STING agonist diABZI induces PANoptosis and DNA mediated acute respiratory distress syndrome (ARDS) - PubMed (original) (raw)

STING agonist diABZI induces PANoptosis and DNA mediated acute respiratory distress syndrome (ARDS)

Yasmine Messaoud-Nacer et al. Cell Death Dis. 2022.

Abstract

Stimulator of interferon genes (STING) contributes to immune responses against tumors and may control viral infection including SARS-CoV-2 infection. However, activation of the STING pathway by airway silica or smoke exposure leads to cell death, self-dsDNA release, and STING/type I IFN dependent acute lung inflammation/ARDS. The inflammatory response induced by a synthetic non-nucleotide-based diABZI STING agonist, in comparison to the natural cyclic dinucleotide cGAMP, is unknown. A low dose of diABZI (1 µg by endotracheal route for 3 consecutive days) triggered an acute neutrophilic inflammation, disruption of the respiratory barrier, DNA release with NET formation, PANoptosis cell death, and inflammatory cytokines with type I IFN dependent acute lung inflammation. Downstream upregulation of DNA sensors including cGAS, DDX41, IFI204, as well as NLRP3 and AIM2 inflammasomes, suggested a secondary inflammatory response to dsDNA as a danger signal. DNase I treatment, inhibition of NET formation together with an investigation in gene-deficient mice highlighted extracellular DNA and TLR9, but not cGAS, as central to diABZI-induced neutrophilic response. Therefore, activation of acute cell death with DNA release may lead to ARDS which may be modeled by diABZI. These results show that airway targeting by STING activator as a therapeutic strategy for infection may enhance lung inflammation with severe ARDS. STING agonist diABZI induces neutrophilic lung inflammation and PANoptosis A, Airway STING priming induce a neutrophilic lung inflammation with epithelial barrier damage, double-stranded DNA release in the bronchoalvelolar space, cell death, NETosis and type I interferon release. B, 1. The diamidobenzimidazole (diABZI), a STING agonist is internalized into the cytoplasm through unknown receptor and induce the activation and dimerization of STING followed by TBK1/IRF3 phosporylation leading to type I IFN response. STING activation also leads to NF-kB activation and the production of pro-inflammatory cytokines TNFα and IL-6. 2. The activation of TNFR1 and IFNAR1 signaling pathway results in ZBP1 and RIPK3/ASC/CASP8 activation leading to MLKL phosphorylation and necroptosis induction. 3. This can also leads to Caspase-3 cleavage and apoptosis induction. 4. Self-dsDNA or mtDNA sensing by NLRP3 or AIM2 induces inflammsome formation leading to Gasdermin D cleavage enabling Gasdermin D pore formation and the release mature IL-1β and pyroptosis. NLRP3 inflammasome formation can be enhanced by the ZBP1/RIPK3/CASP8 complex. 5. A second signal of STING activation with diABZI induces cell death and the release of self-DNA which is sensed by cGAS and form 2'3'-cGAMP leading to STING hyper activation, the amplification of TBK1/IRF3 and NF-kB pathway and the subsequent production of IFN-I and inflammatory TNFα and IL-6. This also leads to IFI204 and DDX41 upregulation thus, amplifying the inflammatory loop. The upregulation of apoptosis, pyroptosis and necroptosis is indicative of STING-dependent PANoptosis.

© 2022. The Author(s).

