oral gavage with 100-200 μl phosphate-buffered saline (PBS). All bacterial strains were administered at 2x108 colony-forming units (CFU) per mouse with the exception of pYV-, yopETH, and yopEH which were administered at 2x109 CFU per mouse.
Antibody-mediated depletions
Mice were given daily rat IgG2 isotype control or depletion antibodies in 100 μl PBS by intraperitoneal injections from day −1 to 4 post infection. To deplete monocytes, mice were given 20 μg Rat anti-mouse CCR259 (clone MC-21 AK). To deplete neutrophils, mice were given 200 μg rat anti-mouse Ly-6G (1A8; Bio X Cell) followed by a secondary anti-Rat kappa Ig light chain (MAR 18.5; Bio X Cell) 8 hours later to enhance depletion efficiency60. To deplete neutrophils in CCR2-deficient animals, mice were given 200 μg rat anti-mouse Gr-1 (RB6-8C5; Bio X Cell).
Protein quantifications
Cytokines were measured in supernatants from homogenized tissue using Cytometric Bead Array (BD Biosciences) according to manufacturer’s instructions with the following modification: the amount of capture beads, detection reagents, and sample volumes was scaled down tenfold. Data were collected on an LSRFortessa flow cytometer (BD Biosciences) with FACSDiva v9.0 (BD Biosciences) and analyzed with FlowJo v10 (BD Biosciences).
Tissue preparation and cell isolation
Blood was harvested by cardiac puncture upon euthanasia and collected in 250 U/ml Heparin solution (Millipore Sigma) prior to erythrocyte lysis with Red Blood Cell Lysing Buffer (Millipore Sigma).
Lymph nodes and spleens were homogenized through a 70 μm cell strainer (Fisher Scientific), then flushed with R10 buffer consisting of RPMI 1640 (Millipore Sigma) supplemented with 10 mM HEPES (Millipore Sigma), 10% FBS (Omega Scientific), 1 mM sodium pyruvate (Thermo-Fisher Scientific) and 100 U/ml penicillin + 100 μg/ml streptomycin (Thermo Fisher Scientific).
Intestines were excised, flushed luminally with sterile PBS to remove the feces, opened longitudinally along the mesenteric side and placed luminal side down. Small-intestinal tissue containing macroscopically visible pyogranulomas (PG+), adjacent non-granulomatous areas (PG−) and uninfected control tissue (uninf) were excised using a 2 mm-ø dermal punch-biopsy tool (Keyes). Biopsies within each mouse were pooled, suspended in epithelial-dissociation buffer consisting of calcium and magnesium-free HBSS (Thermo Fisher Scientific) supplemented with 15 mM HEPES, 10 mg/ml BSA (Millipore Sigma), 5 mM EDTA (Millipore Sigma) and 100 U/ml penicillin + 100 μg/ml streptomycin, then incubated for 30 minutes at 37°C under continuous agitation. To isolate immune cells from the lamina propria, the tissue was enzymatically digested in R10 buffer, along with 0.5 Wünsch units/ml liberase TM (Roche), 30 μg/ml DNase I (Roche), and 5 mM CaCl2 for 20 min at 37°C under continuous agitation. The resulting cell suspensions were filtered through 100 μm cell strainers (Fisher Scientific) and subjected to density-gradient centrifugation using Percoll (GE Healthcare). Briefly, cells were suspended in 40% Percoll and centrifuged over a 70% Percoll layer for 20 min at 600 × g with lowest brake at room temperature. Cells collected between the layers were washed with R10 for downstream analysis.
Flow cytometry
Unspecific Fc binding was blocked for 10 minutes on ice with anti-CD16/CD32 (93; Thermo-Fisher Scientific). Cells were subsequently stained for 30 minutes on ice with the following antibodies and reagents: PE-conjugated rat anti-mouse Siglec-F (E50-2440; BD Biosciences), PE-TxR or PE-Cy5-conjugated rat anti-mouse CD11b (M1/70.15; Thermo Fisher Scientific), PE-Cy5-conjugated mouse anti-mouse NK1.1 (PK136; BioLegend), PE-Cy5.5 or PE-Cy7-conjugated rat anti-mouse CD4 (RM4-5; Thermo Fisher Scientific), PE-Cy7-conjugated rat anti-mouse CD3 (17A2; BioLegend), BV510-conjugated rat anti-mouse CD3e (145-2C11; BioLegend), FITC-conjugated Armenian hamster anti-mouse CD11c (N418; BioLegend), PerCP-Cy5.5-conjugated rat anti-mouse Ly-6C (HK1.4; Thermo Fisher Scientific), PB-conjugated rat anti-mouse CD90.2 (53-2.1; BioLegend), BV510-conjugated rat anti-mouse CD19 (1D3; BD Biosciences), BV605-conjugated Armenian hamster anti-mouse TCRβ (H57-597; BD Biosciences), BV650-conjugated rat anti-mouse I-A/I-E (M5/114.15.2; BD Biosciences), BV711-conjugated rat anti-mouse CD8α (53-6.7; BD Biosciences), BV785-conjugated rat anti-mouse Ly-6G (1A8; Thermo Fisher Scientific), AF647-conjugated mouse anti-mouse CD64 (X54-5/7.1; BD Biosciences), AF700-conjugated mouse anti-mouse CD45.2 (104; BioLegend), PE-CF594-conjugated rat anti-mouse CD45R/B220 (RA3-6B2; BD Biosciences) along with eF780 viability dye (BioLegend) diluted in PBS. Antibodies were used at 1:200 dilution and viability dye at 1:1500 dilution.
