Abstract
Bacteria of the genus Shigella replicate in intestinal epithelial cells and cause shigellosis, a severe diarrheal disease that resolves spontaneously in most healthy individuals. During shigellosis, neutrophils are abundantly recruited to the gut, and have long been thought to be central to Shigella control and pathogenesis. However, how shigellosis resolves remains poorly understood due to the longstanding lack of a tractable and physiological animal model. Here, using our newly developed Nlrc4−/−Casp11−/− mouse model of shigellosis, we unexpectedly find no major role for neutrophils in limiting Shigella or in disease pathogenesis. Instead, we uncover an essential role for macrophages in the host control of Shigella. Macrophages respond to Shigella via TLRs to produce IL-12, which then induces IFN-γ, a cytokine that is essential to control Shigella replication in intestinal epithelial cells. Collectively, our findings reshape our understanding of the innate immune response to Shigella.
eTOC
Eislmayr et al. reveal a previously unrecognized role of macrophages in controlling the intracellular pathogen Shigella. By sensing bacteria and triggering IFN-γ production, macrophages, not neutrophils, help gut epithelial cells fight off infection. This study redefines our understanding of the innate immune response during this severe gastrointestinal infection.
Graphical abstract
Introduction
Shigella species are the causative agents of shigellosis, a severe inflammatory gastrointestinal infection characterized by symptoms that range from fever, cramps, and nausea to aggressive, mucoid, and bloody diarrhea (dysentery)1. Shigellosis is a leading cause of pediatric and geriatric diarrheal-associated mortality, with an estimated 212,000 annual deaths2–4. Shigella is typically acquired via the fecal-oral route upon ingestion of contaminated food or water5. After initial colonization of the gut, Shigella is believed to cross the epithelial barrier through M-cells, a specialized epithelial cell-type sampling the gut lumen. Shigella then invades intestinal epithelial cells (IECs) by using its Type III Secretion System (T3SS)6–8. This needle-like structure enables the cytosolic delivery of ~30 effectors that facilitate bacterial entry into and replication within the cytosol of IECs, the preferred intracellular niche for Shigella7,9. Shigella also uses actin-based motility to spread directly from cell-to-cell within the epithelium10–12.
Mice are naturally highly resistant to Shigella and are able to resist infectious doses 10,000-fold higher than those sufficient to cause disease in humans13,14. Recently, we reported that mice lacking the NAIP–NLRC4 and Caspase-11 inflammasomes are highly susceptible to Shigella infection15,16. The NAIP–NLRC4 inflammasome detects components of the Shigella T3SS17–19, and the Caspase-11 inflammasome senses lipopolysaccharide (LPS) of intracellular Shigella20–23. Susceptibility of Nlrc4−/−Casp11−/− mice to Shigella requires pre-treatment of the mice with antibiotics (e.g., streptomycin) in order to facilitate Shigella gut colonization, as is also the case for colonization of mice with other gut pathogens24–26. Inflammasomes appear to protect mice from Shigella via a highly efficient mechanism in which infected intestinal epithelial cells selectively undergo pyroptotic cell death and expulsion into the gut lumen27,28. Although human NAIP and NLRC4 are functional and can detect Shigella19, the human NAIP–NLRC4 inflammasome does not appear sufficient to protect humans, perhaps due to its poor expression in IECs29. Additionally, Shigella encodes effectors to block host cell death. For example, the Shigella effector IpaH7.8 inactivates human—but not mouse—GSDMD, thus preventing pyroptotic cell death30. The Shigella effector OspC3 potently blocks CASP4 (the human homolog of mouse Caspase-11)31, and partially inhibits mouse Caspase-1123,31,32. Furthermore, we recently reported that the Shigella effector OspF disrupts p38 MAPK-dependent priming of the NAIP–NLRC4 inflammasome, a process essential for rapid inflammasome activation in cells with low basal expression of NAIP–NLRC433. Mice lacking NAIP–NLRC4 and Caspase-11 (Nlrc4−/−Casp11−/−) thus recapitulate the lack of inflammasome responses in human IECs, and represent the only genetically tractable animal model that exhibits all the key manifestations of human shigellosis, including oral infection, efficient bacterial replication in IECs, diarrhea that can be bloody, and neutrophilic inflammation16.
Shigella infection provokes an acute immune response that involves the secretion of proinflammatory factors such as IL-1β and CXCL1, both of which stimulate the recruitment of myeloid cells34–36. As a result, extensive infiltration of neutrophils and monocytes is found in biopsies obtained from humans with active shigellosis, and in infected guinea pigs37–39. However, the role of myeloid cells during shigellosis remains poorly understood. Surprisingly, loss of IL-1 signaling did not impact the progression of shigellosis in Nlrc4−/− mice16. Moreover, Shigella largely avoids interacting with phagocytes by replicating within and spreading directly within the cytosol of intestinal epithelial cells. Encounters of Shigella with macrophages result in the rapid death of macrophages8,34,40–42. For all these reasons, macrophages are not believed to be major participants in bacterial control during shigellosis7. By contrast, the massive influx of neutrophils to the gut lumen—one of the primary hallmarks of shigellosis—is believed both to promote bacterial clearance and to cause intestinal damage and severe disease symptoms7,43. However, direct experimental assessment of the roles of neutrophils and macrophages during infection is lacking. In particular, a possible role for bystander (uninfected) macrophages has not generally been considered, despite the fact these cells are abundant in the gut and would not be subject to Shigella-mediated killing.
Here, we demonstrate that Shigella infection of Nlrc4−/−Casp11−/− mice exhibits a self-limiting and spontaneously resolving course of disease very similar to that typically seen in human shigellosis. We unexpectedly discover that in this model, neutrophil depletion has no discernable effect on bacterial replication or disease progression, whereas macrophage depletion markedly exacerbates shigellosis symptoms and bacterial burdens in IECs. Our data suggest that macrophages orchestrate the innate response to Shigella by initiating a protective cytokine circuit. We propose that this circuit begins with bystander (uninfected) macrophages that detect Shigella via Toll-like receptors (TLRs), leading to the production of IL-12. IL-12 stimulates the production of IFN-γ in Nkp46+ cells and γδT cells, and we demonstrate IFN-γ is an essential factor in resolving the infection in vivo and limiting bacterial replication in human colonic organoids. Mice lacking the ability to produce or sense IFN-γ are incapable of controlling Shigella and succumb to the infection. Overall, our study underscores the essential role of pathogen-sensing by macrophages and highlights the pivotal role of IFN-γ in resolving shigellosis in vivo.
Results
We previously demonstrated that mice deficient in NLRC4 and Caspase-11 (Nlrc4−/−Casp11−/−) are susceptible to oral Shigella infection15,16. In humans, Shigella infection is typically self-limiting and resolves within 5–7 days. To assess whether disease also resolves in Nlrc4−/−Casp11−/− mice, we orally infected streptomycin-pretreated Nlrc4−/−Casp11−/− mice with Shigella flexneri strain 2457T and tracked body weight and fecal CFU (Fig. 1A–B). As expected, after 48h of infection, mice lost over 10% of their initial weight and shed over 109 Shigella CFU/g feces. However, by 72h, mice began to regain weight, coinciding with a decrease in fecal Shigella CFU. By day 5, most mice regained or exceeded their initial weight and stopped shedding bacteria by day 8 post-infection.
Figure 1: IFN-γ signaling in intestinal epithelial cells is associated with the resolution of Shigella infection.
(A) Body weight and (B) Shigella colony forming units (CFU) in the feces of Nlrc4−/−Casp11−/− mice orally infected with Shigella flexneri 2457T. (C) Differential expression analysis of bead-enriched EpCAM+ colonic-epithelial cells from infected versus non-infected mice (n=6/group). Genes with log2 fold change > 1 and padj < 0.001 in red. (D) Gene ontology (GO) enrichment analysis of significantly differently expressed genes for activated or suppressed biological processes with a gene set size <150. (E) In vitro infection of mouse CT26 cells pre-treated with IFN-γ or left untreated for 16h and infected with Shigella (MOI=1) that constitutively express mCherry and an arabinose-inducible GFP. After gentamycin treatment and arabinose addition, viable Shigella (GFP+mCherry+) was assessed by flow cytometry 3h later. (F) Human colonic organoids were pre-treated with IFN-γ or left untreated for 16 hours and infected with Shigella. (A) n=5, (B) LoD=Limit of detection, (E-F) combined data from replicate experiments with n=8, ****p<0.0001 in t-test and median shown.
To identify the signaling pathways involved in restricting Shigella replication, we performed bulk RNA sequencing of bead-enriched EpCAM+ epithelial cells isolated from the colon and cecum of mice 48h post-infection. We focused on the cecum and colon since bacteria are almost exclusively detected in IECs from these tissues (Fig. S1A). We found 474 genes were significantly upregulated 48h after the infection when compared to naïve mice (log2 fold change (lfc) >2, padj < 0.001, Fig. 1C, Table S1). Among the top differentially expressed genes were Igtp, Tgtp1, Nos2 and Gbp2, all of which are known to be induced by IFN-γ. Additional interferon-stimulated genes (ISGs) upregulated by infection included Stat1 and Ciita (complete list of canonical ISGs in Table S2). Indeed, gene set enrichment analysis identified “responses to type II interferon” as the top activated biological process (FDR<0.05, Fig. 1D and S1B) for genes with |lfc| > 1, padj <0.05 and a max GO set size <120.
IFN-γ restricts Shigella replication in vivo and protects against shigellosis disease
Consistent with previous results demonstrating that IFN-γ inhibits the growth of Shigella in mouse embryonic fibroblasts44, we found that pretreatment of CT26 cells (a mouse colon carcinoma cell line) with IFN-γ for 16h resulted in substantially fewer cells harboring metabolically active Shigella 3h after infection (Fig. 1E, Fig. S1C). Similarly, 16h pretreatment of human colonic organoids with IFN-γ significantly reduced the proportion of Shigella-infected cells (Fig. 1F, Fig. S1D).
In vivo, we observed IFN-γ levels rise in the lamina as early as 8h after infection, and continue to increase throughout the infection (Fig. 2A). To test whether IFN-γ is important to resolve Shigella infection, we treated Nlrc4−/−Casp11−/− mice with an IFN-γ-neutralizing antibody every 24h throughout the infection (starting simultaneously with infection). Anti-IFN-γ-treated mice lost significantly more weight, and harbored >100-fold more Shigella CFU in IECs with more pronounced colonic and cecal atrophy—a hallmark of intestinal inflammation—compared to isotype-injected controls (Fig. 2B–D, S2A). Five out of 8 anti-IFN-γ treated mice exhibited high levels of occult blood and the remaining 3 tested faintly positive for occult blood, while only half of the control animals were faintly positive (Fig. 2E). Of note, mice treated with anti-IFN-γ also harbored higher levels of bacteria in their mesenteric lymph nodes and spleens (Fig. 2F), increased fecal calprotectin, lipocalin and myeloperoxidase (MPO, Fig. 2G, H) as well as elevated inflammatory cytokines in the lamina propria (Fig. 2I). Notably, Shigella CFU within the feces was similar between anti-IFN-γ and isotype-treated mice, indicating that altered intestinal luminal colonization did not explain the effects of IFN-γ neutralization (Fig. 2J). From these results, we conclude that IFN-γ restricts intracellular Shigella replication and prevents severe disease in Nlrc4−/−Casp11−/− mice in vivo.
Fig. 2: IFN-γ is essential to protect from severe disease and limit bacterial replication.
(A) ELISA quantification of IFN-γ levels in the lamina propria of infected Nlrc4−/−Casp11−/− mice at indicated time points (n=3 per time point). Mice treated with anti-IFN-γ antibody or isotype were assessed for (B) body weight, (C) intracellular CFU in cecum/colon IECs, (D) length of cecum and colon normalized to the initial weight, (E) fecal occult blood score (see Methods), (F) bacterial dissemination into the mesenteric lymph nodes (MLN) and the spleen, (G-I) clinical markers of gut inflammation (calprotectin, lipocalin and MPO in the feces) as well as the pro-inflammatory cytokines IL-1β and CXCL1 levels in lamina propria. PBS-treated non-infected mice steady indicate basal levels. (J) Quantification of shed Shigella bacteria in the fecal matter of infected animals. Data from two independent experiments with n = 6 PBS and 8 infected mice per treatment group. *p < 0.05, **p < 0.01, ***p < 0.001 in t-test with mean and SD shown (B,D,G,H,I) or Mann-Whitney-test with median and interquartile range (C,E,F,J). ns = not significant (p>0.05).
We subsequently evaluated whether neutralization of IFN-γ renders wild-type (Nlrc4- and Casp11-sufficient) mice susceptible to shigellosis. In undiluted lysates of isolated IECs, we detected only a few (0–5) colonies, regardless of IFN-γ neutralization (Fig. S2B). These low CFU counts are comparable to those observed in mice infected with the virulence plasmid-cured, non-invasive BS103 strain. Therefore, we conclude that Nlrc4 and Casp11 dominantly protect the intestinal epithelial cell from Shigella, and IFN-γ is essential for protecting that niche if the primary Nlrc4/Casp11-mediated defense is absent or circumvented.
IFN-γ is essential for Shigella control and acts predominantly on non-hematopoietic cells
To provide a genetic test of the role of IFN-γ in Shigella control, we generated Nlrc4−/−Casp11−/− mice lacking the IFN-γ-receptor (Ifngr1). Consistent with the effects of IFN-γ neutralization (Fig. 2), infected Ifngr1−/−Nlrc4−/−Casp11−/− mice experienced greater weight loss burdens than IFN-γ-sufficient Nlrc4−/−Casp11−/− animals 48h after infection (Fig. 3A–B). Strikingly, all Ifngr1−/−Nlrc4−/−Casp11−/− mice reached predefined humane endpoints (e.g., <75% starting body weight, severe signs of morbidity) 72h post-infection, while all Nlrc4−/−Casp11−/− mice survived and regained their initial weight. Notably, anti-IFN-γ treatment of Ifngr1−/−Nlrc4−/−Casp11−/− mice did not further enhance susceptibility to infection (Fig. S3A). Together, our data indicate a critical role for IFN-γ in restricting Shigella and in promoting recovery.
Figure 3. The responsiveness of non-hematopoietic cells to IFN-γ is crucial for controlling Shigella infection.
