Type I IFN exacerbates disease in tuberculosis-susceptible mice by inducing neutrophil-mediated lung inflammation and NETosis

Lúcia Moreira-Teixeira, Philippa J Stimpson, Evangelos Stavropoulos, Sabelo Hadebe, Probir Chakravarty, Marianna Ioannou, Iker Valle Aramburu, Eleanor Herbert, Simon L Priestnall, Alejandro Suarez-Bonnet, Jeremy Sousa, Kaori L Fonseca, Qian Wang, Sergo Vashakidze, Paula Rodríguez-Martínez, Cristina Vilaplana, Margarida Saraiva, Venizelos Papayannopoulos, Anne O'Garra, Lúcia Moreira-Teixeira, Philippa J Stimpson, Evangelos Stavropoulos, Sabelo Hadebe, Probir Chakravarty, Marianna Ioannou, Iker Valle Aramburu, Eleanor Herbert, Simon L Priestnall, Alejandro Suarez-Bonnet, Jeremy Sousa, Kaori L Fonseca, Qian Wang, Sergo Vashakidze, Paula Rodríguez-Martínez, Cristina Vilaplana, Margarida Saraiva, Venizelos Papayannopoulos, Anne O'Garra

Abstract

Tuberculosis (TB) is a leading cause of mortality due to infectious disease, but the factors determining disease progression are unclear. Transcriptional signatures associated with type I IFN signalling and neutrophilic inflammation were shown to correlate with disease severity in mouse models of TB. Here we show that similar transcriptional signatures correlate with increased bacterial loads and exacerbate pathology during Mycobacterium tuberculosis infection upon GM-CSF blockade. Loss of GM-CSF signalling or genetic susceptibility to TB (C3HeB/FeJ mice) result in type I IFN-induced neutrophil extracellular trap (NET) formation that promotes bacterial growth and promotes disease severity. Consistently, NETs are present in necrotic lung lesions of TB patients responding poorly to antibiotic therapy, supporting the role of NETs in a late stage of TB pathogenesis. Our findings reveal an important cytokine-based innate immune effector network with a central role in determining the outcome of M. tuberculosis infection.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. GM-CSF controls lung bacterial growth…
Fig. 1. GM-CSF controls lung bacterial growth and necrotic inflammation.
a Schematic of experimental design for GM-CSF blockade during M. tuberculosis infection: intraperitoneal (i.p.) injection of 1 mg anti-GM-CSF (αGM-CSF; open circles) or isotype control (labelled as Ctrl Ab; closed circles) monoclonal antibodies (mAbs) the day prior to aerosol infection with HN878 strain, followed by i.p. injections of 0.3 mg of αGM-CSF or Ctrl mAbs, twice weekly for 3 weeks. b Change in body weight at various days post-infection (n = 8 mice/group; mean ± SD). ***P < 0.0001. c Viable bacterial loads determined in the lungs at day 21 post-infection. **P = 0.0079. d Representative hematoxylin and eosin (H&E) staining of infected lung sections (n = 3 mice/group). Scale bars from top to bottom: 1 mm; 200 µm; 50 µm, 10 µm. e Relative lung lesion burden from H&E stained sections (Supplementary Data 1). 0 = no lesions, 1 = focal lesion, 2 = multiple focal lesions, 3 = one or more focal severe lesions, 4 = multiple focal lesions that are extensive and coalesce, 5 = extensive lesions that occupy the majority of the lung lobe. ***P < 0.0001. f Relative necrosis (left) and intra-alveolar necrotic debris (right) scores from H&E stained lung sections (Supplementary Data 1). 0 = not present, 1 = minimal, 2 = mild, 3 = moderate, 4 = marked. ***P < 0.0001. g Representative Ziehl–Neelsen staining of infected lung sections (n = 3 mice/group). Scale bars, 20 µm. Data representative (bd, g) or pooled (e, f) of five biological experiments. Represented is the mean ± SD; each dot represents an individual mouse: n = 5 mice/group (c) or n = 15 mice/group (e, f). Source data are provided as a Source Data file. Statistical analysis was performed using two-way ANOVA (b) or two-tailed Mann–Whitney test (c, e, f).
Fig. 2. GM-CSF controls transcriptional signatures associated…
Fig. 2. GM-CSF controls transcriptional signatures associated with TB pathogenesis.