For intracellular staining, cells were incubated for 3 hours at 37°C with 5% CO2 in R10 buffer supplemented with 0.33 μl/ml GolgiStop (BD Biosciences) and 15 μg/ml DNase I. Surface proteins were stained as above, then cells were fixed for 20 minutes on ice with Cytofix/Cytoperm Fixation/ Permeabilization solution (BD Biosciences). Lipocalin-2 was stained using biotin-conjugated rat anti-mouse lipocalin-2 (NGAL; BioLegend) on ice for one hour followed by BV711-conjugated streptavidin (BD Biosciences) at 4°C overnight. Intracellular cytokines were stained at 4°C overnight with PerCP-e710-conjugated rat anti-mouse IL-1β (NJTEN3; Thermo Fisher Scientific), eF450-conjugated rat anti-mouse TNF (MP6-XT22; Thermo Fisher Scientific), PE-conjugated Armenian hamster anti-mouse IL-1α (ALF-161; BioLegend). All intracellular antibodies were diluted 1:200 in Perm/Wash Buffer (BD Biosciences). Streptavidin was diluted 1:400 in Perm/Wash Buffer. Cells were acquired on an LSRFortessa flow cytometer with FACSDiva v9.0 and data was analyzed with FlowJo v10. Dead and clustered cells were removed from all analyses.
Histology
Tissues were fixed in 10% neutral-buffered formalin (Fisher Scientific) and stored at 4°C until further processed. Tissue pieces were embedded in paraffin, sectioned by standard histological techniques and stained with hematoxylin and eosin for subsequent histopathological disease scoring by blinded board-certified pathologists. Tissue sections were given scores between 0-4 (healthy-severe) for multiple parameters, including degree of inflammatory cell infiltration, necrosis, and free bacterial colonies along with tissue-specific parameters such as villus blunting and crypt hyperplasia. Healthy mice were characterized by and subsequently scored as having none or low levels of the parameters described, whereas severely afflicted mice presented with high amounts of the respective parameters.
Fluorescence microscopy
Small-intestinal tissue was dissected and flushed with PBS. Intestines were opened longitudinally and ~0.5 cm tissue pieces containing macroscopically visible lesions were excised. Tissues were fixed in 1% paraformaldehyde overnight then blocked for two hours at room temperature in blocking solution containing 10% BSA, 1 μg/ml anti-CD16/32, and 0.5% normal rat IgG in PBS. AF647-conjugated anti-Ly-6G antibody (1A8; BioLegend) was added at 0.01 mg/ml in 100 μl PBS per sample, then whole tissue was stained for 24 hours at 4°C. Samples were washed three times with PBS, mounted whole onto slides in Prolong Glass Antifade Mountant (Thermo Fisher Scientific) and cured for two days at room temperature. Images were acquired on a DMI 6000 laser-scanning confocal microscope (Leica) with a 20x NA 0.75 oil-immersion objective. The center of the sample was determined in the Z direction, then imaged.
Images were analyzed using ImageJ v2.1. Adjustments for brightness and contrast were applied to the entire image. No threshold manipulation was performed. Green and magenta channels were pseudo-colored.
RNA sequencing
Intestinal punch biopsies were collected as described above. Five biopsies per sample type were pooled for each mouse. RNA was extracted using the RNeasy Plus Mini Kit (Qiagen). Sequence-ready libraries were prepared using the Illumina TruSeq Stranded Total RNA kit with Ribo-Zero Gold rRNA depletion (Illumina). Quality assessment and quantification of RNA preparations and libraries were carried out using an Agilent 4200 TapeStation and Qubit 3, respectively. Samples were sequenced on an Illumina NextSeq 500 to produce 150-base pair single end reads with a mean sequencing depth of 9 million reads per sample. Raw reads from this study were mapped to the mouse reference transcriptome (Ensembl; Mus musculus GRCm38) using Kallisto v0.46.261. Raw sequence data are available on the Gene Expression Omnibus (GEO; accession no. GSE194334).
All subsequent analyses were carried out using the statistical computing environment R v4.0.3 in RStudio v1.2.5042 and Bioconductor62. Briefly, transcript quantification data were summarized to genes using the tximport package63 and normalized using the trimmed mean of M values (TMM) method in edgeR64. Genes with <1 CPM in 3 samples were filtered out. Normalized filtered data were variance-stabilized using the voom function in limma65, and differentially expressed genes were identified with linear modeling using limma (FDR ≤ 0.05; absolute log2FC ≥ 1) after correcting for multiple testing using Benjamini-Hochberg.
Statistics
Statistical analyses were performed using Prism v9.0 (GraphPad Software). Independent groups were compared by Mann-Whitney U test or Kruskal-Wallis test with Dunn’s multiple comparisons test. Survival curves were compared by Mantel-Cox test. Statistical significance is denoted * (p<0.05), ** (p<0.01), *** (p<0.001), **** (p<0.0001), NS (not significant).
Extended Data
Extended Data Figure 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 from 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 circle 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 Figure 2. Non-pyogranuloma tissue does not undergo inflammation.
(a) Heatmap 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 Figure 3. CCR2-deficient mice cannot control intestinal Yersinia.
(a) Quantification of total number of intestinal lesions at day 3 post infection. Each circle represents one mouse (n = 9-11). Line represents median. Pooled data 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 of two independent experiments.