(A) Change of body weight over time and (B) the percentage of mice exceeding humane endpoint criteria (indicated with circled symbols) of infected Ifngr1−/−Nlrc4−/−Casp11−/− or Nlrc4−/−Casp11−/− mice. The dashed line in (A) represents the predefined 75% weight retention limit set as a humane endpoint. (C) Experimental design of reciprocal BM transplantation, created in BioRender. Eislmayr, K. (2025) https://BioRender.com/1dzqp93 (D) change in weight and IEC CFU in bone marrow chimeras 48 hours after infection. Donor and irradiated recipient genotypes are as indicated. n=8–15 (A, B) t-test (A) and Log-rank (Mantel-Cox) test (B). n=5–6/group from two replicate experiments in (D) with one-way ANOVA with Tukey’s multiple comparison for weight with median and Kruskal-Wallis test with Dunn’s multiple comparison for CFU. *p<0.05, **p<0.01, ns = not significant (p>0.05).
Next, we sought to determine whether IFN-γ acts on hematopoietic or non-hematopoietic cells by generating reciprocal bone marrow chimeras between Ifngr1−/−Nlrc4−/−Casp11−/−and Nlrc4−/−Casp11−/− mice (schematic in Fig. 3C). After 8 weeks of reconstitution, the chimeric mice were infected. We found resistance to Shigella depends on the genotype of the bone marrow recipient, regardless of the genotype of the donor (Fig. 3D). Recipients lacking the IFN-γ receptor phenocopied full-body IFN-γ receptor deficiency, regardless of the genotype of the donor bone marrow. Conversely, Shigella replication in IECs was restricted in irradiated Nlrc4−/−Casp11−/− mice reconstituted with either bone marrow. From these results, we conclude that IFN-γ predominantly acts on radioresistant (likely non-hematopoietic) cells to restrict Shigella replication. Considering the observed profound upregulation of ISGs in IECs during infection (Fig. 1C), our data are consistent with the hypothesis that IFN-γ restricts Shigella replication by eliciting an anti-bacterial response in IECs.
Given that type I IFNs and IFN-γ induce a similar set of ISGs, we assessed the role of type-I IFNs during Shigella infection. An antibody targeting the Interferon-alpha/beta receptor 1 (αIFNAR) was administered every 24 hours throughout the infection. No significant differences in weight loss or IEC CFU counts were observed between the anti-IFNAR-treated and isotype-treated Nlrc4−/−Casp11−/− or Ifngr1−/−Nlrc4−/−Casp11−/− animals (Fig. S3B). Thus, type-I IFNs appear to be dispensable in controlling Shigella replication in our mouse model, in contrast to the pivotal and essential role of IFN-γ.
Gbp2 and Gbp5 were among the top upregulated ISGs in IECs during infection (Fig. 1C). Indeed, GBPs have been implicated in the cell-intrinsic restriction of intracellular pathogens, including Shigella45,46. However, Shigella expresses the effector IpaH9.8, which has been demonstrated to counteract the activity of GBPs47,48. Therefore, we next evaluated the role of GBPs in controlling the replication of Shigella within CT26 cells. IFN-γ-pretreated Gbp1/2−/− and Gbp5−/− cells had a significantly higher percentage of GFP+ cells than IFN-γ-treated WT cells (Fig. S3C). Gbp1/2/5−/− cells were even more susceptible, suggesting that GBPs act combinatorially to limit Shigella. Importantly, it is evident that additional ISGs contribute to IFN-γ-mediated restriction, since IFN-γ is still able to reduce Shigella burdens in Gbp1/2/5−/− (Fig. S3C). Infection of IFN-γ-treated WT cells with an IpaH9.8-deficient Shigella strain resulted in a markedly reduced number of infected cells compared to the infection with the WT Shigella strain. Notably, in the absence of GBP1/2/5, there was no discernible difference between infections with the WT and IpaH9.8-deficient strain. Collectively, these data indicate that IpaH9.8 partially mitigates the effect exerted by GBPs. In support of this, IpaH9.8-deficient Shigella colonized IECs of Nlrc4−/−Casp11−/− mice less efficiently, and caused less weight loss, as compared to WT Shigella (Fig. S3D). Moreover, the attenuation of the IpaH9.8 mutant is exclusively observable in the context of an IFN-γ response.
Macrophages are essential for Shigella control, but neutrophils are dispensable
Although IFN-γ did not act on hematopoietic cells to restrict Shigella, we hypothesized that immune cells might still be critical for IFN-γ production. We observed a 15-fold increase in neutrophils (Ly6G+Ly6Cmed) and a 5-fold increase in inflammatory monocytes (Ly6G−Ly6ChighMHCII+) in the lamina propria during Shigella infection (Fig. S4A). To determine if myeloid cells contribute to Shigella clearance, we generated Nlrc4−/−Casp11−/− mice with a Cre-inducible diphtheria toxin receptor expressed by LysM+ (myeloid) cells (Lyz2Cre/+iDTRlsl/lslNlrc4−/−Casp11−/−). In these mice, diphtheria toxin (DT) results in the depletion of myeloid cells—including primarily macrophages and neutrophils (Figure S4B–C)—and subsequent Shigella infection resulted in a significantly higher weight loss and ~100-fold more CFUs in IECs, as compared to non-depleted mice (Fig. 4A). In addition, myeloid cell depletion also increased infection pathology, resulting in increased inflammatory cytokines (Figure S4D) and enhanced levels of occult/visible blood in the feces (Fig. 4B). Thus, myeloid cells critically contribute to the control of shigellosis.
Figure 4. LysM+ and CD64+ myeloid cells, but not neutrophils, are essential to control shigellosis.
(A) Change in body weight, IEC CFUs and (B) fecal occult blood score of infected Lyz2Cre/+iDTRlsl/lslNlrc4−/−Casp11−/− mice in myeloid cell-depleted (DT-treated) or non-depleted (PBS-treated) mice. (C) IEC CFU counts, weight loss and (D) fecal MPO levels of Shigella-infected Nlrc4−/−Casp11−/− mice treated with a neutrophil depleting antibody (αLy6G) or an isotype control. (E) Weight loss and IEC CFU counts obtained 48 hours after infection from irradiated Nlrc4−/−Casp11−/− mice reconstituted with Mrp8Cre/+iDTRlsl/lsl bone marrow and treated with DT (neutrophil depletion), or with PBS. (F) Body weight and IEC CFUs of irradiated Nlrc4−/−Casp11−/− mice engrafted with Lyz2Cre/+iDTRlsl/lsl, or CD64Cre/+iDTRlsl/lsl bone marrow, respectively, treated with DT or PBS (n=4/group). Data from two independent experiments with n = 7 (A-B), n = 4 (C-D), n = 3–5 (E), n = 4–5 (F). *p<0.05, **p<0.01, ***p<0.001, ns = not significant according to t-test (A,D, weight data in E) with mean and SD sown or Mann-Whitney-test (CFU data in A,B,C,E) with median and interquartile range, one-way ANOVA with Tukey’s multiple comparisons for weight (F) and median and Kruskal-Wallis test with Dunn’s multiple comparison for CFU (F).
The recruitment of neutrophils into the gut tissue is believed to play an important role both in controlling bacterial burdens and in causing pathology during shigellosis. Therefore, we assessed the impact of neutrophil depletion, using anti-Ly6G antibody (clone 1A8), during infection. Unexpectedly, we observed that neutrophil depletion did not affect bacterial burdens within IECs, weight loss, or the induction of inflammatory markers during Shigella infection, compared to non-treated animals (Fig. 4C and Fig. S4E). Consistent with successful neutrophil depletion (Fig. S4F), 1A8-treated mice exhibited reduced pus and MPO levels in the cecum and colon (Fig. 4D, representative images in Fig. S4H). Treatment with 1A8 had no effect on the luminal levels of Shigella (Fig. S4G).
To verify these results, we crossed mice expressing a neutrophil-specific Cre (Mrp8Cre/+)49 with iDTRlsl/lsl mice, and used bone marrow from these mice to reconstitute irradiated Nlrc4−/−Casp11−/− mice. Treatment of the chimeric mice with DT did not alter weight loss or IEC CFU burdens during Shigella infection as compared to PBS controls (Fig. 4E). However, DT treatment efficiently depleted neutrophils in the blood and lamina propria of infected mice (Fig. S4I, J). Thus, our results indicate that neutrophil depletion has no major effect on the pathogenesis of Shigella infection.
Based on the significant impact of myeloid cell depletion on IEC colonization and the negligible role of neutrophils, we investigated the role of macrophages and monocytes in controlling Shigella infection. Scott et al. recently described the use of CD64Cre mice for macrophage-specific Cre expression50. Bone marrow from CD64Cre/+iDTRlsl/lsl mice was transplanted into irradiated Nlrc4−/−Casp11−/− mice. DT-treated (CD64+ cell-depleted), infected mice experienced a more pronounced weight loss compared to PBS-treated mice (Fig. 4F). Moreover, CD64+ cell-depletion increased Shigella IEC CFU burdens similar to LysM+ cell-depletion. Notably, the lamina propria of infected and DT-injected CD64Cre/+iDTRlsl/lsl chimeras exhibited a significant absence of neutrophils in addition to the depletion of CD64+ macrophages, in both the lamina propria and blood (Fig. S4I, J), with no effect of DT treatment on blood neutrophils in naïve mice (Fig. S4M). Hence, although CD64Cre is specific for monocytes and macrophages under non-inflamed conditions50, it may be expressed by neutrophils during inflammation. In our experiments, however, we rule out an essential role for neutrophils with neutrophil-specific depletion with 1A8 treatment and Mrp8Cre/+iDTRlsl/lsl mice. In addition, specific dendritic cells depletion (using bone marrow chimeras with Zbtb46DTR/DTR donors into Nlrc4−/−Casp11−/− recipients) did not affect the bacterial load in IECs to the extent observed in CD64+ cell-depleted animals (Fig. S4K,L,N). Therefore, we concluded that CD64+ macrophages, and not neutrophils or dendritic cells, play a central role in resistance to Shigella infection.
IFN-γ production is dependent on macrophage TLR sensing.
Since both IFN-γ neutralization and depletion of macrophages/monocytes led to heightened bacterial IEC colonization, we wondered whether macrophages/monocytes might play a role in IFN-γ induction during infection. Indeed, IFN-γ levels were significantly reduced in the lamina propria upon myeloid cell-depletion (Fig. 5A).
Figure 5: TLR and IL-12 signaling are essential for the production of IFN-γ in Nkp46+ and γδT cells to control Shigella infection.
(A) IFN-γ levels in the lamina propria (LAP) of infected Lyz2Cre/+iDTRlsl/lslNlrc4−/−Casp11−/− mice with or without DT-induced myeloid cell-depletion. (B) IFN-γ in the LAP and IEC CFU 48 hours post-infection in irradiated Nlrc4−/−Casp11−/− bone marrow chimeras receiving Tlr2/4-Unc−/− or CD45.1-WT bone marrow and (C) the corresponding fecal occult blood score. (D) IEC CFU from irradiated Nlrc4−/−Casp11−/− engrafted with either Unc−/−, Tlr2/4−/−, Tlr2/4-Unc−/− or CD45.1-WT bone marrow. (E) Effect of IL-12p40 neutralization (αIL-12) or isotype antibody treatment during Shigella infection of Nlrc4−/−Casp11−/− mice on IEC CFU counts, weight loss and (F) IFN-γ levels in LAP. (G) IEC CFU counts and IFN-γ levels in the LAP of αIL-12 or isotype treated Nlrc4−/−Casp11−/− or Ifngr−/−Nlrc4−/−Casp11−/− mice. (H) Mean fluorescent intensity (MFI) of the GREAT-IFN-γ-YFP reporter in different cell-types isolated from the LAP of Shigella-infected (S.f.) or mock-treated (PBS) animals that had been irradiated Nlrc4−/−Casp11−/− and received bone marrow from an IFN-γ reporter strain. Mean and t-test (A,F,H), median and Mann-Whitney-test (B CFU, C, F CFU and weight loss), median and Dunn’s multiple comparisons test (CFU data in G) and mean and Tukey’s multiple comparisons test (IFN-γ data in G). Combined data from replicate experiments with n=5 (A), n=9–12 (B), n=7 (F,G), n=8–10 (D), n=5–16 (G).
Macrophages sense bacterial infections via Toll-like receptors (TLRs) and produce pro-inflammatory cytokines, including the IFN-γ-inducing cytokine, IL-12. It has been demonstrated that Unc93b1-deficient cells no longer respond to TLR3, TLR7, TLR9, TLR11, TLR12, and TLR13 ligands51. By crossing these mice to Tlr2−/− and Tlr4−/− mice, Sivick et al generated mice incapable of TLR signaling52, while still retaining the capacity for IL-1R and IL-18R signaling. Bone marrow chimeras in which Nlrc4−/−Casp11−/− recipients were reconstituted with Tlr2/4/Unc−/− donor marrow exhibited blunted IFN-γ induction compared to WT(CD45.1) into Nlrc4−/−Casp11−/− control chimeras (Fig. 5B). Moreover, loss of TLR signaling by hematopoietic cells also resulted in elevated IEC CFU and enhanced pathology (Fig. 5C and S5A). Of note, the IEC CFU counts in either Tlr2/4−/− or Unc−/− bone marrow chimeras were intermediate between those observed in CD45.1 or Tlr2/4/Unc−/− engrafted Nlrc4−/−Casp11−/− animals (Fig. 5D). Consequently, the restriction of Shigella appears to depend on a combinatorial Unc-dependent and Unc-independent TLRs response.
To determine whether TLRs function within macrophages to orchestrate the protective response to Shigella, we generated bone marrow chimeras in which Nlrc4−/−Casp11−/− mice were reconstituted with a mix of CD64Cre/+ iDTRlsl/lsl and Tlr2/4/Unc−/− donor bone marrow. In these chimeras, half of the bone marrow-derived CD64+ macrophages are DT-sensitive, while the other half lacks TLR signaling. Upon DT-treatment, only CD64− cells in these mice will express functional TLRs. Consistent with a critical role for TLRs in CD64+ cells, DT-treatment of the mixed chimeras elevated the bacterial load within IECs to levels comparable to those of 100% CD64Cre/+ iDTRlsl/lsl or 100% Tlr2/4/Unc−/− chimeras, and similar to Ifngr1−/−Nlrc4−/−Casp11−/− mice (Fig. S5B). Notably, without DT-treatment, the mixed chimeras exhibited IEC CFU counts comparable to non-irradiated Nlrc4−/−Casp11−/− mice. Collectively, our data demonstrate the significant role of TLR-mediated sensing in CD64+ macrophages to initiate the protective IFN-γ response to Shigella.
