Abstract
Yersinia are gram-negative zoonotic bacteria that use a type III secretion system (T3SS) to inject Yersinia outer proteins (Yops) into the host cytosol to subvert essential components of innate immune signaling. However, Yersinia virulence activities can elicit activation of inflammasomes, which lead to inflammatory cell death and cytokine release to contain infection. Yersinia activation and evasion of inflammasomes have been characterized in murine macrophages but remain poorly defined in human cells, particularly intestinal epithelial cells (IECs), a primary site of intestinal Yersinia infection. In contrast to murine macrophages, we find that in both human IECs and macrophages, Yersinia pseudotuberculosis T3SS effectors enable evasion of the caspase-4 inflammasome, which senses cytosolic lipopolysaccharide (LPS). The antiphagocytic YopE and YopH, as well as the translocation regulator YopK, were collectively responsible for evading inflammasome activation, in part by inhibiting Yersinia internalization mediated by YadA and β1-integrin signaling. These data provide insight into the mechanisms of Yersinia-mediated inflammasome activation and evasion in human cells, and reveal species-specific differences underlying regulation of inflammasome responses to Yersinia.
Introduction
Innate immunity is a critical component of host defense and employs pattern recognition receptors (PRRs) to detect pathogen-associated molecular patterns (PAMPs) (1, 2). Specific cytosolic PRRs in the Nucleotide-binding domain and Leucine-rich Repeat-containing protein (NLR) family induce the formation and activation of multimeric immune complexes known as inflammasomes in response to cytosolic PAMPs and virulence-associated activities (3–5). The NLRP3 inflammasome is activated by a variety of stimuli, including potassium efflux downstream of bacterial-induced pore formation (5–9), whereas the NAIP-NLRC4 inflammasome responds to bacterial flagellin and components of bacterial type III secretion systems (T3SS) (10–18). Inflammasomes recruit and activate caspase-1, which in turn processes members of the IL-1 family of cytokines and the pore-forming protein GSDMD into their active forms (19–23). In addition to these canonical inflammasome pathways, a non-canonical caspase-4 inflammasome in humans, and orthologous caspase-11 inflammasome in mice, responds to cytosolic lipopolysaccharide (LPS) during gram-negative bacterial infection to initiate GSDMD processing, pore formation, and cytokine release (24–31). Consequently, inflammasome activation leads to release of active IL-1 family cytokines, specifically IL-1β and IL-18, and an inflammatory form of cell death known as pyroptosis, that collectively amplify immune signaling and promote anti-bacterial defense (32). In parallel, bacterial infection triggers other programmed cell death pathways, including apoptosis and necroptosis, thereby providing multiple layers of defense against bacterial pathogens (33, 34).
The enteric pathogenic Yersiniae, Yersinia pseudotuberculosis and Y. enterocolitica, express a conserved T3SS that injects Yersinia outer proteins (Yops) into target cells (35). T3SS-injected Yops manipulate host cellular pathways to promote infection, but also activate and inhibit effector-triggered cell death responses. For example, in murine macrophages, YopJ-mediated suppression of NF-kB signaling induces apoptosis, whereas YopE-mediated disruption of cytoskeletal dynamics activates the pyrin inflammasome and hyper-translocation of T3SS components in the absence of YopK activates the NLRP3 and caspase-11 inflammasomes (36–45). Notably, Yersinia utilizes two other effectors, YopM and YopK, to evade YopE-triggered pyrin and T3SS-triggered NLRP3/caspase-11-inflammasome activation respectively and promote infection (42, 43, 45, 46). Collectively, these virulence strategies manipulate the host in order to facilitate colonization and evade ensuing effector-triggered immunity (47).
Inflammasome responses to Yersinia have primarily been studied in murine macrophages. However, fundamental differences exist between human and murine inflammasome responses to bacterial infection that may impact pathogenesis and disease severity in the host, including ligand recognition, virulence-driven immune suppression, and inflammasome component expression (48–68). Furthermore, beyond phagocytic cells, inflammasomes are expressed in multiple cell types, including intestinal epithelial cells (IECs) (69), which are a primary site of infection and interact significantly with enteric pathogens during intestinal colonization. While specialized IECs known as M cells, which overlie intestinal immune compartments called Peyer’s patches, are considered the primary point of bacterial invasion of the intestinal epithelial barrier (70)(71)(72)(73), Yersinia microcolonies are frequently found outside of Peyer’s patches within submucosal pyogranulomas and in close proximity to non-M cell IECs (74). Importantly, many enteric bacterial pathogens use secreted virulence factors to inhibit IEC death in order to preserve their replicative niche during early infection (75, 76). Y. enterocolitica uses two anti-phagocytic Yops, YopE and YopH, to evade caspase-1 and NLRP3-dependent inflammasome activation in human IECs (77). However, the role of other Yops or other inflammasomes in IECs and human cells broadly is unknown.
Here, we find that in contrast to prior findings in murine macrophages (38, 44, 78, 79), the Y. pseudotuberculosis (Yptb) effector YopJ does not induce death of human IECs or macrophages. In contrast, in human cells, Yptb evades caspase-4- and GSDMD-dependent cell death and inflammasome-dependent cytokine release. Notably, the type III secreted effectors YopE, YopH, and YopK, collectively enable Yptb to evade caspase-4 activation in human cells. Mechanistically, YopE and YopH blockade of bacterial internalization prevented accumulation of intracellular bacteria and correlated with reduced inflammasome activation. Furthermore, the Yptb adhesin YadA and host β1-integrins were required for Yptb internalization into human IECs and subsequent caspase-4 activation in the absence of YopE, YopH, and YopK. These findings demonstrate a key role for disruption of actin-mediated internalization in Yersinia evasion of the noncanonical inflammasome in human IECs, and uncover important species and cell-type-specific differences in inflammasome responses to Yersinia infection.
Results
Yptb effectors evade T3SS-dependent inflammasome activation in human cells
During infection of murine macrophages, Yersinia injects Yops to evade inflammasome activation (42, 43, 45, 46). However, wild type (WT) Yersinia induces rapid cell death (Fig. 1A) and caspase-8 and caspase-1 activation due to YopJ-mediated inhibition of NF-κB and MAPK signaling, enabling antibacterial defense despite effector-mediated immune modulation (38, 44, 78, 79). In contrast, human macrophages and IECs do not undergo YopJ-dependent cell death early during Y. enterocolitica infection or at all during Y. pseudotuberculosis infection (56, 77, 80). In agreement with these findings, infection of the human colorectal cell line Caco-2 (Fig. 1B) or primary human monocyte-derived macrophages (hMDMs) (Fig. 1C) with WT Yptb failed to induce release of lactate dehydrogenase (LDH), a marker of cell lysis, into the supernatant. Furthermore, WT Yptb infection of the human monocytic cell line THP-1 macrophages similarly failed to induce LDH release (Fig. S1A), collectively suggesting that in contrast to murine cells, human cells do not undergo cell death in response to WT Yptb infection.
Figure 1.

Yptb effectors evade T3SS-dependent inflammasome activation in human cells. BMDMs (A), Caco-2 cells (B, D-E) or hMDMs (C, F-G) were infected with PBS (Mock), WT Yptb, or D6 Yptb. (A-C) Cell death was measured as percent cytotoxicity normalized to cells treated with 2% Triton at 6hpi. (D) Release of IL-18 into the supernatant was measured by ELISA at 6hpi. (E) Lysates and supernatants collected 6hpi were immunoblotted for IL-18 and β-actin. (F) Release of IL-1b into the supernatant was measured by ELISA at 6hpi. (G) Lysates and supernatants collected 6hpi were immunoblotted for IL-1β and β-actin. ns — not significant, * p < 0.05, ** p < 0.01 by t-test (A-C) or one-way ANOVA (D, F). Shown are averages and error bars representing the standard deviation from at least three pooled experiments.
Lack of cell death in Yptb-infected human cells could be due to a failure to initiate programmed cell death pathways, or due to active evasion or suppression of programmed cell death by the bacteria. Y. enterocolitica deploys two injected effectors, YopH and YopE, to evade inflammasome activation in human IECs (77). Critically, Yptb lacking all six of its injected effectors (Δ6 Yptb) triggered significantly higher levels of cell death compared to either mock or WT Yptb-infected cells in Caco-2 IECs (Fig. 1B), hMDMs (Fig. 1C) and THP-1 macrophages (Fig. S1A). Further, Δ6 Yptb-infected polarized and non-polarized Caco-2 cells robustly cleaved and released the inflammasome-dependent cytokine IL-18 compared to mock or WT Yptb-infected cells (Fig. S1B, Fig. 1D–E). IL-18 release following Δ6 Yptb infection occurred at a range of increasing doses, whereas WT Yptb failed to induce IL-18 release even at the highest doses (Fig. S1C). Consistent with our and others’ findings that expression and release of the inflammasome-dependent cytokine IL-1β is very low in human IECs (48, 49, 77), we did not detect IL-1β release during Δ6 Yptb infection (Fig. S1D). While IECs do not produce IL-1β, this cytokine is released by macrophages undergoing pyroptosis. Indeed, hMDMs and THP-1 macrophages infected with Δ6 Yptb robustly cleaved and secreted IL-1β in contrast to mock or WT Yptb-infected cells (Fig. 1F–G, Fig. S1E), collectively indicating that in the absence of its injected effectors, Yersinia triggers inflammasome activation in multiple human cell types. As expected, inflammasome activation in response to Δ6 Yptb infection is T3SS-dependent, as an isogenic Yptb strain cured of its virulence plasmid encoding the T3SS did not induce IL-18 release in human IECs (Fig. 1D). Collectively, these results indicate that Yops enable WT Yptb to evade inflammasome activation in human cells, a fundamentally distinct outcome from the induction of apoptosis and pyroptosis triggered in murine macrophages by WT Yersinia (38, 44, 78, 79).
Caspase-4 is required for Δ6 Yptb-induced inflammasome activation in human cells
Caspases play important roles in cleaving and activating inflammasome-dependent cytokines and executing cell death (34). Pretreatment of Caco-2 cells with the pan-caspase inhibitor ZVAD prior to infection with Δ6 Yptb completely abrogated IL-18 release and cell death compared to the vehicle control (Fig. 2A–B), indicating that caspases mediate inflammasome activation downstream of Δ6 Yptb infection in human IECs. As ZVAD broadly inhibits multiple caspases, we next sought to determine which specific caspases are required for Δ6 Yptb-induced inflammasome activation in human IECs. Caspase-1 is critical for inflammasome-dependent cytokine release and pyroptosis during bacterial infection (34, 44, 64, 81), and is activated during Y. enterocolitica infection of Caco-2 cells (77). Δ6 Yptb infection of two independent clones of CASP1−/− Caco-2 cells (49) resulted in a partial loss of IL-18 release compared to WT Caco-2 cells (Fig. S2A). Similarly, WT Caco-2s pretreated with the caspase-1 inhibitor YVAD exhibited incomplete loss of IL-18 release after Δ6 Yptb infection compared to DMSO vehicle-treated cells (Fig. S2B), suggesting that caspase-1 contributes to, but is not absolutely required for, Δ6 Yptb-induced inflammasome activation. Caspase-8 is activated in murine macrophages and IECs in response to infection by multiple pathogens, including Yersinia (44, 82, 83), and can process caspase-1 substrates such as IL-1β and GSDMD to mediate pyroptosis in the absence of caspase-1 (64, 82, 84). Notably, siRNA knockdown of CASP8 and pretreatment with the caspase-8 inhibitor IETD resulted in a partial reduction in IL-18 release following Δ6 Yptb infection (Fig. S2C–E), suggesting that like caspase-1, caspase-8 partially contributes to inflammasome activation during Δ6 Yptb infection. However, CASP8 siRNA knockdown in CASP1−/− Caco-2 cells did not abrogate inflammasome activation during Δ6 Yptb infection (Fig. S2F–G), indicating that additional caspase activation pathways likely mediate cell death during Δ6 Yptb infection of human IECs.