(c) Bacterial burdens in small intestinal PG− and PG+ tissue at day 3 post infection. Each circle represents the mean Yp-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 circle represents one mouse (n = 29-30). Lines represent median. Pooled data from four independent experiments.
(e) Bacterial burdens in small intestinal PG− and PG+ tissue at day 5 post infection. Each circle represents the mean Yp-CFU of 3-5 pooled punch biopsies from one mouse (n = 21-30). Lines represent geometric mean. Pooled data from four independent experiments.
(f) Bacterial burdens in small intestinal PG− and PG+ tissue at day 3 post infection. Each circle represents the mean Yp-CFU of 3-5 pooled punch biopsies from one mouse (n = 10-13). Lines represent 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 Figure 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 circle represents 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 circle 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 circle 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 circle 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 circle 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 circle 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 Figure 5. CCR2-deficient mice cannot control systemic Yersinia.
(a) Bacterial burdens in indicated organs at day 3 post infection. Each circle 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 circle 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 circle 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 Figure 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 circle 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 Figure 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)
Extended Data Table 1.
Gene ontology analysis of PG+ vs PG− samples
p_value | term_id | term_name |
---|---|---|
2.16113E-06 | GO:0009617 | response to bacterium |
6.32193E-06 | GO:0006952 | defense response |
7.52444E-06 | KEGG:04657 | IL-17 signaling pathway |
2.20616E-05 | REAC:R-MMU-6798695 | Neutrophil degranulation |
2.94164E-05 | GO:0006954 | inflammatory response |
7.19842E-05 | GO:0005576 | extracellular region |
7.26469E-05 | REAC:R-MMU-168249 | Innate Immune System |
0.000111741 | GO:0051707 | response to other organism |
0.000111741 | GO:0043207 | response to external biotic stimulus |
0.000111741 | GO:0009607 | response to biotic stimulus |
0.000111741 | GO:0052548 | regulation of endopeptidase activity |
0.00011503 | REAC:R-MMU-6799990 | Metal sequestration by antimicrobial proteins |
0.000130727 | GO:0052547 | regulation of peptidase activity |
0.000132537 | GO:0044419 | biological process involved in interspecies interaction between organisms |
0.000132537 | GO:0060326 | cell chemotaxis |
0.000190598 | GO:0030162 | regulation of proteolysis |
0.000220253 | GO:0006508 | proteolysis |
0.000288993 | GO:0006950 | response to stress |
0.000291628 | GO:0032496 | response to lipopolysaccharide |
0.000319678 | GO:0030595 | leukocyte chemotaxis |
0.000319678 | GO:0097529 | myeloid leukocyte migration |
0.000319678 | GO:0002237 | response to molecule of bacterial origin |
0.000344762 | GO:0030593 | neutrophil chemotaxis |
0.000395608 | GO:0005615 | extracellular space |
0.000401486 | REAC:R-MMU-5621480 | Dectin-2 family |
0.000438686 | GO:0070488 | neutrophil aggregation |
0.000673081 | GO:1990266 | neutrophil migration |
0.000682844 | GO:0071621 | granulocyte chemotaxis |
0.001021883 | REAC:R-MMU-168256 | Immune System |
0.001021883 | REAC:R-MMU-5686938 | Regulation of TLR by endogenous ligand |
0.001304093 | GO:0097530 | granulocyte migration |
0.001304093 | GO:0010727 | negative regulation of hydrogen peroxide metabolic process |
0.001412884 | GO:0035662 | Toll-like receptor 4 binding |
0.001798466 | GO:0009605 | response to external stimulus |
0.001982425 | KEGG:05134 | Legionellosis |
0.001993156 | GO:0035425 | autocrine signaling |
0.002051465 | REAC:R-MMU-5668599 | RHO GTPases Activate NADPH Oxidases |
0.002194029 | GO:0050900 | leukocyte migration |
0.002194029 | GO:0006935 | chemotaxis |
0.002215455 | GO:0042330 | taxis |
0.00255511 | GO:0045861 | negative regulation of proteolysis |
0.002892713 | KEGG:05417 | Lipid and atherosclerosis |
0.002892713 | KEGG:05323 | Rheumatoid arthritis |
0.003588317 | GO:0038094 | Fc-gamma receptor signaling pathway |
0.004193849 | CORUM:2552 | Il3rb1-Shc complex, IL-3 stimulated |
0.004193849 | CORUM:3202 | Il3rb1-Shc-Ship complex, IL-3 stimulated |
0.004796173 | GO:0005102 | signaling receptor binding |
0.004796173 | GO:0048306 | calcium-dependent protein binding |
0.004796173 | GO:0016209 | antioxidant activity |
0.004796173 | GO:0045236 | CXCR chemokine receptor binding |
0.004796173 | GO:0035325 | Toll-like receptor binding |
0.004831801 | GO:0071222 | cellular response to lipopolysaccharide |
0.00509277 | GO:0018119 | peptidyl-cysteine S-nitrosylation |
0.00509277 | GO:0071219 | cellular response to molecule of bacterial origin |
0.005161487 | KEGG:04668 | TNF signaling pathway |
0.005255007 | GO:0098869 | cellular oxidant detoxification |
0.005255007 | GO:0010310 | regulation of hydrogen peroxide metabolic process |
0.005255007 | GO:0017014 | protein nitrosylation |
0.005516248 | KEGG:04060 | Cytokine-cytokine receptor interaction |
0.005686316 | GO:0070486 | leukocyte aggregation |