WT mice were infected and/or treated as in Fig. 1a. Blood and lungs were collected from M. tuberculosis HN878-infected and uninfected mice for RNA-Seq analysis (n = 4–5 mice/group). a Blood modules of co-expressed genes derived using WGCNA from human TB datasets are shown for blood RNA-Seq datasets obtained from infected mice treated with αGM-CSF or Ctrl mAbs, compared to uninfected Ctrl Ab treated mice (Supplementary Data 3). c Lung modules of co-expressed genes derived using WGCNA from mouse lung disease modules are shown for lung RNA-Seq datasets obtained from infected mice treated with αGM-CSF or Ctrl mAbs, compared to uninfected Ctrl Ab treated mice (Supplementary Data 5). a, c Fold enrichment scores derived using QuSAGE are depicted, with red and blue indicating modules over- or under-abundant, compared to the controls. Colour intensity and size of the dots represents the degree of perturbation, indicated by the colour scale, with the largest dot representing the highest degree of perturbation within the plot. Only modules with fold enrichment scores with FDR P-value < 0.05 were considered significant and depicted here. Module name indicates biological processes associated with the genes within the module. b, d Boxplots depicting the module eigengene expression, i.e., the first principal component for all genes within the module, are shown for uninfected Ctrl Ab (light green) or αGM-CSF (light purple) and infected Ctrl Ab (dark green) or αGM-CSF (dark purple) treated groups (n = 4–5 mice/group), for (b) blood modules: Inflammasome/Granulocyte (HB3), Innate/hemopoietic mediators (HB5), Innate Immunity/PRR/C’/Granulocyte (HB8), Interferon/PRR (HB12) and Interferon/C’/Myeloid (HB23); and (d) lung modules: Type I IFN/Ifit/Oas (L5), Myeloid/granulocyte function (L10), IL-17 pathway/granulocytes (L11), Inflammation/IL-1 signalling/myeloid cells (L12), and Myeloid cells/Il1b/Tnf (L13). Gene expression profiles are shown using boxplots, where the upper and lower box limits show the interquartile range (limits of second and third quartiles), a thick horizontal bar within the box shows the median, whiskers show the minimum and maximum values, and each dot represents an individual mouse (n = 4–5 mice/group). C’, complement.; PRR, pathogen recognition receptor. GCC, glucocorticoid; K-channel, potassium channel; TM, transmembrane; Ubiq, ubiquitination.
Fig. 3. Disease exacerbation upon GM-CSF blockade…
Fig. 3. Disease exacerbation upon GM-CSF blockade is neutrophil-dependent and correlates with NETosis.
a GSEA of immune cell-type associated gene sets showing normalized enrichment scores (NES) for lung RNA-Seq data from infected αGM-CSF versus infected Ctrl Ab treated mice. b Heatmap showing relative expression of genes in the neutrophil-associated gene set for individual lung samples from uninfected Ctrl Ab (light green) or αGM-CSF (light purple) and infected Ctrl Ab (dark green) or αGM-CSF (dark purple) treated mice (n = 4–5 mice/group). Gene expression values were averaged and scaled across the row to indicate the number of standard deviations above (red) or below (blue) the mean, denoted as row Z-score. The dendrogram shows unsupervised hierarchical clustering of genes. c WT mice were infected and treated with Ctrl Ab (closed circles) or αGM-CSF (open circles) as in Fig. 1a. Lung cell suspensions were prepared and stained for the detection of myeloid cells (Supplementary Fig. 5). Cell numbers for neutrophils (CD11b+Ly6G+), Ly6C+ monocytes (CD11b+Ly6G−Ly6C+) and alveolar macrophages (CD11blowCD11c+) are shown. *P = 0.0317. d WT mice were infected and treated with Ctrl Ab (closed symbols) or αGM-CSF (open symbols) as in Fig. 1a with the exception that from day 7 post-infection, mice also received 0.2 mg of anti-Ly6G (αLy6G; squares) or isotype control (labelled as Isotype; circles) mAbs every other day by i.p. injection. Lung viable bacterial loads were determined. **P = 0.0079. e Representative images of lung sections stained with citrullinated histone H3 (citH3; red), MPO (green), and DAPI (blue). NETs are visualized by colocalization of citH3 and DAPI staining (merged images; n = 3 mice/group). Bottom rows show detail from the top rows for each group. Scale bars, 50 µm. f Percentage of NET area in the lung normalized to MPO positive signal. **P = 0.0019. Data representative of one (a, b), two (d) or five (c, e) biological experiments; or data pooled of three independent experiments (f). Represented is the mean ± SD (c, f) or mean ± SEM (d); each dot represents an individual mouse: n = 5 mice/group (c, d) or n = 9 mice/group (f). Source data are provided as a Source Data file. Statistical analysis was performed using two-tailed Mann–Whitney test.