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interests

Figures

None

STING agonist diABZI induces neutrophilic lung inflammation and PANoptosis A, Airway STING priming induce a neutrophilic lung inflammation with epithelial barrier damage, double-stranded DNA release in the bronchoalvelolar space, cell death, NETosis and type I interferon release. B, 1. The diamidobenzimidazole (diABZI), a STING agonist is internalized into the cytoplasm through unknown receptor and induce the activation and dimerization of STING followed by TBK1/IRF3 phosporylation leading to type I IFN response. STING activation also leads to NF-kB activation and the production of pro-inflammatory cytokines TNFα and IL-6. 2. The activation of TNFR1 and IFNAR1 signaling pathway results in ZBP1 and RIPK3/ASC/CASP8 activation leading to MLKL phosphorylation and necroptosis induction. 3. This can also leads to Caspase-3 cleavage and apoptosis induction. 4. Self-dsDNA or mtDNA sensing by NLRP3 or AIM2 induces inflammsome formation leading to Gasdermin D cleavage enabling Gasdermin D pore formation and the release mature IL-1β and pyroptosis. NLRP3 inflammasome formation can be enhanced by the ZBP1/RIPK3/CASP8 complex. 5. A second signal of STING activation with diABZI induces cell death and the release of self-DNA which is sensed by cGAS and form 2′3′-cGAMP leading to STING hyper activation, the amplification of TBK1/IRF3 and NF-kB pathway and the subsequent production of IFN-I and inflammatory TNFα and IL-6. This also leads to IFI204 and DDX41 upregulation thus, amplifying the inflammatory loop. The upregulation of apoptosis, pyroptosis and necroptosis is indicative of STING-dependent PANoptosis.

Fig. 1

Fig. 1. Endotracheal cGAMP induces neutrophilic inflammation, protein extravasation, and self-dsDNA release in the airways.

A cGAMP (1, 3, or 10 µg, i.t.) or saline were administered daily in WT mice for 3 consecutive days, and parameters analyzed on day 4. B Concentration of CXCL1/KC in bronchoalveolar lavage fluid (BALF) determined by ELISA. C Neutrophils counts in BAL. D, E Myeloperoxidase (MPO) concentration in BALF (D) and in the lung (E) determined by ELISA. F Concentration of proteins in BALF. G Concentration of extracellular dsDNA in the acellular fraction of BALF. H, I. Concentration of IFNα and IFNβ in BALF determined by Luminex immunoassay. JN. Lung tissue histology PAS staining (J), with pathology scoring of the presence of epithelial injury (K), peribronchial infiltration of inflammatory cells (L), alveolitis (M), and emphysema (N). Bars, left panels: 2.5 mm, right panels: 250 µm. O Immunoblots of STING pathway activation in the lung tissue in response to cGAMP, including STING dimer, STING, phospho-TBK1, TBK1, IRF3, cGAS, phosphorylated NF-κB p65 (p-p65- NF-κB), and p65 NF-κB, with β-actin as a reference. Graph data were presented as mean ± SEM with n = 6 mice per group. Each point represents an individual mouse. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 (Nonparametric Kruskal–Wallis with Dunn post test).

Fig. 2

Fig. 2. cGAMP-induced type I IFN macrophage response and DNA sensor expression.

Bone marrow-derived macrophages from wild-type (AL) and STING−/− (MQ) mice were unstimulated or stimulated with cGAMP (14 µM) for 4 or 16 h as indicated. AC Protein concentrations of IFNα (A) and IFNβ (B) in macrophage culture supernatant determined by multiplex immunoassay, and of CXCL10 quantified by ELISA (C). DJ Ifnb, Mb21d1, Aim2, Ifi204, Ddx41, Nlrp3, and Tmem173 transcripts measured by real-time PCR. K, L CXCL1 (K) and IL-10 (L) protein concentration quantified by ELISA. MO Ifnβ1, Ifnα2, and Ifnα4 transcripts measured by real-time PCR in macrophages from WT and STING−/−. P, Q Comparison of protein levels of IFNα (P) and IFNβ (Q), in the supernatant of WT and STING−/− macrophages. Data were presented as mean ± SEM with n = 4 mice. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 (AL Nonparametric Kruskal–Wallis with Dunn post test. MQ Two-way Anova with Tukey post test).

Fig. 3

Fig. 3. DiABZI-induced type I IFN response in human alveolar epithelial cells.