Macrophage-derived IL-12 induces IFN-γ production in Nkp46+ and γδT cells
IFN-γ can be induced by different cytokines, including IL-12, IL-15, IL-18, and type I IFN53,54. Interperitoneal injection of IL-18 neutralizing antibody during Shigella infection of Nlrc4−/−Casp11−/− mice had no effect on IEC CFU burdens or pathology (Fig. S5C). By contrast, IL-12p40 neutralizing antibody significantly increased IEC CFU burdens (Fig. 5E), exacerbated weight loss (Fig. S5D), and aggravated the inflammatory symptoms of shigellosis (Fig. S5E–G), as compared to isotype control. Notably, IL-12p40 neutralization resulted in a significant reduction of IFN-γ in the lamina propria (Fig. 5F), and produced symptoms comparable to Ifngr1−/−Nlrc4−/−Casp11−/− mice. Moreover, treatment with anti-IL-12p40 had no effect on the severity of Shigella infection in the absence of IFN-γR (Fig. 5G, S5D–G), consistent with a model in which the main role of IL-12 is to mediate resistance to Shigella via induction of IFN-γ.
Next, we examined whether macrophages are the source of IL-12 in our infection model. As ELISA or flow cytometry-based assays were not sensitive enough, we generated mixed bone marrow chimeras using CD64Cre/+ iDTRlsl/lsl and Il12b−/− donor mice. In these chimeras, half of the hematopoietic cells are incapable of producing IL-12p40 (due to Il12b deficiency), but any CD64+ cells able to produce IL-12 are sensitive to DT-depletion. Thus, upon DT treatment, IL-12p40 can only originate from CD64− cells. Without DT, mixed chimeras exhibited IEC CFU counts akin to those of Nlrc4−/−Casp11−/− mice (Fig S5H). In stark contrast, DT treatment resulted in a 100-fold increase in the IEC bacterial loads, and additional administration of either IL-12p40 or IFN-γ neutralizing antibodies caused only a modest and insignificant additional elevation. The elevated bacterial counts were comparable to DT-treated CD64Cre/+ iDTRlsl/lslNlrc4−/−Casp11−/− bone marrow chimeras or irradiated Nlrc4−/−Casp11−/− mice receiving Il12b−/− bone marrow. We therefore infer that CD64+ cells are a critical source of IL-12p40 during Shigella infection. In an analogous set of experiments, we generated bone marrow chimeras in which irradiated Nlrc4−/−Casp11−/− were engrafted with either a mix of Tlr2/4/Unc−/− and Il12b−/− bone marrow, or a mix of CD45.1 and Il12b−/− bone marrow as a control. In the Tlr2/4/Unc−/−:Il12b−/− mixed chimeras, cells with functional TLR signaling cannot produce IL-12p40, whereas cells that can produce functional IL-12p40 cannot sense Shigella via TLRs. Thus, despite the presence of both TLR-responsive or IL-12p40-producing cells, there are no cells that can both detect Shigella via TLRs and produce IL-12p40. Infection of these chimeras with Shigella resulted in much higher IEC CFU burdens as compared to the control Tlr2/4/Unc−/−:CD45.1 chimeras, indicating that the production of IL-12p40 depends on active TLR signaling within the same cell (Fig. S5I). Collectively, our data indicate that TLR sensing in CD64+ macrophages induces IL-12 in a cell-autonomous manner, which is essential for orchestrating the IFN-γ-mediated restriction of Shigella.
Finally, to identify the IFN-γ-producing immune cells in our infection model, we utilized the GREAT IFN-γ reporter mice55. These transgenic mice carry a bicistronic IFN-γ-IRES-eYFP construct under the control of the endogenous Ifng promoter and poly(A) tail, with unaltered induction and expression levels of IFN-γ56. Shigella infection induced an increase in the mean fluorescence intensity (MFI) of YFP within various different immune cell populations in the colon of Nlrc4−/−Casp11−/− animals engrafted with bone marrow from GREAT mice (Fig. 5H; gating in Fig. S5J). We identified NK cells (Lin−Rorγt−Nkp46+Eomes+), ILC1 (Lin−Rorγt−Nkp46+,Eomes−), Nkp46+ILC3 (Lin−Rorγt+Nkp46+) and γδT cells (Lin+γδTCR+) as the cell-types producing IFN-γ. In contrast, IFN-γ induction was not detected in conventional T cells (Lin+TCRβ+γδTCR−) or other Lineage+TCRβ−γδTCR− cells (Fig. 5H, representative histogram in Fig. S5K). Based on these findings, we conclude that NK cells, Nkp46+ ILCs, and γδT cells are likely responsible for providing the protective IFN-γ in our mouse model of shigellosis.
Discussion
Bacillary dysentery is a self-limiting disease in most healthy adults. However, the lack of suitable animal models has hindered our ability to determine the key processes that control Shigella replication and disease symptoms in vivo. We previously showed that Shigella successfully invades and replicates in epithelial cells of mice lacking the NLRC4 inflammasome14,15. Mice deficient in both NLRC4 and Caspase-11 are even more susceptible to Shigella, and exhibit all pathological aspects of human shigellosis16. Here, we show that infected Nlrc4−/−Casp11−/− mice recover from dysentery within five days, similar to the typical disease course in humans (Fig. 1A,B).
The swift recovery of infected mice within a few days indicates that the processes that ultimately curtail Shigella growth must already be activated in the early stages of the disease, consistent with a pivotal role of the innate immune response. Furthermore, as epithelial cells of the large intestine are the predominant niche for Shigella, these innate responses must ultimately restrict or prevent intracellular bacterial replication in IECs. The top differentially expressed genes in IECs after in vivo infection are characteristic of an IFN-γ response (Fig. 1C, D, S1B). Indeed, mice failing to mount a response to IFN-γ are incapable of recovering from a Shigella infection (Fig. 3A, B) and exhibit severe diarrhea, exacerbated inflammation, and 1,000 times higher CFUs within IECs compared to infected control animals (Fig. 2B–I, S2A, S3A).
Using reciprocal bone marrow chimeras, we found that IFN-γ exerts its effects directly on radioresistant (non-hematopoietic) cells (Fig. 3D). Given that Shigella replicates within IECs, the most parsimonious explanation for our data is that IFN-γ is acting directly on infected IECs. Notably, in WT B6 mice, the rapid NAIP–NLRC4-mediated expulsion of infected epithelial cells obviates the need for IFN-γ (Fig. S2B), in line with reports from our lab and others15,27,57,58. Notably, it has been previously reported that in human IECs, the NAIP–NLRC4 inflammasome is incapable of mounting a protective response against Shigella29,33. Therefore, we postulate that IFN-γ is likely to play an important role in humans. This hypothesis is underscored by the restrictive effect of IFN-γ pretreatment on the intracellular replication of Shigella in human colonic organoids (Fig. S1D). In addition, more indirect mechanisms of IFN-γ-mediated protection are also possible. Ultimately, specific tissue-specific deletion of Ifngr1 from IECs (on an Nlrc4−/−Casp11−/− background) will be necessary to confirm whether IFN-γ signaling in IECs is necessary for Shigella control in vivo.
IFN-γ has been identified as a key factor in restricting several intracellular pathogens, including Mycobacterium tuberculosis, Listeria monocytogenes, and Legionella pneumophila59–65, and previous studies have implicated a potential protective activity against Shigella in vitro and in a non-physiological intranasal infection model66–72. IFN-γ induces the expression of hundreds of interferon-stimulated genes (ISGs) to promote pathogen clearance. Several ISGs have been shown to confer antibacterial functions during Shigella infection in vitro45,68,70,73,74. The most prominent example is the family of guanylate-binding proteins (GBPs). GBPs bind and liberate LPS to promote CASP11- and CASP4-mediated pyroptotic cell death45,46,48,66,69. In addition, the binding of GBPs to LPS forms a cage-like structure, trapping intracellular bacteria and inhibiting actin-dependent motility75,76. However, Shigella disrupts the GBP coat by secretion of an E3 ubiquitin ligase effector protein, IpaH9.8, which ubiquitylates GBPs and targets them for proteasome-dependent degradation48,68,76. Our findings underscore the dynamic interplay between Shigella virulence factors and host immune defenses. Specifically, the reduced responsiveness to IFN-γ in GBP-deficient CT26 cells indicates a role for GBPs in mediating the protective effects of IFN-γ (Fig. S3C). However, this host defense mechanism is partially subverted by the Shigella effector IpaH9.8, as evidenced by the attenuated virulence of IpaH9.8-deficient bacteria in vitro and in vivo (Fig. S3C, D). In line with previous reports, our results suggest that IpaH9.8 antagonizes GBP-dependent immunity. Of note, it is also evident from our data that additional ISGs contribute to the IFN-γ-mediated restriction, as Gbp1/2/5 deficiency only partially abolishes the IFN-γ-mediated protection in CT26 cells (Fig. S3C). Indeed, other ISGs, such as Viperin, Apolipoprotein 3 (APOL3), and the E3 ligase RNF213, have been suggested to restrict intracellular Shigella to some degree70,71,73,77. However, Shigella expresses effector proteins that counteract these activities78. For instance, IpaH1.4 directly antagonizes RNF213 by mediating its proteasomal degradation71,77. Despite Shigella antagonism of ISGs, our study in mice identified IFN-γ responses as central in restricting Shigella in vivo and in human colonic organoids, with a partial contribution of GBPs.
Despite the widely held belief that neutrophil recruitment and transepithelial migration into the gut lumen are significant drivers of pathology during shigellosis1,7, we find no major role for neutrophils during Shigella infection in vivo (Fig. 4C, D, F, Fig. S4D, E). Importantly, we observed extensive recruitment of neutrophils into the lamina propria (Fig. S4A), and the presence of the neutrophil-specific enzyme myeloperoxidase in the feces. These observations suggest that neutrophils are recruited to the intestine in our model, and cross the epithelial layer, as in humans, in which the presence of neutrophils in the stool is a clinical marker for shigellosis1,79,80. Nonetheless, the only noticeable effects of neutrophil depletion were the absence of pus (Fig. S4F) and undetectable MPO levels in the gut lumen, without any impact on weight loss or the bowel inflammation markers lipocalin and calprotectin (Fig. 4C, D, F and S4D, E). Importantly, neutrophil depletion during infection did not affect IEC bacterial counts. It is intriguing that neutrophils appear neither to exacerbate disease nor restrict the growth of Shigella. While numerous studies highlight the critical role of neutrophils in eliminating invading microbes and resolving inflammation (as reviewed by Fournier et al.81), in the case of Shigella infection, neutrophils seem to have a negligible impact on bacterial restriction. This might be explained by the ability of Shigella to effectively induce cell death of phagocytic cells7,79 or by Shigella evasion of phagocytes by replication in epithelial cells. Alternatively, neutrophils may enhance bacterial clearance, but this anti-bacterial function is offset by a distinct pro-bacterial (e.g., immunoregulatory) effect of neutrophils. It may also be the case that neutrophils play important roles in preventing systemic spread of Shigella, or in the late stages of the infection, neither of which were examined in our study.
Shigella rapidly induces macrophage cell death at early stages of infection43,82,83. It is generally accepted that Shigella induces macrophage death to enable the subsequent infection of epithelial cells7,8. Rapid induction of macrophage death is also believed to limit the ability of macrophages to restrict bacterial replication. However, a recent study using a zebrafish model of Shigella infection and experiments with autophagy-deficient (Atg16l1−/−) bone marrow-derived macrophages found a host-protective role for macrophages84,85. Thus, it remains unclear whether Shigella-mediated macrophage cell death is beneficial to the pathogen or to the host and whether macrophages participate in host defense against Shigella intestinal infections. In experiments with mice lacking monocytes and macrophages, we detected increased disease severity and colonization of the intestinal epithelium (Fig. 4A, B, E, and S4C). We also noted similar pathology in mice deficient in TLR signaling (Fig. 5B, C). TLRs sense extracellular-derived microbial ligands, and since Shigella is a pathogen that replicates in the cytosol, it is generally believed that cytosolic sensing pathways (e.g., NOD1/286 and ALPK187) are more important in infected cells than TLRs in initiating innate immunity to Shigella. However, it is likely that microbial ligands are released extracellularly during infection, and thus, bystander (uninfected) macrophages may sense and respond to Shigella. In accord with the results of our Tlr2/4/Unc−/− and CD64Cre/+ iDTRfl/fl mixed bone marrow experiment (Fig. S5B), we propose a model in which the release of bacterial products from infected, pyroptotic macrophages facilitates the release of protective cytokines from noninfected bystander cells—most likely macrophages—in a TLR-dependent manner. Together, our data indicate a beneficial role for macrophages in host defense during Shigella infection.
Finally, our results demonstrate that macrophage activation is pivotal in the induction of adequate IFN-γ to restrict Shigella (Fig. 5A). Although some reports suggest that macrophages can directly produce IFN-γ, our experiments using IL-12p40 neutralizing antibodies suggest that the secretion of IFN-γ is indirect and reliant on IL-12p40 (Fig. 5G), a cytokine subunit known to be induced in macrophages downstream of TLR activation. Our mixed Tlr2/4/Unc−/−:Il12b−/− bone marrow chimera experiments further suggest that TLR signaling and IL-12p40 secretion occur within the same cell (Fig. S5I). IL-12 is a heterodimeric cytokine composed of the IL-12p35 and IL-12p40 (encoded by Il12b) subunits, the latter of which also heterodimerizes with IL-23p19 to form IL-2388. Given this shared subunit, we cannot exclude the possibility that IL-23 contributes to the effects of IL-12p40 in our model. However, the functional profiles of IL-12 and IL-23 diverge significantly: IL-12 is classically associated with the induction of TH1 and NK cell responses and robust IFN-γ production, while IL-23 predominantly promotes the expansion and stabilization of TH17 cells, leading to secretion of IL-17, IL-21, IL-22, and GM-CSF89–95. Given that we observe a critical role for IL-12p40 in IFN-γ induction, rather than in type 17 responses, our data suggest that IL-12, rather than IL-23, is the more likely driver of the immune response observed in our system. Additionally, using the transgenic GREAT IFN-γ reporter chimeras, we identified Nkp46+ ILCs and NK cells, along with γδ T cells, as potential sources of IFN-γ (Fig. 5H).