Figure 2.
Caspase-4 is required for D6 Yptb-induced inflammasome activation in human cells. (A, B) One hour prior to infection, WT Caco-2 cells were treated with 20 μM ZVAD or DMSO as a vehicle control. Cells were then infected with PBS (Mock), WT Yptb or D6 Yptb. At 6hpi (A) release of IL-18 into the supernatant was measured by ELISA and (B) cell death was measured as percent cytotoxicity normalized to cells treated with 2% triton. (C-E) WT or two independent single cell clones of CASP4−/− Caco-2 cells were infected with PBS (Mock), WT Yptb or D6 Yptb. (C) Release of IL-18 into the supernatant was measured by ELISA at 6hpi. (D) Lysates and supernatants collected 6hpi were immunoblotted for IL-18 and β-actin. (E) Cell death was measured at 6hpi as percent cytotoxicity normalized to cells treated with 2% triton. ** p < 0.01, **** p < 0.0001 by two-way ANOVA. Shown are averages and error bars representing the standard deviation from at least three pooled experiments.
Caspase-4 plays a critical role in inflammasome responses to a variety of enteric pathogens in human IECs (48, 49, 60, 67), and its activation triggers both IEC death and IL-18 release (48). To test whether caspase-4 contributes to inflammasome responses to Δ6 Yptb, we infected two independent single-cell clones of CASP4−/− Caco-2 cells (49) with either WT or Δ6 Yptb. As expected, WT Yptb infection did not elicit inflammasome activation in either WT or CASP4−/− Caco-2 cells, and Δ6 Yptb infection of WT Caco-2 cells resulted in robust release of cleaved IL-18 and cell death. Notably, CASP4 deficiency abrogated cleavage and release of active IL-18 and cell death in response to Δ6 Yptb, indicating that caspase-4 is absolutely required for inflammasome responses to Δ6 Yptb infection in human IECs (Fig. 2C–E). Further, CASP4 deficiency in THP-1 macrophages (85) resulted in a partial but significant decrease in inflammasome activation (Fig. S3A), suggesting that Yop-mediated evasion of the caspase-4 inflammasome is a conserved evasion mechanism across human cell types. Loss of inflammasome activation in THP-1 macrophages lacking caspase-4 largely mirrored our findings with pretreatment with the pan-caspase inhibitor ZVAD (Fig. S3B), further supporting caspase-4 contribution to inflammasome activation in human macrophages. Caspase-5 contributes to Salmonella-induced inflammasome activation in Caco-2 cells (49). To test whether caspase-5 also contributes to Δ6 Yptb-induced inflammasome activation in Caco-2 cells, we treated WT Caco-2 cells with either a control scramble siRNA or CASP5 siRNA. Knockdown of CASP5 resulted in a partial decrease in IL-18 release (Fig. S3C and S3D), suggesting that caspase-5 contributes but is not absolutely required for inflammasome activation. Collectively, these data indicate for the first time that Yptb deploys its Yops to evade caspase-4 inflammasome activation in human macrophages and IECs.
GSDMD is required for Δ6 Yptb-induced inflammasome-dependent cytokine release and cell death in human cells
Inflammasome activation leads to cleavage of the protein GSDMD and liberation of its active pore-forming N-terminal domain, leading to its oligomerization into a large ungated pore (19, 25, 86). Formation of the GSDMD pore in the plasma membrane leads to release of IL-1 family cytokines as well as cell lysis and death, collectively termed “pyroptosis” (87–90). Caspase-4 cleaves and activates GSDMD via release of its N-terminal domain (25, 91). Notably, Δ6 Yptb infection led to robust GSDMD cleavage in WT Caco-2 cells, which was completely absent in CASP4−/− Caco-2 cells, indicating that caspase-4 is required for GSDMD cleavage in human IECs in response to Yersinia lacking its secreted effectors (Fig. 3A). In contrast, consistent with a lack of observed cell death and IL-18 release, WT Yptb infection did not elicit GSDMD cleavage in either WT or CASP4−/− IECs (Fig. 3A). To test whether GSDMD is required for cell death and inflammasome-dependent cytokine release during Δ6 Yptb infection, we pretreated Caco-2 cells with disulfiram, a chemical inhibitor of GSDMD pore formation (92). Critically, disulfiram treatment completely abrogated IL-18 release and cell death downstream of inflammasome activation in Δ6 Yptb-infected cells compared to infected vehicle control treated cells (Fig. 3B–C). Consistent with this observation in human IECs, disulfiram treatment of human THP-1 macrophages resulted in an abrogation of IL-1β release (Fig. 3D). Collectively, these results indicate that Yptb Yops enable evasion of the caspase-4 inflammasome and GSDMD activation.
Figure 3.
GSDMD is required for D6 Yptb-induced inflammasome-dependent cytokine release and cell death in human cells. (A) WT or two independent single cell clones of CASP4−/− Caco-2 cells were infected with PBS (Mock), WT Yptb or D6 Yptb. Lysates and supernatants were collected at 6hpi and immunoblotted for GSDMD and β-actin. One hour prior to infection, (B, C) WT Caco-2 cells or (D) THP-1 macrophages were treated with 30 μM disulfiram or DMSO as a vehicle control. Cells were then infected with PBS (Mock), WT Yptb or D6 Yptb. Release of (B) IL-18 or (D) IL-1β into the supernatant and (C) percent cytotoxicity normalized to cells treated with 2% triton were measured at 6hpi. ***p<0.001, **** p < 0.0001 by two-way ANOVA. Shown are averages and error bars representing the standard deviation from at least three pooled experiments.
The NLRP3 inflammasome can be activated by a variety of stimuli during infection, including potassium efflux downstream of caspase-4-dependent GSDMD activation and pore formation (5–9). Previous studies of human IEC responses during Y. enterocolitica infection identified a critical role for the NLRP3 inflammasome (77). However, studies of human IECs found that NLRP3 does not play a role in inflammasome activation in response to Salmonella infection, potentially due to very low levels of NLRP3 expression in human IECs as compared to human macrophages (49, 93, 94). In agreement, WT Caco-2 cells pretreated with a chemical inhibitor of the NLRP3 inflammasome, MCC950, underwent comparable levels of inflammasome activation in response to Δ6 Yptb infection as infected vehicle control-treated Caco-2 cells (Fig. S4A). Caco-2 cells stimulated with LPS and nigericin, a known agonist of the NLRP3 inflammasome, also failed to induce IL-18 release, further suggesting a lack of NLRP3 inflammasome activity in Caco-2 cells. The NAIP/NLRC4 inflammasome, which senses and responds to flagellin and T3SS ligands (10–18) (Fig. S4B), and the inflammasome adaptor protein ASC (Fig. S4C) were also dispensable for Δ6 Yptb-induced inflammasome activation, consistent with prior findings that expression of these proteins is very low in human IECs (49). Collectively, these results indicate that the canonical NLRP3 and NAIP/NLRC4 inflammasomes, as well as broadly ASC-dependent inflammasomes, are not activated during Δ6 Yptb infection of Caco2 cells, and that GSDMD cleavage and activation occurs downstream of caspase-4 and is required for cell death and cytokine release.
YopE, YopH, and YopK synergistically enable Yptb to evade human inflammasome responses
Our findings demonstrate that Yptb lacking its entire repertoire of injected effectors induce inflammasome activation in human IECs (Fig. 1). In contrast, Caco-2 cells infected with a panel of Yptb mutant strains each lacking one of the six Yops failed to elicit IL-18 secretion (Fig. 4A), indicating that loss of any single secreted Yop was insufficient to alleviate inflammasome evasion and that several Yops likely have overlapping functions in evading inflammasome activation. Notably, single loss of YopK and YopM failed to induce inflammasome activation (Fig. 4A), despite their roles in evading the NLRP3/caspase-11 and pyrin inflammasomes respectively in murine macrophages (40, 43, 45, 46). Y. enterocolitica was previously reported to regulate NLRP3 inflammasome activation in Caco-2 cells by a combination of YopE and YopH-mediated blockade of integrin signaling (77). Despite a lack of a role for the NLRP3 inflammasome during Δ6 Yptb infection of human IECs (Fig. S4A), Yptb lacking both YopE and YopH (ΔyopEH Yptb) elicited significantly elevated IL-18 release in Caco-2 cells, indicating that combinatorial loss of both YopE and YopH was sufficient to induce inflammasome activation in human IECs (Fig. 4B). Nonetheless, IL-18 levels during ΔyopEH Yptb infection were significantly lower than IL-18 levels released during Δ6 Yptb infection (Fig. 4B), suggesting that additional Yops contribute to inflammasome evasion during Yersinia infection of human IECs.
Figure 4.
YopE, YopH, and YopK synergistically enable Yptb to evade human inflammasome responses. (A, B) WT Caco-2 cells or (C, D) WT THP-1 macrophages were infected with PBS (Mock) or indicated strain of Yptb. Release of (A, B) IL-18 or (C) IL-1β into the supernatant and (D) percent cytotoxicity normalized to cells treated with 2% triton was measured at 6hpi. (E, F) WT or two independent single cell clones of CASP4−/− Caco-2 cells were infected with PBS (Mock) or the indicated strain of Yptb. (E) Release of IL-18 into the supernatant and (F) percent cytotoxicity normalized to cells treated with 2% triton were measured at 6hpi. * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001 by one-way ANOVA (A-D) or two-way ANOVA (E, F). Shown are averages and error bars representing the standard deviation from at least three pooled experiments.
