Skip to main content
PLOS Pathogens logoLink to PLOS Pathogens
. 2025 May 13;21(5):e1013167. doi: 10.1371/journal.ppat.1013167

Caspase-1-licensed pyroptosis drives dsRNA-mediated necroptosis and dampens host defense against bacterial pneumonia

Qinyu Luo 1, Lihua Shen 1, Shiyue Yang 2, Yan Zhang 1, Yihang Pan 1, Zehua Wu 1, Qiang Shu 1,*, Qixing Chen 1,*
Editor: Andrew J Monteith3
PMCID: PMC12121916  PMID: 40359428

Abstract

Bacterial lung infections cause severe host responses. Here, we showed that global deficiency of caspase-1 can protect against lethal pulmonary Escherichia coli infection by reducing the necroptosis of infiltrated neutrophils, which are key players in immune responses in the lung. Mechanistically, neutrophil necroptosis was not directly triggered in a cell-intrinsic manner by invading bacteria but was triggered by bacteria-stimulated pyroptotic epithelial cell supernatants in vitro. In validation experiments, chimeric mice with nonhematopoietic caspase-1 or GSDMD knockout were protected from lung E. coli infection and exhibited decreased neutrophil death. Nonhematopoietic pyroptosis facilitates the release of dsRNAs and contributes to neutrophil ZBP1-related necroptosis. Moreover, blocking dsRNA or depleting ZBP1 ameliorated the pathophysiological process of pulmonary E. coli infection. Overall, our results demonstrate a paradigm of communication between necroptosis and pyroptosis in different cell types in cooperation with microbes and hosts and suggest that therapeutic targeting of the pyroptosis or necroptosis pathway may prevent pulmonary bacterial infection.

Author summary

Bacterial lung infection has caused many deaths globally, leading to a major economic burden on health systems. The emergence of drug-resistant bacteria has reduced the efficacy of widely used antibiotics. Bacterial invasion induces multiple host responses, including regulated cell death; these pathways participate in the pathological process of lung infection and could be potential targets in the treatment of disease. However, the precise mechanism underlying this type of regulated cell death remains unclear. Here, using a bacterial lung infection model combined with several genetically engineered mice and chimeric mice, we show that two types of regulated cell death, pyroptosis and necroptosis, contribute to the pathobiology of bacterial lung infections. Furthermore, pyroptotic cells promote neutrophil necroptosis through a double-stranded RNA-related pathway. We also found that intervention in the pyroptosis–necroptosis communication pathway ameliorated bacterial lung infection in a mouse model. Our study provides new insights into the triggering mechanisms of regulated cell death and suggests novel targets for the treatment of bacterial lung infection.

Introduction

Lower respiratory tract infections, such as pneumonia, lead to global health issues and cause more deaths worldwide than other infectious diseases [1]. Moreover, these infections are the leading cause of death in children among all diseases [2]. In the USA, community-acquired pneumonia accounts for 1.5 million hospitalizations annually and has a mortality rate of 30.6% [3]. Bacteria are the most commonly identified pathogens in patients with pneumonia [4,5]. Bacterial pneumonia leads to substantial morbidity and mortality and results in a major clinical and economic burden on health care systems worldwide [6]. More importantly, the emergence of drug-resistant bacteria has reduced the efficacy of antibiotics [7]. Thus, understanding the molecular and cellular mechanisms involved in the pathogenesis of bacterial pneumonia is urgently needed for the development of novel clinical therapeutic strategies.

The mechanisms underlying bacterial pneumonia remain unclear. Recently, inflammasomes have been proposed to participate in the pathogenesis of bacterial pneumonia [8]. The inflammasome is a cytoplasmic multiprotein complex that is expressed primarily in immune cells and barrier epithelial cells. As a first-line response in host‒pathogen interactions, inflammasomes detect various sterile or infectious stimuli within the cell and induce subsequent cellular responses. During this process, multiple cytosolic pattern recognition receptors (PRRs) are responsible for stimulus sensing and the assembly of their inflammasomes. For example, the nucleotide-binding oligomerization domain-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome senses pore-forming toxin-induced potassium efflux, the nucleotide-binding oligomerization domain-like receptor family caspase recruitment domain containing 4 (NLRC4) inflammasome senses bacterial flagellin, and the NLRP6 inflammasome senses lipoteichoic acid in gram-positive bacteria. Nevertheless, these canonical inflammasomes all assemble and recruit the same adaptor protein, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and the same effector protein, pro-caspase-1. Upon inflammasome activation, caspase-1 cleaves pro-interleukin (IL)-1β and gasdermin D (GSDMD), leading to inflammatory cytokine release and lytic cell death [9]. Many studies have investigated the potential roles of these canonical inflammasomes in pneumonia. However, the conclusions are inconsistent and complicated. Cohen et al. and Kebaier et al. reported that depletion of NLRP3 or inhibition of caspase-1 increased the bacterial killing ability of macrophages and conferred resistance to Staphylococcus aureus pneumonia in mice [10,11]. Cohen et al. also reported that depletion of NLRC4 or inhibition of caspase-1 alleviated Pseudomonas aeruginosa pneumonia in mice [12]. However, in the case of pulmonary Streptococcus pneumoniae infection, McNeela et al. and Witzenrath et al. reported that the depletion of canonical inflammasomes worsens the disease, leading to poorer bacterial clearance and vital signs [13,14]. These findings suggest that additional processes interact with canonical inflammasome activation in a pathogen-specific manner to further control the pathogenesis of bacterial pneumonia.

Recently, necroptosis has been found to be involved in the pathogenesis of bacterial pneumonia. Necroptosis is a type of regulated cell death involving necrosis that occurs during development, inflammation and disease in multiple cell types, including neurons, epithelial cells, hepatocytes and immune cells [15,16]. Unlike pyroptosis or apoptosis, which are regulated by proteins from the caspase family, necroptosis is regulated mainly by the receptor-interacting protein kinase (RIPK) family [17]. Upon activation, the Toll/IL-1 receptor domain-containing adaptor protein inducing interferon-β (TRIF), Z-DNA binding protein 1 (ZBP1), or RIPK1 hetero-oligomerizes with RIPK3 and induces the assembly of an amyloid-like structure of RIPK3 homo-oligomers. These homo-oligomers act as docking stations for the recruitment of the pseudokinase mixed-lineage kinase domain-like (MLKL), the pore-forming executioner of necroptosis [18]. Necroptosis plays an important role in both noninfectious and infectious diseases. With respect to bacterial pneumonia, Gonzalez-Juarbe et al. reported that the administration of an MLKL inhibitor reduced bacterial titers and increased macrophage numbers during pulmonary Serratia marcescens infection in mice [19]. Kitur et al. reported that treatment with the RIPK1 inhibitor NEC-1s or depletion of RIPK3 increased bacterial clearance and facilitated immune cell infiltration during pulmonary S. aureus infection in mice [20]. However, how and through which pathways necroptosis is triggered during bacterial pneumonia remain unclear. Moreover, necroptosis is associated with inflammasome activation in several ways. Lee et al. reported that upon infection with certain viruses or bacteria, MLKL can be activated by absent in melanoma 2 (AIM2) and caspase-1-associated protein complexes termed PANoptosomes [21]. Whether necroptosis communicates with other cell death processes, such as inflammasome-related pyroptosis, during bacterial pneumonia is unclear.

Since caspase-1 represents the core component of various inflammasomes and Escherichia coli is a typical gram-negative bacterium that activates caspase-1 by stimulating various inflammasomes [2224], we used a model of pulmonary E. coli infection in mice globally deficient in caspase-1 to address the aforementioned questions. We showed that the activation of caspase-1 contributed to the pathophysiology of pulmonary E. coli infection and facilitated lung neutrophil necroptosis. We found that neutrophil necroptosis was triggered not directly by bacteria but also by pyroptotic nonhematopoietic cells during E. coli infection. We further demonstrated that double-stranded RNA (dsRNA) and ZBP1 mediated neutrophil necroptosis and exacerbated pulmonary E. coli infection. Our results provide a previously undescribed paradigm of communication between necroptosis and pyroptosis in different cell types in response to microbial infection.

Results

Lung neutrophils are critical for defense against E. coli pneumonia in Casp1-/- mice

We first investigated the role of canonical inflammasomes in an E. coli pneumonia model using caspase-1 knockout (Casp1-/-) mice and Casp1+/+ mice. Compared with the Casp1+/+ mice, the Casp1-/- mice presented an increased survival rate during the 72-hour observation period after E. coli challenge (Fig 1A), suggesting that the caspase-1 inflammasome participates in the pathogenesis of E. coli pneumonia. We then monitored vital pulmonary signs of infection in the Casp1-/- and Casp1+/+ mice at 12 hours after E. coli challenge. Consistent with the survival data, the Casp1-/- mice maintained their body temperature and respiratory rate (S1A and S1B Fig) and did not exhibit severe tissue injury (Fig 1B). The Casp1-/- mice also presented a decreased protein concentration, indicating less injury to the alveolar capillary barrier and a lower bacterial load in whole lung tissue (S1C and S1D Fig). In addition, significantly lower levels of two major proinflammatory cytokines, IL-1β and TNF-α, were observed in the Casp1-/- mice (S1E and S1F Fig).

Fig 1. Lung neutrophils are critical for defense against E. coli pneumonia in Casp1-/- mice.

Fig 1

(A) Survival of Casp1-/- mice (n = 9) and Casp1+/+ mice (n = 13) after pulmonary E. coli infection. This experiment was conducted across three independent replicates, with each replicate including 3–5 mice. (B) Representative images of H&E-stained lungs from Casp1-/- mice (n = 8) and Casp1+/+ mice (n = 7) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each) and quantification of lung injury scores. Scale bars, 100 µm. (C) Counts of leukocytes in the BALF of Casp1-/- mice (n = 11) and Casp1+/+ mice (n = 8) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each). (D and E) Counts of macrophages and neutrophils in the BALF of Casp1-/- mice (n = 11) and Casp1+/+ mice (n = 8) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each). (F) Survival of Casp1-/- mice treated with anti-Ly6G antibody and IgG control antibody after pulmonary E. coli infection (n = 6 each, conducted across two independent replicates, with each replicate including 3 mice). (G) Survival of Casp1-/- mice treated with clodronate liposomes (n = 6) or control liposomes (n = 5) after pulmonary E. coli infection. This experiment was conducted across two independent replicates, with each replicate including 2–3 mice. The data are shown as the means ± SDs in (B, C, D and E). Statistical differences were determined by the Mantel‒Cox test (A, F and G) and two-way ANOVA (B, C, D and E). *P < 0.05; **P < 0.01; ***P < 0.001; ns, not significant. See also S1 Fig.

Leukocytes constitute the first-line immune defense against pathogen invasion, and they infiltrate infected tissue and perform immune functions. We then counted the number of leukocytes in the bronchoalveolar lavage fluid (BALF) at 12 hours after E. coli infection. We observed many more leukocytes in the Casp1-/- mice than in the Casp1+/+ mice (Fig 1C). We further used flow cytometry to characterize the immune cell composition of these leukocytes. While macrophages, especially SiglecF+ tissue-resident macrophages, are the predominant cell type in the physiological state, neutrophils become more prominent at 12 hours after E. coli challenge (Fig 1D and 1E). Moreover, the number of neutrophils in the Casp1-/- mice was significantly greater than that in the Casp1+/+ mice. Although we observed more tissue-resident macrophages in the Casp1-/- mice in the physiological state, these macrophages decreased dramatically at 12 hours post-E. coli infection. The number of recruited monocytes was comparable between the two groups after E. coli challenge (S1G and S1H Fig).

We also investigated whether the bactericidal function of neutrophils in Casp1-/- mice differed from that in Casp1+/+ mice. We used flow cytometry to measure the production of reactive oxygen species (ROS), an essential procedure for intracellular bacterial killing, in neutrophils upon E. coli challenge. We found that the ROS levels in Casp1-/- neutrophils were comparable to those in Casp1+/+ neutrophils after E. coli challenge (S1I Fig). Moreover, RNA sequencing (RNA-seq) analysis revealed that antibacterial defense signals were expressed at similar levels in Casp1-/- and Casp1+/+ neutrophils (S1J Fig).

To further determine the role of neutrophils and macrophages in protecting against E. coli-induced pneumonia in Casp1-/- mice, we then used clodronate liposomes and an anti-Ly6G antibody to eliminate macrophages and neutrophils, respectively. Strikingly, the absence of neutrophils severely impaired host defense against E. coli infection in the Casp1-/- mice, whereas the deletion of macrophages had no effect (Fig 1F and 1G). These results indicate that neutrophils play a pivotal role in the pathogenesis of E. coli pneumonia in Casp1-/- mice.

Decreased lung pyroptosis and infiltrating neutrophil necroptosis in Casp1-/- mice during pulmonary E. coli infection

We then investigated whether the difference in neutrophil number between Casp1-/- and Casp1+/+ mice resulted from differential neutrophil chemotaxis in the experimental mice after E. coli challenge. We determined the neutrophil number in BALF at an earlier infection stage (4 hours after infection) and found no significant increase in the total cell count or neutrophil count in the Casp1-/- mice (S2A and S2B Fig). Additionally, at 12 hours after E. coli challenge, the numbers of peripheral white blood cells and neutrophils in the Casp1-/- and Casp1+/+ mice were comparable (S2C and S2D Fig). We also determined the levels of the chemokines C-X-C motif ligand 1 (CXCL1) and CXCL2, both of which are important for neutrophil attraction during bacterial infection [25,26]. Unexpectedly, we found increased levels of both CXCL1 and CXCL2 in the BALF of the Casp1+/+ mice (S2E and S2F Fig). These results indicate that the increased number of neutrophils in the lungs of Casp1-/- mice during E. coli infection is not dependent on chemotaxis.

We next investigated cell death, which may account for the difference in the number of neutrophils in the lungs of Casp1-/- and Casp1+/+ mice after E. coli infection. We used propidium iodide (PI), a membrane-permeabilized fluorescent dye, to stain the nuclei of neutrophils undergoing lytic cell death. We found that, compared with the Casp1+/+ mice, the Casp1-/- mice had a lower proportion of PI-stained neutrophils following instillation of E. coli (Fig 2A), suggesting that pulmonary neutrophils were preserved in a much more stable and living state in the Casp1-/- mice after E. coli challenge. A pathogenic bacterial strain is crucial in the study of human disease, as it closely mimics the pathogens responsible for human infection. Thus, we included a clinically isolated E. coli strain in vivo. We observed lower neutrophil death in the presence of this pathogenic E. coli (Fig 2B). We also measured lactate dehydrogenase (LDH) and high mobility group protein 1 (HMGB-1) levels in the BALF, both of which are released during lytic cell death, and observed significantly higher levels of LDH and HMGB-1 in the BALF of the Casp1+/+ mice after E. coli infection (Figs 2C and S2G). However, these cell death-related substances can be released not only from dying neutrophils but also from other cells, such as lung parenchymal cells. Using terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining, we detected many dead cells in the lung tissue of the Casp1+/+ mice, which may consist of both parenchyma cells and immune cells (S3A Fig). We performed TUNEL staining for various cell markers in lung sections from the Casp1+/+ and Casp1-/- mice (S3B Fig). Our findings indicated that both Casp1+/+ and Casp1-/- mice presented comparable TUNEL signals in macrophages (F4/80+), with minimal signals observed in monocytes (CD14+) and type II epithelial cells (SPC+). Notably, the Casp1+/+ mice presented significantly greater TUNEL signals in epithelial cells (CD326+), endothelial cells (CD31+), and neutrophils (Ly6G+), suggesting that type I epithelial cells, endothelial cells, and neutrophils are the primary cell types affected in Casp1+/+ mice compared to Casp1-/- mice. These findings indicated that during pulmonary E. coli infection, both recruited neutrophils and lung parenchymal cells undergo certain types of programmed cell death. To verify this hypothesis, we then performed immunoblot analysis on cells collected from the BALF (containing >95% neutrophils) and remnant lung tissue after bronchoalveolar lavage to determine the related cell death modes using antibodies against the MLKL protein and GSDMD, which are hallmarks of necroptosis and pyroptosis [27,28]. We found that the BALF cells from Casp1+/+ mice had substantially higher levels of phosphorylated MLKL (p-MLKL) than did the BALF cells from Casp1-/- mice, with a negative control from Mlkl-/- BALF cells (Fig 2D). In contrast, lung tissue from Casp1+/+ mice presented elevated levels of cleaved GSDMD and activated caspase-1 (Fig 2D). Additionally, flow cytometry and immunofluorescence analysis confirmed that neutrophils from Casp1+/+ mice presented higher levels of p-MLKL than did neutrophils from Casp1-/- mice (Figs 2E and S4A). We also assessed the location of p-MLKL in BALF-collected cells and found that it colocalized with PKH-26 (a probe indicating cell membranes; S4B Fig) on neutrophils (Ly6G+), which corresponds to its membrane pore-forming function and induction of neutrophil necroptosis. These findings further support the involvement of caspase-1 in neutrophil necroptosis during pulmonary E. coli infection.

Fig 2. Decreased lung pyroptosis and infiltrating neutrophil necroptosis in Casp1-/- mice during pulmonary E. coli infection.