In sum, our results necessitate revisions to the generally accepted model of Shigella pathogenesis. In particular, our results identify a critical function for macrophages, but not neutrophils, in host defense against Shigella. Given that infected macrophages are rapidly killed by Shigella, we propose that bystander (uninfected) macrophages are vital in orchestrating anti-Shigella innate immunity. Instead of directly killing Shigella by phagocytosis, we propose that a TLR–IL-12 circuit induces the expression of IFN-γ, which then acts on IECs to restrict Shigella replication. Ultimately, understanding how host immunity coordinates clearance of Shigella may be essential for the development of an effective Shigella vaccine.
Resource availability:
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Russell Vance (rvance@berkeley.edu).
Materials availability
Materials used in this study will be provided upon request and available upon publication.
Data and code availability
Raw bulk RNA-sequencing data are deposited in the NCBI Gene Expression Omnibus: GSE288567.
Star Methods
EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS
Mice were maintained under specific pathogen-free conditions and housed with a 12-hour light-dark cycle and standard chow diet (Harlan irradiated laboratory animal diet) ad libitum in accorsssdance with the regulatory standards of the University of California Berkeley Institutional Animal Care and Use Committee. All mice were sex- and age-matched and were 6–12 weeks old when infected (except mice receiving BM transplantation, see details below). Both male and female mice were used in all experiments. Littermate controls were used or, if not possible, mice were co-housed for at least three weeks prior to infection.
C57BL/6J-Nlrc4542stopCasp4em13JLR/J (Nlrc4−/−Casp11−/−) animals were previously generated and described by Roncaioli et al.16 via targeted CRISPR-Cas9 mutagenesis of Casp11 in existing Nlrc4−/− (C57BL/6J-Nlrc4542stop/J)96 mice. 129P2-Lyz2tm1(cre)Ifo/J (Lyz2Cre/+)97, B6.Cg-Tg(S100A8-cre,-EGFP)1Ilw/J (Mrp8Cre)49, C57BL/6-Gt(ROSA)26Sortm1(HBEGF)Awai/J (iDTRlsl/lsl)98, B6.129S7-Ifngr1tm1Agt/J (Ifngr1−/−)99, B6.129S1-Il12btm1Jm/J100, B6(Cg)-Zbtb46tm1(HBEGF)Mnz/J101, B6.129S4-Ifngtm3.1Lky/J55 mice were purchased from Jackson Laboratories. The Ifngr1−/−Nlrc4−/−Casp11−/− mouse line was generated by mating Ifngr1−/− and Nlrc4−/−Casp11−/− mice. B6-Fcgr1tm2Ciphe (CD64Cre/+)50 mice were generated by Bernard Malissen at Centre d’Immunologie de Marseille-Luminy and provided by Yasmine Belkaid at the National Institutes of Health. iDTRlsl/lslNlrc4−/−Casp11−/− were generated by crossing iDTRlsl/lsl mice, obtained from the Jackson Laboratory, with our Nlrc4−/−Casp11−/− mice. Subsequently, Lyz2Cre/+iDTRlsl/lslNlrc4−/−Casp11−/−, CD64Cre/+iDTRlsl/lslNlrc4−/−Casp11−/− and Mrp8Cre/+ iDTRlsl/lsl Nlrc4−/−Casp11−/− lines were generated by crossing iDTRlsl/lsl Nlrc4−/−Casp11−/− mice to animals form the Lyz2Cre/+, CD64Cre/+ or Mrp8Cre/+ line, respectively. C57BL/6N-Unc93b1tm1(KOMP)Vlcg/Mmucd (Unc93b1−/−) mice were obtained from the Mutant Mouse Resource and Research Center (MMRRC) at the University of California, Davis, and originally donated to the MMRRC by David Valenzuela of Regeneron Pharmaceuticals102 and after crossing to Tlr2−/−Tlr4−/− mice, Tlr2−/−Tlr4−/−Unc−/− animals were generously provided by Gregory Barton at the University of California, Berkeley.
Shigella cultivation and preparation for in vivo infections.
If not stated otherwise, infection experiments were conducted with a natural streptomycin-resistant strain of Shigella flexneri serovar 2a 2457T32 (S.f.). S.f. was grown on tryptic soy broth (TSB, BD Bacto # DF0370–07-5) agar plates containing 0.01% congo red (CR, Sigma-Aldrich # C6767) and 100 μg/mL streptomycin sulfate at 37°C. For infections, a single CR-positive colony was picked from a streak not older than a week, inoculated into 5 mL TSB supplemented with 100 μg/mL streptomycin, and incubated overnight under constant agitation at 37°C. 16 hours later, the culture was back-diluted 1:100 in 5 mL fresh TSB + 100 μg/mL streptomycin and incubated for another ~3 hours under constant shaking at 37°C. Upon reaching an OD600 of 1, bacteria were pelleted by centrifugation at 3,000×g for 8 minutes, washed twice with PBS, and resuspended in the same volume of pharmaceutical-grade PBS (USP) for infection by oral gavage. From several experiments that tracked the correlation between OD600 and colony-forming units (CFU), we can estimate that an OD600 equals roughly an infection dose of 1–2×108 CFU/mL. Nevertheless, the actual infectious dose was determined for each experiment by serial dilution and plating on TSB agar plates containing 0.01% CR.
Shigella flexneri ΔipaH9.8 StrepR was generated by transferring the K88R mutation of the rpsL gene from Shigella flexneri WT (streptomycin resistant)103 into the ΔipaH9.8 strain (Piro et al 72) using PCR and the λ red recombinase-mediated recombination system104. In short, the rpsL gene was amplified from the WT strain with PCR using Q5® Hot Start High-Fidelity 2X Master Mix with forward (TTTACGCTGACCAATGACGC) and reverse (CGGCATCGCCCTAAAATTCG) primers following the manufacturer’s protocol and subsequent purification with the Monarch® Spin PCR & DNA Cleanup Kit (NEB # T1130S). Shigella flexneri ΔipaH9.8 was transformed with the recombinase-carrying pKD46 (Datsenko et al.104) plasmid and selected on 0.01% CR TSB plates with 100μg/mL Ampicillin and 0.1% Glucose at 30°C. An overnight culture (TSB + Ampicillin + 0.1% Glucose and 30°C) was back-diluted 1:100 in 5mL TSB + Ampicillin and 0.1% L-Arabinose and incubated for another ~3 hours under constant shaking at 30°C. 1mL of bacteria were centrifuged at 13,000×g for 5min, washed three times with sterile H2O and resuspend in 200μL ice-cold H2O with 10% glycerol. 50μL of these competent cells were mixed with an oligonucleotide containing the point mutation and electroporated using a Gene Pulser Cuvette with 0.1cm gap size (BioRad #165–2089) and 1.8kV, 25μF (200Ω). Transformed bacteria were resuspended in 450μl SOC media (Thermo Fisher #15544034) and recovered at 37°C for 1h before being plated on CR TSB plates with 100μg/mL streptomycin.
In vivo infection
One day before infection, littermates or cohoused mice were deprived of food and water in the morning and 4–6 hours later orally gavaged with 100 μL of 250 mg/mL streptomycin sulfate dissolved in pharmaceutical-grade PBS, after which mice were again given food and water. On the day of infection, food and water were removed from the cage again, and 4–6 hours later, mice were orally gavaged with 100 μL of 108 CFU/mL S.f., prepared as described above. In compliance with our Animal Use Protocol, infected mice were visually examined twice per day and their change in weight was monitored every 24 hours. A loss of more than 25% of the initial weight measured on the day of infection (day 0), or signs of severe sickness such as pronounced hunching, combined with shivering or breathing distress, were considered humane endpoints at which mice were euthanized.
For in vivo antibody-mediated cytokine neutralization, 500 μg of anti-IFN-γ antibody (clone XMG1.2, Bio X Cell #BE055), anti-IL12 (clone C17.8, Bio X Cell #BE0051), anti-IL-18 (clone YIGIF74–1G7, Bio X Cell #BE0237), anti-IFNAR-1 (clone MAR1–5A3, Bio X Cell #BE0241) and InVivoPlus rat IgG1 isotype control (anti-HRP, Bio X Cell #BE0088) or InVivoPlus rat IgG2a isotype control (Bio X Cell #BE0089) depending on the isotype of the neutralizing antibody, were administered by daily intraperitoneal injection starting on the day of infection. For antibody-mediated depletion of neutrophils, 500μg of anti-Ly6G (clone 1A8, Bio X Cell #BE0075–1) was injected daily by intraperitoneal injection starting one day before infection (at the time of streptomycin administration).
In experiments involving diphtheria toxin-mediated cell-depletion, mice were injected i.p. with 30ng/g of DT (Sigma #D0564) or an equal volume of pharmacological PBS daily, starting one day before infection.
METHOD DETAILS
Assessment of inflammation
If not indicated otherwise, 48 hours after infection, mice were sacrificed, their colon and cecum were isolated, and their lengths were recorded. Subsequently, the tissue was cut longitudinally, and the most distal fecal matter from the colon was collected, some of which was spread on detection tabs from a Hemoccult blood testing kit (Pro Advantage #P080018). The rest was then transferred into a pre-weighed 2 mL tube. The fecal matter was homogenized in 1 mL of PBS containing protease inhibitors (Roche #04693159001) using a polytron homogenizer. To determine the fecal CFU, serial dilutions were made in PBS and plated on TSB containing 0.01% CR and 100 μg/mL streptomycin sulfate. For lipocalin, calprotectin, and MPO ELISAs, samples were centrifuged at 13,000×g for 5 minutes and supernatants were analyzed in duplicates with R&D sandwich ELISA kits according to the manufacturer protocol.
A four-step scale was applied to quantify the presence of occult blood. A value of 0 indicates that the Hemoccult blood test was negative, 1 indicates a faint blue staining, and 2 indicates an intense staining. Mice that experienced severe hemorrhagic conditions with clearly visible, macroscopic blood in the lumen of the colon were assigned a score value of 3. Assessments were made on blinded samples.
IEC CFU
To determine the levels of intra-epithelial bacteria, the colon and cecum were isolated from infected mice 48 hours after infection as described above. After taking the fecal sample, the tissue was cleared of any remaining luminal content by washing in PBS. The tissue was collected in 5 mL RPMI (Thermo Fisher #21870–76) with 5% FBS, 2mM GlutaMax (Thermo Fisher #35050–61), 25mM HEPES (Thermo Fisher #15630–080), and 400 μg/mL gentamicin (Thermo Fisher #15710–064), and incubated for 1–2 hours at 4°C. Subsequently, tissue was washed six times in PBS, minced into approximately 1 cm pieces, and placed in 12 mL IEC stripping solution (HBSS, 25 HEPES, 2mM Glutamax, 50μg/mL Gentamicin, 2mM DTT, and 5mM EDTA) within a 50 mL Erlenmeyer flask and incubated for 30 minutes at 37°C and constant stirring at low speed (220rpm). The supernatant was passed through a 100 μm cell strainer, and the retained tissue was transferred back into the flask and mixed with 10 mL ice-cold PBS. The flask was sealed with a rubber stopping, shaken vigorously for 20 seconds, and combined with the previous extraction by passing through the same cell strainer. The remaining tissue was saved for further processing (see lamina propria ELISA or Flow staining). Next, the obtained IEC fraction was incubated with 50 μg/mL gentamicin on ice for 20 minutes, centrifuged 500×g for 5 minutes at 4°C, and washed twice with ice-cold PBS. Before the last centrifugation, a small aliquot of cells was taken for cell counting. Finally, the pellet was resuspended in 1 mL of 1% TritonX-100 (Fisher #BP151–100) and IEC-CFU was determined by plating serial dilutions on TSB agar plates with 0.01% CR and 100 μg/mL streptomycin sulfate.
Tissue ELISA
After extracting IEC cells, the remaining tissue was transferred into 14 ml round-bottom tubes containing 1 mL PBS with proteinase inhibitors (Roche #04693159001). After homogenization with a polytron homogenizer at 20,000 rpm, the suspension was centrifuged at 13,000×g for 5 minutes and the supernatant was analyzed in duplicates with R&D sandwich ELISA kits following the manufacturer protocol and normalized to the total protein concentration determined with the Pierce BCA protein assay kit (Thermo scientific #23225) according to the manufacturer’s protocol.
CFU Spleen/MLN
The spleen and mesenteric lymph nodes were isolated from infected mice 48 hours after infection. Collected tissue was placed in RPMI with 5% FBS, 2mM GlutaMax, 25mM HEPES, and 400 μg/mL gentamicin for 1h. After washing 5× with PBS, tissue was homogenized using a polytron homogenizer and serial dilutions plated on TSB agar plates with 0.01% CR and 100 μg/mL streptomycin sulfate.