YopK is a translocated effector that negatively regulates the translocation of other Yops and T3SS components (42, 43, 45). In murine macrophages, YopK evades inflammasome activation, whereas YopE and YopH do not contribute to inflammasome evasion (42, 43, 45). Given that deletion of yopK alone failed to elicit IL-18 release in human IECs (77) (Fig. 4A), we considered that YopK could modulate inflammasome activation in human IECs in a manner that is masked by YopE and YopH, perhaps because these effectors are hypertranslocated in a ΔyopK mutant. Indeed, IL-18 release during ΔyopEHK Yptb infection was substantially elevated compared to ΔyopEH Yptb, and fully recapitulated levels of IL-18 release observed during Δ6 Yptb infection (Fig. 4B), indicating that YopE, YopH and YopK function together to enable Yptb evasion of inflammasome activation during infection. Critically, individual loss of YopK, dual loss of YopK and YopE (ΔyopEK) or dual loss of YopK and YopH (ΔyopHK) all failed to induce inflammasome activation. Only in a yopEH mutant background did additional deletion of YopK lead to an increase in IL-18 release (Fig. 4B). Consistent with infection of human IECs, ΔyopEHK infection also triggered higher levels of inflammasome activation than either ΔyopEH or ΔyopK infection in human THP-1 macrophages (Fig. 4C–D). Further, as with Δ6 Yptb infection, ΔyopEHK-induced inflammasome activation in Caco-2 cells was fully dependent on caspase-4 (Fig. 4E–F). Taken together, these data suggest that YopE, YopH, and YopK act in concert to evade the noncanonical inflammasome in both human IECs and macrophages. These results are distinct from murine macrophages, in which YopE instead activates the pyrin inflammasome, and YopK and YopM contribute to inflammasome evasion (42, 43, 45, 46).
YopE and YopH inhibit caspase-4-dependent inflammasome activation in human IECs by blocking actin-dependent bacterial internalization
During Y. enterocolitica infection of human IECs, YopE and YopH evade NLRP3 inflammasome activation by disrupting IL-18 transcriptional priming downstream of invasin-β1-integrin signaling (77). However, Yersinia adhesin-β1-integrin-mediated signaling also triggers host cytoskeletal rearrangements to facilitate bacterial internalization (95–98). YopE and YopH disruption of focal adhesion complexes and actin filamentation consequently inhibits Yersinia uptake into host cells (98–108). Notably, consistent with both Δ6 and ΔyopEHK Yptb infection, we found that caspase-4 was absolutely required for inflammasome activation induced by ΔyopEH Yptb (Fig. 5A and 5B). Considering YopE and YopH’s known roles in inhibiting bacterial internalization, we hypothesized that YopE- and YopH-mediated inflammasome evasion in human IECs might be linked to their antiphagocytic activity, which would limit bacterial internalization, subsequent cytosolic delivery of LPS, and caspase-4 inflammasome activation. Consistent with previous reports (98, 100–102, 104, 108), levels of Yptb internalization into human cells 2 hours post-infection were lowest in WT Yptb-infected cells, while ΔyopEH-infected cells had significantly elevated levels of intracellular bacteria, as assessed by colony forming units (CFUs) (Fig. S5A–B). ΔyopEH and Δ6-infected cells had comparable levels of intracellular bacteria at 2 hpi, indicating that YopE and YopH regulate internalization into human cells, while YopK limits inflammasome activation through a mechanism distinct from bacterial internalization. In agreement with our CFU data, microscopic analysis of Caco-2 cells infected with GFP-expressing bacteria demonstrated that ΔyopEH and Δ6 Yptb-infected cells had significantly higher levels of intracellular bacteria (i.e. GFP-only bacteria) than WT Yptb-infected cells (Fig. S5C–D). Furthermore, coinfection of Caco-2 cells with Δ6 Yptb and increasing doses of WT Yptb resulted in reduced bacterial internalization (Fig. 5C), which corresponded to a dose-dependent decrease in inflammasome activation (Fig. 5D) as compared to Δ6 Yptb alone infected cells. These results suggest that WT Yptb-injected Yops act in trans and can complement inhibition of bacterial uptake and inflammasome activation during Δ6 Yptb infection. As YopE and YopH block bacterial uptake via disruption of the actin cytoskeleton (98, 101–107), we asked whether pharmacological inhibition of the actin cytoskeleton would prevent inflammasome activation in response to ΔyopEH Yptb infection. Notably, cytochalasin D, an inhibitor of actin polymerization previously demonstrated to block bacterial internalization (102, 109), significantly reduced both intracellular bacterial numbers and inflammasome activation during ΔyopEH infection (Fig. S5E–F). Collectively, these results highlight a role for YopE and YopH-mediated internalization inhibition in contributing to inflammasome evasion in human IECs.
Figure 5.
YopE and YopH evade caspase-4-dependent inflammasome activation and actin-dependent bacterial internalization in human IECs. (A, B) WT or two independent single cell clones of CASP4−/− Caco-2 cells were infected with PBS (Mock) or the indicated strain of Yptb. (A) Release of IL-18 into the supernatant and (B) percent cytotoxicity normalized to cells treated with 2% triton were measured at 6hpi. (C, D) WT Caco-2 cells were infected with the indicated MOI and strain of Yptb. (C) Cells were lysed at 2 hpi and bacteria were plated on Yersinia-selective agar to calculate CFUs. (D) Release of IL-18 into the supernatant was measured at 6hpi. (E, F) WT Caco-2 cells were incubated for one hour with or without a 1:50 dilution of antibody against β−1 integrin and then infected with PBS (Mock) or the indicated strain of Yptb at an MOI of 20 (E) or 60 (F). (E) Cells were lysed at 2 hpi and bacteria were plated on Yersinia-selective agar to calculate CFUs. (F) Release of IL-18 into the supernatant was measured at 6hpi. (G, H) WT Caco-2 cells were infected with PBS (Mock) or the strain of Yptb at an MOI of 20 (G) or 60 (H). (G) Cells were lysed at 2 hpi and bacteria were plated on Yersinia-selective agar to calculate CFUs. (H) Release of IL-18 into the supernatant was measured at 6hpi. * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001 by two-way ANOVA (A, B) or one-way ANOVA (C-H). Shown are averages and error bars representing the standard deviation from at least three pooled experiments.
YadA and β1-integrin are necessary for bacterial internalization and inflammasome activation
Because Cytochalasin D has pleiotropic inhibitory effects, we sought to evaluate the role of bacterial internalization in triggering inflammasome activation by manipulating components of β1-integrin signaling. As macrophages are naturally phagocytic and do not require β1-integrin signaling to induce bacterial uptake, we focused on human IECs. Yersinia-β1-integrin interactions initiate an intracellular signaling cascade, including phosphorylation of the serine/threonine kinase Akt, that ultimately regulates actin-dependent cellular changes necessary for bacterial internalization (110–115). Consistent with previous reports, chemical inhibition of Akt isoforms 1 and 2 diminished Δ6 Yptb internalization in IECs (Fig. S6A) (116). Critically, inflammasome activation induced by Δ6 Yptb was also significantly blunted in the presence of Akt inhibitors (Fig. S6B). Further, pretreatment with an anti-β1-integrin blocking antibody previously found to reduce Yersinia entry into cells (117), also significantly decreased bacterial internalization and inflammasome activation during both ΔyopEH and Δ6 Yptb infection (Fig. 5E–F). Moreover, siRNA knockdown of α5β1 integrin similarly reduced levels of internalized bacteria and inflammasome activation (Fig. S6C–E). Together, these data suggest that disruption of β1-integrin receptor engagement and downstream components of β1-integrin signaling prevents Yersinia internalization and limits inflammasome activation in IECs.
The Yersinia adhesin, invasin, binds to β1-integrin to facilitate efficient bacterial internalization into mammalian cells, particularly during early intestinal invasion and translocation through M cells (70, 118). Inflammasome activation during Y. enterocolitica infection of human IECs was found to be dependent on invasin-β1-integrin signaling that upregulates Il18 transcript levels (77). However, unlike Ye, Yptb does not express high levels of invasin under T3SS-expressing conditions (117, 119, 120). Consistently, deletion of invasin had a minor impact on ΔyopEH and Δ6 Yptb-induced internalization and inflammasome activation (Fig. S6F–G) suggesting the possibility that other adhesins may mediate T3SS-dependent inflammasome activation by Yptb. YadA is encoded on the Yersinia virulence plasmid, and its expression is co-regulated with the T3SS and Yops (121). As YadA mediates efficient cellular entry under conditions where invasin expression is repressed (117), we hypothesized that YadA-mediated bacterial internalization might drive caspase-4-dependent inflammasome activation. Indeed, deletion of YadA in both ΔyopEH and Δ6 Yptb significantly reduced bacterial internalization to levels comparable to WT infection (Fig. 5G) and completely abrogated inflammasome activation (Fig. 5H). Notably, deletion of YadA alone largely phenocopied deletion of both invasin and YadA with respect to internalization and inflammasome activation (Fig. 5G–H), implicating YadA as the primary adhesin responsible for bacterial internalization and inflammasome activation during Yptb infection of human IECs.
Discussion
In this study, we report for the first time that the Yptb T3SS-injected effectors YopE, YopH and YopK collectively enable evasion of caspase-4 inflammasome activation and pyroptosis in human cells (Fig. 1–4, S1, S3). Mechanistically, YopE and YopH prevented bacterial internalization and, together with YopK, limited caspase-4 inflammasome activation downstream of YadA-β1-integrin-mediated uptake of Yptb into IECs (Fig. 5, S5–6).
The NLRP3 inflammasome is reported to be activated by Y. enterocolitica in human IECs (77). However, we found that NLRP3 was dispensable for inflammasome responses to Yptb in Caco-2 cells (Fig. S4A). NLRP3 expression is very low in Caco-2 IECs and primary human epithelium (49, 93, 94), and other enteric pathogens and known NLRP3 stimuli fail to activate the NLRP3 inflammasome in Caco-2 cells (49) (Fig. S4A). Differences in cell culture conditions or between Y. enterocolitica and Y. pseudotuberculosis may account for the differential role of NLRP3 during infection of human IECs. Moreover, the adaptor protein ASC, which is necessary for function of the NLRP3, pyrin, and other inflammasomes, was dispensable for inflammasome responses to Δ6 Yptb (Fig. S4C), indicating that canonical inflammasomes are unlikely to be involved in this response. Notably, although YopE-mediated cytoskeletal disruption activates the pyrin inflammasome, which is inhibited by YopM during infection of murine macrophages (40, 46), YopM had no impact on inflammasome activation in human IECs even in the presence of YopE (Fig. 4A), potentially due in part to low expression of pyrin in IECs (122, 123).
We found instead for the first time that Yptb activates the caspase-4 inflammasome in human epithelial cells (Fig. 2C–E, 4E–F, 5A–B). Caspase-4 is more highly expressed in human IECs than other inflammasome components (49), and a broad range of intracellular enteric pathogens engage or inhibit caspase-4/11 in human and murine IECs (48, 49, 76, 60, 62–67, 75). We also found a partial role for caspase-1 and caspase-8 in inflammasome activation in Caco-2 cells (Fig. S2A–B, S2D–E). Caspase-8 and caspase-1 can be recruited to the same inflammasome complexes and have compensatory or sequential roles (44, 64, 81). Simultaneous knockdown of caspase-1 and caspase-8 still allowed for release of IL-18 during Δ6 Yptb infection (Fig. S2F), suggesting that they are unlikely to play overlapping roles and may act sequentially. Whether caspase-8 and capase-1 are activated downstream of caspase-4 or are acting in a parallel pathway during Yersinia infection of human IECs is unknown. CASP4−/− THP-1 macrophages also had reduced levels of inflammasome activation during Δ6 Yptb infection (Fig S3A–B), suggesting that like in human IECs, caspase-4 contributes to Δ6 Yptb-induced inflammasome activation in human macrophages.