Fig 2

(A and B) Death of neutrophils in the BALF of Casp1-/- mice and Casp1+/+ mice at 12 hours after infection with E. coli ATCC25922 (n = 6) or a clinically isolated E. coli strain (n = 8-10). Cell viability was measured with PI staining and analyzed using flow cytometry. (C) HMGB1 levels in the BALF of Casp1-/- mice and Casp1+/+ mice at 12 hours after E. coli infection. Each lane represents one mouse (n = 3). (D) BALF cells (containing > 95% neutrophils) and lung tissue without BALF cells from the indicated mice were collected and immunoblotted for programmed cell death-related proteins. (E) BALF neutrophils from the indicated mice were harvested at 12 hours after E. coli lung infection and stained with an anti-p-MLKL antibody, and the mean fluorescence was measured by flow cytometry. The data are shown as the means ± SDs in (A, B, C, D and E). Statistical differences were determined by Student’s t test (A, B and C) and one-way ANOVA (D and E). *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. See also S2, S3 and S4 Figs.

Intervention with MLKL decreased neutrophil death and protected mice against E. coli pneumonia

The aforementioned results suggest that via necroptotic death, neutrophils may participate in the pathogenesis of E. coli-induced pneumonia. Next, we intervened the core execution protein of necroptosis, MLKL, to verify these findings. We treated mice with an intraperitoneal injection of GW806742X [29] (an ATP mimetic that inhibits necroptosis by delaying MLKL membrane translocation) 1 hour before E. coli challenge. GW806742X administration significantly prolonged the survival period and increased the survival rate of wild-type (WT) mice (Fig 3A). However, we found no benefit for Casp1-/- mice after infection with an equal dose of E. coli, whereas a mild benefit was observed when they were infected with twice the dose of E. coli (Fig 3B). These findings suggest a potential alternative pathway independent of caspase-1 in regulating neutrophil necroptosis. Additionally, the administration of GW806742X markedly alleviated lung tissue injury, decreased the total protein concentration and bacterial load (S5AS5C Fig), increased the total leukocyte number and neutrophil count and decreased BALF neutrophil death in WT mice (Fig 3C3E), which mostly reproduced the phenomenon observed in infected Casp1-/- mice.

Fig 3. Intervention with MLKL decreased neutrophil death and protected mice against E. coli pneumonia.

Fig 3

(A) Survival of WT mice treated with the MLKL inhibitor GW806742X or control solvent after pulmonary E. coli infection (n = 9 each, conducted across three independent replicates, with each replicate including 3 mice). (B) Survival of Casp1-/- mice treated with the MLKL inhibitor GW806742X (n = 7) or control solvent (n = 8) after pulmonary E. coli infection with a single dose of E. coli (3 × 106 CFUs), and survival of Casp1-/- mice treated with the MLKL inhibitor GW806742X (n = 8) or control solvent (n = 7) after pulmonary infection with a double dose of E. coli (6 × 106 CFUs). This experiment was conducted across three independent replicates, with each replicate including 2–3 mice. (C-E) BALF from WT mice treated with the MLKL inhibitor GW806742X or control solvent was harvested at 12 hours after intratracheal PBS instillation (n = 5 each) or pulmonary E. coli infection (n = 6 each), and the total leukocyte count (C), neutrophil number (D) and PI+ neutrophil proportion (E) were determined. (F) Survival of Mlkl-/- mice (n = 16) and Mlkl+/+ mice (n = 12) after pulmonary E. coli infection. This experiment was conducted across three independent replicates, with each replicate including 4–6 mice. (G-H) BALF from Mlkl-/- mice and Mlkl+/+ mice was harvested at 12 hours after pulmonary E. coli infection, and the neutrophil number (H) and PI+ neutrophil proportion (I) were determined (n = 6 each). The data are shown as the means ± SDs in (C, D, E, G and H). Statistical differences were determined by the Mantel‒Cox test (A, B and F), two-way ANOVA (C, D and E) and Student’s t test (G and H). *P < 0.05; **P < 0.01; ***P < 0.001. ns, not significant. See also S5 Fig.

To further elucidate the role of MLKL in murine E. coli pneumonia, we utilized Mlkl-/- mice to confirm the role of neutrophil necroptosis in E. coli-induced pneumonia. Similarly, deletion of MLKL rescued the mice from lethal pneumonia, reduced overall lung injury, decreased the lung bacterial burden, increased neutrophil counts and decreased neutrophil death (Figs 3F3H, S5D and S5E). These results indicated that inhibition of necroptosis can protect the mice against E. coli-induced lethal pneumonia, suggesting that neutrophil necroptosis underlies disease pathogenesis.

Nonhematopoietic pyroptosis contributes to neutrophil necroptosis

Recent studies have shown that certain pathogens can trigger the formation of the PANoptosome, a multiprotein complex. This complex is composed of caspase-1, caspase-8, RIPK1, RIPK3, Fas-associated protein with death domain, apoptosis-related spot-like protein, AIM2, pyrin, and ZBP1. The PANoptosome induces PANoptosis, a process that includes apoptosis, pyroptosis, and necroptosis [21]. We next speculated that the observed neutrophil necroptosis is a cell-intrinsic mechanism associated with PANoptosis upon the sensing of certain external stimuli. To test this hypothesis, we used live E. coli, lipopolysaccharide (LPS) and heat-inactivated E. coli to stimulate neutrophils and mimic possible infection conditions in vivo. However, we did not find any significant difference in cell death between Casp1+/+ and Casp1-/- neutrophils after stimulation (S6A Fig). Immunoblot analysis also revealed no difference in p-MLKL levels between Casp1+/+ and Casp1-/- neutrophils (S6B Fig). These findings suggested that during E. coli infection, pulmonary neutrophil necroptosis is not caused by an intracellular mechanism associated with caspase-1-dependent PANoptosis.

Necroptosis can also potentially be triggered by inflammatory mediators such as TNF-α, IL-1β or HMGB-1. Among these mediators, IL-1β is the cytokine most relevant to caspase-1 because caspase-1 cleaves pro-IL-1β into its mature form. The activation of IL-1R, the receptor of IL-1β, reportedly limits necroptosis in certain kidney diseases [30]. TNF-α is associated with necroptosis since TNF-α triggers necroptosis when proapoptotic caspase-8 is inhibited [31]. HMGB-1 has also been reported to affect the triggering of necroptosis by pathogen toxins [32]. We then investigated whether the elevated levels of these mediators were responsible for neutrophil necroptosis during pulmonary E. coli infection. We used these mediators as stimuli for neutrophil necroptosis and found that none of the mediators could induce significant cell death regardless of their concentration (S6C Fig). We also administered the IL-1 receptor antagonist anakinra to competitively inhibit the function of IL-1β in vivo or, alternatively, used an anti-IL-1β antibody to neutralize IL-1β during E. coli infection. These results confirmed that blocking IL-1β did not mitigate the severity of lethal pneumonia in mice (S6D–S6F Fig). Thus, these results exclude the possible role of inflammatory mediators in neutrophil necroptosis during pulmonary E. coli infection.

Since western blot analysis of lung tissue revealed that caspase-1 and GSDMD were activated in Casp1+/+ mouse lungs (Fig 2D), we further postulated that neutrophil necroptosis may be a consequence of lung parenchyma cell pyroptosis. We then incubated a single-cell suspension from the lung tissue of E. coli-infected WT mice with FLICA, a probe used to assess caspase-1 activity, to determine which type of cell is associated with highly active caspase-1. We found that among the cells in the lung, the cell type with the highest proportion of active caspase-1 (marked by FLICA+) was epithelial cells and endothelial cells (Figs 4A and S7A). These results indicated that these nonhematopoietic cells are inflammasome-activated cells during lung E. coli infection.

Fig 4. Nonhematopoietic pyroptosis contributes to neutrophil necroptosis.

Fig 4

(A) The proportions of neutrophils, macrophages, endothelial cells, epithelial cells and other cells with activated caspase-1 in the lungs of WT and Mlkl-/- mice at 12 hours after pulmonary E. coli infection were assessed by FLICA staining using flow cytometry (n = 4). (B) Neutrophil death after stimulation with MLE12 lysate supernatant (MLE12 lysate sup), E. coli culture supernatant (bac. sup), a mixture of these supernatants, and pyroptotic supernatant (pyrop. sup) or control medium (n = 3). (C) Immunoblot of neutrophils stimulated with MLE12 cell lysate sup or bac. sup and a mixture of these supernatants and pyrop. sup as well as control medium. (D) BALF cells and lung tissue without BALF cells from Casp1+/+ and Casp1-/- mice were collected 12 hours after E. coli infection and immunoblotted for RIPK3 and p-RIPK3. Lysate of L929 cell line treated with TNF-⍺ and Z-VAD was used as a positive control for p-RIPK3. (E) Immunoblotting for caspase-1 and GSDMD in MLE-12 cells treated with caspase-1 or GSDMD siRNA. (F) Supernatants from PBS- or E. coli-stimulated MLE-12 cells pretreated with caspase-1 or GSDMD siRNA were collected and used to stimulate neutrophils, and cell death was evaluated by PI staining and flow cytometry (n = 3). (G) Neutrophils were pretreated with GSK872 and GW806742X at the indicated concentrations and then stimulated with pyrop. sup and control media, and cell death was assayed by using flow cytometry (n = 3). (H and I) Chimeric mice were generated via reciprocal bone marrow transplantation of WT, Casp1-/- and Gsdmd-/- mice as indicated, after which the mice were subjected to E. coli-induced pneumonia. Mouse survival was monitored (H). BALF cells were collected at 12 hours, and neutrophil death was determined by PI staining and flow cytometry (I). The survival experiment was conducted across three independent replicates, with each replicate including 3–6 mice. The data are shown as the means ± SDs in (A, B, F, G and I) and are representative of 3 independent experiments in (C, D, and E). Statistical differences were determined by two-way ANOVA (A, F and G), one-way ANOVA (B and I) or the Mantel‒Cox test (H). *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. ns, not significant. See also S6 and S7 Figs.

Inflammasomes in barrier epithelial cells can sense certain pathogens and then trigger pyroptosis. In this study, we chose MLE-12 murine lung epithelial cells as an in vitro model. MLE-12 cells were first incubated with live E. coli, and pyroptosis was assessed by detecting the activation of caspase-1 and GSDMD in the culture supernatant (S7B Fig). We defined the supernatant from live E. coli-stimulated MLE-12 cells as ‘pyroptotic supernatant’ and used it as a subsequent stimulus for neutrophils. In parallel, we also tested supernatant from freeze-thawed MLE-12 cell lysates, supernatant from cultured E. coli, and a mixture of these supernatants to stimulate neutrophils. A substantial amount of neutrophil death was induced by the pyroptotic supernatant from live E. coli-stimulated MLE-12 cells (Fig 4B), whereas neither the supernatant from the cell lysate nor that from cultured E. coli alone induced neutrophil death. Interestingly, although these are distinct stimuli, combining the two supernatants resulted in significant neutrophil death (Fig 4B). Notably, this pyroptotic supernatant also induced comparable cell death in Casp1-/- neutrophils (S7C Fig). Immunoblot analysis confirmed elevated p-MLKL levels and phosphorylated levels of RIPK3 (p-RIPK3) during the stimulation of neutrophils with pyroptotic supernatant (Fig 4C). Concurrently, increased p-RIPK3 levels were also detected in pulmonary BALF cells after E. coli infection (Fig 4D, including L929 cells treated with TNF-⍺ and Z-VAD as a positive control of p-RIPK3).

We next utilized caspase-1 and GSDMD siRNAs to suppress their activity in the epithelial cell line MLE-12 during E. coli challenge in vitro and then collected the culture supernatant to stimulate neutrophils. As expected, the supernatants from the caspase-1- and GSDMD siRNA-treated MLE-12 cells were unable to induce neutrophil death (Fig 4E and 4F). These results suggest that during E. coli infection, pulmonary neutrophil necroptosis may not arise from an intracellular mechanism associated with caspase-1 but may result from certain stimuli released from pathogen-induced caspase-1-mediated pyroptotic epithelial cells.

We further explored the possible mechanism involved in this model. Inhibitors, including NEC-1, GSK-872 and GW806742X, were used to inhibit the activity of the critical molecules RIPK1, RIPK3 and MLKL, respectively, in the necroptotic pathway upon stimulation with pyroptotic supernatant. Additionally, the pan-caspase inhibitor Z-VAD-FMK was used. Strikingly, cell death was markedly suppressed by GSK872 and GW806742X in both WT and Casp1-/- neutrophils but not in Mlkl-/- neutrophils (Figs 4G, S7C and S7D). However, the inhibition of NEC-1 or pan-caspases did not affect neutrophil necroptosis (S7E Fig). To validate these findings, we employed a chimeric mouse model comprising WT hematopoietic cells and Casp1-/- or Gsdmd-/- nonhematopoietic hosts in the context of E. coli pneumonia. We found that, compared with the mice with WT nonhematopoietic cells, the mice with Casp1-/- or Gsdmd-/- nonhematopoietic cells exhibited a greater survival rate and significantly lower neutrophil death rate (Fig 4H and 4I). Taken together, these data indicate that during E. coli infection, pulmonary neutrophil death is induced by nonhematopoietic pyroptotic cells such as epithelial cells.

Pyroptosis-released dsRNAs induce neutrophil necroptosis via ZBP1 during pulmonary E. coli infection

During necroptosis, RIPK3 is phosphorylated and activated by proteins containing the RIP homotypic interaction motif (RHIM). These RHIM-containing proteins include TRIF and ZBP1. Additionally, RIPK1, another RHIM-containing protein, contributes to RIPK3 activation [33,34]. We then investigated whether TRIF and ZBP1 mediate neutrophil necroptosis upon E. coli infection. RNA-seq analysis revealed that ZBP1 transcripts were abundant in BALF neutrophils from both Casp1+/+ and Casp1-/- mice following pulmonary E. coli infection, whereas TRIF transcripts were not detectable (Fig 5A). These results were further confirmed by immunoprecipitation (IP) analysis, which not only validated ZBP1 expression but also demonstrated that ZBP1 physically interacts with RIPK3 in BALF neutrophils. This interaction suggests that ZBP1 plays a direct role in mediating neutrophil necroptosis during pulmonary E. coli infection (Fig 5B). ZBP1 is a known nucleic acid sensor that can recognize both microbial and host-derived nucleic acids, including dsRNA [35]. To determine whether dsRNA is involved in neutrophil necroptosis, we performed IP analysis and found that ZBP1 binds dsRNA in BALF neutrophils upon E. coli infection (Fig 5C). We next investigated whether pyroptotic death of epithelial cells during E. coli-induced pneumonia contributes to the release of dsRNA. We found that supernatants from E. coli-stimulated MLE-12 cells contained abundant dsRNA (Fig 5D). Similarly, we found lower levels of dsRNA in the BALF of the E. coli-infected mice generated from WT hematopoietic cells from Casp1-/- or Gsdmd-/- hosts (Fig 5E). These results demonstrate that nonhematopoietic pyroptosis plays a critical role in the release of dsRNA.

Fig 5. Pyroptosis-released dsRNAs induce neutrophil necroptosis via ZBP1 during pulmonary E. coli infection.

Fig 5

(A) RNA sequencing of the gene expression of neutrophils in the BALF of Casp1-/- mice and Casp1+/+ mice at 12 hours after pulmonary E. coli infection. FPKM, fragments per kilobase per million mapped fragments. n = 2 for each group. (B) IP assay using anti-ZBP1 antibody or IgG control antibody in BALF cells from WT mice at 12 hours after pulmonary E. coli infection. (C) IP assay using anti-dsRNA antibody or IgG control antibody in BALF cells from WT mice at 12 hours after pulmonary E. coli infection. (D) Dot blot of MLE12 lysate sup, bac. Sup and pyrop. sup, as well as the control medium, via the dsRNA antibody. (E) Dot blot of BALF from the indicated chimeric mice at 12 hours after pulmonary E. coli infection using an anti-dsRNA antibody. (F) Neutrophil interactions of FITC-conjugated poly(I:C) and neutrophil death (PI+) were measured by flow cytometry after stimulation with E. coli culture supernatant (bac. sup) or control medium with/without FITC-conjugated poly(I:C) (n = 3). (G) Representative immunofluorescence image of neutrophils cocultured with control/bac. sup and FITC-conjugated poly(I:C) using PKH26 and anti-ZBP1 antibodies and quantification of the percentage of AF647/FITC colocalized cells (n = 3 biologically independent samples). Scale bars, 10 μm. (H) Neutrophils were pretreated with an anti-dsRNA antibody or a control IgG antibody and stimulated with pyrop. sup or control medium, and cell death was assayed by using flow cytometry (n = 3). (I) Survival of mice pretreated intratracheally with anti-dsRNA antibody (n = 7) or IgG control antibody (n = 6) 1 hour before pulmonary E. coli infection. This experiment was conducted across three independent replicates, with each replicate including 2–3 mice. (J and K) Neutrophil numbers (J) and proportions of PI+ neutrophils (K) in the BALF of WT mice treated with anti-dsRNA antibody (n = 9) or IgG control antibody (n = 10) at 12 hours after pulmonary E. coli infection. The data are shown as the means ± SDs in (F, G, H, J and K) and are representative of 3 independent experiments in (B, C, D, E and G). Statistical differences were determined by the Mantel‒Cox test (I), two-way ANOVA (F and H) and Student’s t test (G, J and K). *P < 0.05; **P < 0.01; ****P < 0.0001. ns, not significant. See also S8 Fig.