AG Bone marrow-derived macrophages (BMDM) from wild-type and STING−/− mice were unstimulated or stimulated with cGAMP (14 µM) or diABZI (0.3 or 1 µM) for 16 h. Protein concentrations of IFNα (A) and IFNβ (B) in macrophage culture supernatant determined by multiplex immunoassay, of CXCL10 (C), IL-6 (D), TNFα (E), CXCL1 (F), and IL-10 (G) quantified by ELISA. H, I Immunoblots of STING axis (H), including phospho-STING, STING, phospho-TBK1, TBK1, phospho-STAT1, STAT1, DNA damage as revealed by phospho-γH2AX, γH2AX and of cell death axis (I) including Caspase 3, cleaved Gasdermin D, Gasdermin D, phospho-MLKL, MLKL, and ZBP1 in WT and STING −/− macrophages, with β-actin as a reference. JL Human alveolar epithelial cells (hAEC) were unstimulated or stimulated with diABZI (1, 3, or 10 µM) for 24 h. Protein levels of IFNβ (J), IL-1β (K), and IL-8 (L) were released in the culture supernatant. M Immunoblots of STING axis, including phospho-STING, STING, phospho-TBK1, cGAS, and cell death markers cleaved Caspase 3, Caspase 3, and phospho-γH2AX with β-actin as a reference. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, (AG; two-way Anova with Tukey post test. JL; nonparametric Kruskal–Wallis with Dunn post test). Data were presented as mean ± SEM and are representative of two independent experiments with n = 4 (AG) and n = 3 (JL) independent cultures.

Fig. 4

Fig. 4. STING agonist diABZI triggers neutrophilic airway inflammation.

A STING agonist diABZI (0.01, 0.1, or 1 µg, i.t.) DMSO (0.25%) vehicle or saline were administered daily in WT mice for 3 consecutive days and parameters were analyzed on day 4. B Concentration of CXCL1/KC in the bronchoalveolar lavage fluid (BALF) determined by ELISA. C Neutrophils counts in BAL. D Concentration of myeloperoxidase (MPO) in BALF determined by ELISA. E Concentration of proteins in BALF. F Concentration of extracellular dsDNA in the acellular fractions of BAL. G, H Mitochondrial DNA (mtDNA) (G) and nuclear DNA (nDNA) (H) in the BALF after diABZI or cGAMP administration. IM IFNα (I) and IFNβ (J) determined by multiplex immunoassay, and CXCL10 (K), IL-6 (L), and TNFα (M) quantified by ELISA in the BALF. N Visualization of NETs in BAL and lung with the staining of DNA dye DAPI (cyan), MPO (green), and citrullinated Histone 3 (red). Bars, 20 µm. O, P Quantification of Cit-H3 staining intensity (O) and MPO staining intensity (P) in the lung. Q, R Correlation between MPO and Cit-H3 after diABZI administration at 0.1 µg (Q) and 1 µg (R). SW Lung tissue histology PAS staining (S), with pathology scoring of the presence of epithelial injury (T), peribronchial infiltration of inflammatory cells (U), alveolitis (V), and emphysema (W). Bars, left panels: 2.5 mm, right panels: 250 µm. Graph data were presented as mean ± SEM with n = 5–6 mice per group. Each point represents an individual mouse. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 (Nonparametric Kruskal–Wallis with Dunn post test).

Fig. 4

Fig. 4. STING agonist diABZI triggers neutrophilic airway inflammation.

A STING agonist diABZI (0.01, 0.1, or 1 µg, i.t.) DMSO (0.25%) vehicle or saline were administered daily in WT mice for 3 consecutive days and parameters were analyzed on day 4. B Concentration of CXCL1/KC in the bronchoalveolar lavage fluid (BALF) determined by ELISA. C Neutrophils counts in BAL. D Concentration of myeloperoxidase (MPO) in BALF determined by ELISA. E Concentration of proteins in BALF. F Concentration of extracellular dsDNA in the acellular fractions of BAL. G, H Mitochondrial DNA (mtDNA) (G) and nuclear DNA (nDNA) (H) in the BALF after diABZI or cGAMP administration. IM IFNα (I) and IFNβ (J) determined by multiplex immunoassay, and CXCL10 (K), IL-6 (L), and TNFα (M) quantified by ELISA in the BALF. N Visualization of NETs in BAL and lung with the staining of DNA dye DAPI (cyan), MPO (green), and citrullinated Histone 3 (red). Bars, 20 µm. O, P Quantification of Cit-H3 staining intensity (O) and MPO staining intensity (P) in the lung. Q, R Correlation between MPO and Cit-H3 after diABZI administration at 0.1 µg (Q) and 1 µg (R). SW Lung tissue histology PAS staining (S), with pathology scoring of the presence of epithelial injury (T), peribronchial infiltration of inflammatory cells (U), alveolitis (V), and emphysema (W). Bars, left panels: 2.5 mm, right panels: 250 µm. Graph data were presented as mean ± SEM with n = 5–6 mice per group. Each point represents an individual mouse. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 (Nonparametric Kruskal–Wallis with Dunn post test).