0.00618356 | GO:0038093 | Fc receptor signaling pathway |
0.006324423 | GO:0071216 | cellular response to biotic stimulus |
0.006363274 | GO:1900048 | positive regulation of hemostasis |
0.006363274 | GO:0030194 | positive regulation of blood coagulation |
0.006785708 | GO:1990748 | cellular detoxification |
0.006976822 | GO:0019538 | protein metabolic process |
0.006976822 | GO:0010951 | negative regulation of endopeptidase activity |
0.006976822 | GO:0050820 | positive regulation of coagulation |
0.006976822 | GO:0061844 | antimicrobial humoral immune response mediated by antimicrobial peptide |
0.006976822 | GO:0051336 | regulation of hydrolase activity |
0.006976822 | GO:0097237 | cellular response to toxic substance |
0.006976822 | GO:0002523 | leukocyte migration involved in inflammatory response |
0.006976822 | GO:0010466 | negative regulation of peptidase activity |
0.007587123 | GO:0030546 | signaling receptor activator activity |
0.007587123 | GO:0048018 | receptor ligand activity |
0.008029604 | GO:0098754 | detoxification |
0.008029604 | GO:0070887 | cellular response to chemical stimulus |
0.008491456 | GO:1901564 | organonitrogen compound metabolic process |
0.008491456 | GO:0043280 | positive regulation of cysteine-type endopeptidase activity involved in apoptotic process |
0.008871636 | REAC:R-MMU-380108 | Chemokine receptors bind chemokines |
0.009751859 | GO:0050729 | positive regulation of inflammatory response |
0.010252954 | GO:0007166 | cell surface receptor signaling pathway |
0.01137206 | GO:2001056 | positive regulation of cysteine-type endopeptidase activity |
0.011871608 | GO:0019730 | antimicrobial humoral response |
0.012088564 | GO:0050727 | regulation of inflammatory response |
0.01368439 | GO:0050896 | response to stimulus |
0.013729547 | GO:0033993 | response to lipid |
0.013729547 | GO:0002376 | immune system process |
0.014330714 | GO:0050790 | regulation of catalytic activity |
0.015486453 | GO:0042060 | wound healing |
0.015486453 | GO:0006955 | immune response |
0.015667249 | GO:0007596 | blood coagulation |
0.01569326 | GO:1902913 | positive regulation of neuroepithelial cell differentiation |
0.01569326 | GO:1901331 | positive regulation of odontoblast differentiation |
0.01569326 | GO:1904843 | cellular response to nitroglycerin |
0.01569326 | GO:0050817 | coagulation |
0.01569326 | GO:0061408 | positive regulation of transcription from RNA polymerase II promoter in response to heat stress |
0.01569326 | GO:0035490 | regulation of leukotriene production involved in inflammatory response |
0.01569326 | GO:2000296 | negative regulation of hydrogen peroxide catabolic process |
0.01569326 | GO:0007599 | hemostasis |
0.01569326 | GO:0035491 | positive regulation of leukotriene production involved in inflammatory response |
0.01569326 | GO:0009405 | obsolete pathogenesis |
0.01569326 | GO:0014002 | astrocyte development |
0.01569326 | GO:0010950 | positive regulation of endopeptidase activity |
0.017653698 | GO:0006953 | acute-phase response |
0.018417791 | GO:0010952 | positive regulation of peptidase activity |
0.018858959 | GO:0032268 | regulation of cellular protein metabolic process |
0.019282879 | GO:0018198 | peptidyl-cysteine modification |
0.019829956 | GO:2000378 | negative regulation of reactive oxygen species metabolic process |
0.020378866 | GO:0042743 | hydrogen peroxide metabolic process |
0.020384657 | GO:0042742 | defense response to bacterium |
0.021431689 | GO:0098542 | defense response to other organism |
0.021452884 | GO:0004857 | enzyme inhibitor activity |
0.021452884 | GO:0061770 | translation elongation factor binding |
0.021452884 | GO:0008009 | chemokine activity |
0.021651519 | GO:0005509 | calcium ion binding |
0.022590954 | GO:0051346 | negative regulation of hydrolase activity |
0.022590954 | GO:0090303 | positive regulation of wound healing |
0.022999507 | GO:0043281 | regulation of cysteine-type endopeptidase activity involved in apoptotic process |
0.022999507 | GO:0044092 | negative regulation of molecular function |
0.023149342 | GO:0005544 | calcium-dependent phospholipid binding |
0.023526513 | GO:0072737 | response to diamide |
0.023526513 | GO:0051246 | regulation of protein metabolic process |
0.023526513 | GO:0062026 | negative regulation of SCF-dependent proteasomal ubiquitin-dependent catabolic process |
0.023526513 | GO:0072738 | cellular response to diamide |
0.023526513 | GO:0062025 | regulation of SCF-dependent proteasomal ubiquitin-dependent protein catabolic process |
0.023526513 | GO:0072593 | reactive oxygen species metabolic process |
0.023526513 | GO:2000295 | regulation of hydrogen peroxide catabolic process |
0.023526513 | GO:1900138 | negative regulation of phospholipase A2 activity |
0.023526513 | GO:1904842 | response to nitroglycerin |
0.023526513 | GO:0043086 | negative regulation of catalytic activity |
0.023526513 | GO:0035606 | peptidyl-cysteine S-trans-nitrosylation |
0.023526513 | GO:2000098 | negative regulation of smooth muscle cell-matrix adhesion |
0.023526513 | GO:0030887 | positive regulation of myeloid dendritic cell activation |
0.023526513 | GO:0010332 | response to gamma radiation |
0.02358878 | REAC:R-MMU-6803157 | Antimicrobial peptides |
0.02359003 | GO:0004866 | endopeptidase inhibitor activity |
0.023704517 | GO:0050789 | regulation of biological process |