To determine whether extracellular dsRNA can enter neutrophils, we cultured neutrophils with fluorescein isothiocyanate-labeled poly(I:C), a synthetic analog of dsRNA, in the presence of E. coli supernatant. Flow cytometric analysis revealed that poly(I:C) effectively interacted with neutrophils under these conditions and induced cell death (Fig 5F). Furthermore, immunocytochemical analysis revealed the colocalization of poly(I:C) with ZBP1 (Fig 5G), suggesting that dsRNA interacts with ZBP1 in neutrophils. To further assess the role of dsRNA in neutrophil death, we preincubated the pyroptotic supernatant with a dsRNA antibody before stimulating neutrophils. This treatment significantly reduced the ability of the pyroptotic supernatant to induce neutrophil death (Fig 5H). Moreover, administration of the dsRNA antibody significantly increased WT mice survival after E.coli infection (Fig 5I). Compared with the control antibody-treated mice, the mice treated with the dsRNA antibody also presented higher total cell counts and neutrophil numbers in the BALF (Figs 5J and S8A), a lower percentage of neutrophil death (Fig 5K), reduced protein leakage, increased bacterial clearance and decreased lung injury (S8B-S8D Fig). To study the redundancies between these pathways, we also treated Casp1-/-, Mlkl-/- or Zbp1-/- mice with an anti-dsRNA antibody before E. coli pneumonia and found that dsRNA neutralization did not reduce neutrophil death in these mice after infection (S8E-S8G Fig). These results indicate that dsRNA neutralization ameliorates lung injury and promotes bacterial clearance by reducing neutrophil death and inflammation, but its protective effects depend on intact caspase-1-, MLKL-, and ZBP1-mediated pathways.

To confirm the role of ZBP1 in mediating neutrophil necroptosis in vivo, we conducted pulmonary E. coli infection in Zbp1-/- mice. Twelve hours post-infection, Zbp1-/- mice exhibited significantly reduced neutrophil death in the BALF (Fig 6A), along with increased BALF neutrophil counts and mild increases in total cell numbers (Fig 6B and 6C). Importantly, compared with their Zbp1+/+ littermates, Zbp1-/- mice presented a significantly lower pulmonary bacterial burden and decreased lung injury (Figs 6D and S8H), similar to the effects observed in mice treated with the dsRNA antibody during pulmonary E. coli infection. We also used supernatants from E. coli-stimulated epithelial cells to validate the roles of ZBP1 and MLKL in vitro. As expected, significantly fewer Zbp1-/- and Mlkl1-/- neutrophils died after challenge with pyroptotic supernatants derived from epithelial cells (Fig 6E). TLR3 is a critical receptor for recognizing dsRNA, which plays a central role in initiating immune responses to viral infections. As shown by the RNA-seq data (Fig 5A) and western blot analysis (S9A Fig), the neutrophils from these mice did not express TLR3. We further used TLR3-deficient mice to conduct in vivo experiments. We found that TLR3 deficiency did not prevent neutrophil death or decrease the bacterial burden (S9B-S9E Fig). These results suggest that TLR3 may not be involved in this neutrophil necroptosis pathway. LPS is actively secreted during bacterial growth, contributing to the pathogenicity of E. coli infections. To investigate the role of LPS in this model, we treated purified neutrophils with LPS (1 μg/ml) and poly(I:C) (1 μg/ml). We found that this combination could trigger neutrophil death (Fig 6F). However, treatment with poly(I:C) in combination with bacterial supernatant resulted in significantly greater neutrophil death (Fig 5F). Thus, we believe that in addition to LPS, other components facilitate dsRNA-related cell death under such circumstances. Taken together, these data demonstrate that dsRNA can be released from E. coli-stimulated pyroptotic nonhematopoietic cells, such as epithelial cells, and interact with neutrophil ZBP1 to facilitate cell death. Furthermore, ZBP1 knockout or in vivo neutralization of dsRNA alleviated E. coli pneumonia in mice.

Fig 6. ZBP1 deficiency protected against pulmonary E. coli infection.

Fig 6

(A-D) Twelve hours after E. coli infection, the neutrophil number (A), proportion of PI+ neutrophils (B), total leukocyte count (C) and bacterial burden (D) in the BALF of Zbp1+/+ mice (n = 11) and Zbp1-/- mice (n = 10) were determined. (E) WT, Mlkl-/-, and Zbp1-/- neutrophils (n = 3) were treated with E. coli-stimulated epithelial or endothelial cell supernatant, and cell death was evaluated via flow cytometry. (F) WT neutrophils were treated with control medium, LPS (1 μg/ml) or LPS+poly(I:C) (1 μg/ml) for 4 hours, and neutrophil death was determined by PI staining and flow cytometry. The data are shown as the means ± SDs. Statistical differences were determined by Student’s t test (A, B, C and D), two-way ANOVA (E) and one-way ANOVA (F). *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. ns, not significant. CFU, colony-forming unit. See also S9 Fig.

Discussion

In the present study, we found that the activation of caspase-1 exacerbated the pathophysiology of pulmonary E. coli infection by driving infiltrating neutrophil necroptosis. Neutrophil necroptosis is not directly triggered in a cell-intrinsic manner by bacteria but is indirectly triggered by pyroptotic lung epithelium during E. coli infection. Moreover, pyroptotic epithelial cell-released dsRNA induced neutrophil necroptosis in a ZBP1-dependent manner. These findings revealed for the first time that two types of programmed cell death, controlled by distinct death-executing molecules, can communicate intercellularly during pathogenic infection.

Previous studies have reported that pyroptosis and necroptosis participate in the pathogenesis of bacterial pneumonia. Moreover, some studies have identified crosstalk between inflammasome activation and necroptosis that is mediated in a cell-intrinsic manner by a mechanism in which membrane-associated MLKL causes potassium efflux and induces NLRP3 activation [36]. However, the connection between these two types of programmed cell death in different types of cells remains to be determined. Our results showed that nonhematopoietic cell inflammasome-activated pyroptosis triggered necroptosis in infiltrating neutrophils. This result demonstrated the intercellular communication between pyroptosis and necroptosis, which contributes to the understanding of programmed cell death regulation and provides additional insights for future studies.

Cell death may be associated with many other immune events during pathogenic insult. LPS from gram-negative bacteria can induce cell death. Our results showed that LPS induced neutrophil p-MLKL but only very mild cell death. LPS combined with other substances in bacteria and host cells may collaborate to further promote neutrophil death. The release of damage-associated molecular patterns, including IL-1β, IL-18, HMGB1 and adenosine triphosphate, from cell rupture potentially amplifies inflammatory responses and mediates cell death [32,37,38]. Furthermore, programmed cell death-released substrates may support the growth of invasive microbes [39]. These paracrine effects of dead cells have important impacts on the development of various diseases. Moreover, we found that sterile epithelial cell content promoted the necroptotic membrane permeabilization in neutrophils with costimulation of bacterial secretion. These findings demonstrated that the epithelial cell content resulting from pyroptosis could induce necroptosis in neutrophils in a paracrine manner. Moreover, our results showed that the release of dsRNA by lung epithelial cells is one of the possible extracellular triggers of neutrophil necroptosis. We found that extracellular dsRNA and ZBP1-regulated neutrophil necroptosis could be potential targets for the treatment of these cell death-related diseases, including infectious diseases, cancer [40], neurological diseases [41], ischemic diseases and cardiovascular diseases [42].

This study has several limitations. First, a direct way to prove this cellular crosstalk in vivo is to utilize cell-specific gene manipulation in mice, including conditional knockout with the Cre-loxP system. We only used chimeric reconstructed mice in our in vivo bacterial pneumonia disease model. Second, bacterial load and inflammation are critical factors that can influence mouse survival during disease. Although caspase-1 modulates bacterial clearance and inflammation status, it remains unclear how bacterial load, inflammation, and neutrophil necroptosis individually contribute to mouse survival. Third, the initiating inflammasome upstream of caspase-1 in this model has not been investigated. Inflammasomes are crucial activators of caspase-1 and pyroptosis. Understanding this mechanism could provide valuable insights for manipulating the pathophysiology of bacterial pneumonia. Fourth, the mechanisms of dsRNA-triggered neutrophil ZBP1-regulated necroptosis are not fully detailed in this study due to technical limitations. This may involve one or a combination of the following processes: first, bacterial secretions form membrane pores to promote the uptake of dsRNA by neutrophils and ZBP1 sensing of extracellular substances, including dsRNA [43]; second, bacterial secretion stimulation upregulates the expression of neutrophil dsRNA receptors, such as SID-1 transmembrane family member 1 and SID1 transmembrane family member 2 [44,45], leading to greater dsRNA uptake and ZBP1 sensing. Third, according to our results, dsRNA uptake and sensing may not be the sole mechanism underlying neutrophil necroptosis, as dsRNA neutralization does not fully inhibit neutrophil death in vitro. There could be other regulatory mechanisms, including those involving Toll-like receptors or ATP sensing, that may also participate. Future studies are warranted to fully elucidate the crosstalk between programmed cell death pathways.

In summary, we showed that caspase-1 inflammasome-licensed pyroptosis could drive necroptosis in a paracrine manner during pulmonary E. coli infection. This communication is mediated by pyroptotic epithelial cell-released intracellular dsRNA, which is recognized by neutrophils and serves as a trigger for ZBP1-dependent necroptosis. These findings not only demonstrate a paradigm of communication between necroptosis and pyroptosis in different cell types during microbe invasion but also provide directions for manipulating immune defenses against infectious diseases.

Materials/Subjects and methods

Ethics statement

The animal experimental protocols were approved by the Laboratory Animal Welfare and Ethics Committee of Zhejiang University School of Medicine and were in compliance with institutional guidelines. This study did not involve human participants, human data or human tissue.

Mice

C57BL/6 WT mice aged 6–8 weeks were purchased from Shanghai SLAC Laboratory Animal Corporation (Shanghai, China). Casp1-/- mice were kindly provided by Professor Di Wang of Zhejiang University School of Medicine. Mlkl-/- mice and Zbp1-/- mice were obtained from Cyagen Biosciences (Suzhou, China), Inc. Tlr3-/- mice were gifts from Professor Chun-Feng Wang of Jilin Agricultural University [46]. Experiments involving gene-edited mice were performed with littermate controls, whereas experiments involving antibody or drug treatments were conducted using purchased, cohoused control mice. All the mice were housed in a specific pathogen-free and temperature-controlled standard environment in accordance with the National Institutes of Health Guide for Care and Use of Laboratory Animals. The animal experimental protocols were approved by the Laboratory Animal Welfare and Ethics Committee of Zhejiang University School of Medicine and were in compliance with institutional guidelines.

Bacterial strains

E. coli bacteria (ATCC 25922) were obtained from the American Type Culture Collection (ATCC). The clinical strain of E. coli was isolated from a hospitalized patient with infectious arthritis of the knee at the Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China. The strain was only used in a murine infection model to assess pathophysiology.

Reagents and antibodies

For the inhibition experiments, the NEC-1 (HY-15760, 4311-88-0), GSK-872 (HY-101872, 1346546-69-7), z-VAD-FMK (HY-16658B, 161401-82-7), Ac-YVAD-cmk (HY-16990, 178603-78-6), GW806742X (HY-112292, 579515-63-2), and disulfiram (HY-B0240, 97-77-8) inhibitors were purchased from MedChemExpress. For immunoblot analysis, the following primary antibodies were used: caspase-1 (Abcam, ab179515), GSDMD (Abcam, ab209845), caspase-3 (Cell Signaling Technology, 14220), MLKL (Abcam, ab243142), MLKL (phospho S345; Abcam, ab196436), RIPK3 (Novus Biologicals, NBP1–77299), RIPK3 (phospho T231 + S232; Abcam, ab205421), HMGB-1 (Abcam, ab79823), dsRNA (SCICONS, 10010200) and β-actin (Sigma‒Aldrich, A5316). The following substances were used as indicated: LPS (Sigma‒Aldrich, L2630), TNF-α (BioLegend, 575202), Il-1β (PeproTech, 211-11b), and HMGB-1 (R&D Systems, 1690-HMB-050).

Pulmonary E. coli infection model

E. coli bacteria were seeded on Luria–Bertani agar plates and cultured overnight, and a single clone was selected for culture in liquid Luria–Bertani medium and incubated at 200 rpm at 37 °C for another 12 hours. The bacterial suspension was then prepared at a concentration of 3 × 106 colony formation units (CFUs) per 50 μl of PBS. For pulmonary bacterial infection, 50 μl of live E. coli suspension was intratracheally administered to the mice. Mortality was assessed hourly. In some experiments, the mice were intraperitoneally injected with GW806742X (100 μM, 100 μl in PBS) or anakinra (100 mg/kg, 100 μl in saline; Bioxell, BE0246) or intratracheally injected with anakinra (200 μg/kg, 20 μl in PBS), an anti-dsRNA antibody (500 μg/kg, 20 μl in PBS), or related control solvent or control IgG (Bioxell, BE0260; Absin, abs20038) 1 hour before the induction of pulmonary E. coli infection.

Cell count, protein level and bacterial load in BALF

Mouse lungs were washed with 0.5 ml of cold PBS three times, and the total white blood cell count was immediately calculated using a hemocytometer. The protein concentration was determined using a commercially available BCA protein assay kit (Thermo Fisher Scientific, 23225) according to the manufacturer’s instructions. The bacterial load in the BALF was determined by adding the diluted BALF to Luria–Bertani agar plates and incubating at 37 °C overnight. The bacterial load in CFU per unit was calculated by multiplying the countable bacterial colony units by the number of dilutions.

Collection of BALF cells and lung samples for western blot analysis

The mice were sacrificed 12 hours after E. coli infection. BALF was collected by performing three washes with 0.5 ml of PBS, followed by centrifugation at 800 × g for 5 minutes to isolate BALF cells. The lungs were subsequently harvested and ground in liquid nitrogen. For protein extraction, lysis buffer (10 mM Tris-HCl, pH 7.4, 1 mM EDTA, 150 mM NaCl, 0.5% NP-40, 1 mM Na3VO4, and 1 mM PMSF) was added to both the BALF cells and the lung tissue. The samples were then subjected to ultrasonic fragmentation at 4 °C and 50 W for 3 minutes. Next, the samples were centrifuged at 12,000 rpm for 10 minutes, and the supernatants were collected for protein concentration analysis using a BCA protein assay kit. The samples were then subjected to SDS–PAGE and transferred to a polyvinylidene fluoride membrane (Millipore). The western blots were probed with the indicated antibodies and visualized via an EZ-ECL kit (Biological Industries, 20-500-120). Images were taken via a Clinx ChemiScope3300.

Histology

Mouse lungs were harvested and fixed in 4% paraformaldehyde for at least 24 hours, embedded in paraffin wax and sectioned at a thickness of 5 μm. The sections were stained with hematoxylin and eosin (H&E) to analyze cellular and tissue morphological changes. Images were taken using an Olympus VS120 microscope (Shinjuku). Assessment of histological lung injury was performed by grading as previous described and the details are as follows: 1, normal; 2, focal (<50% lung section) interstitial congestion and inflammatory cell infiltration; 3, diffuse (>50% lung section) interstitial congestion and inflammatory cell infiltration; 4, focal (<50% lung section) consolidation and inflammatory cell infiltration; 5, diffuse (>50% lung section) consolidation and inflammatory cell infiltration [47,48]. For each section, at least five random high-power fields were selected for scoring. The mean score was used for comparison between groups. For TUNEL staining, mouse lungs were fixed in 4% paraformaldehyde for 12 hours at 4 °C, incubated in sucrose solution until the sample dropped to the bottom of the tube, embedded in optimal cutting temperature compound (Sakura, 4583) and sectioned at a 10-μm thickness. The sections were stained using a TUNEL kit (Beyotime, C1086) according to the manufacturer’s instructions. For immunofluorescence staining of p-MLKL, BALF cells were air-dried on L-lysine-coated slides and stained with PKH26 (Sigma‒Aldrich, MIDI26–1KT) according to the manufacturer’s instructions. Then, the cells were fixed with 4% formaldehyde at room temperature for 5 minutes, permeabilized with 1% Triton X-100 for another 5 minutes, blocked with 10% bovine serum albumin (Sigma‒Aldrich, A2153) at room temperature for 1 hour, and incubated with a primary antibody against p-MLKL at 4 °C overnight. The cells were then incubated with an AF647-conjugated secondary antibody (Thermo Fisher Scientific, A-31573) at room temperature for 1 hour. Nuclei were stained with 4’,6-diamidino-2-phenylindole (Beyotime Biotechnology, P0131). Images were taken with an Olympus FV3000 microscope (Olympus), and the data were analyzed with ImageJ software (NIH).