Fig. 4. NETs are detected in lung…
Fig. 4. NETs are detected in lung lesions of TB-susceptible C3HeB/FeJ mice.
a C3HeB/FeJ mice were aerosol infected with M. tuberculosis HN878 treated by i.p. injection with 0.2 mg of anti-Ly6G (αLy6G; open circles) or isotype control (labelled as Isotype; closed circles) mAbs every other day, from day 7 post-infection until harvest on day 26 post-infection. Lung viable bacterial loads were determined. **P = 0.0079. Represented is the mean ± SD; each dot represents an individual mouse: n = 5 mice/group. Source data are provided as a Source Data file. Statistical analysis was performed using two-tailed Mann–Whitney test. b C3HeB/FeJ mice were aerosol infected with M. tuberculosis HN878 and their lungs harvested at day 35 post-infection. Formalin-fixed paraffin-embedded lung sections were stained with citrullinated histone H3 (citH3; red), MPO (green), and DAPI (blue). NETs are visualized by colocalization of citH3 and DAPI staining (merged image; n = 3 mice/group). Scale bars, 50 μm. Data from one experiment.
Fig. 5. NETs are detected in necrotic…
Fig. 5. NETs are detected in necrotic lung lesions of human pulmonary TB.
Formalin-fixed paraffin-embedded lung sections from a total of thirteen patients with pulmonary TB (1 section per TB patient; Supplementary Table 1) were labelled with antibodies specific for citrullinated histone H3 (citH3; red) MPO (green) and DAPI (blue). NETs, visualized by colocalization of citH3 and DAPI staining (merged images, left), are shown for 6 TB patients. Scale bars, 50 µm. (All thirteen slides were stained and scanned together. See also Supplementary Fig. 6a).
Fig. 6. IFNAR deletion abrogates disease exacerbation…
Fig. 6. IFNAR deletion abrogates disease exacerbation prompted by GM-CSF blockade during infection.
WT (circles) and Ifnar-/- (triangles) mice were infected and treated with Ctrl Ab (closed symbols) or αGM-CSF (open symbols) as in Fig. 1a. a Viable bacterial loads determined in the lungs at day 21 post-infection. **P < 0.0079. Represented is the mean ± SEM; each dot represents an individual mouse: n = 5 mice/group. Source data are provided as a Source Data file. Data representative of two biological experiments. Statistical analysis was performed using the two-tailed Mann–Whitney test. b Representative H&E staining of infected lung sections (n = 2–4 mice/group). Scale bars from top to bottom: 1 mm; 200 µm; 50 µm; 10 µm. c Relative lung lesion burden from H&E stained sections (Supplementary Data 1). 0 = no lesions, 1 = focal lesion, 2 = multiple focal lesions, 3 = one or more focal severe lesions, 4 = multiple focal lesions that are extensive and coalesce, 5 = extensive lesions that occupy the majority of the lung lobe. ***P < 0.0001. d Relative necrosis and intra-alveolar necrotic debris scores from H&E stained lung sections (Supplementary Data 1). 0 = not present, 1 = minimal, 2 = mild, 3 = moderate, 4 = marked. ***P < 0.0001; *P = 0.0294. c, d Data pooled from four biological experiments. Represented is the mean ± SD; each dot represents an individual mouse: n = 11–13 mice/group. Source data are provided as a Source Data file. Statistical analysis was performed using one-way ANOVA Tukey’s multiple comparisons test. e Representative Ziehl–Neelsen staining of infected lung sections (n = 2–4 mice/group). Scale bars, 20 µm. Data representative of four biological experiments (b, e).
Fig. 7. IFNAR deletion abrogates NET formation…
Fig. 7. IFNAR deletion abrogates NET formation prompted by GM-CSF blockade during infection.
WT (circles) and Ifnar−/− (triangles) mice were infected and treated with Ctrl Ab (closed symbols) or αGM-CSF (open symbols) as in Fig. 1a. a Representative images of lung sections stained with citrullinated histone H3 (citH3; red), MPO (green), and DAPI (blue). NETs are visualized by colocalization of citH3 and DAPI staining (merged images; n = 3 mice/group). Scale bars, 50 µm. Data representative of two biological experiments. b Percentage of NET area in the lung normalized to MPO positive signal. **P = 0.0022. Data pooled from two biological experiments. Represented is the mean ± SD; each dot represents an individual mouse: n = 6 mice/group. Source data are provided as a Source Data file. Statistical analysis was performed using the two-tailed Mann–Whitney test.