0.023715466 | GO:0030414 | peptidase inhibitor activity |
0.023715466 | GO:0046899 | nucleoside triphosphate adenylate kinase activity |
0.023715466 | GO:0061135 | endopeptidase regulator activity |
0.023891517 | HP:0040245 | Reduced alpha-2-antiplasmin activity |
0.023891517 | HP:0040228 | Decreased level of plasminogen |
0.023891517 | HP:0040248 | Reduced plasminogen activator inhibitor 1 activity |
0.023891517 | HP:0040224 | Abnormality of fibrinolysis |
0.023891517 | HP:0040230 | Decreased level of tissue plasminogen activator |
0.023891517 | HP:0040249 | Reduced plasminogen activator inhibitor 1 antigen |
0.023891517 | HP:0100310 | Epidural hemorrhage |
0.023891517 | HP:0032312 | Decreased circulating globulin level |
0.023891517 | HP:0011854 | Hemoperitoneum |
0.023891517 | HP:0040184 | Oral bleeding |
0.025295615 | GO:0030193 | regulation of blood coagulation |
0.025816626 | GO:1900046 | regulation of hemostasis |
0.025816626 | GO:2001244 | positive regulation of intrinsic apoptotic signaling pathway |
0.025816626 | GO:2000116 | regulation of cysteine-type endopeptidase activity |
0.025816626 | GO:0031638 | zymogen activation |
0.025816626 | GO:0009636 | response to toxic substance |
0.026199211 | GO:0050818 | regulation of coagulation |
0.026878816 | GO:0004869 | cysteine-type endopeptidase inhibitor activity |
0.026878816 | GO:0005125 | cytokine activity |
0.027315059 | GO:0009611 | response to wounding |
0.027561199 | HP:0031029 | Elevated carcinoembryonic antigen level |
0.027561199 | HP:0030657 | Umbilical cord hematoma |
0.027561199 | HP:0025391 | Crazy paving pattern on pulmonary HRCT |
0.028727626 | GO:0042379 | chemokine receptor binding |
0.028727626 | GO:0061134 | peptidase regulator activity |
0.028845751 | GO:0042509 | regulation of tyrosine phosphorylation of STAT protein |
0.029344406 | GO:1990911 | response to psychosocial stress |
0.029344406 | GO:1903852 | positive regulation of cristae formation |
0.029344406 | GO:1904845 | cellular response to L-glutamine |
0.029344406 | GO:1990910 | response to hypobaric hypoxia |
0.029344406 | GO:0031349 | positive regulation of defense response |
0.029344406 | GO:0007260 | tyrosine phosphorylation of STAT protein |
0.029344406 | GO:1901329 | regulation of odontoblast differentiation |
0.029344406 | GO:0002292 | T cell differentiation involved in immune response |
0.029344406 | GO:0002540 | leukotriene production involved in inflammatory response |
0.029344406 | GO:1903036 | positive regulation of response to wounding |
0.029344406 | GO:0045204 | MAPK export from nucleus |
0.030446796 | GO:0051716 | cellular response to stimulus |
0.030446796 | GO:0007165 | signal transduction |
0.030913215 | REAC:R-MMU-5621481 | C-type lectin receptors (CLRs) |
0.031668575 | GO:0006919 | activation of cysteine-type endopeptidase activity involved in apoptotic process |
0.031841638 | HP:0030879 | Interlobular septal thickening |
0.031841638 | HP:0033711 | Pulmonary interstitial thickening |
0.032646273 | GO:1990444 | F-box domain binding |
0.032646273 | GO:0030246 | carbohydrate binding |
0.032646273 | GO:0001691 | pseudophosphatase activity |
0.03353745 | GO:0070098 | chemokine-mediated signaling pathway |
0.03353745 | GO:1901700 | response to oxygen-containing compound |
0.034461909 | GO:0031347 | regulation of defense response |
0.035643318 | GO:1903850 | regulation of cristae formation |
0.035643318 | GO:1901491 | negative regulation of lymphangiogenesis |
0.035643318 | GO:1900365 | positive regulation of mRNA polyadenylation |
0.035643318 | GO:0006172 | ADP biosynthetic process |
0.035643318 | GO:1904844 | response to L-glutamine |
0.035643318 | GO:0002538 | arachidonic acid metabolite production involved in inflammatory response |
0.035643318 | GO:0050794 | regulation of cellular process |
0.035997936 | GO:0048708 | astrocyte differentiation |
0.036002466 | REAC:R-MMU-3371511 | HSF1 activation |
0.036269445 | GO:0044267 | cellular protein metabolic process |
0.037888022 | GO:0071396 | cellular response to lipid |
0.038461896 | REAC:R-MMU-168898 | Toll-like Receptor Cascades |
0.038648573 | GO:1900034 | regulation of cellular response to heat |
0.038648573 | GO:1900004 | negative regulation of serine-type endopeptidase activity |
0.038648573 | GO:1900003 | regulation of serine-type endopeptidase activity |
0.038648573 | GO:1901342 | regulation of vasculature development |
0.038648573 | GO:0061044 | negative regulation of vascular wound healing |
0.038648573 | GO:0065009 | regulation of molecular function |
0.038648573 | GO:0009136 | purine nucleoside diphosphate biosynthetic process |
0.038648573 | GO:0009180 | purine ribonucleoside diphosphate biosynthetic process |
0.038648573 | GO:1902572 | negative regulation of serine-type peptidase activity |
0.038648573 | GO:1902571 | regulation of serine-type peptidase activity |
0.038648573 | GO:1990868 | response to chemokine |
0.038648573 | GO:1902512 | positive regulation of apoptotic DNA fragmentation |
0.038648573 | GO:1903936 | cellular response to sodium arsenite |
0.038648573 | GO:1990869 | cellular response to chemokine |
0.038648573 | GO:0045765 | regulation of angiogenesis |
0.038648573 | GO:2000097 | regulation of smooth muscle cell-matrix adhesion |
0.038648573 | GO:0009188 | ribonucleoside diphosphate biosynthetic process |