Flow cytometry for cell classification and cell death

For determination of the proportions of neutrophils and macrophages in the BALF, the cells were separated from the BALF by centrifugation at 800 × g for 5 minutes and resuspended in 100 μl of PBS. The cells were labeled with FITC-conjugated anti-mouse Ly6G/Ly6C (BioLegend, 108405), PE-CY7-conjugated anti-mouse F4/80 (eBioscience, 25-4801-82), APC-conjugated anti-mouse SiglecF (BioLegend, 155508), and BV421-conjugated anti-mouse Ly6C (BioLegend, 128031) for 25 minutes at 4 °C. The cells were subsequently washed and resuspended in flow cytometry buffer and stained with 2.5 μg/ml PI (Beyotime, ST511) for analysis.

For the caspase-1 activation assay in the lung parenchyma, the lungs of the mice were prepared as previously described. Briefly, mouse lungs were washed with 1 ml of HBSS with Liberase (100 μg/ml final concentration; Roche, 5401119001) and digested in 4 ml of HBSS digestion medium for 40 minutes at 37 °C with vortexing every 10 minutes. The resulting samples were filtered through a 70-μm cell strainer, washed with Dulbecco’s modified Eagle’s medium and treated with red blood cell lysis buffer (eBioscience, 00-4333-57) to lyse red blood cells. The cell suspensions were then stained with FITC-conjugated anti-mouse Ly6G/Ly6C, BV421-conjugated anti-mouse F4/80 (BioLegend, 123132), BV510-conjugated anti-mouse CD326 (BioLegend, 118231), APC-conjugated anti-mouse CD31 (BioLegend, 102509), FLICA 660-YVAD-FMK caspase-1 detection probes (ImmunoChemistry Technologies, 9122), and fixable viability dye 780 (BD Biosciences, 565388). Control isotype antibodies were used to determine the cutoff between negative and positive fluorescent populations. The data were collected with a BD LSRFortessa flow cytometer (BD Biosciences) and analyzed using FlowJo v.10 software (BD Biosciences).

Macrophage or neutrophil depletion in vivo

For depletion of macrophages in vivo, the mice were intratracheally injected with 100 µl of clodronate liposomes or control liposomes (FormuMax Scientific, F70101C-NC-10) 48 hours before induction of the pneumonia model. For depletion of neutrophils in vivo, mice were intraperitoneally injected with 200 μg of anti-ly6G antibody (BioXCell, BE0075–1) or isotype control (BioXCell, BE0089) 2 days before the experiment and were intratracheally injected with 100 μg of anti-ly6G antibody or isotype control on the day of the experiment. The depletion efficacy was confirmed by flow cytometry.

Bone marrow chimeric mice

WT, Casp1-/- and Gsdmd-/- mice were given gentamycin (0.08 g/200 ml) in the drinking water 1 week before radiation, which was stopped at 1 week after radiation. All the mice received 7 Gy of radiation for 20 minutes in an RS2000 Pro X-ray Biological Irradiator (Rad Source, US). The mice were retro-orbitally injected with bone marrow cells (5 × 106) within 24 hours after radiation. Six to eight weeks later, the chimeric mice were used for further experiments.

Cell culture

MLE12 cells (American Type Culture Collection, CRL-2110) were cultured in DMEM/F12 1:1 medium (Cytiva, SH30023.01) supplemented with 2% fetal bovine serum (FBS; Biological Industries, 04-001-1ACS) in an atmosphere with 5% CO2 at 37 °C. The cells were seeded at 4 × 105/ml for further experiments. For siRNA silencing, MLE-12 cells were seeded at 1 × 105 in a 24-well plate and cultured. On the second day, Casp1 siRNA, GSDMD siRNA or control siRNA (10 pmol of each) was preincubated with Lipofectamine RNAiMAX Reagent (3 µl) for 5 minutes in Opti-MEM at room temperature as instructed. The mixture was then added to the cells, and the cells were incubated at 37°C for 72 hours for further analysis.

Isolation of bone marrow neutrophils

For bone marrow neutrophils, the mice were sacrificed by cervical dislocation, and the femurs and tibias were harvested. The bone marrow cells were flushed from the femurs and tibias with DMEM supplemented with 10% FBS using a syringe with a 25-gauge needle. The cell suspension was collected and centrifuged at 800 × g for 5 minutes at room temperature. Discontinuous Percoll (65% and 78%; Absin, abs9102) gradients were prepared and added to a 15-ml centrifuge tube according to the manufacturer’s instructions. Pelleted cells were then resuspended in 3 ml of DMEM, gently added to the top of 65% Percoll solution and subjected to centrifugation at 800 × g for 30 minutes. The cell layer between 65% and 78% was bone marrow neutrophils, and the purity was > 95% according to flow cytometry verification. The neutrophils were immediately used for subsequent experiments without further culture.

Preparation of infectious cell supernatant, cell lysate and bacterial supernatant

For preparation of the infectious cell supernatant, live E. coli were added to MLE12 cells at a multiplicity of infection (MOI) of 2. The cell-free supernatant was collected 4 hours later and centrifuged at 3000 × g for 5 minutes to remove cell debris and bacteria. The supernatant was subsequently filtered through a 0.2 μm syringe filter. This infectious cell supernatant or complete medium control was frozen at -20 °C for later use. For preparation of cell lysates, MLE12 cells were digested and resuspended in complete medium in 2-ml cryogenic storage vials and subjected to freeze‒thaw cycles. For each freeze‒thaw cycle, the cells were frozen in liquid nitrogen and thawed in a 37 °C water bath. The supernatant was subsequently collected and adjusted to the appropriate culture volume using complete medium, after which it was centrifuged and filtered as described above. For preparation of the bacterial supernatant, E. coli was cultured at a concentration of 8 × 105 CFU/ml in complete medium as MLE12 cells. Four hours later, the supernatant was collected and processed as described above.

In vitro stimulation and cell death analysis of neutrophils

E. coli (MOI = 2), LPS (1 μg/ml), heated E. coli (MOI = 20) or the collected supernatants were added to the neutrophils 1 hour after inhibitor (GSK-872, GW806742X, NEC-1 or Z-VAD FMK) treatment. Four hours after stimulation, the cells were centrifuged, and the lysates were collected for western blot analysis. For some experiments, the nuclei were stained with 2.5 μg/ml PI, and the data were collected with a Beckman Coulter DxFLEX flow cytometer (Beckman Coulter Life Sciences) and analyzed with FlowJo v.10 software. For immunofluorescence analysis, purified neutrophils were treated with LPS (1 μg/ml) and poly(I:C) (1 μg/ml) for 4 hours, and the cells were collected, stained with PKH26 and distributed on slides. After drying, fixation, and permeabilization, the cells were stained with anti-ZBP1 and anti-AF647 secondary antibodies (MedChemExpress) for analysis. Images were taken with an Olympus FV3000 microscope (Olympus), and the data were analyzed with ImageJ software (NIH).

Cell death induction assays

Inhibitors (Ac-YVAD-cmk 40 μM, disulfiram 50 μM) and the control (dimethyl sulfoxide) were added to MLE12 cells 1 hour before bacterial infection. Live E. coli was added to MLE12 cells at an MOI of 2. At the indicated time points, the supernatant was collected and processed as described. PBS was added, and the nuclei were stained with 2.5 μg/ml PI. Images were taken with an Olympus IX73 inverted microscope. The red masks used for quantification of the PI stain are shown in the representative images.

Enzyme-linked immunosorbent assay (ELISA)

The levels of the cytokines Il-1β (MultiSciences, EK201B) and TNF-α (MultiSciences, EK282) and the chemokines CXCL-1 (MultiSciences, EK296) and CXCL-2 (MultiSciences, EK2142) were measured using commercially available kits according to the manufacturers’ instructions.

Transcriptomic RNA-seq

Total RNA was isolated from BALF neutrophils from Casp1-/- mice and Casp1+/+ mice using TRIzol reagent (Life Technologies, 155896018) following the manufacturer’s protocol. RNA-seq was performed by LC-Bio Technology Co., Ltd. (Hangzhou, China) according to the manufacturer’s recommendations. Briefly, total RNA quantity and purity were analyzed using a Bioanalyzer 2100 and an RNA 6000 Nano LabChip kit (Agilent), with RNA integrity values >7.0. Poly(A) RNA was purified from total RNA (5 µg) via two rounds of purification with poly-T oligo-attached magnetic beads. Following purification, the mRNA was fragmented into small pieces using divalent cations under elevated temperature. The cleaved RNA fragments were subsequently reverse-transcribed to create the final cDNA library in accordance with the protocol for TruSeq RNA Sample Preparation v.2 (Illumina, RS-122–2001 and RS-122–2002); the average insert size for the paired-end libraries was 300 bp (±50 bp). Paired-end sequencing was carried out on an Illumina NovaSeq 6000 following the manufacturer’s recommended protocol. Before assembly, low-quality reads were removed using Fastp software (https://github.com/OpenGene/fastp). HISAT2 (https://ccb.jhu.edu/software/hisat2) was subsequently used to map reads to the reference genome of Mus musculus GRCm39. After the final transcriptome was generated, StringTie (https://ccb.jhu.edu/software/stringtie) was used to estimate the expression levels of all the transcripts. The differentially expressed mRNAs with a fold change > 2 or < 0.5 and with a parametric F test comparing nested linear models (P value < 0.05) were selected via the R package edgeR (https://bioconductor.org/packages/release/bioc/html/edgeR.html).

Statistical analysis

The data are shown as the means ± SDs. Differences were analyzed by Student’s t test or ANOVA. The Mantel‒Cox test and log-rank comparison were performed for survival experiments. A P value < 0.05 was considered statistically significant. Statistical analysis was carried out using GraphPad Prism 9.3 (GraphPad Software).

Supporting information

S1 Fig. Lung neutrophils are critical for defense against E. coli pneumonia in Casp1-/- mice.

(A and B) Respiratory rates and body temperatures of Casp1-/- mice (n = 11) and Casp1+/+ mice (n = 8) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each). (C) BALF protein concentrations in Casp1-/- mice (n = 11) and Casp1+/+ mice (n = 8) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each). (D) BALF bacterial burden in Casp1-/- mice (n = 11) and Casp1+/+ mice (n = 8) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each) was determined and is expressed as CFU/ml. (E and F) IL-1β and TNF-α levels in the BALF of Casp1-/- mice (n = 11) and Casp1+/+ mice (n = 8) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each) were measured by ELISAs. (G and H) Resident macrophages (SiglecF+Ly6C-F4/80+) and recruited monocyte (SiglecF-Ly6C+F4/80+) in the BALF of Casp1-/- mice (n = 11) and Casp1+/+ mice (n = 8) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each) were counted by flow cytometry. (I) Total cellular ROS (determined by CellROX) and mitochondrial ROS (determined by MitoSOX staining) in neutrophils and macrophages from the BALF of Casp1-/- mice and Casp1+/+ mice at 12 hours after pulmonary E. coli infection were analyzed by flow cytometry (n = 7). (J) Pathway enrichment of expressed genes in neutrophils from the BALF of Casp1-/- mice and Casp1+/+ neutrophils at 12 hours after pulmonary E. coli infection was analyzed using RNA-seq transcriptomes (n = 2). The data are shown as the means ± SDs (A-I). Statistical differences were determined by two-way ANOVA (A‒H), Student’s t test (I) and the parametric F test comparing nested linear models (J). *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. ns, not significant. CFU, colony-forming unit.

(TIF)

S2 Fig. Chemotaxis of neutrophils in the lung during pulmonary E. coli infection.

BALF cells (A) and BALF neutrophils (B) from Casp1-/- mice (n = 6) and Casp1+/+ mice (n = 6) were counted at 4 hours after bacterial instillation. Peripheral white blood cells (C) and neutrophils (D) from Casp1-/- mice and Casp1+/+ mice were counted at 12 hours after E. coli challenge (n = 11 each) or PBS instillation (n = 5 each). BALF levels of CXCL1 (E) and CXCL2 (F) in Casp1-/- mice and Casp1+/+ mice were measured at 12 hours after E. coli challenge (n = 11 each) or PBS instillation (n = 5 each). (G) LDH levels in the BALF of Casp1-/- mice and Casp1+/+ mice at 12 hours after E. coli infection (n = 6) or intratracheal PBS instillation (n = 5). The data are shown as the means ± SDs in (A-G). Statistical differences were determined by Student’s t test (A and B) and one-way ANOVA (C, D, E, F and G). ***P < 0.001; ****P < 0.0001. ns, not significant.

(TIF)

ppat.1013167.s002.tif (426.1KB, tif)
S3 Fig. TUNEL staining of lung sections during pulmonary E. coli infection.

(A) TUNEL staining of lung sections from Casp1-/- mice and Casp1+/+ mice at 12 hours after E. coli infection and quantification of the TUNEL-positive area relative to the DAPI-stained area (n = 4). Scale bars, 100 μm. (B) Casp1+/+ and Casp1-/- mouse lungs were harvested at 12 hours after E. coli infection. TUNEL staining of lung sections was performed with different markers including the epithelial marker CD326, the endothelial marker CD31, the type II epithelial marker SPC, the macrophage marker F4/80, the neutrophil marker Ly-6G, and the monocyte marker CD14. Quantification of the percentage of TUNEL+ cells of different cell type (n = 3 biologically independent samples). Scale bars, 50 μm. The data are shown as the means ± SDs and are representative of 3 independent experiments. Statistical differences were determined by Student’s t test. *P < 0.05. HPF, high-power field.

(TIF)

ppat.1013167.s003.tif (1.9MB, tif)
S4 Fig. p-MLKL staining of lung sections and BALF cells during pulmonary E. coli infection.

(A) Casp1+/+ and Casp1-/- mouse lungs were harvested at 12 hours after E. coli infection. p-MLKL staining of lung sections with the neutrophil marker Ly6G was performed by immunofluorescence. Scale bars, 50 μm. (B) BALF cells from Casp1-/- mice and Casp1+/+ mice were fixed on slides, stained with PKH26 and p-MLKL antibodies and visualized by using confocal microscopy. Scale bars, 10 μm. The data are representative of 3 independent experiments.

(TIF)

ppat.1013167.s004.tif (1.3MB, tif)
S5 Fig. Intervention with MLKL decreased neutrophil death and protected mice against E. coli pneumonia.

(A) Representative images of lungs with H&E staining and quantification of lung injury scores in GW806742X (n = 5)- or control solvent (n = 6)-treated WT mice at 12 hours after pulmonary E. coli infection or intratracheal PBS instillation (n = 5 each). Scale bars, 100 µm. BALF protein concentration (B) and bacterial burden (C) in WT mice treated with GW806742X (n = 6) or control solvent (n = 6) at 12 hours after pulmonary E. coli infection or intratracheal PBS instillation (n = 5 each). (D) Representative images of lungs with HE staining and quantification of lung injury scores in Mlkl-/- (n = 9) or Mlkl+/+ (n = 6) mice at 12 hours after pulmonary E. coli infection. Scale bars, 100 µm. (E) Twelve hours after E. coli infection, mouse lungs were harvested from Mlkl-/- or Mlkl+/+ mice, and the bacterial burdens in whole-lung tissue were determined. The data are shown as the means ± SDs (A, B, C, D and E). Statistical differences were determined by Student’s t test (D and E) and two-way ANOVA (A, B and C). *P < 0.05; **P < 0.01; ****P < 0.0001. CFU, colony-forming unit.

(TIF)

ppat.1013167.s005.tif (1.6MB, tif)
S6 Fig. Neutrophil necroptosis is independent of bacterial stimuli and inflammatory stimuli.

(A) Proportion of PI+ neutrophils from Casp1-/- and Casp1+/+ mice after stimulation with LPS, live E. coli, heat-inactivated E. coli or PBS (n = 3). (B) Immunoblot analysis of cell death-related proteins in neutrophils stimulated with LPS, live E. coli or heat-inactivated E. coli. (C) Proportion of PI+ neutrophils from WT mice treated with IL-1β, TNF-α, HMGB-1 or control solvent at the indicated concentrations (n = 3). (D and E) Survival of WT mice intraperitoneally (n = 5 each) or intratracheally (n = 6 each) pretreated with the IL-1β antagonist anakinra or saline after pulmonary E. coli infection. (F) Survival of WT mice intraperitoneally (n = 6 each) pretreated with the anti-IL-1β antibody or control IgG antibody after pulmonary E. coli infection. The data are shown as the means ± SDs in (A and C) and are representative of 3 independent experiments in (B). Statistical differences were determined by the Mantel‒Cox test (D, E and F). ns, not significant.

(TIF)

ppat.1013167.s006.tif (493.4KB, tif)
S7 Fig. Pyroptotic epithelial cells trigger neutrophil necroptosis.

(A) Flow cytometry gating strategy for identifying caspase-1-activated cells in mouse lungs via FLICA staining. (B) MLE-12 epithelial cells were stimulated with E. coli, and then, the cell lysate and culture supernatant were collected and immunoblotted for pyroptosis-related proteins. (C and D) Casp1-/- neutrophils and Mlkl-/- neutrophils were pretreated with GSK872, GW806742X or control solvent and then stimulated with pyroptotic supernatant (pyrop. sup) or control medium, and cell death was assayed by flow cytometry (n = 3). (E) WT neutrophils were pretreated with NEC-1, Z-VAD or control solvent and then stimulated with pyrop. sup or control medium, and cell death was assayed by flow cytometry (n = 3). The data are shown as the means ± SDs in (C, D and E) and are representative of 3 independent experiments in (B). Statistical differences were determined by two-way ANOVA (C and D) or one-way ANOVA (E). ****P < 0.0001.