IEC bead enrichment and RNAseq
The IEC faction was obtained as described above and further digested with Dispase-II to generate a single-cell suspension, as described by Gracz et al105. In brief, the pelleted IEC fraction was washed with 10 mL PBS with 10% PBS and resuspended in 10 mL of pre-warmed (37°C) HBSS containing 8 mg Dispase-II (Sigma #D4693). Tubes were incubated in a 37°C water bath for 10 minutes with vigorous shaking every 2 minutes. Next, 1 mL FBS with 500 μg DNase (Roche #11284932001) was added and incubated 3 minutes on ice. After washing with RPMI containing 10% FBS, cells were passed through a 40 μm cell strainer. Epithelial cells were enriched using the MojoSort™ Mouse CD326 (Ep-CAM) Selection Kit (BioLegend Cat #480141) following the manufacturer’s protocol for Positive Selection. Retained CD326+ cells were washed with 4mL RPMI containing 10% FBS and centrifuged at 1,000×g for 5 minutes at 4°C and lysed in 250 μL Trizol (Thermo Fisher #10296028). Samples were topped up with 100 μL DNase/RNase-free water and mixed with 200 μL chloroform (Fisher Scientific #C298–500). The aqueous phase and mixed with an equal volume of ethanol. Subsequently, the RNA was isolated using the Monarch Total RNA Miniprep Kit (NEB #T2010S) following the manufacturer’s protocol with on-column DNase treatment. The RNA obtained was sent to Azenta for quality assessment, automated PolyA selection, library preparation, multiplexing, and paired-end sequencing with a read length of 150 bases. After demultiplexing, raw data were pre-processed (including quality control as well as barcode, adaptor, and quality trimming) using FastQC (www.bioinformatics.babraham.ac.uk/projects/fastqc/) and cutadapt, mapped to the Mus_musculus.GRCm38.96 genome assembly using Kalisto (0.44.0 via Bioconda). Overall, each sample contained between 24 million and 32 million reads. Differential expression analysis was performed using DESeq2 (V1.44.0)106 with lfc shrinkage correction in R project version 4.4.1 with RStudio version 2024.04.2+764 (Foundation for Statistical Computing, Vienna, Austria, www.R-project.org/). For gene ontology (GO) enrichment analysis genes with differential expression values padj < 0.001 and log2 fold change > 1 were selected and enrichment analysis was performed using gseGO of the clusterProfiler (4.12.2) package for R with a gene set size <150.
Flow cytometry of lamina propria cells
To prepare cells from the lamina propria for flow cytometry, the colon and cecum were isolated from infected mice and stripped of IEC as described above. The collected remaining tissue was minced thoroughly with scissors and transferred into 50 mL Erlenmeyer flasks. After adding 10 mL of pre-warmed HBSS with 100 μg/mL Liberase TM (Sigma #5401127001) and 5 μg/mL DNase, the tissue was digested for 45 minutes under constant stirring at 37°C. The cell suspension was passed through a 70 μm strainer and washed twice with cold RPMI containing 10% FBS. Epithelial and dead cells were removed using a 70% and 40% two-phase Percoll gradient. Cells from the interface were subsequently collected, washed with PBS and stained with Ghost Dye™ Red 780 (Tonbo #SKU 13–0865-T100) before blocking the FcγII/III receptor with an anti-CD16/anti-CD32 antibody (BioLegend, #156604). Samples were stained for 45 minutes to an hour at room temperature in FACS buffer (PBS with 5g/L BSA and 2g/L sodium azid) with the following antibodies: BV785-labeled CD45 (Clone 104, BioLegend #109839), BUV496-labeled CD3 (Clone 145–2C11, BD #612955), APC-eFlour-labeled B220 (Clone RA3–6B2, Thermo Fisher #47–0452-82), PE-labeled CD11b (Clone M1/70 Thermo Fisher #12–0112-82), FITC-labeled Ly6G (Clone 1A8, BD #551460), APC-labeled Ly6C (Clone HK1.4, BioLegend #128016), MHCII (Colon M5/114.15.2, BioLegend #107626), BUV605-labeled CD64 (Clone H1.2F3, BioLegend #104530), BV421-labeled F4/80 (Clone BM8, BioLegend #123137) and APC-Cy7-labeled EpCAM (Clone G8.8, BioLegend #118218). Stained samples were washed twice and fixed with cytofix/cytoperm (BD biosciences #554722) for 10 minutes at room temperature before measured on an Aurora (Cytek) flow cytometer. Data were analyzed with Flowjo version 10 (BD Biosciences).
Blood Staining
Terminal blood sampling was performed by retro-orbital sinus puncture of anesthetized mice with a heparinized capillary (#22–260950 Fisher scientific). 15μL of the collected blood was immediately mixed with 100μL PBS containing 100U/mL heparin (#H0878–100KU Sigma) and 25μL counting beads (AccuCheck Counting Beads, Thermo Fisher #PCB100). After centrifugation and washing with FACS buffer, cell pellet was stained with 25μL of antibody mix (APC-labeled Ly6C (Clone HK1.4, BioLegend), FITC-labeled Ly6G (Clone 1A8, BD #551460), PE-labeled CD3 (ebio #12–0031-81 Clone 14–2011)) along with the viability dye (Tonbo #13–0658-T) and FcγII/III-block. Subsequently, cells were fixed cytofix/cytoperm (BD biosciences #554722) for 10 min and red blood cells were lysed with ACK buffer (Thermo fisher #A1049201). After washing twice with FACS buffer, samples were analyzed with an Aurora (Cytek) or a BD LSR Fortessa flow cytometer.
CT26 CRISPR-Cas9 Knockout and infection
CT26 were purchased from the Berkeley Cell Culture Facility and cultured in RPMI with 10% FBS, 2 mM Glutamax (Thermo Fisher #35050061), 10 mM HEPES (Thermo Fisher # 15630106), 1 mM sodium pyruvate (Thermo Fisher #11360070) and Penicillin-Streptomycin (Thermo Fisher #15070063). For gene disruption, CT26 cells were trypsinized, washed with media and electroporated with Cas9 2 NLS nuclease (Synthego) complexed with two gRNAs per gene (Synthego, sgRNA EZ kits), and Alt-R® Cas9 Electroporation Enhancer (IDT, 1075916), in Lonza Cell Line Solution SE (Lonza, V4XC-10323) with buffer supplement 1 according to manufacturer’s protocol. Electroporation was performed with a Lonza 4D-Nucleofector Core Unit (AAF-1002B) using the program DS-120. Electroporated cells were immediately plated in media and knockout efficiency were determined by TIDE analysis of PCR products of the modified gnomic locus using Primestar PCR reagents according to manufacturer’s instructions and primers for Gbp1 (Fw: CAGCATTGGATGTTCTGCACTC and Rev: CTCATCATTTCCATAGGCTCACAC), Gbp2 (Fw: AGACTGTCAACATAGGAGGAACA and Rev: GGCTGATCCGCTGCTATTCT), Gbp5 (Fw: TGCTCCGGACAAGAAACTCC and Rev: CTTCACCAGAACTGCCTCGT) and Stat1 (Fw: TCTCGTTTGCGACCATCCG and Rev: ACCATCAGGGCCAGCATTAGG). All genotypes had a knockout efficiency of above 91%.The gRNA sequences used are the following (two per gene): Gbp1/2: CCTCAGTGTCAAGCAGAACC, GAGACTGCACAGTGGAGCCC, Gbp5: GCUUCCAACACUCAGCAACG, CUCAAACAUUCAAUCUACCG, Stat1: UUAAUGACGAGCUCGUGGAG, GGAUAGACGCCCAGCCACUG
For in vitro infections, 106 CT26 cells were seeded into one well of a 6-well tissue culture treated plate in media without Penicillin-Streptomycin one day before the infection. Shigella flexneri - pFCcGi (constitutively expressing mCherry and an arabinose inducible GFP) was grown as described above. CT26 cells were pre-treated with 10 ng/mI IFN-γ (Peprotech #315–05-100UG) or left untreated for 16 hours and were spin-infected (600×g 10min at 37°C) with mid-log phase Shigella at an MOI of 1. After 45 minutes, cells were washed twice with PBS and medium exchanged to CT26 medium supplemented with 100 μg/ml gentamicin (Thermo fisher # 15710064) and 0.4% L-Arabinose (Sigma #A3256–25G). After 3 hours cells were isolated by trypsinization, washed, passed through a 70μm cell strainer, stained with Ghost Dye™ Red 780 (Tonbo #SKU 13–0865-T100), fixed with cytofix/cytoperm (BD biosciences #554722) for 10 min and analyzed with a BD LSR Fortessa flow cytometer.
Human organoid maintenance and infection
Deidentified human colonic organoids were a gift from Scott B. Snapper and established from a rectal biopsy obtained during a routine diagnostic endoscopy in a pediatric subject under Boston Children’s Hospital IRB protocol and cultured with methods modified from Sato et al.107. Briefly, organoids were maintained in 50μL Matrigel domes with human IntestiCult™ Organoid Growth Medium (STEMCELL #6010) supplemented with 10 μM of the ROCK1/ROCK2 inhibitor Y-27632 and Penicillin-Streptomycin. 16 hours before infection, organoids were stimulated with 10 ng/ml IFN-γ (Peprotech # 300–02-100UG). Before infection domes were washed with PBS and disrupted with a pipette tip and the Matrigel was digested with 0.25% Trypsin at 37°C for 5min. After washing with media, organoids were infected with mid-log phase Shigella flexneri - pFCcGi at an MOI of approximately 1. After 45 minutes, organoids were washed twice with PBS and medium exchanged to IntestiCult medium supplemented with 100 μg/ml gentamicin (Thermo fisher # 15710064) and 0.4% L-Arabinose (Sigma #A3256–25G). After 3 hours organoids were washed and, stained with Ghost Dye™ Red 780 (Tonbo #SKU 13–0865-T100). After single-cell digestion with TrypeLE (Thermo fisher #12604013) at 37°C for 5 minutes, cells were fixed with cytofix/cytoperm (BD biosciences #554722) for 10 min, passed through a cell strainer and analyzed with a BD LSR Fortessa flow cytometer.
QUANTIFICATION AND STATISTICAL ANALYSIS
Data were analyzed using GraphPad Prism10 software and as indicated in the figure legends. Statistical parameters are reported in the figure legends and supplementary material. Statistical tests employed include unpaired Student’s t-test, Mantel-Cox test, 1-way or 2-way ANOVA with Tukey’s multiple comparison or Kruskal-Wallis test with Dunn’s corrections, to obtain the indicated p-values with ****p<0.0001, ***p<0.001, **p<0.01, ****p<0.05, ns = not significant (p>0.05).
Supplementary Material
Supplemental Videos and Spreadsheets_Table1: Differential expression IEC naïve vs infected, related to Figure 1c.
Supplemental Videos and Spreadsheets_Table2: Differential expression of ISGs in IEC naïve vs infected, related to Figure 1c.
Key resources table.
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
Antibodies | ||
anti-CD16/anti-CD32 antibody | BioLegend | Cat# 156604; RRID:AB_2783138 |
APC CD11c | BioLegend | Cat# 117310; RRID:AB_313779 |
APC CD19 | BioLegend | Cat# 152409; RRID:AB_2629838 |
APC CD3 | Thermo Fisher Scientific | Cat# 17-0031-82; RRID:AB_469315 |
APC CD5 | BioLegend | Cat# 100625; RRID:AB_2563928 |
APC F4/80 | Thermo Fisher Scientific | Cat# 17-4801-80; RRID:AB_2784647 |
APC Ly6C | BioLegend | Cat# 128016; RRID:AB_1732076 |
APC Ly6G | BioLegend | Cat# 127614; RRID:AB_2227348 |
APC NKp46 | BioLegend | Cat# 137608; RRID:AB_10612758 |
APC-Cy7 EpCAM | BioLegend | Cat# 118218 RRID:AB_2098648 |
APC-Cy7 MHCII | BioLegend | Cat# 107627; RRID:AB_1659252 |
APC-eFlour780™ CD45R | Thermo Fisher Scientific | Cat# 47-0452-82; RRID:AB_1518810 |
APC-eFlour780™ KLRG1 | Thermo Fisher Scientific | Cat# 100625; RRID:AB_2573987 |
BUV395 CD45 | BD | Cat# 749889; RRID:AB_2874129 |
BUV496 CD3 | BD | Cat# 612955 RRID:AB_2870231 |
BUV496 TCRb | BD | Cat# 749915; RRID:AB_2874154 |
BV421 CX3CR1 | BioLegend | Cat# 149023; RRID:AB_2565706 |
BV421 F4/80 | BioLegend | Cat# 123137; RRID:AB_2563102 |
BV421 NKp46 | BioLegend | Cat# 137612; RRID:AB_2563104 |
BV605 CD127 | BioLegend | Cat# 135041; RRID:AB_2572047 |
BV605 CD64 | BioLegend | Cat# 104530; RRID:AB_2629778 |
BV605 EpCAM | BioLegend | Cat# 118227; RRID:AB_2563984 |
BV785 CD45 | BioLegend | Cat# 109839; RRID:AB_2562604 |
cytofix/cytoperm | BD | Cat# 554722; RRID:AB_2869010 |
FITC CD11c | Thermo Fisher Scientific | Cat# 11-0114-82; RRID:AB_464940 |
FITC Ly6G | BD | Cat# 551460; RRID:AB_394207 |
Ghost Dye™ Red 780 | Tonbo | Cat# SKU 13-0865-T100 |
Ghost Dye™ Violet 540 | Tonbo | Cat# SKU 13-0879-T100 |
PE CD11b | Thermo Fisher Scientific | Cat# 12-0112-82; RRID:AB_2734869 |
PE CD64 | BioLegend | Cat# 139304; RRID:AB_10612740 |
PE Eomes | BioLegend | Cat# 157705; RRID:AB_2888891 |
PE-Cy5 CD11b | BioLegend | Cat# 101209; RRID:AB_312792 |
PE-Cy7 EpCAM | BioLegend | Cat# 118216; RRID:AB_1236471 |
PerCP-Cy5.5 Ly6C | Thermo Fisher Scientific | Cat# 45-5932-80; RRID:AB_2723342 |
PerCP-Cy5.5 MHCII | BioLegend | Cat# 107626; RRID:AB_2191071 |
PerCP-Cy5.5 TCRgd | BioLegend | Cat# 118117; RRID:AB_10612572 |
R718 RORyt | BD | Cat# 567362; RRID:AB_2916571 |
InVivoPlus anti-mouse IFNγ | Bio X Cell | Cat# BE0055; RRID:AB_1107694 |
InVivoPlus rat IgG1 isotype control (anti-HRP) | Bio X Cell | Cat# BE0088; RRID:AB_1107775 |
InVivoPlus anti-mouse IL-12 p40 | Bio X Cell | Cat# BE0051; RRID:AB_1107698 |
InVivoMab anti-mouse IL-18 | Bio X Cell | Cat# BE0237; RRID:AB_2687719 |
InVivoPlus anti-mouse Ly6G | Bio X Cell | Cat# BE0075-1; RRID:AB_1107721 |
InVivoMAb anti-mouse IFNAR-1 | Bio X Cell | Cat# BE0241, RRID:AB_2687723 |