0.039982864 | GO:0065007 | biological regulation |
0.041518754 | GO:0005126 | cytokine receptor binding |
0.042452241 | GO:0071347 | cellular response to interleukin-1 |
0.043080047 | GO:0016477 | cell migration |
0.043080047 | GO:1903626 | positive regulation of DNA catabolic process |
0.043080047 | GO:0097187 | dentinogenesis |
0.043080047 | GO:0010757 | negative regulation of plasminogen activation |
0.043080047 | GO:0071895 | odontoblast differentiation |
0.043080047 | GO:0051918 | negative regulation of fibrinolysis |
0.043080047 | GO:1904528 | positive regulation of microtubule binding |
0.043080047 | GO:0010519 | negative regulation of phospholipase activity |
0.043080047 | GO:0045113 | regulation of integrin biosynthetic process |
0.043080047 | GO:2001033 | negative regulation of double-strand break repair via nonhomologous end joining |
0.04419202 | GO:0023052 | signaling |
0.04419202 | GO:0007159 | leukocyte cell-cell adhesion |
0.044654435 | GO:0032269 | negative regulation of cellular protein metabolic process |
0.045893648 | GO:0009408 | response to heat |
0.046945217 | GO:0002526 | acute inflammatory response |
0.046945217 | GO:0030885 | regulation of myeloid dendritic cell activation |
0.046945217 | GO:0045112 | integrin biosynthetic process |
0.046945217 | GO:1903935 | response to sodium arsenite |
0.046945217 | GO:0061302 | smooth muscle cell-matrix adhesion |
0.046945217 | GO:1901490 | regulation of lymphangiogenesis |
0.046945217 | GO:1902075 | cellular response to salt |
0.046945217 | GO:0007154 | cell communication |
0.048811625 | GO:0045862 | positive regulation of proteolysis |
0.048811625 | GO:0002286 | T cell activation involved in immune response |
0.049030752 | GO:0021782 | glial cell development |
0.049030752 | GO:0050878 | regulation of body fluid levels |
0.049326198 | KEGG:04061 | Viral protein interaction with cytokine and cytokine receptor |
0.049360301 | REAC:R-MMU-3371568 | Attenuation phase |
Extended Data Table 2.
Gene ontology analysis of PG− vs uninfected samples Supplementary code file
p_value | term_id | term_name |
---|---|---|
0.00243412 | REAC:R-MMU-71387 | Metabolism of carbohydrates |
0.00243412 | REAC:R-MMU-9033658 | Blood group systems biosynthesis |
0.00243412 | REAC:R-MMU-9037629 | Lewis blood group biosynthesis |
0.00276581 | KEGG:00603 | Glycosphingolipid biosynthesis - globo and isoglobo series |
0.00341096 | GO:0005976 | polysaccharide metabolic process |
0.00372835 | KEGG:00601 | Glycosphingolipid biosynthesis - lacto and neolacto series |
0.00855723 | GO:0005975 | carbohydrate metabolic process |
0.02242855 | KEGG:01230 | Biosynthesis of amino acids |
0.02874227 | GO:0000139 | Golgi membrane |
0.02918355 | REAC:R-MMU-8964540 | Alanine metabolism |
0.03100142 | GO:0044264 | cellular polysaccharide metabolic process |
0.03100142 | GO:0006022 | aminoglycan metabolic process |
0.03100142 | GO:0000272 | polysaccharide catabolic process |
0.03361355 | GO:0044262 | cellular carbohydrate metabolic process |
0.03466317 | GO:0102148 | N-acetyl-beta-D-galactosaminidase activity |
0.03466317 | GO:0001632 | leukotriene B4 receptor activity |
0.03466317 | GO:0008107 | galactoside 2-alpha-L-fucosyltransferase activity |
0.03466317 | GO:0051734 | ATP-dependent polynucleotide kinase activity |
0.03466317 | GO:0051733 | polydeoxyribonucleotide kinase activity |
0.03466317 | GO:0051731 | polynucleotide 5’-hydroxyl-kinase activity |
0.03466317 | GO:0047635 | alanine-oxo-acid transaminase activity |
0.03466317 | GO:0046404 | ATP-dependent polydeoxyribonucleotide 5’-hydroxyl-kinase activity |
0.03466317 | GO:0031127 | alpha-(1,2)-fucosyltransferase activity |
0.03466317 | GO:0016740 | transferase activity |
0.03466317 | GO:0003872 | 6-phosphofructokinase activity |
0.03466317 | GO:0004021 | L-alanine:2-oxoglutarate aminotransferase activity |
0.03466317 | GO:0015016 | [heparan sulfate]-glucosamine N-sulfotransferase activity |
0.03466317 | GO:0004974 | leukotriene receptor activity |
0.03500403 | REAC:R-MMU-9033807 | ABO blood group biosynthesis |
0.03932616 | KEGG:01200 | Carbon metabolism |
0.03932616 | KEGG:01100 | Metabolic pathways |
0.04042288 | GO:0070095 | fructose-6-phosphate binding |
0.04128752 | GO:0030262 | apoptotic nuclear changes |
0.04128752 | GO:0006921 | cellular component disassembly involved in execution phase of apoptosis |
0.04167664 | GO:0004519 | endonuclease activity |
0.04167664 | GO:0070061 | fructose binding |
0.04167664 | GO:0004563 | beta-N-acetylhexosaminidase activity |
0.04426833 | GO:0098791 | Golgi apparatus subcompartment |
0.04857165 | REAC:R-MMU-391906 | Leukotriene receptors |
Supplementary Material
Acknowledgements
We thank Dr. James Bliska for generously providing plasmids for Yop mutant Yp strains, as well as Dr. Kimberly 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 Dr. David Christian and Dr. Andrea Stout for key advice on confocal microscopy methods, and Dr. Sunny Shin for constructive editorial comments and scientific discussion. This work was supported by NIH Awards R01AI128530 (IEB), R0AI1139102A1 (IEB), R01DK123528 (IEB) and a BWF Investigator in the Pathogenesis of Infectious Disease Award (IEB); the Foundation Blanceflor Postdoctoral Scholarship (DS), the Swedish Society for Medical Research postdoctoral fellowship (DS) and the Sweden-America Foundation J. Sigfrid Edström award (DS); NIH NRSA F31AI160741-01 (RM); NIH T32 AI141393-2 in Microbial Pathogenesis and Genomics (RM); Mark Foundation Grant 19-011MIA (IEB), F32 AI164655 (JPG); and NSF GRFP Award (SP). We thank members of the Brodsky lab for scientific discussion and Dr. Daniel Grubaugh for comments on the manuscript.