(TIF)

ppat.1013167.s007.tif (1.1MB, tif)
S8 Fig. Pyroptosis-released dsRNAs induce neutrophil necroptosis via ZBP1 during pulmonary E. coli infection.

The total BALF cell number (A), protein concentration (B) and bacterial burden (C) were measured in the mice pretreated with the anti-dsRNA antibody (n = 9) or the IgG control antibody (n = 10) at 12 hours after pulmonary E. coli infection. (D) Representative images of lungs with HE staining and quantification of lung injury scores in anti-dsRNA antibody (n = 6)- or control IgG (n = 6)-treated WT mice at 12 hours after pulmonary E. coli infection. Scale bars, 100 µm. (E‒G) Proportion of PI+ neutrophils in the BALF of Casp1-/- (L), Mlkl-/- (M) and Zbp1-/- (N) mice treated with an anti-dsRNA antibody (n = 6‒8) or an IgG control antibody (n = 7‒8) at 12 h after pulmonary E. coli infection. (H) Representative images of lungs with HE staining and quantification of lung injury scores in Zbp1-/- (n = 6) or Zbp1+/+ littermate (n = 6) mice at 12 hours after pulmonary E. coli infection. Scale bars, 100 µm. The data are shown as the means ± SDs. Statistical differences were determined by Student’s t test. *P < 0.05; **P < 0.01. ns, not significant. CFU, colony-forming unit.

(TIF)

ppat.1013167.s008.tif (1.4MB, tif)
S9 Fig. TLR3 may not be involved in the neutrophil necroptosis pathway during pulmonary E. coli infection.

(A) Western blot analysis of BALF neutrophils and spleen cells derived from E. coli-infected Casp1-/- and Casp1+/+ mice at 12 hours. (B-E) Twelve hours after E. coli infection, the proportions of PI+ neutrophils (B), neutrophil numbers (C), total leukocytes (D) and bacterial burdens (E) in the BALF of Tlr3+/+ mice (n = 8) and Tlr3-/- mice (n = 7) were determined. The data are shown as the means ± SDs (B-E) and are representative of 2 independent experiments in (A). Statistical differences were determined by Student’s t test (B-D). ns, not significant. CFU, colony-forming unit.

(TIF)

ppat.1013167.s009.tif (239.5KB, tif)
S1 Data. Quantitative data used in calculations corresponding to primary figures.

Numeric values used to generate graphs, means, and standard deviations for primary figures are included on tabs, with each tab indicating the relevant figure panel.

(XLSX)

ppat.1013167.s010.xlsx (49.9KB, xlsx)
S2 Data. Quantitative data used in calculations corresponding to supplemental figures.

Numeric values used to generate graphs, means, and standard deviations for supplemental figures are included on tabs, with each tab indicating the relevant figure panel.

(XLSX)

ppat.1013167.s011.xlsx (60KB, xlsx)

Acknowledgments

We thank Yanwei Li, Zhaoxiaonan Lin, Chun Guo and Jiajia Wang from the Core Facilities, Zhejiang University School of Medicine, for their technical support. We also thank Dr. Yang Yang from Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, for help for Tlr3-/- mice.

Data Availability

The authors confirm that all data underlying the findings are fully available without restriction. All relevant data are within the paper and its Supporting Information files.

Funding Statement

This study is supported by a program from the National Natural Science Foundation of China (82370013 to Q.C.). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Hansen V, Oren E, Dennis LK, Brown HE. Infectious disease mortality trends in the United States, 1980-2014. JAMA. 2016;316(20):2149–51. [DOI] [PubMed] [Google Scholar]
  • 2.Rudan I, Boschi-Pinto C, Biloglav Z, Mulholland K, Campbell H. Epidemiology and etiology of childhood pneumonia. Bull World Health Organ. 2008;86(5):408–16. doi: 10.2471/blt.07.048769 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ramirez J. Adults hospitalized with pneumonia in the United States: Incidence, epidemiology, and mortality. Clin Infect Dis. 2017;65(11):1806–12. [DOI] [PubMed] [Google Scholar]
  • 4.Heath PT. Epidemiology and bacteriology of bacterial pneumonias. Paediatr Respir Rev. 2000;1(1):4–7. doi: 10.1053/prrv.2000.0001 [DOI] [PubMed] [Google Scholar]
  • 5.Carugati M, Aliberti S, Reyes LF, Franco Sadud R, Irfan M, Prat C, et al. Microbiological testing of adults hospitalised with community-acquired pneumonia: an international study. ERJ Open Res. 2018;4(4):00096–2018. doi: 10.1183/23120541.00096-2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Peyrani P, Mandell L, Torres A, Tillotson GS. The burden of community-acquired bacterial pneumonia in the era of antibiotic resistance. Expert Rev Respir Med. 2019;13(2):139–52. doi: 10.1080/17476348.2019.1562339 [DOI] [PubMed] [Google Scholar]
  • 7.Laxminarayan R, Matsoso P, Pant S, Brower C, Røttingen J-A, Klugman K, et al. Access to effective antimicrobials: a worldwide challenge. Lancet. 2016;387(10014):168–75. doi: 10.1016/S0140-6736(15)00474-2 [DOI] [PubMed] [Google Scholar]
  • 8.McVey MJ, Steinberg BE, Goldenberg NM. Inflammasome activation in acute lung injury. Am J Physiol Lung Cell Mol Physiol. 2021;320(2):L165–78. doi: 10.1152/ajplung.00303.2020 [DOI] [PubMed] [Google Scholar]
  • 9.Zheng D, Liwinski T, Elinav E. Inflammasome activation and regulation: toward a better understanding of complex mechanisms. Cell Discov. 2020;6:36. doi: 10.1038/s41421-020-0167-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kebaier C, Chamberland RR, Allen IC, Gao X, Broglie PM, Hall JD, et al. Staphylococcus aureus α-hemolysin mediates virulence in a murine model of severe pneumonia through activation of the NLRP3 inflammasome. J Infect Dis. 2012;205(5):807–17. doi: 10.1093/infdis/jir846 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cohen T, Boland ML, Boland BB, Takahashi V, Tovchigrechko A, Lee Y, et al. S. aureus evades macrophage killing through NLRP3-dependent effects on mitochondrial trafficking. Cell Rep. 2018;22(9):2431–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Cohen TS, Prince AS. Activation of inflammasome signaling mediates pathology of acute P. aeruginosa pneumonia. J Clin Invest. 2013;123(4):1630–7. doi: 10.1172/JCI66142 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Witzenrath M, Pache F, Lorenz D, Koppe U, Gutbier B, Tabeling C, et al. The NLRP3 inflammasome is differentially activated by pneumolysin variants and contributes to host defense in pneumococcal pneumonia. J Immunol. 2011;187(1):434–40. doi: 10.4049/jimmunol.1003143 [DOI] [PubMed] [Google Scholar]
  • 14.McNeela EA, Burke A, Neill DR, Baxter C, Fernandes VE, Ferreira D, et al. Pneumolysin activates the NLRP3 inflammasome and promotes proinflammatory cytokines independently of TLR4. PLoS Pathog. 2010;6(11):e1001191. doi: 10.1371/journal.ppat.1001191 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res. 2019;29(5):347–64. doi: 10.1038/s41422-019-0164-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Weinlich R, Oberst A, Beere HM, Green DR. Necroptosis in development, inflammation and disease. Nat Rev Mol Cell Biol. 2017;18(2):127–36. doi: 10.1038/nrm.2016.149 [DOI] [PubMed] [Google Scholar]
  • 17.Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature. 2015;517(7534):311–20. doi: 10.1038/nature14191 [DOI] [PubMed] [Google Scholar]
  • 18.Riebeling T, Kunzendorf U, Krautwald S. The role of RHIM in necroptosis. Biochem Soc Trans. 2022;50(4):1197–205. doi: 10.1042/BST20220535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.González-Juarbe N, Gilley RP, Hinojosa CA, Bradley KM, Kamei A, Gao G, et al. Pore-Forming Toxins Induce Macrophage Necroptosis during Acute Bacterial Pneumonia. PLoS Pathog. 2015;11(12):e1005337. doi: 10.1371/journal.ppat.1005337 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kitur K, Parker D, Nieto P, Ahn DS, Cohen TS, Chung S, et al. Toxin-induced necroptosis is a major mechanism of Staphylococcus aureus lung damage. PLoS Pathog. 2015;11(4):e1004820. doi: 10.1371/journal.ppat.1004820 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lee S, Karki R, Wang Y, Nguyen LN, Kalathur RC, Kanneganti T-D. AIM2 forms a complex with pyrin and ZBP1 to drive PANoptosis and host defence. Nature. 2021;597(7876):415–9. doi: 10.1038/s41586-021-03875-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Gao Y, Yu S, Chen M, Wang X, Pan L, Wei B, et al. cFLIPS regulates alternative NLRP3 inflammasome activation in human monocytes. Cell Mol Immunol. 2023;20(10):1203–15. doi: 10.1038/s41423-023-01077-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Yang J, Zhao Y, Shi J, Shao F. Human naip and mouse naip1 recognize bacterial type iii secretion needle protein for inflammasome activation. Proc Natl Acad Sci U S A. 2013;110(35):14408–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yang J, Hwang I, Lee E, Shin SJ, Lee EJ, Rhee JH, et al. Bacterial outer membrane vesicle-mediated cytosolic delivery of flagellin triggers host NLRC4 canonical inflammasome signaling. Front Immunol. 2020;11:581165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lee J, Cacalano G, Camerato T, Toy K, Moore MW, Wood WI. Chemokine binding and activities mediated by the mouse IL-8 receptor. J Immunol. 1995;155(4):2158–64. doi: 10.4049/jimmunol.155.4.2158 [DOI] [PubMed] [Google Scholar]
  • 26.Zlotnik A, Yoshie O. Chemokines: a new classification system and their role in immunity. Immunity. 2000;12(2):121–7. doi: 10.1016/s1074-7613(00)80165-x [DOI] [PubMed] [Google Scholar]
  • 27.Sun L, Wang H, Wang Z, He S, Chen S, Liao D, et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell. 2012;148(1–2):213–27. doi: 10.1016/j.cell.2011.11.031 [DOI] [PubMed] [Google Scholar]
  • 28.Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526(7575):660–5. doi: 10.1038/nature15514 [DOI] [PubMed] [Google Scholar]
  • 29.Hildebrand JM, Tanzer MC, Lucet IS, Young SN, Spall SK, Sharma P, et al. Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic cell death. Proc Natl Acad Sci U S A. 2014;111(42):15072–7. doi: 10.1073/pnas.1408987111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yang B, Fu L, Privratsky JR, Lu X, Ren J, Mei C, et al. Interleukin-1 receptor activation aggravates autosomal dominant polycystic kidney disease by modulating regulated necrosis. Am J Physiol Renal Physiol. 2019;317(2):F221–8. doi: 10.1152/ajprenal.00104.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N, et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol. 2005;1(2):112–9. doi: 10.1038/nchembio711 [DOI] [PubMed] [Google Scholar]
  • 32.Meng R. High mobility group box 1 enables bacterial lipids to trigger receptor-interacting protein kinase 3 (RIPK3)-mediated necroptosis and apoptosis in mice. J Biol Chem. 2019;294(22):8872–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kaiser WJ, Sridharan H, Huang C, Mandal P, Upton JW, Gough PJ, et al. Toll-like receptor 3-mediated necrosis via TRIF, RIP3, and MLKL. J Biol Chem. 2013;288(43):31268–79. doi: 10.1074/jbc.M113.462341 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Upton JW, Kaiser WJ, Mocarski ES. DAI/ZBP1/DLM-1 complexes with RIP3 to mediate virus-induced programmed necrosis that is targeted by murine cytomegalovirus vIRA. Cell Host Microbe. 2012;11(3):290–7. doi: 10.1016/j.chom.2012.01.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Jiao H, Wachsmuth L, Kumari S, Schwarzer R, Lin J, Eren RO, et al. Z-nucleic-acid sensing triggers ZBP1-dependent necroptosis and inflammation. Nature. 2020;580(7803):391–5. doi: 10.1038/s41586-020-2129-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Conos S, Chen KW, De Nardo D, Hara H, Whitehead L, Núñez G, et al. Active mlkl triggers the nlrp3 inflammasome in a cell-intrinsic manner. Proc Natl Acad Sci U S A. 2017;114(6):E961–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Schmitt M, Ceteci F, Gupta J, Pesic M, Böttger TW, Nicolas AM, et al. Colon tumour cell death causes mTOR dependence by paracrine P2X4 stimulation. Nature. 2022;612(7939):347–53. doi: 10.1038/s41586-022-05426-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wooff Y, Man SM, Aggio-Bruce R, Natoli R, Fernando N. IL-1 Family Members Mediate Cell Death, Inflammation and Angiogenesis in Retinal Degenerative Diseases. Front Immunol. 2019;10:1618. doi: 10.3389/fimmu.2019.01618 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Anderson CJ, Medina CB, Barron BJ, Karvelyte L, Aaes TL, Lambertz I, et al. Microbes exploit death-induced nutrient release by gut epithelial cells. Nature. 2021;596(7871):262–7. doi: 10.1038/s41586-021-03785-9 [DOI] [PubMed] [Google Scholar]
  • 40.Wang Y, Gao W, Shi X, Ding J, Liu W, He H, et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature. 2017;547(7661):99–103. doi: 10.1038/nature22393 [DOI] [PubMed] [Google Scholar]
  • 41.Yuan J, Amin P, Ofengeim D. Necroptosis and ripk1-mediated neuroinflammation in cns diseases. Nat Rev Neurosci. 2019;20(1):19–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Shi H, Gao Y, Dong Z, Yang JE, Gao R, Li X, et al. GSDMD-Mediated Cardiomyocyte Pyroptosis Promotes Myocardial I/R Injury. Circ Res. 2021;129(3):383–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lemon JK, Weiser JN. Degradation products of the extracellular pathogen Streptococcus pneumoniae access the cytosol via its pore-forming toxin. mBio. 2015;6(1):e02110-14. doi: 10.1128/mBio.02110-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Nguyen TA, Smith BRC, Elgass KD, Creed SJ, Cheung S, Tate MD, et al. SIDT1 Localizes to Endolysosomes and Mediates Double-Stranded RNA Transport into the Cytoplasm. J Immunol. 2019;202(12):3483–92. doi: 10.4049/jimmunol.1801369 [DOI] [PubMed] [Google Scholar]
  • 45.Nguyen TA, Smith BRC, Tate MD, Belz GT, Barrios MH, Elgass KD, et al. SIDT2 Transports Extracellular dsRNA into the Cytoplasm for Innate Immune Recognition. Immunity. 2017;47(3):498-509.e6. doi: 10.1016/j.immuni.2017.08.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Zhang R. Tlr3/trif and mavs signaling is essential in regulating mucosal t cell responses during rotavirus infection. J Immunol. 2024;213(7):1008–22. [DOI] [PubMed] [Google Scholar]
  • 47.D’Alessio FR, Tsushima K, Aggarwal NR, West EE, Willett MH, Britos MF, et al. CD4+CD25+Foxp3+ Tregs resolve experimental lung injury in mice and are present in humans with acute lung injury. J Clin Invest. 2009;119(10):2898–913. doi: 10.1172/JCI36498 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Eckle T, Grenz A, Laucher S, Eltzschig HK. A2b adenosine receptor signaling attenuates acute lung injury by enhancing alveolar fluid clearance in mice. J Clin Invest. 2008;118(10):3301–15. [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Andrew Monteith

20 May 2024

Dear Professor Chen,

Thank you very much for submitting your manuscript "Caspase-1-licensed pyroptosis drives dsRNA mediated necroptosis and dampen host defense against bacterial pneumonia" for consideration at PLOS Pathogens. As with all papers reviewed by the journal, your manuscript was reviewed by members of the editorial board and by several independent reviewers. In light of the reviews (below this email), we would like to invite the resubmission of a significantly-revised version that takes into account the reviewers' comments.

We cannot make any decision about publication until we have seen the revised manuscript and your response to the reviewers' comments. Your revised manuscript is also likely to be sent to reviewers for further evaluation.

When you are ready to resubmit, please upload the following:

[1] A letter containing a detailed list of your responses to the review comments and a description of the changes you have made in the manuscript. Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

[2] Two versions of the revised manuscript: one with either highlights or tracked changes denoting where the text has been changed; the other a clean version (uploaded as the manuscript file).

Important additional instructions are given below your reviewer comments.

Please prepare and submit your revised manuscript within 60 days. If you anticipate any delay, please let us know the expected resubmission date by replying to this email. Please note that revised manuscripts received after the 60-day due date may require evaluation and peer review similar to newly submitted manuscripts.

Thank you again for your submission. We hope that our editorial process has been constructive so far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Andrew J Monteith, Ph.D.