InVivoPlus rat IgG2a isotype control | Bio X Cell | Cat# BE0089; RRID:AB_1107769 |
MojoSort™ Mouse CD326 (Ep-CAM) selection kit | BioLegend | Cat# 480142 |
AccuCheck Counting Beads | Thermo Fisher Scientific | Cat# PCB100 |
Bacterial and virus strains | ||
Shigella flexneri: serovar 2a 2457T strain, natural streptomycin-resistant | Mitchell et al.15 | N/A |
Shigella flexneri: serovar 2a 2457T strain ΔipaH9.8 | Lesser Lab Piro et al.72 | N/A |
Shigella flexneri: serovar 2a 2457T strain ΔipaH9.8, streptomycin-resistant | This study | N/A |
Shigella flexneri: serovar 2a 2457T strain Virulence-plasmid cured | Lesser Lab Lampel et al.5 | N/A |
Biological samples | ||
Deidentified human colonic organoids | Scott B. Snapper (see acknowledgments) | N/A |
Chemicals, peptides, and recombinant proteins | ||
Diphtheria toxin | Sigma-Aldrich | Cat# D0564 |
cOmplete™, Mini, EDTA-free Protease Inhibitor Cocktail | Sigma-Aldrich | Cat# 04693159001 |
Mouse IFN-gamma Quantikine ELISA Kit | R&D Systems | Cat# DY485-05 |
Mouse S100A8/S100A9 Heterodimer DuoSet ELISA | R&D Systems | Cat# DY8596-05 |
Mouse Lipocalin-2/NGAL DuoSet ELISA | R&D Systems | Cat# DY1857-05 |
Mouse Myeloperoxidase DuoSet ELISA | R&D Systems | Cat# DY3667 |
Mouse IL-1 beta/IL-1F2 DuoSet ELISA | R&D Systems | Cat# DY401-05 |
Mouse CXCL1/KC DuoSet ELISA | R&D Systems | Cat# DY453-05 |
Dispase-II | Sigma-Aldrich | Cat# D4693 |
DNase | Sigma-Aldrich | Cat# 11284932001 |
Liberase™ | Sigma-Aldrich | Cat# 5401127001 |
Heparin | Sigma-Aldrich | Cat# #H0878-100KU |
ACK buffer | Thermo Fisher Scientific | Cat# A1049201 |
SpCas9 2 NLS nuclease | Synthego | |
Alt-R® Cas9 Electroporation Enhancer | IDT | Cat# 1075916 |
Recombinant mouse IFN-γ | Peprotech | Cat# 315-05-100UG |
Recombinant human IFN-γ | Peprotech | Cat# 300-02-100UG |
IntestiCult™ Organoid Growth Medium | StemCell | Cat# 6010 |
Y-27632 | StemCell | Cat# 72304 |
Critical commercial assays | ||
Hemoccult blood testing kit | Pro Advantage | Cat# P080018 |
Pierce BCA protein assay kit | Thermo Fisher Scientific | Cat# 23225 |
Monarch Total RNA Miniprep Kit | NEB | Cat# T2010S |
Lonza Cell Line Solution SE | Lonza | V4XC-10323 |
Deposited data | ||
Raw and analyzed RNA-Seq data from EpCAM enriched cecum/colon cells | This paper | GEO: GSE288567 |
Experimental models: Cell lines | ||
Mouse: CT26 | Berkeley Cell Culture Facility | RRID:CVCL_7254 |
Experimental models: Organisms/strains | ||
Mouse: Nlrc4−/−: C57BL/6J-Nlrc4542stop/J | Tenthorey et al.96 | Jax# 039847 (about to be publicly released from Jax) |
Mouse: Nlrc4−/−Casp11−/−: C57BL/6J- Nlrc4542stop Casp4em13JLR/J | Roncaioli et al.16 | N/A |
Mouse: Lyz2Cre: B6.129P2-Lyz2tm1(cre)Ifo/J | The Jackson Laboratory | RRID:IMSR_JAX:004781 |
Mouse: Mrp8Cre: B6.Cg-Tg(S100A8-cre,-EGFP)1Ilw/J | The Jackson Laboratory | RRID:IMSR_JAX:021614 |
Mouse: CD64Cre: B6-Fcgr1tm2Ciphe | Bernard Malissen Scott et al.50 | N/A |
Mouse: iDTRlsl/lsl: C57BL/6-Gt(ROSA)26Sortm1(HBEGF)Awai/J | The Jackson Laboratory | RRID:IMSR_JAX:007900 |
Mouse: Ifngr1−/−: B6.129S7-Ifngr1tm1Agt/J | The Jackson Laboratory | RRID:IMSR_JAX:003288 |
Mouse: Tlr2−/−Tlr4−/−Unc93b1−/− (C57BL/6N-Unc93b1tm1(KOMP)Vlcg/Mmucd) | Barton Lab Sivick et al.52 | N/A |
Mouse: Il12−/−: B6.129S1-Il12btm1Jm/J | The Jackson Laboratory | RRID:IMSR_JAX:002693 |
Mouse: Zbtb46DTR/DTR: B6(Cg)-Zbtb46tm1(HBEGF)Mnz/J | The Jackson Laboratory | RRID:IMSR_JAX:019506 |
Mouse: GREAT reporter: B6.129S4-Ifngtm3.1Lky/J | The Jackson Laboratory | RRID:IMSR_JAX:017581 |
Oligonucleotides | ||
sgRNA targeting sequence: Gbp1 & Gbp2 #1: CCTCAGTGTCAAGCAGAACC | This study Ordered form Synthego | N/A |
sgRNA targeting sequence: Gbp1 & Gbp2 #2: GAGACTGCACAGTGGAGCCC | This study Ordered form Synthego | N/A |
sgRNA targeting sequence: Gbp5 #1: GCUUCCAACACUCAGCAACG | This study Ordered form Synthego | N/A |
sgRNA targeting sequence: Gbp5 #2: CUCAAACAUUCAAUCUACCG | This study Ordered form Synthego | N/A |
sgRNA targeting sequence: Stat1 #2: UUAAUGACGAGCUCGUGGAG | This study Ordered form Synthego | N/A |
sgRNA targeting sequence: Stat1 #2: GGAUAGACGCCCAGCCACUG | This study Ordered form Synthego | N/A |
Recombinant DNA | ||
p46 (carries the araBAD promoter driven λ red recombinase genes) | Lesser Lab Datsenko et al.104 | N/A |
pFPV-mCherry | Olivia Steele-Mortimer109 | RRID:Addgene_20956 |
pFCcGi | Sophie Helaine & David Holden108 | RRID:Addgene_59324 |
Software and algorithms | ||
FastQC | Andrews, S | www.bioinformatics.babraham.ac.uk/projects/fastqc/ |
cutadapt | Martin, M | https://cutadapt.readthedocs.io/en/stable/ |
Kallisto | Nicolas L Bray | https://pachterlab.github.io/kallisto/about |
DESeq2 | Love et al.106 | https://bioconductor.org/packages/release/bioc/html/DESeq2.html |
R | Version 4.4.1 | https://cran.rstudio.com/ |
clusterProfiler | Version 4.12.2 | https://bioconductor.org/packages/devel/bioc/html/clusterProfiler.html |
GraphPad Prism 9 | GraphPad Software | https://www.graphpad.com/ |
BioRender | BioRender Company | https://BioRender.com/64uv53d https://BioRender.com/1dzqp93 |
Other | ||
Highlights.
Neutrophils are dispensable for Shigella control in vivo.
Macrophages sense Shigella via TLRs and produce IL-12.
IL-12 induces IFN-γ, essential for controlling Shigella replication.
IFN-γ limits Shigella in intestinal epithelial cells to resolve infection in vivo.
Acknowledgment
We thank Scott B. Snapper, David Breault, Daniel Zeve, Richelle Bearup and the Organoid Core (NIH grant P30DK034854), Division of Gastroenterology, Hepatology & Nutrition, Boston Children’s Hospital, for providing deidentified human colonic organoids. Graphical abstract and model figures were created with BioRender.com
R.E.V. and G.M.B. are Howard Hughes Medical Institute Investigators and supported by Investigator Awards and Emerging Pathogens Initiative Awards from the HHMI. We also acknowledge NIH grants AI075039 and AI155634 (to R.E.V.), AI072429 (to G.M.B.), and AI169795 (to C.F.L.).
Footnotes
Declaration of interests
R.E.V. and G.M.B. are SAB members of X-biotix. The other authors declare no competing interests.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Kotloff KL, Riddle MS, Platts-Mills JA, Pavlinac P, and Zaidi AKM (2018). Shigellosis. Lancet 391, 801–812. 10.1016/S0140-6736(17)33296-8. [DOI] [PubMed] [Google Scholar]
- 2.Khalil IA, Troeger C, Blacker BF, Rao PC, Brown A, Atherly DE, Brewer TG, Engmann CM, Houpt ER, Kang G, et al. (2018). Morbidity and mortality due to Shigella and enterotoxigenic Escherichia coli diarrhoea: the Global Burden of Disease Study 1990–2016. The Lancet Infectious Diseases 18, 1229–1240. 10.1016/S1473-3099(18)30475-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Troeger C, Colombara DV, Rao PC, Khalil IA, Brown A, Brewer TG, Guerrant RL, Houpt ER, Kotloff KL, Misra K, et al. (2018). Global disability-adjusted life-year estimates of long-term health burden and undernutrition attributable to diarrhoeal diseases in children younger than 5 years. Lancet Glob Health 6, e255–e269. 10.1016/S2214-109X(18)30045-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Libby TE, Delawalla MLM, Al-Shimari F, MacLennan CA, Vannice KS, and Pavlinac PB (2023). Consequences of Shigella infection in young children: a systematic review. Int J Infect Dis 129, 78–95. 10.1016/j.ijid.2023.01.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Lampel KA, Formal SB, and Maurelli AT (2018). A Brief History of Shigella. EcoSal Plus 8, 10.1128/ecosalplus.ESP-0006-2017. https://doi.org/10.1128/ecosalplus.ESP-0006-2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Sansonetti PJ, Arondel J, Cantey JR, Prévost MC, and Huerre M (1996). Infection of rabbit Peyer’s patches by Shigella flexneri: effect of adhesive or invasive bacterial phenotypes on follicle-associated epithelium. Infection and Immunity 64, 2752–2764. 10.1128/iai.64.7.2752-2764.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Schnupf P, and Sansonetti PJ (2019). Shigella Pathogenesis: New Insights through Advanced Methodologies. Microbiology Spectrum 7. 10.1128/MICROBIOLSPEC.BAI-0023-2019/ASSET/56EB7020-056E-4F29-ADB0-9A3AC62ED9AE/ASSETS/GRAPHIC/BAI-0023-2019-FIG4.GIF. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Schroeder GN, and Hilbi H (2008). Molecular pathogenesis of Shigella spp.: controlling host cell signaling, invasion, and death by type III secretion. Clin Microbiol Rev 21, 134–156. 10.1128/CMR.00032-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ashida H, Mimuro H, and Sasakawa C (2015). Shigella Manipulates Host Immune Responses by Delivering Effector Proteins with Specific Roles. Front Immunol 6, 219. 10.3389/fimmu.2015.00219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Bernardini ML, Mounier J, d’Hauteville H, Coquis-Rondon M, and Sansonetti PJ (1989). Identification of icsA, a plasmid locus of Shigella flexneri that governs bacterial intra- and intercellular spread through interaction with F-actin. Proc Natl Acad Sci U S A 86, 3867–3871. 10.1073/pnas.86.10.3867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Goldberg MB, and Theriot JA (1995). Shigella flexneri surface protein IcsA is sufficient to direct actin-based motility. Proc Natl Acad Sci U S A 92, 6572–6576. 10.1073/pnas.92.14.6572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mattock E, and Blocker AJ (2017). How do the virulence factors of Shigella work together to cause disease? Frontiers in Cellular and Infection Microbiology 7. 10.3389/fcimb.2017.00064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Formal SB (1989). Inoculum Size in Shigellosis and Implications For Expected Mode of Transmission. Journal of Infectious Diseases 159, 1126–1128. 10.1093/infdis/159.6.1126. [DOI] [PubMed] [Google Scholar]
- 14.Alphonse N, and Odendall C (2023). Animal models of shigellosis: a historical overview. Curr Opin Immunol 85, 102399. 10.1016/j.coi.2023.102399. [DOI] [PubMed] [Google Scholar]
- 15.Mitchell PS, Roncaioli JL, Turcotte EA, Goers L, Chavez RA, Lee AY, Lesser CF, Rauch I, and Vance RE (2020). NAIP-NLRC4-deficient mice are susceptible to shigellosis. eLife 9, 1–25. 10.7554/ELIFE.59022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Roncaioli JL, Babirye JP, Chavez RA, Liu FL, Turcotte EA, Lee AY, Lesser CF, and Vance RE (2023). A hierarchy of cell death pathways confers layered resistance to shigellosis in mice. eLife 12, e83639. 10.7554/eLife.83639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kofoed EM, and Vance RE (2011). Innate immune recognition of bacterial ligands by NAIPs dictates inflammasome specificity. Nature 477, 592–595. 10.1038/nature10394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Suzuki S, Franchi L, He Y, Muñoz-Planillo R, Mimuro H, Suzuki T, Sasakawa C, and Núñez G (2014). Shigella Type III Secretion Protein MxiI Is Recognized by Naip2 to Induce Nlrc4 Inflammasome Activation Independently of Pkcδ. PLoS Pathog 10, e1003926. 10.1371/journal.ppat.1003926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Yang J, Zaho Y, Shi J, and Shao F (2013). Human NAIP and mouse NAIP1 recognize bacterial type III secretion needle protein for inflammasome activation. Proceedings of the National Academy of Sciences of the United States of America 110. 10.1073/pnas.1306376110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kayagaki N, Stowe IB, Lee BL, O’Rourke K, Anderson K, Warming S, Cuellar T, Haley B, Roose-Girma M, Phung QT, et al. (2015). Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 526, 666–671. 10.1038/nature15541. [DOI] [PubMed] [Google Scholar]
- 21.Hagar JA, and Miao EA (2014). Detection of cytosolic bacteria by inflammatory caspases. Curr Opin Microbiol 0, 61–66. 10.1016/j.mib.2013.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Oh C, Verma A, Hafeez M, Hogland B, and Aachoui Y (2021). Shigella OspC3 suppresses murine cytosolic LPS sensing. iScience 24, 102910. 10.1016/j.isci.2021.102910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Li Z, Liu W, Fu J, Cheng S, Xu Y, Wang Z, Liu X, Shi X, Liu Y, Qi X, et al. (2021). Shigella evades pyroptosis by arginine ADP-riboxanation of caspase-11. Nature 599, 290–295. 10.1038/s41586-021-04020-1. [DOI] [PubMed] [Google Scholar]