In addition to blocking entry into epithelial cells (98, 101–104) (Fig. S5), YopE and H also block bacterial internalization into M cells (77), specialized follicular epithelial cells overlying Peyer’s patches that are considered the primary site of entry across the intestinal epithelium for Yersinia (70–73, 104). As such, invasin-dependent entry into M cells may occur prior to full T3SS upregulation, thereby allowing the bacteria to cross the epithelium without triggering inflammasome activation. Intriguingly, we found that D6 Yptb induces inflammasome activation during both apical and basolateral infection of polarized Caco-2 cells, suggesting a role for non-M cell IECs in inflammasome-mediated host defense. While integrins are predominantly expressed apically on M cells and basolaterally on non-M cell IECs, intestinal pathogens can interact with basolateral host receptors at cellular junctions either due to depolarization events leading to apical receptor relocation or to focal discontinuities leading to luminal exposure of basolateral elements (124–126). Furthermore, Yptb microcolonies are found outside Peyer’s patches within submucosal pyogranulomas in close proximity basolaterally to IECs (74). YopE and YopH may prevent apical or basolateral uptake by non M-cell IECs that would otherwise trigger inflammasome-mediated responses within the intestinal epithelium.
Y. entercolitica YopE and YopH were previously reported to inhibit inflammasome activation by blocking a priming signal downstream of integrin signaling that upregulates IL18 transcript levels (77). However, integrin signaling is also crucial for bacterial internalization into IECs. Using a variety of orthogonal approaches, we show here that disruption of integrin signaling reduced inflammasome activation commensurate with reduced levels of Yptb internalization by IECs (Fig. 5, S6). Additionally, YadA, rather than invasin, was required for Δ6 Yptb-induced inflammasome activation and bacterial internalization (Fig. 5G–H), consistent with Yptb downregulation of invasin and YadA co-expression with the T3SS (117, 121). Our findings lead us to propose a model in which YopE/H-dependent blockade of YadA-mediated bacterial uptake by IECs limits delivery of LPS into the host cell, thereby allowing bacteria to evade the noncanonical inflammasome. Because adhesin-integrin binding and signal transduction mediate a variety of cellular outcomes, it is difficult to fully exclude the impact of additional factors on inflammasome activation and evasion facilitated by YopE and YopH.
While YopE and YopH interfere with inflammasome responses during Y. enterocolitica infection of human IECs, no role was previously found for the Y. enterocolitica homolog of YopK, YopQ (77). Consistently, while loss of YopK alone had no effect on inflammasome responses to Yptb, combinatorial loss of YopK, YopE, and YopH recapitulated the levels of inflammasome activation observed with Δ6 Yptb infection, suggesting that YopK evades a component of an inflammasome pathway masked by YopE and YopH (Fig. 4). In murine macrophages, YopK prevents LPS-mediated inflammasome activation by preventing destabilization of the Yersinia-containing vacuole (42, 43). In the absence of YopE, YopH, and YopK, Yersinia may be more readily taken up and exposed to the host cell cytosol, potentially due to vacuolar damage as a result of T3SS-mediated pore formation (42, 127). We found that loss of YopK augments ΔyopEH-induced inflammasome activation in both human macrophages and IECs, potentially pointing to a conserved mechanism by which YopK, E, and H enable Yptb to evade inflammasome responses across human cell types. Further studies are needed to determine precisely how the T3SS and YopK evade cytosolic Yersinia LPS exposure and caspase-4 activation.
The synergistic evasion of the caspase-4 inflammasome by YopE, YopH, and YopK, as well as the absence of YopJ-induced cell death, in both human IECs and THP-1 macrophages is fundamentally distinct from the response to Yptb observed in murine macrophages. Human cells may be intrinsically more resistant to pathogen-induced cell death due to altered expression of prosurvival factors such as cellular FLICE-inhibitory protein (cFLIP) or A20. Furthermore, unlike macrophages, IECs are not phagocytic, potentially allowing contamination of the host cell cytosol by bacterial LPS to be a more specific indicator of virulence activity in IECs and providing a rationale for increased reliance on caspase-4-mediated responses. Further studies dissecting inflammasome responses between murine and human IECs and macrophages will provide additional insight into cell type- and species-specific differences in the mechanisms of inflammasome activation by bacterial pathogens.
Overall, our data demonstrate that Yops E, H, and K, enable Yersinia pseudotuberculosis to evade caspase-4 inflammasome responses downstream of YadA-β1-integrin signaling in human cells, thereby revealing a major difference in interactions between Yops and inflammasomes in murine and human macrophages. Our study further highlights the distinct nature of inflammasome responses and bacterial effector activities in different cell types in mice and humans, which provides insight into how inflammasome responses and bacterial virulence activities shape health and disease in mice and humans.
Materials and Methods
Ethics statement
All studies involving primary human monocyte-derived macrophages (hMDMs) were performed in compliance with the requirements of the US Department of Health and Human Services and the principles expressed in the Declaration of Helsinki. hMDMs from de-identified healthy human donors were obtained from the University of Pennsylvania Human Immunology Core, which holds Institutional Review Board approval. These samples are considered a secondary use of de-identified human specimens and are exempt via Title 55 Part 46, Subpart A of 46.101 (b) of the Code of Federal Regulations.
Bacterial strains and growth conditions
Yersinia strains are described in Table S1 in the supplemental material. ΔyopEHK, ΔyopEK, and ΔyopHK were generated by introducing a frameshift mutation of the yopK open reading frame into the ΔyopEH, ΔyopE and ΔyopH backgrounds respectively using a plasmid provided by Dr. James Bliska and an allelic exchange method (128). ΔyopM was generated by introducing an unmarked deletion of the yopM open reading frame into IP2666 using a plasmid provided by Dr. James Bliska and the same allelic exchange method. ΔyopEHΔinv and Δ6Δinv strains were generated by introducing an unmarked deletion of the invasin open reading frame into ΔyopEH and Δ6 strains respectively using a plasmid provided by Dr. Joan Mecsas and the same allelic exchange method. ΔyopEHΔyadA, ΔyopEHΔinvΔyadA, Δ6ΔyadA, and Δ6ΔinvΔyadA strains were generated by introducing a kanamycin resistance cassette in place of the yadA open reading frame into ΔyopEH, ΔyopEHΔinv, Δ6, and Δ6Δinv strains respectively using a plasmid provided by Dr. Petra Dersch and the same allelic exchange method. Yersiniae were cultured overnight at 26°C with aeration in 2x yeast extract-tryptone (YT) broth. To induce T3SS expression, in the morning, the bacteria were diluted into fresh 2xYT containing 20 mM sodium oxalate and 20 mM MgCl2. Bacteria were grown with aeration for 1 hour at 26°C followed by 2 hour at 37°C prior to infection. All cultures were pelleted at 6000 × g for 3 min and resuspended in phosphate-buffered saline (PBS). Cells were infected at an MOI of 60 unless otherwise indicated, centrifuged at 290 × g for 10 min and incubated at 37°C. At 1 hour post-infection, epithelial cells were treated with 20 ng/ml or 100 ng/ml of gentamicin for 6 hour or 2 hour time points respectively and macrophages were treated with 100 ng/ml of gentamicin for all time points. Infections proceeded at 37°C for the indicated length of time for each experiment. In all experiments control cells were mock infected with PBS.
Cell culture conditions
All cells were grown at 37°C in a humidified incubator with 5% CO2.
Cell culture of Caco-2 cells
Caco-2 cells (HTB-27; American Type Culture Collection) were maintained in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% (vol/vol) heat-inactivated fetal bovine serum (FBS), 100 IU/mL penicillin and 100 μg/mL streptomycin. One day prior to infection, Caco-2 cells were incubated with 0.25% trypsin-EDTA (Gibco) diluted 1:1 with 1 × PBS at 37°C for 15 min to dissociate cells. Trypsin was neutralized with serum-containing medium. Cells were replated in medium without antibiotics in a 24-well plate at a density of 3 × 105 cells/well and unprimed prior to infection as we have not observed differential inflammasome-dependent cytokine release in IECs during infection with priming (49). All Caco-2 knockout cell lines are described previously (49).
Cell culture of polarized Caco-2 cells
Polarized Caco-2 cells were grown on polycarbonate 3 μM pore size cell culture inserts (Corning 3415) in a 24 well plate. Inserts were coated with collagen coating solution containing 30 μg/ml collagen, 10 μg/ml fibronectin and 10 μg/ml BSA in DMEM and incubated for 3 hours. Caco-2 cells were then plated in growth medium containing Corning MITO+ serum extender (Fisher Scientific CB-50006) on inverted (for basolateral infection) or noninverted (for apical infection) inserts. After 24 hours, the growth medium was replaced with Corning enterocyte differentiation medium (Fisher Scientific 355357) with MITO+ serum extender. The media was replaced daily and following three days of incubation in differentiation media, the transepithelial electrical resistance was measured using an Epithelial Volt/Ohm (TEER) Meter (World Precision Instruments) to ensure that resistance was above 250 Ω.cm2 prior to infection. Infections were administered on the apical or basolateral side of cells as indicated.
Cell culture of THP-1 monocyte derived macrophages
THP-1 macrophages (TIB-202; American Type Culture Collection) were maintained in RPMI supplemented with 10% (vol/vol) heat-inactivated fetal bovine serum (FBS), 0.05 nM β-mercaptoethanol, 100 IU/mL penicillin and 100 μg/mL streptomycin. Two days prior to infection, THP-1 cells were replated in medium without antibiotics in a 48-well plate at a density of 2 × 105 cells/well and incubated with phorbol 12-myristate 13-acetate (PMA) for 24 hours to allow differentiation into macrophages. Macrophages were primed with 100 ng/mL Pam3CSK4 (Invivogen) for 16 hours prior to bacterial infections in order to upregulate pro-IL-1β transcript levels.
Cell culture of primary human monocyte-derived macrophages (hMDMs)
Purified human monocytes from de-identified healthy human donors were obtained from the University of Pennsylvania Human Immunology Core. Monocytes were differentiated into macrophages by culturing in RPMI supplemented with 10% (vol/vol) heat-inactivated FBS, 2 mM L-glutamine, 100 IU/mL penicillin, 100 μg/ml streptomycin, and 50 ng/ml recombinant human M-CSF (Gemini Bio-Products) for 6 days. Two days prior to infection, adhered hMDMs were replated in media with 25 ng/ml human M-CSF lacking antibiotics at 1×105 cells/well in a 48 well plate. hMDMs were then primed with 100 ng/ml Pam3CSK4 (Invivogen) for 16 hours prior to bacterial infection in order to upregulate pro-IL-1β transcript levels.