Guest Editor

PLOS Pathogens

Matthew Wolfgang

Section Editor

PLOS Pathogens

Michael Malim

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0002-7699-2064

***********************

Reviewer's Responses to Questions

Part I - Summary

Please use this section to discuss strengths/weaknesses of study, novelty/significance, general execution and scholarship.

Reviewer #1: Luo et al. use a E. coli respiratory infection mouse model of bacterial pneumonia to evaluate the host determinants of pathogenesis. Interestingly, the authors observe that Casp1 knockout (KO) mice experience less disease despite an increase in BALF neutrophils. These observations were largely phenocopied by Zbp1 and Mlkl KO mice deficient in necroptosis, raising the interesting question of how pyroptotic and necroptotic pathways intersect to promote lung disease. A combination of in vivo and in vitro experiments suggests a possible role for host-derived dsRNA in the initiation of Zbp1-dependent necroptosis in neutrophils. By evaluating FLICA and cell death in distinct cell types from infected lungs, the authors observed enhanced Casp1 activity primarily in epithelia (and endothelia) whereas markers of necroptotic death were primarily observed in neutrophils. Based on these findings and some clever in vitro experiments, the authors conclude that E. coli lung infection causes lung epithelial cell Casp1-dependent pyroptosis, which releases dsRNA that is taken up by neutrophils leading to Zbp1-dependent necroptosis, the subsequent loss of neutrophils, and enhanced pathogenesis. In many respects, I find this model of non-intrinsic iterative cell death appealing, and it provides an important alternative mechanism to so called 'PANoptotic' cell-intrinsic pathways to explain how distinct cell death pathways can interact. However, many of the most exciting and new conclusions are largely based on correlative analyses that leave several alternative explanations for the data presented.

Reviewer #2: In “Caspase-1-licensed pyroptosis drives dsRNA mediated necroptosis and dampen host

defense against bacterial pneumonia”, the authors seek to show that caspase-1 mediated death of epithelial cells leads to necroptosis of neutrophils that negatively impacts lung health. Points of scholarship that would benefit from improvement: 1) Current epidemiological and other background (some studies are 20 years out of date and not accurate), 2) there is an insufficient description of E coli pneumonia as a disease, 3) the endotoxin responses in the lung, which has extensive literature that should be cited and discussed, since this has not been excluded as a major driver of inflammation in this model, and is expected to be. The topic is of overall interest with moderate novelty, using mostly appropriate methodology, but lacks support for major claims or relies on indirect evidence, building an overall model that could be incorrect. These are outlined below:

Reviewer #3: The manuscript by Qinyu Luo et al describes how epithelial cells-neutrophils crosstalk leading to the progression of pneumonia. Specifically, it showed during E. coli lung infection (pneumonia), Caspase1-mediated pyroptotic epithelial cells release dsRNA in the bronchoalveolar lavage fluid (BALF). As a result, dsRNA along with E. coli induce neutrophil necroptosis, which is associated with a detrimental impact on the disease outcome. The manuscript is well written except for the last portion of the result section. It used several genetic and pharmaceutical tools to support these findings. For the most part, I found the data supported the authors’ concussions. I think the findings are valuable to the field of lung immunopathology. The data will increase our understanding of how different death pathways are engaged in a complex environment with multiple types of cells to drive disease pathology. However, I have several concerns that could clear some of the ambiguous conclusions that the authors draw. The following are my concerns:

**********

Part II – Major Issues: Key Experiments Required for Acceptance

Please use this section to detail the key new experiments or modifications of existing experiments that should be absolutely required to validate study conclusions.

Generally, there should be no more than 3 such required experiments or major modifications for a "Major Revision" recommendation. If more than 3 experiments are necessary to validate the study conclusions, then you are encouraged to recommend "Reject".

Reviewer #1: One of the major claims is that Casp1-dependent pyroptosis is primarily occurring in epithelial cells, whereas Zbp1/Ripk3/Mlkl-dependent pyroptosis is primarily occurring in neutrophils downstream of epithelial pyroptosis. As the authors correctly point out, the in vivo experiments do not allow for this strong conclusion.

o Cell-type specific KO or KI mice (e.g., deletion of Casp1 and Mlkl in epithelial vs neutrophils) to assess these claims would be welcome and definitive, but I recognize the considerable time and cost associated with such experiments. Nevertheless, the authors focus primarily on epithelia, which ignores the result in Fig. 4A and 4B that Casp1 is active in endothelia. Thus, it is unclear whether the in vivo source of pyroptosis (and the presumed release of dsRNA) is epithelial, endothelia, or both.

o Reciprocal bone marrow chimera experiments would be an appropriate alternative. If the authors choose to do so, they might consider also including Gsdmd KO mice since they formally did not genetically demonstrate the role of pyroptosis in vivo or in vitro.

o While I find the in vitro experiments with MLE12 and neutrophils a useful in vitro model, the authors could much more rigorously support their claims by genetically deleting genes relevant to the inflammasome and necroptotic pathways in both MLE12 and neutrophils. Editing primary neutrophils may not be possible, but presumably neutrophils could be used from KO mice in this study, or alternatively, make use of more genetically tractable cell lines (e.g., Cas9+ER-Hoxb8 neutrophil progenitor). Such a model would also allow for complementation of KO neutrophils with variants of interest (e.g., Zbp1 mutants that lack the capacity to bind dsRNA).

The role for dsRNA in neutrophil necroptosis relies primarily on two observations: 1) that Zbp1 KO mice are protected from disease, and 2) injection of a dsRNA binding antibody attenuates disease. This is a remarkable claim, and is the key mechanistic detail linking the two cell death modalities in a non-cell intrinsic manner. Thus, further validation either in vivo or in vitro is important to substantiate the author’s conclusions:

o A prediction of the model is that the injection of dsRNA binding Ab would have no effect on a Zbp1, Ripk3, or Mlkl KO mouse, or similarly, in the response of neutrophils from those mice ex vivo.

o Epithelial cell death is not sufficient to induce neutrophil death, but an explanation is not provided. The authors should present data to determine if dsRNA is released from non-E. coli infected epithelia, and if so, if dsRNA is being taken up by neutrophils in the absence of bacteria. Similarly, the experiments in Figure S4A (e.g., WT vs Casp1 KO challenged with E. coli, heat-killed E. coli, etc) should be conducted on MLE12 cells, followed by conditioned supernatants from those studies being used to evaluate necroptosis in neutrophils derived from WT and relevant KO animals. Moreover, the authors rule out a role for IL-1B, but it remains possible that IL-18 or other signals released from pyroptotic epithelia are required to stimulate neutrophils for either the uptake of dsRNA or activation of the necroptotic pathway.

Reviewer #2: While E. coli pneumonia can occur, the strain used, ATCC 25922, is a non-pathogen. A relevant virulent clone known to cause pulmonary disease in humans or experimental models should be used.

Furthermore, there are no counts of bacteria in the lung for nearly experiment. This is critical to include throughout, and is very unconventional to exclude. A major limitation to the interpretation that the authors should additionally discuss how the greater survival of casp1-/- mice, and their lessor injury, could be due to them just having fewer bacteria (Fig S1). In this sense, inflammatory responses are typically proportional to bacterial load, and the effects reported could be entirely a consequence of that. Effects on IL-1b, respiration, temperature, neutrophil number, and their necroptosis, would all expect to be consequent to that, and not necessarily through the described casp-1-dependent mechanism.

The authors report that IL-1 is not involved because anakinra could not rescue the mice from lethal pneumonia (line 275). However, the authors have not shown that anakinra as delivered was available in the lung, nor that it inhibited IL-1 signaling like claimed. Actual signaling of IL-1 in the lung should to be measured to know whether the drug is having the stated effect, and this mechanism needs to be confirmed using IL-1R KO mice.

Similarly, the authors need to show that the cell ablations of Fig 1F and 1G are leading to actual depletion of these cells in the experiment.

The experiments of Fig 5F and 5G are an unconvincing demonstration of PolyIC uptake. F shows association with cells, which could be sticking on the surface and not uptake. G is very low magnification, and appears to show staining of the whole cell for some cells, and no staining at all for others, which isn’t a demonstration of colocalization. Higher-magnification imaging, with membrane markers if needed, should be used to show polyIC is intracellular, and colocalization requires that there are areas that there are areas lacking both polyIC and ZBP1, not just the whole cell imaged as positive for both.

Figure 2E compares proteins in BALF cells, which are mostly neutrophils, to the tissue, which is mostly not. For one, the methods for this experiment are unclear, not in the text, the legend, or figure is it stated how and when these samples were derived. I will assume this is just an omission and that they were infected mice, though not stated, and for a similar amount of time as other experiments. However, there are several biases in these comparisons needing controls including uninfected mice and purified neutrophils, and a survivorship bias, as their other data show evidence of significant death by this point, so it is living cells that have p-MLKL, arguing that the dead cells might have died by other means. This major point also needs the good blotting control using the MLKL-/- mice of Fig 3G

3E requires PBS control for infection-independent effects of the drug on neutrophil death

The cytometry of Fig 4A 4B requires additional details on markers, compensation, and gating, total and live/dead cells of each population, and controls of uninfected mice. As it is, it is not clear how abundant the cell populations are, how accurate their identification is, and how reliable the FLICA staining is. In particular, the cell processing method is quite harsh and expected to lead to significant cell death, which can bias these populations, and give false positives/negatives.

A statistician should review the methods, which are applied inconsistently between figures. For example, two-way ANOVA with Tukey-Kramer’s post hoc analysis is done for Fig 3 (C, D, E and F), but some of these are pairwise, others multivariate, and methods are applied inconsistently between different figures of similar data.

There are gaps in the claims on the specificity of each cell death & sensing mechanism to each cell type. This is most cleanly and directly addressed by lineage-specific knockout. It minimum, these limitations should be discussed and redundancies between these pathways should be excluded including the effect of dsRNA antibody on neutrophil death in infected casp1-/- and mlkl-/- mice, the effect of mlkl-/- on FLICA cells, and histological examinations added to the major mechanistic points (not just Fig 1B)

Reviewer #3: 1. The presented data did not directly demonstrate in vivo that pyroptotic epithelial cells are responsible for dsRNA release in BALF. It is also not clear which cell death (Pyroptosis versus necroptosis) occurs first, or whether dead neutrophil causes epithelial cell damage and release of dsRNA. To address these, the authors should use epithelial cell-specific Caspase-1 deficient mice. However, generating such a mouse line will take at least 6 months. Instead, adoptive transfer experiment could provide some support to the conclusions. Bone marrow-derived neutrophils from wild-type mice could be adoptively transferred to UV-irradiated Caspase-1 KO mice and test pneumonia outcomes (mice survival; bacterial and immune cell counts; dsRNA level in the BALF and lungs; neutrophil and epithelial cell death and so on). The mice should be protected if the proposed conclusion is correct (i.e., pyroptotic epithelial cells released dsRNA to induce necroptotic neutrophils). Also, bone marrow-derived neutrophils from Caspase-1 KO mice could be adoptively transferred to UV-irradiated wild-type mice and test pneumonia outcomes. The experiment outcome could assess whether epithelial cell death or neutrophil death occurs first.

2. Which type of epithelial cells are affected? the authors should combine Tunel staining of histology sections from wild-type and Caspase1 KO with different cell markers including type I epithelial, type II epithelial, macrophages, neutrophils, and monocytes.

3. A combination of LPS + poly I:C stimulation to induce necroptotic neutrophils in vitro and in vivo should be tested. This will provide evidence that dsRNA from dying cells along with LPS from bacteria are sufficient to derive necroptotic neutrophils.

4. The authors need to measure bacterial loads in the lungs. Measuring bacterial counts in the BALF is not sufficient to conclude bacterial clearance. Bacterial counts in BALF depend on the efficiency of performing the lavage (do the authors use equal volume to lavage mice and whether end up with equal volumes of BALF from wild type and KOs. Also, immune cell counts in the lungs are a better predictor of disease outcomes than BALF.

5. Fig. 1D showed a massive loss of macrophage during infection. Are they dying? If so, which cell deaths are responsible, and could they contribute to dsRNA release? Why there are lower macrophage counts in uninfected wild-type relative to Caspase-1 KO? Do Caspase-1 KO mice experience some sort of low-grade inflammation, especially in the lungs?

6. All immune cells count, and bacterial burden should be graphed using a log y-axis scale. This way we could see the lower number of immune cells and bacteria that are present in some of the conditions.

7. It is not clear whether BALF cells in Fig. 2C or Fig. 4H that induced necroptosis are macrophages, monocytes, or neutrophils. Although neutrophil counts are higher in BALF, the amount of protein in these cells is very low compared to macrophages and monocytes. The authors should either sort these cells or magnetic purify them. Similarly, the remnant lung tissue after lavage is not only epithelial cells. Many other cells including interstitial macrophages could be the driver for activation of Caspase-1 and GSDMD.

8. Treated mice with control solvents such as DMSO, Saline, control IgG…etc are dying at a faster rate than untreated mice. Examples include in fig. 3A, fig. 3B, fig.5I, supplementary fig. 4D and 4E. Do the authors know why this is the case? Does DMSO or saline treatment make the mice more susceptible to infection? This should be emphasized.

**********

Part III – Minor Issues: Editorial and Data Presentation Modifications

Please use this section for editorial suggestions as well as relatively minor modifications of existing data that would enhance clarity.

Reviewer #1: - It is unclear why preventing neutrophil necroptosis limits pathogenesis. Do the authors consider this to be similar to pathology associated with NETosis?

- Can the authors provide an explanation based on their model for why E. coli CFU is lower in all KOs or interventions that prevent pyroptosis and necroptosis?

- I find it somewhat surprising that Escherichia coli Seattle 1946 strain induces epithelial cell Casp1-dependent death. Presumably this strain is attaching/effacing, and not replicating intracellularly? Can the authors comment on this and the nature of the upstream sensor that induces Casp1 activation in lung epithelia?

- Similarly, while I do not think it is within the scope of the present work, it would be interesting to know the inflammasome-forming sensor upstream of Casp1. This would increase the impact of the manuscript.

- Tracheal infection of E. coli is not very physiologic. Demonstrating this pathway occurs in the context of a respiratory pathogen would increase relevance.

- In Fig. S4B, Casp1 does seem to be active in neutrophils upon challenge with E. coli or LPS. Can the authors speak to this?

- For each mouse experiment, please clarify if mice were littermates, cohoused, or neither.

Reviewer #2: none

Reviewer #3: 1. Impact neutrophil depletion on wild-type mice should be provided. Does neutrophil depletion impact bacterial clearance in wild-type mice?

2. The lung injury score graph (Fig. 1B) should be fixed back to a standard y-axis graph. It is not clear why a two-segment y-axis was used.

3. The last section of the results was poorly written (lines 325-381). There are a lot of missing spaces, the authors used “we next” several times to start a new sentence. It seems that the authors listing experiment findings without rationale or context.

4. In line 41 “conbined” should be fixed to combined.

5. The authors should immunofluorescence stain other BALF and lung cells with pMLKL antibody, like supplement fig. 2H. This could provide evidence that necroptosis impacted only neutrophils and/or other cells.

6. Does pyroptosis still occur in epithelial cells when the mice were treated with GW806742X or in Mlkl KO mice?

7. It is not clear how purified neutrophils are dying in response to E. coli and heat-killed E. coli in supplementary fig. 4A. The authors need to include more detail on how infections were performed. What is the multiplicity of infection (MOI) used and what time point post-infection cell death was analyzed? Are extracellular bacteria washed or kept in the media? Also, LPS-treated purified neutrophils induce activation of MLKL but no cell death. The authors need to explain why LPS stimulation is insufficient to trigger necroptosis.

8. The number of mice used in the supplementary fig. 4D and 4E is low. It is not clear how many times the experiment was repeated on different days. Power analysis should be performed, and I recommend using a biostatistical expert to determine this.

9. For the in vivo data, the authors often list the n number, which represents the number of used mice. However, whether the experiments were done on different days needs to be clarified.

10. The authors should calculate and graph the intensity of bands from immunoblots in Fig. 2D and 2E.

11. What is the stain used in Fig. 4D? The authors should indicate detailed information in the figure legend and the method section.

12. It is not stated how the anti-dsRNA was administered to mice.

13. It is not clear what is the purpose of measuring long terminal repeat elements (LTRs), long interspersed nuclear elements (LINEs), or short interspersed nuclear elements. The authors need to explain the rationale behind doing such an analysis. Is the dsRNA measured by RNAseq originate from epithelial or neutrophil?

14. The authors state “However, GW806742X is not a specific inhibitor of MLKL” What other proteins does the inhibitor target?

**********

PLOS authors have the option to publish the peer review history of their article (what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy .

Reviewer #1: No

Reviewer #2: No

Reviewer #3: No

Figure Files:

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org.

Data Requirements:

Please note that, as a condition of publication, PLOS' data policy requires that you make available all data used to draw the conclusions outlined in your manuscript. Data must be deposited in an appropriate repository, included within the body of the manuscript, or uploaded as supporting information. This includes all numerical values that were used to generate graphs, histograms etc.. For an example see here on PLOS Biology: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5.