- 24.Wlodarska M, Willing B, Keeney KM, Menendez A, Bergstrom KS, Gill N, Russell SL, Vallance BA, and Finlay BB (2011). Antibiotic Treatment Alters the Colonic Mucus Layer and Predisposes the Host to Exacerbated Citrobacter rodentium-Induced Colitis. Infect Immun 79, 1536–1545. 10.1128/IAI.01104-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Becattini S, Littmann ER, Carter RA, Kim SG, Morjaria SM, Ling L, Gyaltshen Y, Fontana E, Taur Y, Leiner IM, et al. (2017). Commensal microbes provide first line defense against Listeria monocytogenes infection. J Exp Med 214, 1973–1989. 10.1084/jem.20170495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Barthel M, Hapfelmeier S, Quintanilla-Martínez L, Kremer M, Rohde M, Hogardt M, Pfeffer K, Rüssmann H, and Hardt W-D (2003). Pretreatment of Mice with Streptomycin Provides a Salmonella enterica Serovar Typhimurium Colitis Model That Allows Analysis of Both Pathogen and Host. Infect Immun 71, 2839–2858. 10.1128/IAI.71.5.2839-2858.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Rauch I, Deets KA, Ji DX, von Moltke J, Tenthorey JL, Lee AY, Philip NH, Ayres JS, Brodsky IE, Gronert K, et al. (2017). NAIP-NLRC4 Inflammasomes Coordinate Intestinal Epithelial Cell Expulsion with Eicosanoid and IL-18 Release via Activation of Caspase-1 and −8. Immunity 46, 649–659. 10.1016/j.immuni.2017.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sellin ME, Müller AA, Felmy B, Dolowschiak T, Diard M, Tardivel A, Maslowski KM, and Hardt W-D (2014). Epithelium-Intrinsic NAIP/NLRC4 Inflammasome Drives Infected Enterocyte Expulsion to Restrict Salmonella Replication in the Intestinal Mucosa. Cell Host & Microbe 16, 237–248. 10.1016/j.chom.2014.07.001. [DOI] [PubMed] [Google Scholar]
- 29.Naseer N, Zhang J, Bauer R, Constant DA, Nice TJ, Brodsky IE, Rauch I, and Shin S (2022). Salmonella enterica Serovar Typhimurium Induces NAIP/NLRC4- and NLRP3/ASC-Independent, Caspase-4-Dependent Inflammasome Activation in Human Intestinal Epithelial Cells. Infection and Immunity 90. 10.1128/iai.00663-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Luchetti G, Roncaioli JL, Chavez RA, Schubert AF, Kofoed EM, Reja R, Cheung TK, Liang Y, Webster JD, Lehoux I, et al. (2021). Shigella ubiquitin ligase IpaH7.8 targets gasdermin D for degradation to prevent pyroptosis and enable infection. Cell Host and Microbe 29, 1521–1530.e10. 10.1016/j.chom.2021.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kobayashi T, Ogawa M, Sanada T, Mimuro H, Kim M, Ashida H, Akakura R, Yoshida M, Kawalec M, Reichhart J-M, et al. (2013). The Shigella OspC3 Effector Inhibits Caspase-4, Antagonizes Inflammatory Cell Death, and Promotes Epithelial Infection. Cell Host & Microbe 13, 570–583. 10.1016/j.chom.2013.04.012. [DOI] [PubMed] [Google Scholar]
- 32.Mou X, Souter S, Du J, Reeves AZ, and Lesser CF (2018). Synthetic bottom-up approach reveals the complex interplay of Shigella effectors in regulation of epithelial cell death. Proceedings of the National Academy of Sciences 115, 6452–6457. 10.1073/pnas.1801310115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Turcotte EA, Kim K, Eislmayr KD, Goers L, Mitchell PS, Lesser CF, and Vance RE (2025). Shigella OspF blocks rapid p38-dependent priming of the NAIP–NLRC4 inflammasome. Preprint at bioRxiv, 10.1101/2025.02.01.636075 https://doi.org/10.1101/2025.02.01.636075. [DOI] [Google Scholar]
- 34.Hilbi H, Chen Y, Thirumalai K, and Zychlinsky A (1997). The interleukin 1beta-converting enzyme, caspase 1, is activated during Shigella flexneri-induced apoptosis in human monocyte-derived macrophages. Infect Immun 65, 5165–5170. 10.1128/iai.65.12.5165-5170.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Raqib R, Wretlind B, Andersson J, and Lindberg AA (1995). Cytokine secretion in acute shigellosis is correlated to disease activity and directed more to stool than to plasma. J Infect Dis 171, 376–384. 10.1093/infdis/171.2.376. [DOI] [PubMed] [Google Scholar]
- 36.Levine MM, Kotloff KL, Barry EM, Pasetti MF, and Sztein MB (2007). Clinical trials of Shigella vaccines: two steps forward and one step back on a long, hard road. Nat Rev Microbiol 5, 540–553. 10.1038/nrmicro1662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Anand BS, Malhotra V, Bhattacharya SK, Datta P, Datta D, Sen D, Bhattacharya MK, Mukherjee PP, and Pal SC (1986). Rectal histology in acute bacillary dysentery. Gastroenterology 90, 654–660. 10.1016/0016-5085(86)91120-0. [DOI] [PubMed] [Google Scholar]
- 38.Skerniskyte J, Mulet C, André AC, Anderson MC, Injarabian L, Buck A, Prade VM, Sansonetti PJ, Reibel-Foisset S, Walch AK, et al. Ascorbate deficiency increases progression of shigellosis in guinea pigs and mice infection models. Gut Microbes 15, 2271597. 10.1080/19490976.2023.2271597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Shim D-H, Suzuki T, Chang S-Y, Park S-M, Sansonetti PJ, Sasakawa C, and Kweon M-N (2007). New Animal Model of Shigellosis in the Guinea Pig: Its Usefulness for Protective Efficacy Studies. The Journal of Immunology 178, 2476–2482. 10.4049/jimmunol.178.4.2476. [DOI] [PubMed] [Google Scholar]
- 40.Zychlinsky A, Prevost MC, and Sansonetti PJ (1992). Shigella flexneri induces apoptosis in infected macrophages. Nature 358, 167–169. 10.1038/358167a0. [DOI] [PubMed] [Google Scholar]
- 41.Zychlinsky A, Thirumalai K, Arondel J, Cantey JR, Aliprantis AO, and Sansonetti PJ (1996). In vivo apoptosis in Shigella flexneri infections. Infection and Immunity 64, 5357–5365. 10.1128/iai.64.12.5357-5365.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Chen Y, Smith MR, Thirumalai K, and Zychlinsky A (1996). A bacterial invasin induces macrophage apoptosis by binding directly to ICE. EMBO J 15, 3853–3860. [PMC free article] [PubMed] [Google Scholar]
- 43.Ashida H, Mimuro H, Ogawa M, Kobayashi T, Sanada T, Kim M, and Sasakawa C (2011). Cell death and infection: a double-edged sword for host and pathogen survival. J Cell Biol 195, 931–942. 10.1083/jcb.201108081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Jehl SP, Nogueira CV, Zhang X, and Starnbach MN (2012). IFNγ Inhibits the Cytosolic Replication of Shigella flexneri via the Cytoplasmic RNA Sensor RIG-I. PLOS Pathogens 8, e1002809. 10.1371/journal.ppat.1002809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kutsch M, Sistemich L, Lesser CF, Goldberg MB, Herrmann C, and Coers J (2020). Direct binding of polymeric GBP1 to LPS disrupts bacterial cell envelope functions. EMBO J 39, e104926. 10.15252/embj.2020104926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dickinson MS, Kutsch M, Sistemich L, Hernandez D, Piro AS, Needham D, Lesser CF, Herrmann C, and Coers J LPS-aggregating proteins GBP1 and GBP2 are each sufficient to enhance caspase-4 activation both in cellulo and in vitro. Proc Natl Acad Sci U S A 120, e2216028120. 10.1073/pnas.2216028120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Nakandakari-Higa S, Walker S, Canesso MCC, van der Heide V, Chudnovskiy A, Kim D-Y, Jacobsen JT, Parsa R, Bilanovic J, Parigi SM, et al. (2024). Universal recording of immune cell interactions in vivo. Nature 627, 399–406. 10.1038/s41586-024-07134-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Li P, Jiang W, Yu Q, Liu W, Zhou P, Li J, Xu J, Xu B, Wang F, and Shao F (2017). Ubiquitination and degradation of GBPs by a Shigella effector to suppress host defence. Nature 551, 378–383. 10.1038/nature24467. [DOI] [PubMed] [Google Scholar]
- 49.Passegué E, Wagner EF, and Weissman IL (2004). JunB deficiency leads to a myeloproliferative disorder arising from hematopoietic stem cells. Cell 119, 431–443. 10.1016/j.cell.2004.10.010. [DOI] [PubMed] [Google Scholar]
- 50.Scott CL, T’Jonck W, Martens L, Todorov H, Sichien D, Soen B, Bonnardel J, De Prijck S, Vandamme N, Cannoodt R, et al. (2018). The Transcription Factor ZEB2 Is Required to Maintain the Tissue-Specific Identities of Macrophages. Immunity 49, 312–325.e5. 10.1016/j.immuni.2018.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Price AE, Shamardani K, Lugo KA, Deguine J, Roberts AW, Lee BL, and Barton GM (2018). A Map of Toll-like Receptor Expression in the Intestinal Epithelium Reveals Distinct Spatial, Cell Type-Specific, and Temporal Patterns. Immunity 49, 560–575.e6. 10.1016/j.immuni.2018.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sivick KE, Arpaia N, Reiner GL, Lee BL, Russell BR, and Barton GM (2014). Toll-like receptor-deficient mice reveal how innate immune signaling influences Salmonella virulence strategies. Cell Host Microbe 15, 203–213. 10.1016/j.chom.2014.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Castro F, Cardoso AP, Gonçalves RM, Serre K, and Oliveira MJ (2018). Interferon-gamma at the crossroads of tumor immune surveillance or evasion. Frontiers in Immunology 9, 847. 10.3389/fimmu.2018.00847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lee AJ, Chen B, Chew MV, Barra NG, Shenouda MM, Nham T, van Rooijen N, Jordana M, Mossman KL, Schreiber RD, et al. (2017). Inflammatory monocytes require type I interferon receptor signaling to activate NK cells via IL-18 during a mucosal viral infection. J Exp Med 214, 1153–1167. 10.1084/jem.20160880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Reinhardt RL, Liang H-E, and Locksley RM (2009). Cytokine-secreting follicular T cells shape the antibody repertoire. Nat Immunol 10, 385–393. 10.1038/ni.1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Reinhardt RL, Liang H-E, Bao K, Price AE, Mohrs M, Kelly BL, and Locksley RM (2015). A Novel Model for IFN-γ–Mediated Autoinflammatory Syndromes. J Immunol 194, 2358–2368. 10.4049/jimmunol.1401992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Fattinger SA, Geiser P, Samperio Ventayol P, Di Martino ML, Furter M, Felmy B, Bakkeren E, Hausmann A, Barthel-Scherrer M, Gül E, et al. (2021). Epithelium-autonomous NAIP/NLRC4 prevents TNF-driven inflammatory destruction of the gut epithelial barrier in Salmonella-infected mice. Mucosal Immunology 14, 615–629. 10.1038/s41385-021-00381-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ventayol PS, Geiser P, Di Martino ML, Florbrant A, Fattinger SA, Walder N, Sima E, Shao F, Gekara NO, Sundbom M, et al. (2021). Bacterial detection by NAIP/NLRC4 elicits prompt contractions of intestinal epithelial cell layers. Proceedings of the National Academy of Sciences of the United States of America 118. 10.1073/pnas.2013963118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ottenhoff TH, Kumararatne D, and Casanova JL (1998). Novel human immunodeficiencies reveal the essential role of type-I cytokines in immunity to intracellular bacteria. Immunol Today 19, 491–494. 10.1016/s0167-5699(98)01321-8. [DOI] [PubMed] [Google Scholar]
- 60.Vordermeier HM, Chambers MA, Cockle PJ, Whelan AO, Simmons J, and Hewinson RG (2002). Correlation of ESAT-6-specific gamma interferon production with pathology in cattle following Mycobacterium bovis BCG vaccination against experimental bovine tuberculosis. Infect Immun 70, 3026–3032. 10.1128/IAI.70.6.3026-3032.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Flynn JL, Chan J, Triebold KJ, Dalton DK, Stewart TA, and Bloom BR (1993). An essential role for interferon gamma in resistance to Mycobacterium tuberculosis infection. J Exp Med 178, 2249–2254. 10.1084/jem.178.6.2249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Dussurget O, Bierne H, and Cossart P (2014). The bacterial pathogen Listeria monocytogenes and the interferon family: type I, type II and type III interferons. Front Cell Infect Microbiol 4, 50. 10.3389/fcimb.2014.00050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Yang C, McDermot DS, Pasricha S, Bedoui S, Lenz LL, van Driel IR, and Hartland EL (2020). Interferon γ receptor downregulation facilitates Legionella survival in alveolar macrophages. J Leukoc Biol 107, 273–284. 10.1002/JLB.4MA1019-152R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Byrd TF, and Horwitz MA (1989). Interferon gamma-activated human monocytes downregulate transferrin receptors and inhibit the intracellular multiplication of Legionella pneumophila by limiting the availability of iron. J Clin Invest 83, 1457–1465. 10.1172/JCI114038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Cai S, Batra S, Langohr I, Iwakura Y, and Jeyaseelan S (2016). IFN-γ induction by neutrophil-derived IL-17A homodimer augments pulmonary antibacterial defense. Mucosal Immunol 9, 718–729. 10.1038/mi.2015.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Wandel MP, Kim B-H, Park E-S, Boyle KB, Nayak K, Lagrange B, Herod A, Henry T, Zilbauer M, Rohde J, et al. (2020). Guanylate-binding proteins convert cytosolic bacteria into caspase-4 signaling platforms. Nat Immunol 21, 880–891. 10.1038/s41590-020-0697-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Way SS, Borczuk AC, Dominitz R, and Goldberg MB (1998). An essential role for gamma interferon in innate resistance to Shigella flexneri infection. Infect Immun 66, 1342–1348. 10.1128/IAI.66.4.1342-1348.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wandel MP, Pathe C, Werner EI, Ellison CJ, Boyle KB, von der Malsburg A, Rohde J, and Randow F (2017). GBPs Inhibit Motility of Shigella flexneri but Are Targeted for Degradation by the Bacterial Ubiquitin Ligase IpaH9.8. Cell Host & Microbe 22, 507. 10.1016/J.CHOM.2017.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Goers L, Kim K, Stedman TC, Canning PJ, Mou X, Ernst NH, Coers J, and Lesser CF (2023). Shigella IpaH9.8 limits GBP1-dependent LPS release from intracytosolic bacteria to suppress caspase-4 activation. Proceedings of the National Academy of Sciences 120, e2218469120. 10.1073/pnas.2218469120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Helbig KJ, Teh MY, Crosse KM, Monson EA, Smith M, Tran EN, Standish AJ, Morona R, and Beard MR (2019). The interferon stimulated gene viperin, restricts Shigella. flexneri in vitro. Sci Rep 9, 15598. 10.1038/s41598-019-52130-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Saavedra-Sanchez L, Dickinson MS, Apte S, Zhang Y, De Jong M, Skavicus S, Heaton NS, Alto NM, and Coers J (2024). The Shigella flexneri effector IpaH1.4 facilitates RNF213 degradation and protects cytosolic bacteria against interferon-induced ubiquitylation. Preprint, 10.1101/2024.09.05.611450 https://doi.org/10.1101/2024.09.05.611450. [DOI] [Google Scholar]