Cell culture of murine bone marrow derived macrophages (BMDMs)
Bone marrow cells were grown in RPMI containing L-cell supernatant, heat-inactivated FBS, penicillin and streptomycin for 8 days. One day prior to infection, differentiated BMDMs were replated into 24-well dishes in media lacking antibiotics at a density of 2 × 105 cells/well.
ELISAs
Supernatants harvested from infected cells were assayed using enzyme-linked immunosorbent assay (ELISA) kits for human IL-18 (R&D Systems) and IL-1β (BD Biosciences).
LDH cytotoxicity assays
Supernatants harvested from infected cells were assayed for cytotoxicity by measuring loss of cellular membrane integrity via lactate dehydrogenase (LDH) assay. LDH release was quantified using an LDH Cytotoxicity Detection Kit (Clontech) according to the manufacturer’s instructions and normalized to mock-infected (min cytotoxicity) and 2% triton-treated cells (max cytotoxicity)
Immunoblot analysis
Cells were replated and infected on serum-free medium to collect supernatant samples. Supernatant samples were centrifuged at 200 × g to pellet any cell debris and treated with trichloroacetic acid (TCA) (25 μL TCA per 500 μL supernatant) overnight at 4°C. The following day, TCA-treated samples were centrifuged at max speed (15,871 × g) for 15 min at 4°C and washed with ice-cold acetone. TCA-precipitated supernatant samples and cell lysates were resuspended in 1 × SDS-PAGE sample buffer and boiled for 5 min. Samples were separated by SDS-PAGE on a 12% (vol/vol) acrylamide gel and transferred to polyvinylidene difluoride (PVDF) Immobilon-P membranes (Millipore). Primary antibodies specific for human IL-18 (MLB International PM014), IL-1β (R&D Systems MAB201), β-actin (4967L; Cell Signaling) and GSDMD (G7422 Sigma-Aldrich) and horseradish peroxidase (HRP)-conjugated secondary antibodies anti-rabbit IgG (7074S; Cell Signaling) and anti-mouse IgG (7076S; Cell Signaling) were used. Enhanced chemiluminescence (ECL) Western blotting substrate or SuperSignal West Femto (Pierce Thermo Scientific) HRP substrate were used for detection.
Inhibitor and antibody blocking experiments
Cells were treated 1 h prior to infection at the indicated concentrations of the following inhibitors: 10 μM MCC950 (Sigma-Aldrich; PZ0280), 20 μM pan-caspase inhibitor Z-VAD(Ome)-FMK (SM Biochemicals; SMFMK001), 20 μM caspase-1 inhibitor Ac-YVAD-cmk (Sigma-Aldrich; SML0429), 30 μM disulfiram (Sigma), 10 μM cytochalasin D (Sigma), and 25 μM Akt inhibitor VIII (EMD Millipore). Cells were treated 1 h prior to infection with a 1:50 dilution of monoclonal antibody 1987 clone P4C10 (EMD Millipore) directed against β1 integrins.
siRNA-mediated gene knockdown
CASP5 (S2417), CASP8 (S2427), ITGA5 (S7549) and two Silencer Select negative-control siRNAs (Silencer Select negative control no. 1 and no. 2 siRNA) were purchased from Ambion (Life Technologies). Three days before infection, 30 nM siRNA was transfected into Caco-2 cells using Lipofectamine RNAiMAX transfection reagent (Thermo Fisher Scientific) following the manufacturer’s protocol.
Quantitative RT-PCR analysis
RNA was isolated using the RNeasy Plus Mini Kit (Qiagen) following the manufacturer’s instructions. Cells were lysed in 350 μL RLT buffer with b-mercaptoethanol and centrifuged through a QIAshredder spin column (Qiagen). cDNA was synthesized from isolated RNA using SuperScript II Reverse Transcriptase (Invitrogen) following the manufacturer’s instructions. Quantitative PCR was conducted with the CFX96 real-time system from Bio-Rad using the SsoFast EvaGreen Supermix with Low ROX (Bio-Rad). For analysis, mRNA levels of siRNA-treated cells were normalized to housekeeping gene HPRT and control siRNA-treated cells using the 2−ΔΔCT (cycle threshold) method to calculate knockdown efficiency (129). The following primers were used:
CASP5 forward: TTCAACACCACATAACGTGTCC
CASP5 reverse: GTCAAGGTTGCTCGTTCTATGG
CASP8 forward: GTTGTGTGGGGTAATGACAATCT
Casp8 reverse: TCAAAGGTCGTGGTCAAAGCC
ITGA5 forward: GGCTTCAACTTAGACGCGGAG
ITGA5 reverse: TGGCTGGTATTAGCCTTGGGT
HPRT forward: CCTGGCGTCGTGATTAGTGAT
HPRT reverse: AGACGTTCAGTCCTGTCCATAA
Bacterial uptake enumeration with colony forming units (CFUs)
Cells were infected with indicated strains of Yersinia at an MOI of 20. 1 hpi, cells were treated with 100 μg/mL of gentamicin to kill extracellular bacteria. 2 hpi the supernatants were aspirated and cells were lysed with PBS containing 0.5% Triton to collect intracellular bacteria. Harvested bacteria were serially diluted in PBS and plated on LB agar plates containing 2 μg/mL Irgasan. Plates were incubated at 28°C for two days and CFUs were counted.
Fluorescence microscopy of intracellular Yersinia
One day before infection 2 × 105 cells/well were plated on glass coverslips in a 24-well plate. Cells were infected with indicated strains of Yersinia constitutively expressing GFP at an MOI of 20. At 2hpi, cells were washed 2 times with PBS, fixed with 4% paraformaldehyde for 10 min at 37°C and stored overnight at 4°C in PBS. The following day, cells were blocked for 30 min at room temperature in blocking solution containing 1% BSA in PBS and incubated for 1 h at room temperature in blocking solution with the polyclonal anti-Yersinia antibody SB349 diluted 1:1000 (kindly provided by Dr. James Bliska) (102). AF594-conjugated goat anti-Rabbit IgG antibody (A-11012 Thermo Fisher Scientific) was diluted 1:500 in blocking solution was added to cells and incubated for 45 min at room temperature. Cells were mounted on glass slides with DAPI mounting medium (Sigma Fluoroshield). Coverslips were imaged on an inverted fluorescence microscope (IX81; Olympus) and images were collected using a high-resolution charge-coupled-devise camera (FAST1394; QImaging) at a magnification of 60x. Images were analyzed and presented using SlideBook (version 5.0) software (Intelligent Imaging Innovations, Inc.). %intracellular bacteria were scored in unblinded fashion by counting 20 captures per coverslip for coverslips across independent triplicate experiments.
Statistical analysis
Prism 9.4.1 (GraphPad Software) was utilized for the graphing of data and all statistical analyses. Statistical significance for experiments were determined using the appropriate test and are indicated in each figure legend. Differences were considered statistically significant if the p value was <0.05.
Supplementary Material
Importance.
Yersinia are responsible for significant disease burdens in humans, ranging from recurrent disease outbreaks (yersiniosis) to pandemics (Yersinia pestis plague). Together with rising antibiotic resistance rates, there is a critical need to better understand Yersinia pathogenesis and host immune mechanisms, as this information will aid in developing improved immunomodulatory therapeutics. Inflammasome responses in human cells are less studied relative to murine models of infection, though recent studies have uncovered key differences in inflammasome responses between mice and humans. Here, we dissect human intestinal epithelial cell and macrophage inflammasome responses to Yersinia pseudotuberculosis. Our findings provide insight into species- and cell type-specific differences in inflammasome responses to Yersinia.
Acknowledgments
We thank members of the Shin and Brodsky laboratories for helpful scientific discussions. We thank Dr. James Bliska, Dr. Joan Mecsas, and Dr. Petra Dersch for generously providing plasmids and Yop mutant strains.
We thank the Human Immunology Core of the Penn Center for AIDS Research and Abramson Cancer Center for providing purified primary human monocytes.
This work is supported by NIH/NIAID grants AI151476, AI118861, AI123243 (S.S.), AI128630, AI163596, and AI139102 (I.E.B). S.S. and I.E.B. are both recipients of the Burroughs-Welcome Fund Investigators in the Pathogenesis of Infectious Disease Award. J.Z. is a recipient of the NIH/NIAID Microbial Pathogenesis and Genomics training grant 5T32AI141393-03.