Reproducibility:

To enhance the reproducibility of your results, we recommend that you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option to publish peer-reviewed clinical study protocols. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

Attachment

Submitted filename: Qinyu Luo et al manuscript review .docx

ppat.1013167.s012.docx (22KB, docx)

Decision Letter 1

Andrew Monteith

23 Dec 2024

PPATHOGENS-D-24-00639R1

Caspase-1-licensed pyroptosis drives dsRNA mediated necroptosis and dampen host defense against bacterial pneumonia

PLOS Pathogens

Dear Dr. Chen,

Thank you for submitting your manuscript to PLOS Pathogens. After careful consideration, we feel that it has merit but does not fully meet PLOS Pathogens's publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

Please submit your revised manuscript within 30 days Feb 21 2025 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plospathogens@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/ppathogens/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

* A rebuttal letter that responds to each point raised by the editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'. This file does not need to include responses to any formatting updates and technical items listed in the 'Journal Requirements' section below.

* A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

* An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, competing interests statement, or data availability statement, please make these updates within the submission form at the time of resubmission. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

We look forward to receiving your revised manuscript.

Kind regards,

Andrew J Monteith, Ph.D.

Guest Editor

PLOS Pathogens

Matthew Wolfgang

Section Editor

PLOS Pathogens

Sumita Bhaduri-McIntosh

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0003-2946-9497

Michael Malim

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0002-7699-2064

Additional Editor Comments :

I appreciate the effort that the authors gave to resolving the issues associated with this manuscript; however, there are still concerns that exist or remain unaddressed.

• I share a similar concern that bacterial burdens in BALF may not be reflective of whole tissue burdens, which was only addressed by citing a reference in the rebuttal letter. However, my larger concern is the lack of burden quantification in the in vivo models, in particular when quantifying in vivo functions. Burden quantifications are critical to ensure that the infection was consistent and that identified phenotypes are not associated with differences in inoculations or infection dynamics. Every in vivo characterization should be coupled with CFU burdens to ensure accuracy of the data.

• Multiple pieces of data were provided in the rebuttal letter to address reviewer concerns, but these were not included in the manuscript (TUNEL staining with different cell populations, stimulation with LPS+ poly I:C, and pMLKL immunofluorescence stain of BALF cells). The point of these data is making the manuscript better not to directly address reviewers. This was an oversight not integrating these data into the manuscript. There were also instances in the text where conclusions are made without showing data (eg: line 161; phagocytosis efficiency…were comparable).

• The interpretation of Figure 5B is confusing. It seems unlikely that simply adding bacterial supernatant and MLE12 cell lysate nearly fully recreates the supernatant from pyroptotic MLE12 cells. These are entirely different stimulations. Also, if the stimulating agent is dsRNA that gets released from the epithelial cells, does pyroptosis cause an abundance of dsRNA? It is unclear why dsRNA (which would likely be in low abundance) would be such a potent stimulation when the abundance of cytokines and other nuclear molecules (dsDNA, ssRNA) would be much higher. In addition, why would dsRNA only be found in the BALF, but not identified in the lung homogenates (Fig. 5E)?

• The authors use an antibody to neutralize dsRNA during infection (Fig. 5I-K), but rather than simply neutralizing the dsRNA, this likely created immune complexes and would skew the inflammatory environment. It's really uncertain what the dsRNA antibody is doing, especially in vivo. A cleaner experiment would be to use TLR3-deficient mice, which are readily available, and prevent signaling by dsRNA. TLR3 seems the most likely receptor mediating neutrophil necroptosis and it is unusual that this was overlooked in the manuscript.

Data quality concerns:

• In Figure 5B the input lane is bands while the IP lanes look highly smeared or dot blot-like (ZBP1 bands). Presumably this was all run on the same gel, so why are the lanes so variable in presentation?

• The fluorescent images provided in Figure 5G are not of sufficient quality to draw any conclusions. The pattern of the dapi stain seems unusual and doesn’t seem to be staining the nucleus. The background of the Poly(I:C) in the bacterial stimulated cells looks excessive and localized to a specific region, which is not present in the mock stimulated. Despite these concerns in the images, providing a single cell is not sufficient to draw conclusions. Higher quality representative images and quantification is required.

• Supplementary Figure 2I includes images of BALF cells with high pMLKL protein at the membrane. Based on the proposed model, the neutrophils are the cell type with high pMLKL protein; however, the nuclear staining (Dapi) of these cells do not look neutrophil-like. This is concerning and requires further validation (Ly6g or neutrophil specific protein staining) to ensure the cell type of the BALF cells.

Journal Requirements:

1) We note that your Data Availability Statement is currently as follows: "All data generated and analyzed during this study are available." Please confirm at this time whether or not your submission contains all raw data required to replicate the results of your study. Authors must share the “minimal data set” for their submission. PLOS defines the minimal data set to consist of the data required to replicate all study findings reported in the article, as well as related metadata and methods (https://journals.plos.org/plosone/s/data-availability#loc-minimal-data-set-definition).

For example, authors should submit the following data: 

1) The values behind the means, standard deviations and other measures reported;

2) The values used to build graphs;

3) The points extracted from images for analysis..

Authors do not need to submit their entire data set if only a portion of the data was used in the reported study.

If your submission does not contain these data, please either upload them as Supporting Information files or deposit them to a stable, public repository and provide us with the relevant URLs, DOIs, or accession numbers. For a list of recommended repositories, please see https://journals.plos.org/plosone/s/recommended-repositories. 

If there are ethical or legal restrictions on sharing a de-identified data set, please explain them in detail (e.g., data contain potentially sensitive information, data are owned by a third-party organization, etc.) and who has imposed them (e.g., an ethics committee). Please also provide contact information for a data access committee, ethics committee, or other institutional body to which data requests may be sent. If data are owned by a third party, please indicate how others may request data access.

Please ensure that the Data Availability Statement provided in the online submission form matches the one mentioned in the manuscript.

2) Please amend your detailed Financial Disclosure statement. This is published with the article. It must therefore be completed in full sentences and contain the exact wording you wish to be published.

1) Please clarify all sources of financial support for your study. List the grants, grant numbers, and organizations that funded your study, including funding received from your institution. Please note that suppliers of material support, including research materials, should be recognized in the Acknowledgements section rather than in the Financial Disclosure.

2)State the initials, alongside each funding source, of each author to receive each grant. For example: "This work was supported by the National Institutes of Health (####### to AM; ###### to CJ) and the National Science Foundation (###### to AM)."

3) State what role the funders took in the study. If the funders had no role in your study, please state: "The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript."

3) Your current Financial Disclosure states, "The author(s) received no specific funding for this work."

However, your funding information on the submission form indicates receiving a fund. Please ensure that the funders and grant numbers match between the Financial Disclosure field and the Funding Information tab in your submission form. Note that the funders must be provided in the same order in both places as well.. 

Please indicate by return email the full and correct funding information for your study and confirm the order in which funding contributions should appear. Please be sure to indicate whether the funders played any role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Comments to the Authors:

Please note that one of the reviews is uploaded as an attachment.

Reviewers' Comments:

Reviewer's Responses to Questions

Part I - Summary

Please use this section to discuss strengths/weaknesses of study, novelty/significance, general execution and scholarship.

Reviewer #1: The authors have addressed my major issues, in particular, the bone marrow chimeras indicating that pyroptosis and necroptosis occurs in distinct compartments (hematopoietic vs non-hematopoietic, respectively). In my opinion, this constitutes a significant advance in our understanding of how distinct cell death pathways in distinct cell types can underlie the host response and pathogenesis in a model of respiratory bacterial infection.

Reviewer #3: The authors addressed some of the first round of reviews. However, there are still many concerns that need to be addressed. I also recommend having English as the first language colleague and working in the same field to edit the manuscript.

**********

Part II – Major Issues: Key Experiments Required for Acceptance

Please use this section to detail the key new experiments or modifications of existing experiments that should be absolutely required to validate study conclusions.

Generally, there should be no more than 3 such required experiments or major modifications for a "Major Revision" recommendation. If more than 3 experiments are necessary to validate the study conclusions, then you are encouraged to recommend "Reject".

Reviewer #1: (No Response)

Reviewer #3: 1) The authors did not include the new data (TUNEL staining, stimulation with LPS+ poly I:C, and pMLKL immunofluorescence stain of BALF cells) in the revised manuscript. Only zoomed-out immunofluorescence images were provided, which made it difficult to analyze. The authors should provide higher-resolution zoomed-in images and quantify the data.

2) Measuring bacterial load in the lungs is still missing. Providing references that used BALF bacterial burden to assess lung pneumonia is weak. The authors should attempt at least to provide CFU data in the lungs of WT and Casp1 KO mice.

**********

Part III – Minor Issues: Editorial and Data Presentation Modifications

Please use this section for editorial suggestions as well as relatively minor modifications of existing data that would enhance clarity.

Reviewer #1: The authors should quantify results shown in figures Fig. 5F and 5G.

Reviewer #3: 1) In the Abstract (line 18), what did the authors mean by “cell reaction”? Need some clarification.

2) In line 38, there is a missing period at the end of the sentence.

3) How is regulated cell death participating in the pathological process as stated in line 39? What do the authors mean by stating “regulated cell death”. Which cell death is not regulated? If cell death is controlled, then it should be beneficial.

4) The sentence in lines 248-253 is too long and needs to be broken into multiple sentences.

5) In lines 191-193, the authors included data regarding the clinical strain of bacteria, but there is no context. The authors should emphasize that the observed phenotype also occurs during infection with pathogenic E.coli. Information about the clinical isolate should be provided. I could not find the information in figure legends and materials/methods. Figures 2A and 2B should be labeled with the corresponding bacterial strain used to infect mice.

6) The authors need to indicate how they performed lung histology scoring.

7) Statistical analysis in Figures 1B, 1C, 1D, and 1E should be performed by two-way ANOVA.

8) In Fig.S2H, the authors need to provide zoomed images of neutrophils from Casp1 KO mice to compare to WT neutrophils.

9) In line 241. No need to start a new sentence.

10) In lines 243-245, the authors stated, “These findings also imply that caspase-1 may not act directly in the upstream of MLKL in this context, suggesting a potential alternative pathway in regulating neutrophil necroptosis during infection”. I am not following this train of thought. What data supports this conclusion?

11) No need for a new sentence in line 279.

12) In Figure S4E, treating mice with Anakinra is protective. The authors only used 6 mice, which may be insufficient to perform statistical analysis. A power analysis to identify sample size is needed. Provide the actual p-value for this graph.

13) The end of the paragraph in line 286 needs to have a conclusion sentence.

14) Two-way ANOVA must perform statistical analysis for figures 3C, 3D, and 3E.

15) Two-way ANOVA must perform statistical analysis for figures 4A, 4F, and 4G.

16) No need for a new sentence in line 320.

17) Two-way ANOVA must perform statistical analysis for figures S5B and S5C.

18) The sentence in lines 330-333 is too long and needs to be broken into multiple sentences.

19) No need for a new sentence in lines 341 and 345.

20) There is an extra period at the end of the sentence in line 342.

21) The end of the paragraph in line 350 needs to have a conclusion sentence.

22) No need for a new sentence in line 358.

23) Weirdly, poly(I:C) does not label control neutrophils in Figure 5G. How long the cells were stimulated?

24) The end of the paragraph in line 370 needs to have a conclusion sentence.

25) Two-way ANOVA must perform statistical analysis for figures 5F and 5H.

26) In Figure 6D, the authors indicated that they used WT control littermates. This should be labeled in the figures. Replace WT with either Zbp1+/+ or Zbp1+/- which is used in the experiment.

27) If control littermates were used in figures as indicated in lines 464-466, the authors should replace "WT" to indicate the WT mice genotype (i.e., Casp1+/+ or Casp1+/-).

28) No need for a new sentence in lines 379 and 383.

29) Two-way ANOVA must perform statistical analysis for Figure 6E.

30) No need for a new sentence in lines 531 and 537.

31) No need for a new sentence in lines 558, 559, 620, and 642.

**********

PLOS authors have the option to publish the peer review history of their article (what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy .

Reviewer #1: No

Reviewer #3: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

Figure resubmission:

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step. If there are other versions of figure files still present in your submission file inventory at resubmission, please replace them with the PACE-processed versions.

Reproducibility:

To enhance the reproducibility of your results, we recommend that authors of applicable studies deposit laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option to publish peer-reviewed clinical study protocols. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

Attachment

Submitted filename: Qinyu Luo et al review comments.docx

ppat.1013167.s014.docx (20.5KB, docx)

Decision Letter 2

Andrew Monteith

3 Apr 2025

PPATHOGENS-D-24-00639R2

Caspase-1-licensed pyroptosis drives dsRNA-mediated necroptosis and dampens host defense against bacterial pneumonia

PLOS Pathogens

Dear Dr. Chen,

Thank you for submitting your manuscript to PLOS Pathogens. After careful consideration, we feel that it has merit but does not fully meet PLOS Pathogens's publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process. At this point no further experiments are required, but we will ask you to address the Minor Comments and Major Comment #7 brought up by Reviewer 3 as these concerns relate to clarity issues and specific statistical tests that were used. 

Please submit your revised manuscript within 30 days Jun 02 2025 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plospathogens@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/ppathogens/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

* A rebuttal letter that responds to each point raised by the editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'. This file does not need to include responses to any formatting updates and technical items listed in the 'Journal Requirements' section below.

* A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

* An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, competing interests statement, or data availability statement, please make these updates within the submission form at the time of resubmission. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

We look forward to receiving your revised manuscript.

Kind regards,

Andrew J Monteith, Ph.D.

Guest Editor

PLOS Pathogens

Matthew Wolfgang

Section Editor

PLOS Pathogens

Sumita Bhaduri-McIntosh

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0003-2946-9497

Michael Malim

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0002-7699-2064

Reviewers' Comments:

Reviewer's Responses to Questions

Part I - Summary

Please use this section to discuss strengths/weaknesses of study, novelty/significance, general execution and scholarship.

Reviewer #1: The authors have made substantial revisions, which markedly improve an already strong manuscript. The work provides an important characterization of non-cell autonomous crosstalk between cell death pathways in a model of bacterial pathogenesis. The major findings are well supported by the data and meaningfully advance the field.

Reviewer #3: Although the authors did a somewhat good job incorporating the new data in the manuscript per previous review comments, several major concerns, especially technical concerns about the immunofluorescence microscopy images, need to be addressed. See below:

**********

Part II – Major Issues: Key Experiments Required for Acceptance

Please use this section to detail the key new experiments or modifications of existing experiments that should be absolutely required to validate study conclusions.

Generally, there should be no more than 3 such required experiments or major modifications for a "Major Revision" recommendation. If more than 3 experiments are necessary to validate the study conclusions, then you are encouraged to recommend "Reject".

Reviewer #1: (No Response)

Reviewer #3: 1) The immunofluorescence staining in Fig. 5G is not convincing to be specific. Why there is only one dot of FITC-poly(I: C) on neutrophils while fluorescent-positive cells can be easily directed by flow cytometry in Fig. 5F. The staining needs to be validated by using other cells like splenocytes or macrophages. The purpose of using PKH26 is to label cell boundaries, but clearly, this dye does not work for neutrophils. Why do DAPI-stained control cells not show classical neutrophil staining (polymorphonuclear)? The authors need to show images of many cells (lower magnification and zoomed). The provided images also appear blurry.

2) CD14-positive cells are not shown in Fig. S3B. The authors should use different areas of the slide where you can find the monocytes. The zoomed images of Casp1 KO mice are missing.

3) The Ly6G staining in Fig. S4 does not seem to be specific. The cells don’t look like neutrophils but epithelial cells. The staining looks different than the images from Fig. S3B, which represent stained neutrophils.

4) The bacterial burdens in the lungs seem to be too high. Mice were inoculated with 10^6 CFUs and yet there were 10^8 CFUs recovered after 12h. This is a two-log increase in bacterial counts in vivo within 12h, which seems abnormal. The authors need to check their calculations.

5) To compare the bactericidal function of Caspase 1 deficient neutrophils to wild-type neutrophils as stated in line 157, It is best to directly measure bactericidal activities.

6) It is not clear how WT and Casp1 KO neutrophils are dying from E.coli infection in Fig. S6A. What is the MOI and how long the infection was carried out? Do the neutrophils die due to bacterial overgrowth? If so, a lower MOI must be used. This needs some clarification and also needs to be mentioned in the main text.

7) Histology scoring criteria were based on the severity of damaged lungs. How the damage was quantified remained unclear. The word “damage” is ambiguous for assessing disease severity.

8) The authors’ conclusion that “the neutrophils from these mice did not express TLR3” needs rigorous assessment. Neutrophils from wild-type mice should express TLR3. Neutrophils response to TLR3 agonist in Fig. 6F and Fig. 5F, which implies that it is expressed. The RNAseq in Fig. 5A showed relative expression of the gene and was not simply present or absent. The data was also performed from n=2, which is insufficient to perform statistics. The immunoblot in Fig. S9 showed there is less TLR3 in Casp1 KO splenocytes compared to the wild-type. The authors need to confirm the specificity and sensitivity of the antibody that they used.

**********

Part III – Minor Issues: Editorial and Data Presentation Modifications

Please use this section for editorial suggestions as well as relatively minor modifications of existing data that would enhance clarity.

Reviewer #1: (No Response)

Reviewer #3: 1) Statements in lines 81-85 must be referenced.