- 72.Piro AS, Hernandez D, Luoma S, Feeley EM, Finethy R, Yirga A, Frickel EM, Lesser CF, and Coers J (2017). Detection of Cytosolic Shigella flexneri via a C-Terminal Triple-Arginine Motif of GBP1 Inhibits Actin-Based Motility. mBio 8, e01979–17. 10.1128/mBio.01979-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Gaudet RG, Zhu S, Halder A, Kim B-H, Bradfield CJ, Huang S, Xu D, Mamiñska A, Nguyen TN, Lazarou M, et al. (2021). A human apolipoprotein L with detergent-like activity kills intracellular pathogens. Science 373, eabf8113. 10.1126/science.abf8113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Santos JC, Boucher D, Schneider LK, Demarco B, Dilucca M, Shkarina K, Heilig R, Chen KW, Lim RYH, and Broz P (2020). Human GBP1 binds LPS to initiate assembly of a caspase-4 activating platform on cytosolic bacteria. Nat Commun 11, 3276. 10.1038/s41467-020-16889-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Randow F, MacMicking JD, and James LC (2013). Cellular self-defense: How cell-autonomous immunity protects against pathogens. Science 340, 701–706. 10.1126/science.1233028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Piro AS, Hernandez D, Luoma S, Feeley EM, Finethy R, Yirga A, Frickel EM, Lesser CF, and Coers J (2017). Detection of Cytosolic Shigella flexneri via a C-Terminal Triple-Arginine Motif of GBP1 Inhibits Actin-Based Motility. mBio 8, e01979–17. 10.1128/mBio.01979-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Naydenova K, Boyle KB, Pathe C, Pothukuchi P, Crespillo-Casado A, Scharte F, Hammoudi P-M, Otten EG, and Randow F (2024). Shigella flexneri evades LPS ubiquitylation through IpaH1.4-mediated degradation of RNF213. Preprint, 10.1101/2024.09.24.614686 https://doi.org/10.1101/2024.09.24.614686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Alphonse N, Dickenson RE, and Odendall C (2021). Interferons: Tug of War Between Bacteria and Their Host. Frontiers in Cellular and Infection Microbiology 11. 10.3389/fcimb.2021.624094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.François M, Le Cabec V, Dupont M-A, Sansonetti PJ, and Maridonneau-Parini I (2000). Induction of Necrosis in Human Neutrophils by Shigella flexneri Requires Type III Secretion, IpaB and IpaC Invasins, and Actin Polymerization. Infect Immun 68, 1289–1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Eilers B, Mayer-Scholl A, Walker T, Tang C, Weinrauch Y, and Zychlinsky A (2010). Neutrophil antimicrobial proteins enhance Shigella flexneri adhesion and invasion. Cellular Microbiology 12, 1134–1143. 10.1111/j.1462-5822.2010.01459.x. [DOI] [PubMed] [Google Scholar]
- 81.Fournier BM, and Parkos CA (2012). The role of neutrophils during intestinal inflammation. Mucosal Immunol 5, 354–366. 10.1038/mi.2012.24. [DOI] [PubMed] [Google Scholar]
- 82.Suzuki T, Franchi L, Toma C, Ashida H, Ogawa M, Yoshikawa Y, Mimuro H, Inohara N, Sasakawa C, and Nuñez G (2007). Differential regulation of caspase-1 activation, pyroptosis, and autophagy via Ipaf and ASC in Shigella-infected macrophages. PLoS pathogens 3. 10.1371/journal.ppat.0030111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Arizmendi O, Picking WD, and Picking WL (2016). Macrophage Apoptosis Triggered by IpaD from Shigella flexneri. Infect Immun 84, 1857–1865. 10.1128/IAI.01483-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Gomes MC, Brokatzky D, and Mostowy S (2024). Shigella Induces Pro-Inflammatory Macrophages that Cooperate with Neutrophils to Protect from Secondary Infection. Preprint at Social Science Research Network, 10.2139/ssrn.4981730 https://doi.org/10.2139/ssrn.4981730. [DOI] [Google Scholar]
- 85.Maculins T, Verschueren E, Hinkle T, Choi M, Chang P, Chalouni C, Rao S, Kwon Y, Lim J, Katakam AK, et al. (2021). Multiplexed proteomics of autophagy-deficient murine macrophages reveals enhanced antimicrobial immunity via the oxidative stress response. eLife 10, e62320. 10.7554/eLife.62320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Girardin SE, Tournebize R, Mavris M, Page AL, Li X, Stark GR, Bertin J, DiStefano PS, Yaniv M, Sansonetti PJ, et al. (2001). CARD4/Nod1 mediates NF-kappaB and JNK activation by invasive Shigella flexneri. EMBO Rep 2, 736–742. 10.1093/embo-reports/kve155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.García-Weber D, Dangeard A-S, Cornil J, Thai L, Rytter H, Zamyatina A, Mulard LA, and Arrieumerlou C (2018). ADP-heptose is a newly identified pathogen-associated molecular pattern of Shigella flexneri. EMBO reports 19, e46943. 10.15252/embr.201846943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Oppmann B, Lesley R, Blom B, Timans JC, Xu Y, Hunte B, Vega F, Yu N, Wang J, Singh K, et al. (2000). Novel p19 Protein Engages IL-12p40 to Form a Cytokine, IL-23, with Biological Activities Similar as Well as Distinct from IL-12. Immunity 13, 715–725. 10.1016/S1074-7613(00)00070-4. [DOI] [PubMed] [Google Scholar]
- 89.Awoniyi M, Miller SI, Wilson CB, Hajjar AM, and Smith KD (2012). Homeostatic Regulation of Salmonella-Induced Mucosal Inflammation and Injury by IL-23. PLoS One 7, e37311. 10.1371/journal.pone.0037311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.El-Behi M, Ciric B, Dai H, Yan Y, Cullimore M, Safavi F, Zhang G-X, Dittel BN, and Rostami A (2011). The encephalitogenicity of T(H)17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nat Immunol 12, 568–575. 10.1038/ni.2031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Zheng Y, Danilenko DM, Valdez P, Kasman I, Eastham-Anderson J, Wu J, and Ouyang W (2007). Interleukin-22, a T(H)17 cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature 445, 648–651. 10.1038/nature05505. [DOI] [PubMed] [Google Scholar]
- 92.Ghoreschi K, Laurence A, Yang X-P, Tato CM, McGeachy MJ, Konkel J, Ramos HL, Wei L, Davidson T, Bouladoux N, et al. (2010). Generation of Pathogenic Th17 Cells in the Absence of TGF-β Signaling. Nature 467, 967–971. 10.1038/nature09447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Moschen AR, Tilg H, and Raine T (2019). IL-12, IL-23 and IL-17 in IBD: immunobiology and therapeutic targeting. Nat Rev Gastroenterol Hepatol 16, 185–196. 10.1038/s41575-018-0084-8. [DOI] [PubMed] [Google Scholar]
- 94.Floss DM, Moll JM, and Scheller J (2020). IL-12 and IL-23—Close Relatives with Structural Homologies but Distinct Immunological Functions. Cells 9, 2184. 10.3390/cells9102184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Teng MWL, Bowman EP, McElwee JJ, Smyth MJ, Casanova J-L, Cooper AM, and Cua DJ (2015). IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nat Med 21, 719–729. 10.1038/nm.3895. [DOI] [PubMed] [Google Scholar]
- 96.Tenthorey JL, Chavez RA, Thompson TW, Deets KA, Vance RE, and Rauch I (2020). NLRC4 inflammasome activation is NLRP3- and phosphorylation-independent during infection and does not protect from melanoma. Journal of Experimental Medicine 217, e20191736. 10.1084/jem.20191736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Clausen BE, Burkhardt C, Reith W, Renkawitz R, and Förster I (1999). Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Research 8, 265–277. 10.1023/A:1008942828960. [DOI] [PubMed] [Google Scholar]
- 98.Buch T, Heppner FL, Tertilt C, Heinen TJAJ, Kremer M, Wunderlich FT, Jung S, and Waisman A (2005). A Cre-inducible diphtheria toxin receptor mediates cell lineage ablation after toxin administration. Nature Methods 2, 419–426. 10.1038/nmeth762. [DOI] [PubMed] [Google Scholar]
- 99.Huang S, Hendriks W, Althage A, Hemmi S, Bluethmann H, Kamijo R, Vilcek J, Zinkernagel RM, and Aguet M (1993). Immune response in mice that lack the interferon-gamma receptor. Science 259, 1742–1745. 10.1126/science.8456301. [DOI] [PubMed] [Google Scholar]
- 100.Magram J, Connaughton SE, Warrier RR, Carvajal DM, Wu C, Ferrante J, Stewart C, Sarmiento U, Faherty DA, and Gately MK (1996). IL-12-Deficient Mice Are Defective in IFNγ Production and Type 1 Cytokine Responses. Immunity 4, 471–481. 10.1016/S1074-7613(00)80413-6. [DOI] [PubMed] [Google Scholar]
- 101.Meredith MM, Liu K, Darrasse-Jeze G, Kamphorst AO, Schreiber HA, Guermonprez P, Idoyaga J, Cheong C, Yao K-H, Niec RE, et al. (2012). Expression of the zinc finger transcription factor zDC (Zbtb46, Btbd4) defines the classical dendritic cell lineage. J Exp Med 209, 1153–1165. 10.1084/jem.20112675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Valenzuela DM, Murphy AJ, Frendewey D, Gale NW, Economides AN, Auerbach W, Poueymirou WT, Adams NC, Rojas J, Yasenchak J, et al. (2003). High-throughput engineering of the mouse genome coupled with high-resolution expression analysis. Nat Biotechnol 21, 652–659. 10.1038/nbt822. [DOI] [PubMed] [Google Scholar]
- 103.Hosaka T, Tamehiro N, Chumpolkulwong N, Hori-Takemoto C, Shirouzu M, Yokoyama S, and Ochi K (2004). The novel mutation K87E in ribosomal protein S12 enhances protein synthesis activity during the late growth phase in Escherichia coli. Mol Genet Genomics 271, 317–324. 10.1007/s00438-004-0982-z. [DOI] [PubMed] [Google Scholar]
- 104.Datsenko KA, and Wanner BL (2000). One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proceedings of the National Academy of Sciences 97, 6640–6645. 10.1073/pnas.120163297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Gracz AD, Puthoff BJ, and Magness ST (2012). Identification, isolation, and culture of intestinal epithelial stem cells from murine intestine. Methods in Molecular Biology 879, 89–107. 10.1007/978-1-61779-815-3_6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Love MI, Huber W, and Anders S (2014). Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 15, 550. 10.1186/s13059-014-0550-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Sato T, Stange DE, Ferrante M, Vries RGJ, van Brink Es, J.H., van den Houdt S., van WJ, Pronk A, Gorp J, Siersema PD, et al. (2011). Long-term Expansion of Epithelial Organoids From Human Colon, Adenoma, Adenocarcinoma, and Barrett’s Epithelium. Gastroenterology 141, 1762–1772. 10.1053/j.gastro.2011.07.050. [DOI] [PubMed] [Google Scholar]
- 108.Figueira R, Watson KG, Holden DW, and Helaine S (2013). Identification of Salmonella Pathogenicity Island-2 Type III Secretion System Effectors Involved in Intramacrophage Replication of S. enterica Serovar Typhimurium: Implications for Rational Vaccine Design. mBio 4, e00065–13. 10.1128/mBio.00065-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Drecktrah D, Levine-Wilkinson S, Dam T, Winfree S, Knodler LA, Schroer TA, and Steele-Mortimer O (2008). Dynamic behavior of Salmonella-induced membrane tubules in epithelial cells. Traffic 9, 2117–2129. 10.1111/j.1600-0854.2008.00830.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Eislmayr K (2025). Graphical Abstract. Created in BioRender. https://BioRender.com/64uv53d [Google Scholar]
- 111.Eislmayr K (2025). Figure 3. The responsiveness of non-hematopoietic cells to IFN-γ is crucial for controlling Shigella infection. Created in BioRender. https://BioRender.com/1dzqp93 [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplemental Videos and Spreadsheets_Table1: Differential expression IEC naïve vs infected, related to Figure 1c.
Supplemental Videos and Spreadsheets_Table2: Differential expression of ISGs in IEC naïve vs infected, related to Figure 1c.
Data Availability Statement
Raw bulk RNA-sequencing data are deposited in the NCBI Gene Expression Omnibus: GSE288567.