Footnotes
Code Availability
Code for RNA sequencing analysis is available in the Supplementary Code File.
Competing interests
The authors declare no competing interests.
Data Availability
Raw RNA sequencing data are available on the Gene Expression Omnibus (GEO; accession no. GSE194334). All other raw data are available upon request to the corresponding author.
References
- 1.Pagán AJ & Ramakrishnan L The Formation and Function of Granulomas. Annu. Rev. Immunol 36, 639–665 (2018). [DOI] [PubMed] [Google Scholar]
- 2.Petersen HJ & Smith AM The Role of the Innate Immune System in Granulomatous Disorders. Front. Immunol 4, 1–11 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Casson CN 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Dunay IR et al. Gr1+ Inflammatory Monocytes Are Required for Mucosal Resistance to the Pathogen Toxoplasma gondii. Immunity 29, 306–317 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Espinosa V. et al. Inflammatory Monocytes Orchestrate Innate Antifungal Immunity in the Lung. PLoS Pathog. 10, e1003940 (2014). [DOI] [PMC free article] [PubMed] [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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Antonelli LR 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.St. John AL et al. S1P-Dependent Trafficking of Intracellular Yersinia pestis through Lymph nodes establishes buboes and systemic infection. Immunity 41, 440–450 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Eisele NA et al. Salmonella require the fatty acid regulator PPARδ for the establishment of a metabolic environment essential for long term persistence. 14, 171–182 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Paff JR, Triplett DA & Saari TN Clinical and Laboratory Aspects of Yersinia pseudotuberculosis Infections, with a Report of Two Cases. Am. J. Clin. Pathol 66, 101–110 (1976). [DOI] [PubMed] [Google Scholar]
- 11.El-Maraghi NRH & Mair NS The Histopathology of Enteric Infection with Yersinia pseudotuberculosis. Am. J. Clin. Pathol 71, 631–639 (1979). [DOI] [PubMed] [Google Scholar]
- 12.Barnes PD, Bergman MA, Mecsas JC & Isberg RR Yersinia pseudotuberculosis disseminates directly from a replicating bacterial pool in the intestine. J. Exp. Med 203, 1591–601 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Lamps LW 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). [DOI] [PubMed] [Google Scholar]
- 14.Zhang Y, Tam JW, Mena P, van der Velden AWM & Bliska JB 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhang Y, Khairallah C, Sheridan BS, van der Velden AWM & Bliska JB 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kuziel WA et al. Severe reduction in leukocyte adhesion and monocyte extravasation in mice deficient in CC chemokine receptor 2. Proc. Natl. Acad. Sci. U. S. A 94, 12053–12058 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Serbina NV & Pamer EG Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat. Immunol 7, 311–317 (2006). [DOI] [PubMed] [Google Scholar]
- 18.Peterson LW et al. RIPK1-dependent apoptosis bypasses pathogen blockade of innate signaling to promote immune defense. J. Exp. Med 214, 3171–3182 (2017). [DOI] [PMC free article] [PubMed] [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). [DOI] [PubMed] [Google Scholar]
- 20.Bliska JB, Brodsky IE & Mecsas J Role of the Yersinia pseudotuberculosis Virulence Plasmid in Pathogen-Phagocyte Interactions in Mesenteric Lymph Nodes. EcoSal Plus 9, eESP-0014-2021 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Davis KM All Yersinia Are Not Created Equal: Phenotypic Adaptation to Distinct Niches Within Mammalian Tissues. Front. Cell. Infect. Microbiol 8, 1–8 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Davis KM, Mohammadi S & Isberg RR 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Clark MA, Hirst BH & Jepson MA 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Fasciano AC 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Nuss AM et al. Tissue dual RNA-seq allows fast discovery of infection-specific functions and riboregulators shaping host-pathogen transcriptomes. Proc. Natl. Acad. Sci. U. S. A 114, E791–E800 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Serbina NV, Salazar-Mather TP, Biron CA, Kuziel WA & Pamer EG TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity 19, 59–70 (2003). [DOI] [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). [DOI] [PubMed] [Google Scholar]
- 28.Grainger JR et al. Inflammatory monocytes regulate pathologic responses to commensals during acute gastrointestinal infection. Nat. Med 19, 713–721 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Davis JM & Ramakrishnan L The Role of the Granuloma in Expansion and Dissemination of Early Tuberculous Infection. Cell 136, 37–49 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Satpathy AT et al. Notch2-dependent classical dendritic cells orchestrate intestinal immunity to attaching-and-effacing bacterial pathogens. Nat. Immunol 14, 937–948 (2013). [DOI] [PMC free article] [PubMed] [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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Borjesson DL, Simon SI, Hodzic E, Ballantyne CM & Barthold SW Kinetics of CD11b/CD18 Up-Regulation During Infection with the Agent of Human Granulocytic Ehrlichiosis in Mice. Lab. Investig 82, 303–311 (2002). [DOI] [PubMed] [Google Scholar]