Fig. 5

Fig. 5. Airway diABZI induces lung tissue PANoptosis and damage and STING pathway activation.

diABZI (0.01, 0.1, or 1 µg, i.t.) or saline were administered daily in WT mice for 3 consecutive days as in Fig. 4 and parameters analyzed on day 4. A Immunoblots of cleaved Caspase 3, Caspase 3, cleaved Gasdermin D, Gasdermin D, phospho-MLKL, MLKL, phospho-γH2AX, citrullinated Histone 3 (Cit-H3), and ZBP1 with β-actin as a reference. B Immunoblot quantification of cleaved Caspase 3, Caspase 3, cleaved Gasdermin D, Gasdermin D, pMLKL, MLKL, phospho-γH2AX, citrullinated Histone 3 (Cit-H3), and ZBP1, normalized to b-actin. C Confocal microscopy showing Caspase 8 (green), ASC (red), RIPK3 (far-red/turquoise blue), and DNA dye DAPI (cyan) in BAL cells of mice exposed to saline or diABZI 1 µg, showing the colocalization of PANoptosome components. Bars, 2 µm. D Immunoblots of STING axis in the lung of WT mice including STING and STING dimer, cGAS, NLRP3, IFI204, DDX41 with b-actin as reference. E Immunoblot quantification of STING and STING dimer, cGAS, NLRP3, IFI204, and DDX41 normalized to β-actin. F Mb21d1, Tmem173, Aim2, Ifi204, Nlrp3, Ddx41, and Cxcl10 transcripts were measured by real-time PCR. *p < 0.05, **p < 0.01, ***p < 0.001. (Nonparametric Kruskal–Wallis test followed by Dunn post test). Graph data from real-time PCR are presented as mean ± SEM with n = 4–6 mice per group. Immunoblots representative of n = 2 samples from two independent experiments, quantified in bar graphs with n = 2.

Fig. 5

Fig. 5. Airway diABZI induces lung tissue PANoptosis and damage and STING pathway activation.

diABZI (0.01, 0.1, or 1 µg, i.t.) or saline were administered daily in WT mice for 3 consecutive days as in Fig. 4 and parameters analyzed on day 4. A Immunoblots of cleaved Caspase 3, Caspase 3, cleaved Gasdermin D, Gasdermin D, phospho-MLKL, MLKL, phospho-γH2AX, citrullinated Histone 3 (Cit-H3), and ZBP1 with β-actin as a reference. B Immunoblot quantification of cleaved Caspase 3, Caspase 3, cleaved Gasdermin D, Gasdermin D, pMLKL, MLKL, phospho-γH2AX, citrullinated Histone 3 (Cit-H3), and ZBP1, normalized to b-actin. C Confocal microscopy showing Caspase 8 (green), ASC (red), RIPK3 (far-red/turquoise blue), and DNA dye DAPI (cyan) in BAL cells of mice exposed to saline or diABZI 1 µg, showing the colocalization of PANoptosome components. Bars, 2 µm. D Immunoblots of STING axis in the lung of WT mice including STING and STING dimer, cGAS, NLRP3, IFI204, DDX41 with b-actin as reference. E Immunoblot quantification of STING and STING dimer, cGAS, NLRP3, IFI204, and DDX41 normalized to β-actin. F Mb21d1, Tmem173, Aim2, Ifi204, Nlrp3, Ddx41, and Cxcl10 transcripts were measured by real-time PCR. *p < 0.05, **p < 0.01, ***p < 0.001. (Nonparametric Kruskal–Wallis test followed by Dunn post test). Graph data from real-time PCR are presented as mean ± SEM with n = 4–6 mice per group. Immunoblots representative of n = 2 samples from two independent experiments, quantified in bar graphs with n = 2.