Data availability
All data are included in the manuscript and supplemental material. Bacterial strains available upon request
References
- 1.Janeway CA. 1989. Approaching the Asymptote? Evolution and Revolution in Immunology. Cold Spring Harb Symp Quant Biol 54:1–13. [DOI] [PubMed] [Google Scholar]
- 2.Janeway CA, Medzhitov R. 2003. Innate Immune Recognition. https://doi.org/101146/annurev.immunol20083001084359 20:197–216. [DOI] [PubMed] [Google Scholar]
- 3.Vance RE, Isberg RR, Portnoy DA. 2009. Patterns of Pathogenesis: Discrimination of Pathogenic and Nonpathogenic Microbes by the Innate Immune System. Cell Host Microbe 6:10–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Brodsky IE, Monack D. 2009. NLR-mediated control of inflammasome assembly in the host response against bacterial pathogens. Semin Immunol 21:199–207. [DOI] [PubMed] [Google Scholar]
- 5.Lamkanfi M, Dixit VM. 2009. Inflammasomes: guardians of cytosolic sanctity. Immunol Rev 227:95–105. [DOI] [PubMed] [Google Scholar]
- 6.Mariathasan S, Weiss DS, Newton K, McBride J, O’Rourke K, Roose-Girma M, Lee WP, Weinrauch Y, Monack DM, Dixit VM. 2006. Cryopyrin activates the inflammasome in response to toxins and ATP. Nat 2006 4407081 440:228–232. [DOI] [PubMed] [Google Scholar]
- 7.Muñoz-Planillo R, Kuffa P, Martínez-Colón G, Smith BL, Rajendiran TM, Núñez G. 2013. K+ Efflux Is the Common Trigger of NLRP3 Inflammasome Activation by Bacterial Toxins and Particulate Matter. Immunity 38:1142–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, Fitzgerald KA, Latz E. 2008. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol 2008 98 9:847–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Franchi L, Kanneganti TD, Dubyak GR, Núñez G. 2007. Differential requirement of P2X7 receptor and intracellular K+ for caspase-1 activation induced by intracellular and extracellular bacteria. J Biol Chem 282:18810–18818. [DOI] [PubMed] [Google Scholar]
- 10.Ren T, Zamboni DS, Roy CR, Dietrich WF, Vance RE. 2006. Flagellin-Deficient Legionella Mutants Evade Caspase-1- and Naip5-Mediated Macrophage Immunity. PLOS Pathog 2:e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhao Y, Yang J, Shi J, Gong Y-N, Lu Q, Xu H, Liu L, Shao F. 2011. The NLRC4 inflammasome receptors for bacterial flagellin and type III secretion apparatus 10.1038/nature10510. [DOI] [PubMed] [Google Scholar]
- 12.Kofoed EM, Vance RE. 2011. Innate immune recognition of bacterial ligands by NAIPs determines inflammasome specificity 10.1038/nature10394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Miao EA, Mao DP, Yudkovsky N, Bonneau R, Lorang CG, Warren SE, Leaf IA, Aderem A. Innate immune detection of the type III secretion apparatus through the NLRC4 inflammasome 10.1073/pnas.0913087107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhao Y, Shi J, Shi X, Wang Y, Wang F, Shao F. 2016. Genetic functions of the NAIP family of inflammasome receptors for bacterial ligands in mice. J Exp Med 213:647–656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Rauch I, Tenthorey JL, Nichols RD, Al Moussawi K, Kang JJ, Kang C, Kazmierczak BI, Vance RE. 2016. NAIP proteins are required for cytosolic detection of specific bacterial ligands in vivo. J Exp Med 213:657–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Yang J, Zhao Y, Shi J, Shao F. 2013. Human NAIP and mouse NAIP1 recognize bacterial type III secretion needle protein for inflammasome activation. Proc Natl Acad Sci U S A 110:14408–14413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Rayamajhi M, Zak DE, Chavarria-Smith J, Vance RE, Miao EA. 2013. Cutting Edge: Mouse NAIP1 Detects the Type III Secretion System Needle Protein. J Immunol 191:3986–3989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Molofsky AB, Byrne BG, Whitfield NN, Madigan CA, Fuse ET, Tateda K, Swanson MS. 2006. Cytosolic recognition of flagellin by mouse macrophages restricts Legionella pneumophila infection. J Exp Med 203:1093–1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y, Cai T, Wang F, Shao F. 2015. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nat 2015 5267575 526:660–665. [DOI] [PubMed] [Google Scholar]
- 20.Li P, Allen H, Banerjee S, Franklin S, Herzog L, Johnston C, Mcdowell J, Paskind M, Rodman L, Salfeld J, Towne E, Tracey D, Wardwell S, Wei F-Y, Wong W, Kamen R, Seshadri T. 1995. Mice Deficient in IL-lp-Converting Enzyme Are Defective in Production of Mature IL-lp and Resistant to Endotoxic Shock. Cell 80:401–411. [DOI] [PubMed] [Google Scholar]
- 21.Kuida K, Lippke JA, Ku G, Harding MW, Livingston DJ, Su MSS, Flavell RA. 1995. Altered Cytokine Export and Apoptosis in Mice Deficient in Interleukin-1β Converting Enzyme. Science (80- ) 267:2000–2003. [DOI] [PubMed] [Google Scholar]
- 22.Cookson BT, Brennan MA. 2001. Pro-inflammatory programmed cell death [2]. Trends Microbiol 9:113–114. [DOI] [PubMed] [Google Scholar]
- 23.Mariathasan S, Monack DM. 2007. Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation. Nat Rev Immunol 2007 71 7:31–40. [DOI] [PubMed] [Google Scholar]
- 24.Kayagaki N, Warming S, Lamkanfi M, Vande Walle L, Louie S, Dong J, Newton K, Qu Y, Liu J, Heldens S, Zhang J, Lee WP, Roose-Girma M, Dixit VM. 2011. Non-canonical inflammasome activation targets caspase-11. Nature 479:117–121. [DOI] [PubMed] [Google Scholar]
- 25.Kayagaki N, Stowe IB, Lee BL, O’Rourke K, Anderson K, Warming S, Cuellar T, Haley B, Roose-Girma M, Phung QT, Liu PS, Lill JR, Li H, Wu J, Kummerfeld S, Zhang J, Lee WP, Snipas SJ, Salvesen GS, Morris LX, Fitzgerald L, Zhang Y, Bertram EM, Goodnow CC, Dixit VM. 2015. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nat 2015 5267575 526:666–671. [DOI] [PubMed] [Google Scholar]
- 26.Rathinam VAK, Vanaja SK, Waggoner L, Sokolovska A, Becker C, Stuart LM, Leong JM, Fitzgerald KA. 2012. TRIF licenses caspase-11-dependent NLRP3 inflammasome activation by gram-negative bacteria. Cell 150:606–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Gurung P, Subbarao Malireddi RK, Anand PK, Demon D, Vande Walle L, Liu Z, Vogel P, Lamkanfi M, Kanneganti TD. 2012. Toll or interleukin-1 receptor (TIR) domain-containing adaptor inducing interferon-β (TRIF)-mediated caspase-11 protease production integrates toll-like receptor 4 (TLR4) protein- and Nlrp3 inflammasome-mediated host defense against enteropathogens. J Biol Chem 287:34474–34483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Broz P, Ruby T, Belhocine K, Bouley DM, Kayagaki N, Dixit VM, Monack DM. 2012. Caspase-11 increases susceptibility to Salmonella infection in the absence of caspase-1. Nat 2012 4907419 490:288–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kayagaki N, Wong MT, Stowe IB, Ramani SR, Gonzalez LC, Akashi-Takamura S, Miyake K, Zhang J, Lee WP, Muszynśki A, Forsberg LS, Carlson RW, Dixit VM. 2013. Noncanonical inflammasome activation by intracellular LPS independent of TLR4. Science 341:1246–1249. [DOI] [PubMed] [Google Scholar]
- 30.Shi J, Zhao Y, Wang Y, Gao W, Ding J, Li P, Hu L, Shao F. 2014. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature 514:187–192. [DOI] [PubMed] [Google Scholar]
- 31.Hagar JA, Powell DA, Aachoui Y, Ernst RK, Miao EA. 2013. Cytoplasmic LPS activates caspase-11: implications in TLR4-independent endotoxic shock. Science 341:1250–1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lamkanfi M, Dixit VM. 2014. Mechanisms and Functions of Inflammasomes. Cell 157:1013–1022. [DOI] [PubMed] [Google Scholar]
- 33.Lamkanfi M, Dixit VM. 2010. Manipulation of Host Cell Death Pathways during Microbial Infections. Cell Host Microbe 8:44–54. [DOI] [PubMed] [Google Scholar]
- 34.Labbé K, Saleh M. 2008. Cell death in the host response to infection. Cell Death Differ 2008 159 15:1339–1349. [DOI] [PubMed] [Google Scholar]
- 35.Cornelis GR, Wolf-Watz H. 1997. The Yersinia Yop virulon: a bacterial system for subverting eukaryotic cells. Mol Microbiol 23:861–867. [DOI] [PubMed] [Google Scholar]
- 36.Bliska JB, Wang X, Viboud GI, Brodsky IE. 2013. Modulation of innate immune responses by Yersinia type III secretion system translocators and effectors. Cell Microbiol 15:1622–1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Viboud GI, Bliska JB. 2005. YERSINIA OUTER PROTEINS: Role in Modulation of Host Cell Signaling Responses and Pathogenesis. https://doi.org/101146/annurev.micro59030804121320 59:69–89. [DOI] [PubMed] [Google Scholar]
- 38.Palmer LE, Hobble S, Galán JE, Bliska JB. 1998. YopJ of Yersinia pseudotuberculosis is required for the inhibition of macrophage TNF-α production and downregulation of the MAP kinases p38 and JNK. Mol Microbiol 27:953–965. [DOI] [PubMed] [Google Scholar]
- 39.Orth K, Palmer LE, Bao ZQ, Stewart S, Rudolph AE, Bliska JB, Dixon JE. 1999. Inhibition of the Mitogen-Activated Protein Kinase Kinase Superfamily by a Yersinia Effector. Science (80- ) 285:1920–1923. [DOI] [PubMed] [Google Scholar]
- 40.Medici NP, Rashid M, Bliska JB. 2019. Characterization of pyrin dephosphorylation and inflammasome activation in macrophages as triggered by the Yersinia effectors YopE and YopT. Infect Immun 87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Wang X, Parashar K, Sitaram A, Bliska JB. 2014. The GAP Activity of Type III Effector YopE Triggers Killing of Yersinia in Macrophages. PLoS Pathog 10:1004346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zwack EE, Feeley EM, Burton AR, Baofeng H, Yamamoto M, Kanneganti T-D, Bliska JB, Coers J, Brodsky IE. 2017. Guanylate Binding Proteins Regulate Inflammasome Activation in Response to. Infect Immun 85:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zwack EE, Snyder AG, Wynosky-Dolfi MA, Ruthel G, Philip NH, Marketon MM, Francis MS, Bliska JB, Brodsky IE. 2015. Inflammasome activation in response to the yersinia type III secretion system requires hyperinjection of translocon proteins YopB and YopD. MBio 6:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Philip NH, Dillon CP, Snyder AG, Fitzgerald P, Wynosky-Dolfi MA, Zwack EE, Hu B, Fitzgerald L, Mauldin EA, Copenhaver AM, Shin S, Wei L, Parker M, Zhang J, Oberst A, Green DR, Brodsky IE. 2014. Caspase-8 mediates caspase-1 processing and innate immune defense in response to bacterial blockade of NF- B and MAPK signaling. Proc Natl Acad Sci 111:7385–7390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Brodsky IE, Palm NW, Sadanand S, Ryndak MB, Sutterwala FS, Flavell RA, Bliska JB, Medzhitov R. 2010. A Yersinia effector protein promotes virulence by preventing inflammasome recognition of the type III secretion system. Cell Host Microbe 7:376–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Chung LK, Park YH, Zheng Y, Brodsky IE, Hearing P, Kastner DL, Chae JJ, Bliska JB. 2016. The Yersinia Virulence Factor YopM Hijacks Host Kinases to Inhibit Type III Effector-Triggered Activation of the Pyrin Inflammasome. Cell Host Microbe 20:296–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Lopes Fischer N, Naseer N, Shin S, Brodsky IE. 2020. Effector-triggered immunity and pathogen sensing in metazoans. Nat Microbiol 5:14–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Knodler LA, Crowley SM, Sham HP, Yang H, Wrande M, Ma C, Ernst RK, Steele-Mortimer O, Celli J, Vallance BA. 2014. Noncanonical inflammasome activation of caspase-4/caspase-11 mediates epithelial defenses against enteric bacterial pathogens. Cell Host Microbe 16:249–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Naseer N, Zhang J, Bauer R, Constant DA, Nice TJ, Brodsky IE, Rauch I, Shin S. 2022. Salmonella enterica Serovar Typhimurium Induces NAIP/NLRC4- And NLRP3/ASC-Independent, Caspase-4-Dependent Inflammasome Activation in Human Intestinal Epithelial Cells. Infect Immun 90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Matsuura M, Takahashi H, Watanabe H, Saito S, Kawahara K. 2010. Immunomodulatory effects of Yersinia pestis lipopolysaccharides on human macrophages. Clin Vaccine Immunol 17:49–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Montminy SW, Khan N, McGrath S, Walkowicz MJ, Sharp F, Conlon JE, Fukase K, Kusumoto S, Sweet C, Miyake K, Akira S, Cotter RJ, Goguen JD, Lien E. 2006. Virulence factors of Yersinia pestis are overcome by a strong lipopolysaccharide response. Nat Immunol 7:1066–1073. [DOI] [PubMed] [Google Scholar]