- 33.Mann BS & Chung KF Blood neutrophil activation markers in severe asthma: lack of inhibition by prednisolone therapy. Respir. Res 7, 1–10 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Yoon JW, Pahl MV & Vaziri ND Spontaneous leukocyte activation and oxygen-free radical generation in end-stage renal disease. Kidney Int. 71, 167–172 (2007). [DOI] [PubMed] [Google Scholar]
- 35.Othman A, Sekheri M & Filep JG Roles of neutrophil granule proteins in orchestrating inflammation and immunity. FEBS J. 289, 3932–3953 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Rijneveld AW, De Vos AF, Florquin S, Verbeek JS & van der Poll T CD11b Limits Bacterial Outgrowth and Dissemination during Murine Pneumococcal Pneumonia. J. Infect. Dis 191, 1755–1760 (2005). [DOI] [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). [DOI] [PubMed] [Google Scholar]
- 38.Portnoy DA, Wolf-Watz H, Bolin I, Beeder AB & 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Balada-Llasat JM & Mecsas JC 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Mukherjee S. et al. Yersinia YopJ Acetylates and Inhibits Kinase Activation by Blocking Phosphorylation. Science 312, 1211–1214 (2006). [DOI] [PubMed] [Google Scholar]
- 41.Brodsky IE & Medzhitov R Reduced Secretion of YopJ by Yersinia Limits In Vivo Cell Death but Enhances Bacterial Virulence. PLoS Pathog. 4, e1000067 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Chung LK 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.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). [DOI] [PubMed] [Google Scholar]
- 44.Green SP, Hartland EL,M, Robins-Browne RM & Phillips WA 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). [DOI] [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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Rolán HG, Durand EA & 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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Logsdon LK & Mecsas JC Requirement of the Yersinia pseudotuberculosis Effectors YopH and YopE in Colonization and Persistence in Intestinal and Lymph Tissues. Infect. Immun 71, 4595–4607 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fisher ML, 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). [DOI] [PMC free article] [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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Stackowicz J, Jönsson F & Reber LL Mouse Models and Tools for the in vivo Study of Neutrophils. Front. Immunol 10, 3130 (2020). [DOI] [PMC free article] [PubMed] [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). [DOI] [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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Durand EA, Maldonado-Arocho FJ, Castillo C, Walsh RL & Mecsas JC The presence of professional phagocytes dictates the number of host cells targeted for Yop translocation during infection. Cell. Microbiol 12, 1064–1082 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Taheri N, Fahlgren A & Fällman M Yersinia pseudotuberculosis Blocks Neutrophil Degranulation. Infect. Immun 84, 3369–3378 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Murphy AJ 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). [DOI] [PubMed] [Google Scholar]
- 56.Simonet M & Falkow S Invasin expression in Yersinia pseudotuberculosis. Infect. Immun 60, 4414–4417 (1992). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Portnoy DA, Moseley SL & Falkow S Characterization of Plasmids and Plasmid-Associated Determinants of Yersinia enterocolitica Pathogenesis. Infect. Immun 31, 775–782 (1981). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zhang Y, Murtha J, Roberts MA, Siegel RM & Bliska JB Type III secretion decreases bacterial and host survival following phagocytosis of Yersinia pseudotuberculosis by macrophages. Infect. Immun 76, 4299–4310 (2008). [DOI] [PMC free article] [PubMed] [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). [DOI] [PubMed] [Google Scholar]
- 60.Boivin G. et al. Durable and controlled depletion of neutrophils in mice. Nat. Commun 11, 1–12 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Bray NL, Pimentel H, Melsted P & Pachter L Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol 34, 525–527 (2016). [DOI] [PubMed] [Google Scholar]
- 62.Huber W. et al. Orchestrating high-throughput genomic analysis with Bioconductor. Nat. Methods 12, 115–121 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Soneson C, Love MI & Robinson MD Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Research 4, 1521 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Robinson MD, McCarthy DJ & Smyth GK edgeR: A Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Ritchie ME et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Raw RNA sequencing data are available on the Gene Expression Omnibus (GEO; accession no. GSE194334). All other raw data are available upon request to the corresponding author.