Fig. 6

Fig. 6. DNase I treatment or NET inhibition reduce diABZI-induced neutrophilic airway inflammation.

A diABZI (0.1 or 1 µg, i.t.) were administered with DNase I (50 µg/mouse, i.t.) daily in WT mice for 3 consecutive days and parameters were analyzed on day 4. B Extracellular dsDNA in acellular fraction of BAL. C Neutrophils counts in BAL. D Concentration of CXCL10 in BALF, determined by ELISA. E, F IFNα (E) and IFNβ (F) in BALF are determined by multiplex immunoassay. G Visualization of NETs with staining of DNA (DAPI, cyan), MPO (green), and Cit-H3 (red) in cells from BAL. Bars 20 µm, magnification x20 upper panels, x40 middle panels, x100 lower panels. H Visualization of PANoptosis with stainings of ZBP1 (vert) and DNA (DAPI, cyan) in cells from lung and BAL. Bars 20 µm, magnification x20 (Lung), x100 (BAL). I, J Annexin V/PI flow cytometry analysis pre-gated on singlet cells, and CD45+ (leukocytes) or CD45+Ly6G+CD11C− (neutrophils). K diABZI (1 µg, i.t.) was administered alone or with Cl-amidine (200 µg/mouse, i.p.) daily in WT mice for 3 consecutive days and parameters were analyzed on day 4. L Neutrophils counts in BAL. M Visualization of NETs with staining of DNA (DAPI, cyan), MPO (green), and Cit-H3 (red) in cells from BAL; Bars 20 µm, magnification x20 upper panels, x40 lower panels. N Immunoblots of Cit-H3, STING, cleaved caspase 3, caspase 3, cleaved Gasdermin D, Gasdermin D, phospho-MLKL, MLKL, phospho-γH2AX, and ZBP1 in the lung of WT mice exposed to diABZI alone or treated with Cl-amidine, with β-actin as reference. O Immunoblot quantification of Cit-H3, STING, cleaved caspase 3, caspase 3, cleaved GSDMD, GSDMD, phospho-γH2AX and ZBP1 normalized to β-actin, and phospho-MLKL normalized to MLKL. P Confocal microscopy showing Caspase 8 (green), ASC (red), RIPK3 (far-red/turquoise blue), and DNA dye DAPI (cyan) in BAL from mice challenged with diABZI at 1 µg and treated with Cl-amidine or saline. PANoptosome formation is illustrated by colocalization of the components of PANoptosome in the merged image (indicated by arrowheads). Bars, 2 µm. Q Concentration of extracellular dsDNA in the acellular fraction of BALF. R Lactate dehydrogenase (LDH) quantification in the acellular fraction of BALF. Graph data were presented as mean ± SEM with n = 5–8 mice/group. Each point represents an individual mouse. *p < 0.05, **p < 0.01, ***p < 0.001. (Nonparametric Kruskal–Wallis test followed by Dunn post test).

Fig. 6

Fig. 6. DNase I treatment or NET inhibition reduce diABZI-induced neutrophilic airway inflammation.