- 52.Hajjar AM, Tsai JH, Wilson CB, Miller SI. 2002. Human Toll-like receptor 4 recognizes host-specific LPS modifications. Nat Immunol 3:354–359. [DOI] [PubMed] [Google Scholar]
- 53.Luchetti G, Roncaioli JL, Chavez RA, Kayagaki N, Vance RE, Dixit Correspondence VM, Schubert AF, Kofoed EM, Reja R, Cheung TK, Liang Y, Webster JD, Lehoux I, Skippington E, Reeder J, Haley B, Tan MW, Rose CM, Newton K, Dixit VM. 2021. In brief Shigella ubiquitin ligase IpaH7.8 targets gasdermin D for degradation to prevent pyroptosis and enable infection The pore-forming protein gasdermin. Cell Host Microbe 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Alexander-Floyd J, Bass AR, Harberts EM, Grubaugh D, Buxbaum JD, Brodsky IE, Ernst RK, Shin S. 2022. Lipid A Variants Activate Human TLR4 and the Noncanonical Inflammasome Differently and Require the Core Oligosaccharide for Inflammasome Activation. Infect Immun 90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Harberts EM, Grubaugh D, Akuma DC, Shin S, Ernst RK, Brodsky IE. 2022. Position-Specific Secondary Acylation Determines Detection of Lipid A by Murine TLR4 and Caspase-11. Infect Immun 90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sarhan J, Liu BC, Muendlein HI, Li P, Nilson R, Tang AY, Rongvaux A, Bunnell SC, Shao F, Green DR, Poltorak A. 2018. Caspase-8 induces cleavage of gasdermin D to elicit pyroptosis during Yersinia infection. Proc Natl Acad Sci U S A 115:E10888–E10897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Valeria MRR, Ramirez J, Naseer N, Palacio NM, Siddarthan IJ, Yan BM, Boyer MA, Pensinger DA, Sauer JD, Shin S. 2017. Broad detection of bacterial type III secretion system and flagellin proteins by the human NAIP/NLRC4 inflammasome. Proc Natl Acad Sci U S A 114:13242–13247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Naseer N, Egan MS, Reyes Ruiz VM, Scott WP, Hunter EN, Demissie T, Rauch I, Brodsky IE, Shin S. 2022. Human NAIP/NLRC4 and NLRP3 inflammasomes detect Salmonella type III secretion system activities to restrict intracellular bacterial replication. PLoS Pathog 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Gram AM, Wright JA, Pickering RJ, Lam NL, Booty LM, Webster SJ, Bryant CE. 2021. Salmonella Flagellin Activates NAIP/NLRC4 and Canonical NLRP3 Inflammasomes in Human Macrophages. J Immunol 206:631–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kobayashi T, Ogawa M, Sanada T, Mimuro H, Kim M, Ashida H, Akakura R, Yoshida M, Kawalec M, Reichhart JM, Mizushima T, Sasakawa C. 2013. The Shigella OspC3 effector inhibits caspase-4, antagonizes inflammatory cell death, and promotes epithelial infection. Cell Host Microbe 13:570–583. [DOI] [PubMed] [Google Scholar]
- 61.Egan MS, Zhang J, Shin S. 2023. Human and mouse NAIP/NLRC4 inflammasome responses to bacterial infection. Curr Opin Microbiol 73:102298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Sellin ME, Müller AA, Felmy B, Dolowschiak T, Diard M, Tardivel A, Maslowski KM, Hardt WD. 2014. Epithelium-intrinsic NAIP/NLRC4 inflammasome drives infected enterocyte expulsion to restrict salmonella replication in the intestinal mucosa. Cell Host Microbe 16:237–248. [DOI] [PubMed] [Google Scholar]
- 63.Hausmann A, Böck D, Geiser P, Berthold DL, Fattinger SA, Furter M, Bouman JA, Barthel-Scherrer M, Lang CM, Bakkeren E, Kolinko I, Diard M, Bumann D, Slack E, Regoes RR, Pilhofer M, Sellin ME, Hardt W-D. 2020. Intestinal epithelial NAIP/NLRC4 restricts systemic dissemination of the adapted pathogen Salmonella Typhimurium due to site-specific bacterial PAMP expression. Mucosal Immunol 10.1038/s41385-019-0247-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Rauch I, Deets KA, Ji DX, von Moltke J, Tenthorey JL, Lee AY, Philip NH, Ayres JS, Brodsky IE, Gronert K, Vance RE. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Mitchell PS, Roncaioli JL, Turcotte EA, Goers L, Chavez RA, Lee AY, Lesser CF, Rauch I, Vance RE. 2020. Naip–nlrc4-deficient mice are susceptible to shigellosis. Elife 9:1–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Roncaioli JL, Babirye JP, Chavez RA, Liu FL, Turcotte EA, Lee AY, Lesser CF, Vance RE. 2022. A hierarchy of cell death pathways confers layered resistance to shigellosis in mice. bioRxiv 2022.09.21.508939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Pallett MA, Crepin VF, Serafini N, Habibzay M, Kotik O, Sanchez-Garrido J, Di Santo JP, Shenoy AR, Berger CN, Frankel G. 2016. Bacterial virulence factor inhibits caspase-4/11 activation in intestinal epithelial cells. Mucosal Immunol 2017 103 10:602–612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Steeghs L, Keestra AM, Van Mourik A, Uronen-Hansson H, Van Der Ley P, Callard R, Klein N, Van Putten JPM. 2008. Differential activation of human and mouse Toll-like receptor 4 by the adjuvant candidate LpxL1 of Neisseria meningitidis. Infect Immun 76:3801–3807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lei-Leston AC, Murphy AG, Maloy KJ. 2017. Epithelial Cell Inflammasomes in Intestinal Immunity and Inflammation. Front Immunol 8:1168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Marra A, Isberg RR. 1997. Invasin-dependent and invasin-independent pathways for translocation of Yersinia pseudotuberculosis across the Peyer’s patch intestinal epithelium. Infect Immun 65:3412–3421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Clark MA, Hirst BH, Jepson MA. 1998. M-cell surface β1 integrin expression and invasin-mediated targeting of Yersinia pseudotuberculosis to mouse Peyer’s patch M cells. Infect Immun 66:1237–1243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Isberg RR, Leong JM. 1990. Multiple beta 1 chain integrins are receptors for invasin, a protein that promotes bacterial penetration into mammalian cells. Cell 60:861–871. [DOI] [PubMed] [Google Scholar]
- 73.Autenrieth IB, Firsching R. 1996. Penetration of M cells and destruction of Peyer’s patches by Yersinia enterocolitica: an ultrastructural and histological study. J Med Microbiol 44:285–294. [DOI] [PubMed] [Google Scholar]
- 74.Sorobetea D, Matsuda R, Peterson ST, Grayczyk JP, Rao I, Krespan E, Lanza M, Assenmacher C-A, Mack M, Beiting DP, Radaelli E, Brodsky IE. 2023. Inflammatory monocytes promote granuloma control of Yersinia infection. Nat Microbiol 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ashida H, Mimuro H, Ogawa M, Kobayashi T, Sanada T, Kim M, Sasakawa C. 2011. Cell death and infection: A double-edged sword for host and pathogen survival. J Cell Biol 195:931–942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Fink SL, Cookson BT. 2007. Pyroptosis and host cell death responses during Salmonella infection. Cell Microbiol 9:2562–2570. [DOI] [PubMed] [Google Scholar]
- 77.Thinwa J, Segovia JA, Bose S, Dube PH. 2014. Integrin-Mediated First Signal for Inflammasome Activation in Intestinal Epithelial Cells. J Immunol 193:1373–1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Denecker G, Declercq W, Geuijen CAW, Boland A, Benabdillah R, Van Gurp M, Sory MP, Vandenabeele P, Cornelis GR. 2001. Yersinia enterocolitica YopP-induced apoptosis of macrophages involves the apoptotic signaling cascade upstream of bid. J Biol Chem 276:19706–19714. [DOI] [PubMed] [Google Scholar]
- 79.Lilo S, Zheng Y, Bliska JB. 2008. Caspase-1 activation in macrophages infected with Yersinia pestis KIM requires the type III secretion system effector YopJ. Infect Immun 76:3911–3923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Spinner JL, Seo KS, O’Loughlin JL, Cundiff JA, Minnich SA, Bohach GA, Kobayashi SD. 2010. Neutrophils Are Resistant to Yersinia YopJ/P-Induced Apoptosis and Are Protected from ROS-Mediated Cell Death by the Type III Secretion System. PLoS One 5:e9279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Man SM, Hopkins LJ, Nugent E, Cox S, Glück IM, Tourlomousis P, Wright JA, Cicuta P, Monie TP, Bryant CE. 2014. Inflammasome activation causes dual recruitment of NLRC4 and NLRP3 to the same macromolecular complex. Proc Natl Acad Sci U S A 111:7403–7408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sarhan J, Liu BC, Muendlein HI, Li P, Nilson R, Tang AY, Rongvaux A, Bunnell SC, Shao F, Green DR, Poltorak A. 2018. Caspase-8 induces cleavage of gasdermin D to elicit pyroptosis during Yersinia infection. Proc Natl Acad Sci U S A 115:E10888–E10897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Weng D, Marty-Roix R, Ganesan S, Proulx MK, Vladimer GI, Kaiser WJ, Mocarski ES, Pouliot K, Chan FKM, Kelliher MA, Harris PA, Bertin J, Gough PJ, Shayakhmetov DM, Goguen JD, Fitzgerald KA, Silverman N, Lien E. 2014. Caspase-8 and RIP kinases regulate bacteria-induced innate immune responses and cell death. Proc Natl Acad Sci U S A 111:7391–7396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Antonopoulos C, Russo HM, El Sanadi C, Martin BN, Li X, Kaiser WJ, Mocarski ES, Dubyak GR. 2015. Caspase-8 as an Effector and Regulator of NLRP3 Inflammasome Signaling. J Biol Chem 290:20167–20184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Bass A. 2020. Elucidating Human Inflammasome Responses To Legionella Pneumophila And Lipopolysaccharide Variants. Publicly Access Penn Diss. [Google Scholar]