2) The statement in lines 152-154 “Although we observed more tissue-resident macrophages in the Casp1-/- mice in the physiological state, these cells almost completely disappeared at 12 hours after E. coli infection” is incorrect. Fig. 1D showed 10^4 macrophages at 12h, They have not completely disappeared as stated. Also, what did the authors mean by recruited macrophages? Macrophages are not recruited, monocytes do and then differentiate into macophages. Provide information in the figure legend on what markers were used to discriminate between different immune cells.

3) It is not clear how the RNAseq in Fig. S1J shows similar antimicrobial defense signals were induced by the two mouse strains (WT and Casp1 KO). There is only one set of data. What is the n=2 represent in there? How many mice per group were used for the RNA sequencing?

4) What cells are referred to in line 165?

5) Need a figure caption to the findings in lines 206-212.

6) For Fig. 2D, the authors must provide quantifications for all the bands, it is not clear why they chose to quantify only selected bands. If other bands are unnecessary, then another blot should be provided.

7) Change the double dose of bacteria in Fig. 3B to actual CFUs

8) Change “depletion” in line 254 to deletion.

9) What is L929+T/Z in Fig. 4D? Please explain why this is done in the main text and needs to be mentioned in the figure legend.

10) Fig. 4F needs a better label to indicate that neutrophils were incubated with supernatants from indicated epithelial cell lines that were left uninfected (PBS) or infected (E.coli).

11) Fig. S8E was mentioned after Fig. S8F to H.

12) The authors concluded that administration of dsRNA antibody during E. coli infection strongly protected the mice in lines 375-376. However, the data (Fig. 5I) showed that the administration of dsRNA antibodies weakly protected mice from infection.

13) There is no panel H in Fig. S2 as stated in the figure legend.

14) The quantification in Fig. S3B must be performed by One-way ANOVA.

15) The histology images in Fig. S5D, Fig. S8D, and Fig. S8E need to be labeled

16) Statistical analysis in Fig. S5 A, B, and C must be performed using Two-way ANOVA.

17) What are the histograms shown in Fig. 7A? The x-axis needs to be labeled.

18) Statistical analysis in Fig. S7D must be performed using Owo-way ANOVA. All statistically significant data needs to be marked.

19) There should not be any statistical test performed on the image in Fig. 9A. Correct the figure legend.

**********

PLOS authors have the option to publish the peer review history of their article (what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy .

Reviewer #1: No

Reviewer #3: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

Figure resubmission:

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step. If there are other versions of figure files still present in your submission file inventory at resubmission, please replace them with the PACE-processed versions.

Reproducibility:

To enhance the reproducibility of your results, we recommend that authors of applicable studies deposit laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option to publish peer-reviewed clinical study protocols. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

Decision Letter 3

Andrew Monteith

29 Apr 2025

Dear Professor Chen,

We are pleased to inform you that your manuscript 'Caspase-1-licensed pyroptosis drives dsRNA-mediated necroptosis and dampens host defense against bacterial pneumonia' has been provisionally accepted for publication in PLOS Pathogens.

Before your manuscript can be formally accepted you will need to complete some formatting changes, which you will receive in a follow up email. A member of our team will be in touch with a set of requests.

Please note that your manuscript will not be scheduled for publication until you have made the required changes, so a swift response is appreciated.

IMPORTANT: The editorial review process is now complete. PLOS will only permit corrections to spelling, formatting or significant scientific errors from this point onwards. Requests for major changes, or any which affect the scientific understanding of your work, will cause delays to the publication date of your manuscript.

Should you, your institution's press office or the journal office choose to press release your paper, you will automatically be opted out of early publication. We ask that you notify us now if you or your institution is planning to press release the article. All press must be co-ordinated with PLOS.

Thank you again for supporting Open Access publishing; we are looking forward to publishing your work in PLOS Pathogens.

Best regards,

Andrew J Monteith, Ph.D.

Guest Editor

PLOS Pathogens

Matthew Wolfgang

Section Editor

PLOS Pathogens

Sumita Bhaduri-McIntosh

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0003-2946-9497

Michael Malim

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0002-7699-2064

***********************************************************

Reviewer Comments (if any, and for reference):

Acceptance letter

Andrew Monteith

Dear Professor Chen,

We are delighted to inform you that your manuscript, "Caspase-1-licensed pyroptosis drives dsRNA-mediated necroptosis and dampens host defense against bacterial pneumonia," has been formally accepted for publication in PLOS Pathogens.

We have now passed your article onto the PLOS Production Department who will complete the rest of the pre-publication process. All authors will receive a confirmation email upon publication.

The corresponding author will soon be receiving a typeset proof for review, to ensure errors have not been introduced during production. Please review the PDF proof of your manuscript carefully, as this is the last chance to correct any scientific or type-setting errors. Please note that major changes, or those which affect the scientific understanding of the work, will likely cause delays to the publication date of your manuscript. Note: Proofs for Front Matter articles (Pearls, Reviews, Opinions, etc...) are generated on a different schedule and may not be made available as quickly.

Soon after your final files are uploaded, the early version of your manuscript, if you opted to have an early version of your article, will be published online. The date of the early version will be your article's publication date. The final article will be published to the same URL, and all versions of the paper will be accessible to readers.

Thank you again for supporting open-access publishing; we are looking forward to publishing your work in PLOS Pathogens.

Best regards,

Sumita Bhaduri-McIntosh

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0003-2946-9497

Michael Malim

Editor-in-Chief

PLOS Pathogens

orcid.org/0000-0002-7699-2064

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. Lung neutrophils are critical for defense against E. coli pneumonia in Casp1-/- mice.

    (A and B) Respiratory rates and body temperatures of Casp1-/- mice (n = 11) and Casp1+/+ mice (n = 8) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each). (C) BALF protein concentrations in Casp1-/- mice (n = 11) and Casp1+/+ mice (n = 8) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each). (D) BALF bacterial burden in Casp1-/- mice (n = 11) and Casp1+/+ mice (n = 8) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each) was determined and is expressed as CFU/ml. (E and F) IL-1β and TNF-α levels in the BALF of Casp1-/- mice (n = 11) and Casp1+/+ mice (n = 8) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each) were measured by ELISAs. (G and H) Resident macrophages (SiglecF+Ly6C-F4/80+) and recruited monocyte (SiglecF-Ly6C+F4/80+) in the BALF of Casp1-/- mice (n = 11) and Casp1+/+ mice (n = 8) at 12 hours after E. coli infection or intratracheal PBS instillation (n = 5 each) were counted by flow cytometry. (I) Total cellular ROS (determined by CellROX) and mitochondrial ROS (determined by MitoSOX staining) in neutrophils and macrophages from the BALF of Casp1-/- mice and Casp1+/+ mice at 12 hours after pulmonary E. coli infection were analyzed by flow cytometry (n = 7). (J) Pathway enrichment of expressed genes in neutrophils from the BALF of Casp1-/- mice and Casp1+/+ neutrophils at 12 hours after pulmonary E. coli infection was analyzed using RNA-seq transcriptomes (n = 2). The data are shown as the means ± SDs (A-I). Statistical differences were determined by two-way ANOVA (A‒H), Student’s t test (I) and the parametric F test comparing nested linear models (J). *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. ns, not significant. CFU, colony-forming unit.

    (TIF)

    S2 Fig. Chemotaxis of neutrophils in the lung during pulmonary E. coli infection.

    BALF cells (A) and BALF neutrophils (B) from Casp1-/- mice (n = 6) and Casp1+/+ mice (n = 6) were counted at 4 hours after bacterial instillation. Peripheral white blood cells (C) and neutrophils (D) from Casp1-/- mice and Casp1+/+ mice were counted at 12 hours after E. coli challenge (n = 11 each) or PBS instillation (n = 5 each). BALF levels of CXCL1 (E) and CXCL2 (F) in Casp1-/- mice and Casp1+/+ mice were measured at 12 hours after E. coli challenge (n = 11 each) or PBS instillation (n = 5 each). (G) LDH levels in the BALF of Casp1-/- mice and Casp1+/+ mice at 12 hours after E. coli infection (n = 6) or intratracheal PBS instillation (n = 5). The data are shown as the means ± SDs in (A-G). Statistical differences were determined by Student’s t test (A and B) and one-way ANOVA (C, D, E, F and G). ***P < 0.001; ****P < 0.0001. ns, not significant.

    (TIF)

    ppat.1013167.s002.tif (426.1KB, tif)
    S3 Fig. TUNEL staining of lung sections during pulmonary E. coli infection.

    (A) TUNEL staining of lung sections from Casp1-/- mice and Casp1+/+ mice at 12 hours after E. coli infection and quantification of the TUNEL-positive area relative to the DAPI-stained area (n = 4). Scale bars, 100 μm. (B) Casp1+/+ and Casp1-/- mouse lungs were harvested at 12 hours after E. coli infection. TUNEL staining of lung sections was performed with different markers including the epithelial marker CD326, the endothelial marker CD31, the type II epithelial marker SPC, the macrophage marker F4/80, the neutrophil marker Ly-6G, and the monocyte marker CD14. Quantification of the percentage of TUNEL+ cells of different cell type (n = 3 biologically independent samples). Scale bars, 50 μm. The data are shown as the means ± SDs and are representative of 3 independent experiments. Statistical differences were determined by Student’s t test. *P < 0.05. HPF, high-power field.

    (TIF)

    ppat.1013167.s003.tif (1.9MB, tif)
    S4 Fig. p-MLKL staining of lung sections and BALF cells during pulmonary E. coli infection.

    (A) Casp1+/+ and Casp1-/- mouse lungs were harvested at 12 hours after E. coli infection. p-MLKL staining of lung sections with the neutrophil marker Ly6G was performed by immunofluorescence. Scale bars, 50 μm. (B) BALF cells from Casp1-/- mice and Casp1+/+ mice were fixed on slides, stained with PKH26 and p-MLKL antibodies and visualized by using confocal microscopy. Scale bars, 10 μm. The data are representative of 3 independent experiments.

    (TIF)

    ppat.1013167.s004.tif (1.3MB, tif)
    S5 Fig. Intervention with MLKL decreased neutrophil death and protected mice against E. coli pneumonia.

    (A) Representative images of lungs with H&E staining and quantification of lung injury scores in GW806742X (n = 5)- or control solvent (n = 6)-treated WT mice at 12 hours after pulmonary E. coli infection or intratracheal PBS instillation (n = 5 each). Scale bars, 100 µm. BALF protein concentration (B) and bacterial burden (C) in WT mice treated with GW806742X (n = 6) or control solvent (n = 6) at 12 hours after pulmonary E. coli infection or intratracheal PBS instillation (n = 5 each). (D) Representative images of lungs with HE staining and quantification of lung injury scores in Mlkl-/- (n = 9) or Mlkl+/+ (n = 6) mice at 12 hours after pulmonary E. coli infection. Scale bars, 100 µm. (E) Twelve hours after E. coli infection, mouse lungs were harvested from Mlkl-/- or Mlkl+/+ mice, and the bacterial burdens in whole-lung tissue were determined. The data are shown as the means ± SDs (A, B, C, D and E). Statistical differences were determined by Student’s t test (D and E) and two-way ANOVA (A, B and C). *P < 0.05; **P < 0.01; ****P < 0.0001. CFU, colony-forming unit.

    (TIF)

    ppat.1013167.s005.tif (1.6MB, tif)
    S6 Fig. Neutrophil necroptosis is independent of bacterial stimuli and inflammatory stimuli.

    (A) Proportion of PI+ neutrophils from Casp1-/- and Casp1+/+ mice after stimulation with LPS, live E. coli, heat-inactivated E. coli or PBS (n = 3). (B) Immunoblot analysis of cell death-related proteins in neutrophils stimulated with LPS, live E. coli or heat-inactivated E. coli. (C) Proportion of PI+ neutrophils from WT mice treated with IL-1β, TNF-α, HMGB-1 or control solvent at the indicated concentrations (n = 3). (D and E) Survival of WT mice intraperitoneally (n = 5 each) or intratracheally (n = 6 each) pretreated with the IL-1β antagonist anakinra or saline after pulmonary E. coli infection. (F) Survival of WT mice intraperitoneally (n = 6 each) pretreated with the anti-IL-1β antibody or control IgG antibody after pulmonary E. coli infection. The data are shown as the means ± SDs in (A and C) and are representative of 3 independent experiments in (B). Statistical differences were determined by the Mantel‒Cox test (D, E and F). ns, not significant.

    (TIF)

    ppat.1013167.s006.tif (493.4KB, tif)
    S7 Fig. Pyroptotic epithelial cells trigger neutrophil necroptosis.

    (A) Flow cytometry gating strategy for identifying caspase-1-activated cells in mouse lungs via FLICA staining. (B) MLE-12 epithelial cells were stimulated with E. coli, and then, the cell lysate and culture supernatant were collected and immunoblotted for pyroptosis-related proteins. (C and D) Casp1-/- neutrophils and Mlkl-/- neutrophils were pretreated with GSK872, GW806742X or control solvent and then stimulated with pyroptotic supernatant (pyrop. sup) or control medium, and cell death was assayed by flow cytometry (n = 3). (E) WT neutrophils were pretreated with NEC-1, Z-VAD or control solvent and then stimulated with pyrop. sup or control medium, and cell death was assayed by flow cytometry (n = 3). The data are shown as the means ± SDs in (C, D and E) and are representative of 3 independent experiments in (B). Statistical differences were determined by two-way ANOVA (C and D) or one-way ANOVA (E). ****P < 0.0001.

    (TIF)

    ppat.1013167.s007.tif (1.1MB, tif)
    S8 Fig. Pyroptosis-released dsRNAs induce neutrophil necroptosis via ZBP1 during pulmonary E. coli infection.

    The total BALF cell number (A), protein concentration (B) and bacterial burden (C) were measured in the mice pretreated with the anti-dsRNA antibody (n = 9) or the IgG control antibody (n = 10) at 12 hours after pulmonary E. coli infection. (D) Representative images of lungs with HE staining and quantification of lung injury scores in anti-dsRNA antibody (n = 6)- or control IgG (n = 6)-treated WT mice at 12 hours after pulmonary E. coli infection. Scale bars, 100 µm. (E‒G) Proportion of PI+ neutrophils in the BALF of Casp1-/- (L), Mlkl-/- (M) and Zbp1-/- (N) mice treated with an anti-dsRNA antibody (n = 6‒8) or an IgG control antibody (n = 7‒8) at 12 h after pulmonary E. coli infection. (H) Representative images of lungs with HE staining and quantification of lung injury scores in Zbp1-/- (n = 6) or Zbp1+/+ littermate (n = 6) mice at 12 hours after pulmonary E. coli infection. Scale bars, 100 µm. The data are shown as the means ± SDs. Statistical differences were determined by Student’s t test. *P < 0.05; **P < 0.01. ns, not significant. CFU, colony-forming unit.

    (TIF)

    ppat.1013167.s008.tif (1.4MB, tif)
    S9 Fig. TLR3 may not be involved in the neutrophil necroptosis pathway during pulmonary E. coli infection.

    (A) Western blot analysis of BALF neutrophils and spleen cells derived from E. coli-infected Casp1-/- and Casp1+/+ mice at 12 hours. (B-E) Twelve hours after E. coli infection, the proportions of PI+ neutrophils (B), neutrophil numbers (C), total leukocytes (D) and bacterial burdens (E) in the BALF of Tlr3+/+ mice (n = 8) and Tlr3-/- mice (n = 7) were determined. The data are shown as the means ± SDs (B-E) and are representative of 2 independent experiments in (A). Statistical differences were determined by Student’s t test (B-D). ns, not significant. CFU, colony-forming unit.

    (TIF)

    ppat.1013167.s009.tif (239.5KB, tif)
    S1 Data. Quantitative data used in calculations corresponding to primary figures.

    Numeric values used to generate graphs, means, and standard deviations for primary figures are included on tabs, with each tab indicating the relevant figure panel.

    (XLSX)

    ppat.1013167.s010.xlsx (49.9KB, xlsx)
    S2 Data. Quantitative data used in calculations corresponding to supplemental figures.

    Numeric values used to generate graphs, means, and standard deviations for supplemental figures are included on tabs, with each tab indicating the relevant figure panel.

    (XLSX)

    ppat.1013167.s011.xlsx (60KB, xlsx)
    Attachment

    Submitted filename: Qinyu Luo et al manuscript review .docx

    ppat.1013167.s012.docx (22KB, docx)
    Attachment

    Submitted filename: point to point response letter1121.pdf

    ppat.1013167.s015.pdf (7.5MB, pdf)
    Attachment

    Submitted filename: Qinyu Luo et al review comments.docx

    ppat.1013167.s014.docx (20.5KB, docx)
    Attachment

    Submitted filename: Response to Reviewers.docx

    ppat.1013167.s016.docx (3.4MB, docx)
    Attachment

    Submitted filename: Point to point response-chen-20250411.pdf

    ppat.1013167.s017.pdf (496KB, pdf)

    Data Availability Statement

    The authors confirm that all data underlying the findings are fully available without restriction. All relevant data are within the paper and its Supporting Information files.


    Articles from PLOS Pathogens are provided here courtesy of PLOS

    RESOURCES