A diABZI (0.1 or 1 µg, i.t.) were administered with DNase I (50 µg/mouse, i.t.) daily in WT mice for 3 consecutive days and parameters were analyzed on day 4. B Extracellular dsDNA in acellular fraction of BAL. C Neutrophils counts in BAL. D Concentration of CXCL10 in BALF, determined by ELISA. E, F IFNα (E) and IFNβ (F) in BALF are determined by multiplex immunoassay. G Visualization of NETs with staining of DNA (DAPI, cyan), MPO (green), and Cit-H3 (red) in cells from BAL. Bars 20 µm, magnification x20 upper panels, x40 middle panels, x100 lower panels. H Visualization of PANoptosis with stainings of ZBP1 (vert) and DNA (DAPI, cyan) in cells from lung and BAL. Bars 20 µm, magnification x20 (Lung), x100 (BAL). I, J Annexin V/PI flow cytometry analysis pre-gated on singlet cells, and CD45+ (leukocytes) or CD45+Ly6G+CD11C− (neutrophils). K diABZI (1 µg, i.t.) was administered alone or with Cl-amidine (200 µg/mouse, i.p.) daily in WT mice for 3 consecutive days and parameters were analyzed on day 4. L Neutrophils counts in BAL. M Visualization of NETs with staining of DNA (DAPI, cyan), MPO (green), and Cit-H3 (red) in cells from BAL; Bars 20 µm, magnification x20 upper panels, x40 lower panels. N Immunoblots of Cit-H3, STING, cleaved caspase 3, caspase 3, cleaved Gasdermin D, Gasdermin D, phospho-MLKL, MLKL, phospho-γH2AX, and ZBP1 in the lung of WT mice exposed to diABZI alone or treated with Cl-amidine, with β-actin as reference. O Immunoblot quantification of Cit-H3, STING, cleaved caspase 3, caspase 3, cleaved GSDMD, GSDMD, phospho-γH2AX and ZBP1 normalized to β-actin, and phospho-MLKL normalized to MLKL. P Confocal microscopy showing Caspase 8 (green), ASC (red), RIPK3 (far-red/turquoise blue), and DNA dye DAPI (cyan) in BAL from mice challenged with diABZI at 1 µg and treated with Cl-amidine or saline. PANoptosome formation is illustrated by colocalization of the components of PANoptosome in the merged image (indicated by arrowheads). Bars, 2 µm. Q Concentration of extracellular dsDNA in the acellular fraction of BALF. R Lactate dehydrogenase (LDH) quantification in the acellular fraction of BALF. Graph data were presented as mean ± SEM with n = 5–8 mice/group. Each point represents an individual mouse. *p < 0.05, **p < 0.01, ***p < 0.001. (Nonparametric Kruskal–Wallis test followed by Dunn post test).

Fig. 6

Fig. 6. DNase I treatment or NET inhibition reduce diABZI-induced neutrophilic airway inflammation.

A diABZI (0.1 or 1 µg, i.t.) were administered with DNase I (50 µg/mouse, i.t.) daily in WT mice for 3 consecutive days and parameters were analyzed on day 4. B Extracellular dsDNA in acellular fraction of BAL. C Neutrophils counts in BAL. D Concentration of CXCL10 in BALF, determined by ELISA. E, F IFNα (E) and IFNβ (F) in BALF are determined by multiplex immunoassay. G Visualization of NETs with staining of DNA (DAPI, cyan), MPO (green), and Cit-H3 (red) in cells from BAL. Bars 20 µm, magnification x20 upper panels, x40 middle panels, x100 lower panels. H Visualization of PANoptosis with stainings of ZBP1 (vert) and DNA (DAPI, cyan) in cells from lung and BAL. Bars 20 µm, magnification x20 (Lung), x100 (BAL). I, J Annexin V/PI flow cytometry analysis pre-gated on singlet cells, and CD45+ (leukocytes) or CD45+Ly6G+CD11C− (neutrophils). K diABZI (1 µg, i.t.) was administered alone or with Cl-amidine (200 µg/mouse, i.p.) daily in WT mice for 3 consecutive days and parameters were analyzed on day 4. L Neutrophils counts in BAL. M Visualization of NETs with staining of DNA (DAPI, cyan), MPO (green), and Cit-H3 (red) in cells from BAL; Bars 20 µm, magnification x20 upper panels, x40 lower panels. N Immunoblots of Cit-H3, STING, cleaved caspase 3, caspase 3, cleaved Gasdermin D, Gasdermin D, phospho-MLKL, MLKL, phospho-γH2AX, and ZBP1 in the lung of WT mice exposed to diABZI alone or treated with Cl-amidine, with β-actin as reference. O Immunoblot quantification of Cit-H3, STING, cleaved caspase 3, caspase 3, cleaved GSDMD, GSDMD, phospho-γH2AX and ZBP1 normalized to β-actin, and phospho-MLKL normalized to MLKL. P Confocal microscopy showing Caspase 8 (green), ASC (red), RIPK3 (far-red/turquoise blue), and DNA dye DAPI (cyan) in BAL from mice challenged with diABZI at 1 µg and treated with Cl-amidine or saline. PANoptosome formation is illustrated by colocalization of the components of PANoptosome in the merged image (indicated by arrowheads). Bars, 2 µm. Q Concentration of extracellular dsDNA in the acellular fraction of BALF. R Lactate dehydrogenase (LDH) quantification in the acellular fraction of BALF. Graph data were presented as mean ± SEM with n = 5–8 mice/group. Each point represents an individual mouse. *p < 0.05, **p < 0.01, ***p < 0.001. (Nonparametric Kruskal–Wallis test followed by Dunn post test).