- 86.Agard NJ, Maltby D, Wells JA. 2010. Inflammatory stimuli regulate caspase substrate profiles. Mol Cell Proteomics 9:880–893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.He WT, Wan H, Hu L, Chen P, Wang X, Huang Z, Yang ZH, Zhong CQ, Han J. 2015. Gasdermin D is an executor of pyroptosis and required for interleukin-1β secretion. Cell Res 2015 2512 25:1285–1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Liu X, Zhang Z, Ruan J, Pan Y, Magupalli VG, Wu H, Lieberman J. 2016. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nat 2016 5357610 535:153–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Sborgi L, Rühl S, Mulvihill E, Pipercevic J, Heilig R, Stahlberg H, Farady CJ, Müller DJ, Broz P, Hiller S. 2016. GSDMD membrane pore formation constitutes the mechanism of pyroptotic cell death. EMBO J 35:1766–1778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Ding J, Wang K, Liu W, She Y, Sun Q, Shi J, Sun H, Wang DC, Shao F. 2016. Poreforming activity and structural autoinhibition of the gasdermin family. Nat 2016 5357610 535:111–116. [DOI] [PubMed] [Google Scholar]
- 91.Aglietti RA, Estevez A, Gupta A, Ramirez MG, Liu PS, Kayagaki N, Ciferri C, Dixit VM, Dueber EC. 2016. GsdmD p30 elicited by caspase-11 during pyroptosis forms pores in membranes. Proc Natl Acad Sci U S A 113:7858–7863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Hu JJ, Liu X, Xia S, Zhang Z, Zhang Y, Zhao J, Ruan J, Luo X, Lou X, Bai Y, Wang J, Hollingsworth LR, Magupalli VG, Zhao L, Luo HR, Kim J, Lieberman J, Wu H. 2020. FDA-approved disulfiram inhibits pyroptosis by blocking gasdermin D pore formation. Nat Immunol 2020 217 21:736–745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kummer JA, Broekhuizen R, Everett H, Agostini L, Kuijk L, Martinon F, Van Bruggen R, Rg Tschopp J. Inflammasome Components NALP 1 and 3 Show Distinct but Separate Expression Profiles in Human Tissues Suggesting a Site-specific Role in the Inflammatory Response 10.1369/jhc.6A7101.2006. [DOI] [PubMed] [Google Scholar]
- 94.Sellin ME, Maslowski KM, Maloy KJ, Hardt WD. 2015. Inflammasomes of the intestinal epithelium. Trends Immunol 36:442–450. [DOI] [PubMed] [Google Scholar]
- 95.Dersch P, Isberg RR. 2000. An Immunoglobulin Superfamily-Like Domain Unique to the Yersinia pseudotuberculosis Invasin Protein Is Required for Stimulation of Bacterial Uptake via Integrin Receptors. Infect Immun 68:2930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Isberg RR. 1989. Determinants for thermoinducible cell binding and plasmid-encoded cellular penetration detected in the absence of the Yersinia pseudotuberculosis invasin protein. Infect Immun 57:1998–2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Isberg RR, Tran Van Nhieu G. 1994. Binding and internalization of microorganisms by integrin receptors. Trends Microbiol 2:10–14. [DOI] [PubMed] [Google Scholar]
- 98.Fallman M, Andersson K, Hakansson S, Magnusson KE, Stendahl O, Wolf- Watz H. 1995. Yersinia pseudotuberculosis inhibits Fc receptor-mediated phagocytosis in J774 cells. Infect Immun 63:3117–3124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Bliska JB, Copass MC, Falkow S. 1993. The Yersinia pseudotuberculosis adhesin YadA mediates intimate bacterial attachment to and entry into HEp-2 cells. Infect Immun 61:3914–3921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Rosqvist R, Forsberg, Rimpiläinen M, Bergman T, Wolf-Watz H. 1990. The cytotoxic protein YopE of Yersinia obstructs the primary host defence. Mol Microbiol 4:657–667. [DOI] [PubMed] [Google Scholar]
- 101.Andersson K, Carballeira N, Magnusson KE, Persson C, Stendahl O, Wolf-Watz H, Fällman M. 1996. YopH of Yersinia pseudotuberculosis interrupts early phosphotyrosine signalling associated with phagocytosis. Mol Microbiol 20:1057–1069. [DOI] [PubMed] [Google Scholar]
- 102.Black DS, Bliska JB. 2000. The RhoGAP activity of the Yersinia pseudotuberculosis cytotoxin YopE is required for antiphagocytic function and virulence. Mol Microbiol 37:515–527. [DOI] [PubMed] [Google Scholar]
- 103.Cornelis GR. 2002. The Yersinia Ysc–Yop “Type III” weaponry. Nat Rev Mol Cell Biol 2002 310 3:742–753. [DOI] [PubMed] [Google Scholar]
- 104.Fasciano AC, Dasanayake GS, Estes MK, Zachos NC, Breault DT, Isberg RR, Tan S, Mecsas J. 2021. Yersinia pseudotuberculosis YopE prevents uptake by M cells and instigates M cell extrusion in human ileal enteroid-derived monolayers. Gut Microbes 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Black DS, Bliska JB. 1997. Identification of p130Cas as a substrate of Yersinia YopH (Yop51), a bacterial protein tyrosine phosphatase that translocates into mammalian cells and targets focal adhesions. EMBO J 16:2730–2744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Persson C, Carballeira N, Wolf-Watz H, Fällman M. 1997. The PTPase YopH inhibits uptake of Yersinia, tyrosine phosphorylation of p130Cas and FAK, and the associated accumulation of these proteins in peripheral focal adhesions. EMBO J 16:2307–2318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Hamid N, Gustavsson A, Andersson K, McGee K, Persson C, Rudd CE, Fällman M. 1999. YopH dephosphorylates Cas and Fyn-binding protein in macrophages. Microb Pathog 27:231–242. [DOI] [PubMed] [Google Scholar]
- 108.Rosqvist R, Forsberg A, Wolf-Watz H. 1991. Intracellular targeting of the Yersinia YopE cytotoxin in mammalian cells induces actin microfilament disruption. Infect Immun 59:4562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Viboud GI, Bliska JB. 2001. A bacterial type III secretion system inhibits actin polymerization to prevent pore formation in host cell membranes. EMBO J 20:5373–5382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Wong KW, Mohammadi S, Isberg RR. 2008. The Polybasic Region of Rac1 Modulates Bacterial Uptake Independently of Self-association and Membrane Targeting. J Biol Chem 283:35954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Wong KW, Isberg RR. 2005. Yersinia pseudotuberculosis Spatially Controls Activation and Misregulation of Host Cell Rac1. PLOS Pathog 1:e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Alrutz MA, Srivastava A, Wong KW, D’Souza-Schorey C, Tang M, Ch’ng LE, Snapper SB, Isberg RR. 2001. Efficient uptake of Yersinia pseudotuberculosis via integrin receptors involves a Rac1-Arp 2/3 pathway that bypasses N-WASP function. Mol Microbiol 42:689–703. [DOI] [PubMed] [Google Scholar]
- 113.Rottner K, Stradal TEB, Wehland J. 2005. Bacteria-host-cell interactions at the plasma membrane: stories on actin cytoskeleton subversion. Dev Cell 9:3–17. [DOI] [PubMed] [Google Scholar]
- 114.Ridley AJ. 2006. Rho GTPases and actin dynamics in membrane protrusions and vesicle trafficking. Trends Cell Biol 16:522–529. [DOI] [PubMed] [Google Scholar]
- 115.Parsons JT. 2003. Focal adhesion kinase: the first ten years. J Cell Sci 116:1409–1416. [DOI] [PubMed] [Google Scholar]
- 116.Uliczka F, Kornprobst T, Eitel J, Schneider D, Dersch P. 2009. Cell invasion of Yersinia pseudotuberculosis by invasin and YadA requires protein kinase C, phospholipase C-γ1 and Akt kinase. Cell Microbiol 11:1782–1801. [DOI] [PubMed] [Google Scholar]
- 117.Eitel J, Dersch P. 2002. The YadA Protein of Yersinia pseudotuberculosis Mediates High-Efficiency Uptake into Human Cells under Environmental Conditions in Which Invasin Is Repressed. Infect Immun 70:4880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Isberg RR, Voorhis DL, Falkow S. 1987. Identification of invasin: a protein that allows enteric bacteria to penetrate cultured mammalian cells. Cell 50:769–778. [DOI] [PubMed] [Google Scholar]
- 119.Maldonado-Arocho FJ, Green C, Fisher ML, Paczosa MK, Mecsas J. 2013. Adhesins and Host Serum Factors Drive Yop Translocation by Yersinia into Professional Phagocytes during Animal Infection. PLoS Pathog 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Simonet M, Falkow S. 1992. Invasin expression in Yersinia pseudotuberculosis. Infect Immun 60:4414–4417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.de Rouvroit CL, Sluiters C, Cornelis GR. 1992. Role of the transcriptional activator, VirF, and temperature in the expression of the pYV plasmid genes of Yersinia enterocolitica. Mol Microbiol 6:395–409. [DOI] [PubMed] [Google Scholar]
- 122.Sharma D, Malik A, Guy CS, Karki R, Vogel P, Kanneganti TD. 2018. Pyrin Inflammasome Regulates Tight Junction Integrity to Restrict Colitis and Tumorigenesis. Gastroenterology 154:948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Saavedra PHV, Huang L, Ghazavi F, Kourula S, Vanden Berghe T, Takahashi N, Vandenabeele P, Lamkanfi M. 2018. Apoptosis of intestinal epithelial cells restricts Clostridium difficile infection in a model of pseudomembranous colitis. Nat Commun 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Pentecost M, Kumaran J, Ghosh P, Amieva MR. 2010. Listeria monocytogenes Internalin B Activates Junctional Endocytosis to Accelerate Intestinal Invasion. PLoS Pathog 6:1000900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.McCormick BA, Nusrat A, Parkos CA, D’Andrea L, Hofman PM, Carnes D, Liang TW, Madara JL. 1997. Unmasking of intestinal epithelial lateral membrane β1 integrin consequent to transepithelial neutrophil migration in vitro facilitates inv- mediated invasion by Yersinia pseudotuberculosis. Infect Immun 65:1414–1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Tafazoli F, Holmstrom A, Forsberg A, Magnusson KE. 2000. Apically exposed, tight junction-associated beta1-integrins allow binding and YopE-mediated perturbation of epithelial barriers by wild-type Yersinia bacteria. Infect Immun 68:5335–5343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Feeley EM, Pilla-Moffett DM, Zwack EE, Piro AS, Finethy R, Kolb JP, Martinez J, Brodsky IE, Coers J. 2017. Galectin-3 directs antimicrobial guanylate binding proteins to vacuoles furnished with bacterial secretion systems. Proc Natl Acad Sci U S A 114:E1698–E1706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Zhang Y, Murtha J, Roberts MA, Siegel RM, Bliska JB. 2008. Type III secretion decreases bacterial and host survival following phagocytosis of Yersinia pseudotuberculosis by macrophages. Infect Immun 76:4299–4310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Livak KJ, Schmittgen TD. 2001. Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2−ΔΔCT Method. Methods 25:402–408. [DOI] [PubMed] [Google Scholar]
- 130.Viboud GI, So SSK, Ryndak MB, Bliska JB. 2003. Proinflammatory signalling stimulated by the type III translocation factor YopB is counteracted by multiple effectors in epithelial cells infected with Yersinia pseudotuberculosis. Mol Microbiol 47:1305–1315. [DOI] [PubMed] [Google Scholar]
- 131.Ryndak MB, Chung H, London E, Bliska JB. 2005. Role of predicted transmembrane domains for Type III translocation, pore formation, and signaling by the Yersinia pseudotuberculosis YopB protein. Infect Immun 73:2433–2443. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All data are included in the manuscript and supplemental material. Bacterial strains available upon request