Fig. 7

Fig. 7. STING is the major sensor of diABZI-induced lung inflammation.

DiABZI (0.1 or 1 µg, i.t.) or saline were administered daily in STING−/−, cGAS−/−, NLRP3−/−, IFNAR−/−, AIM2−/−, TLR9−/−, and WT mice for 3 consecutive days and parameters analyzed on day 4. A–D Neutrophils (A), concentrations of dsDNA (B), IFNα (C), and CXCL10 (D) in BALF in WT and STING−/− mice. E–H. Neutrophils (E), dsDNA (F), IFNα (G), and CXCL10 (H) concentration in BALF in WT and cGAS−/− mice. IL Neutrophils (I), dsDNA (J), IFNα (K), and CXCL10 (L) concentration in BALF in WT and NLRP3−/− and IFNAR−/− mice. M–P Neutrophils (M), dsDNA (N), IFNα (O), and CXCL10 (P) concentration in BALF in WT and AIM2−/− mice. R–U Neutrophils (R), dsDNA (S), IFNα (T), and CXCL10 (U) concentration in BALF in WT and TLR9−/− mice. Graph data were presented as mean ± SEM with n = 4–10 mice/group, each point representing an individual mouse. *p < 0.05, **p < 0.01, ***p < 0.001. (Nonparametric Kruskal–Wallis test followed by Dunn post test).

Similar articles

Cited by

References

    1. Motedayen Aval L, Pease JE, Sharma R, Pinato DJ. Challenges and opportunities in the clinical development of STING agonists for cancer immunotherapy. J Clin Med. 2020;9:3323. - PMC - PubMed
    1. Woo SR, Fuertes MB, Corrales L, Spranger S, Furdyna MJ, Leung MY, et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity. 2014;41:830–42. doi: 10.1016/j.immuni.2014.10.017. - DOI - PMC - PubMed
    1. Le Naour J, Zitvogel L, Galluzzi L, Vacchelli E, Kroemer G. Trial watch: STING agonists in cancer therapy. Oncoimmunology. 2020;9:1777624. doi: 10.1080/2162402X.2020.1777624. - DOI - PMC - PubMed
    1. Humphries F, Shmuel-Galia L, Jiang Z, Wilson R, Landis P, Ng SL, et al. A diamidobenzimidazole STING agonist protects against SARS-CoV-2 infection. Sci Immunol. 2021;6:eabi9002. - PMC - PubMed
    1. Li M, Ferretti M, Ying B, Descamps H, Lee E, Dittmar M, et al. Pharmacological activation of STING blocks SARS-CoV-2 infection. Sci Immunol. 2021;6:eabi9007. - PMC - PubMed

Publication types

MeSH terms

Substances

Grants and funding

LinkOut - more resources