Fig. 8. Type I IFN activates neutrophils…
Fig. 8. Type I IFN activates neutrophils to promote bacterial growth.
Ifnarfl/fl MRP8-Cre negative (Creneg; circles) or positive (Crepos; squares) mice were infected and treated with Ctrl Ab (closed symbols) or αGM-CSF (open symbols) as in Fig. 1a. a Viable bacterial loads determined in the lungs at day 21 post-infection. **P < 0.0022. Data representative of two biological experiments. Represented is the mean ± SEM; each dot represents an individual mouse: n = 4–6 mice/group/experiment. b Representative images of lung sections stained with citrullinated histone H3 (citH3; red), MPO (green), and DAPI (blue). NETs are visualized by colocalization of citH3 and DAPI staining (merged images). Scale bars, 50 µm. Data representative of two biological experiments (n = 3–4 mice/group/experiment). c Percentage of NET area in the lung normalized to MPO positive signal. Data pooled from two biological experiments. Represented is the mean ± SD; each dot represents an individual mouse: n = 7–8 mice/group. Source data are provided as a Source Data file. Statistical analysis was performed using two-tailed Mann–Whitney test.
Fig. 9. IFNAR signalling is required for…
Fig. 9. IFNAR signalling is required for severity of lung NETosis in C3HeB/FeJ mice.
a Schematic of experimental design: C3HeB/FeJ mice were treated by intraperitoneal (i.p.) injection of 0.5 mg of anti-IFNAR (αIFNAR; open circles) or isotype control (labelled as Ctrl Ab; closed circles) mAbs the day prior low dose HN878 aerosol infection, followed by i.p. injections of 0.5 mg of αIFNAR or Ctrl mAbs, three times per weekly until harvest on day 26 post-infection. b Viable bacterial loads were determined. **P = 0.0079. c Representative hematoxylin and eosin (H&E) staining of infected lung sections (n = 3 mice/group). Scale bars from top to bottom: 1 mm; 200 µm; 50 µm, 10 µm. d Relative lung lesion burden from H&E stained sections (Supplementary Data 1). 0 = no lesions, 1 = focal lesion, 2 = multiple focal lesions, 3 = one or more focal severe lesions, 4 = multiple focal lesions that are extensive and coalesce, 5 = extensive lesions that occupy the majority of the lung lobe. *P = 0.0152. e Relative necrosis (left) and intra-alveolar necrotic debris (right) scores from H&E stained lung sections (Supplementary Data 1). 0 = not present, 1 = minimal, 2 = mild, 3 = moderate, 4 = marked. **P = 0.0023. f Representative Ziehl–Neelsen staining of infected lung sections. Scale bars, 20 µm. g Representative images of lung sections labelled with citrullinated histone H3 (citH3; red), MPO (green), and DAPI (blue). NETs are visualized by colocalization of citH3 and DAPI staining (merged images). Scale bars, 50 μm. h Percentage of NET area in the lung normalized to MPO positive signal. **P < 0.0012. b, c, f, g Data representative of two biological replicate experiments. d, e, h Data pooled from two biological experiments. Represented is the mean ± SD; each dot represents an individual mouse: n = 5 mice/group (b) or n = 6–7 mice/group (d, e, h). Source data are provided as a Source Data file. Statistical analysis was performed using the two-tailed Mann–Whitney test.

References

    1. WHO. Global Tuberculosis Report 2019. (World Health Organization, 2019).
    1. Cooper AM. Cell-mediated immune responses in tuberculosis. Annu. Rev. Immunol. 2009;27:393–422. doi: 10.1146/annurev.immunol.021908.132703.
    1. Mayer-Barber KD, Sher A. Cytokine and lipid mediator networks in tuberculosis. Immunol. Rev. 2015;264:264–275. doi: 10.1111/imr.12249.
    1. O’Garra A, et al. The immune response in tuberculosis. Annu. Rev. Immunol. 2013;31:475–527. doi: 10.1146/annurev-immunol-032712-095939.
    1. Singhania A, Wilkinson RJ, Rodrigue M, Haldar P, O’Garra A. The value of transcriptomics in advancing knowledge of the immune response and diagnosis in tuberculosis. Nat. Immunol. 2018;19:1159–1168. doi: 10.1038/s41590-018-0225-9.
    1. Berry MP, et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature. 2010;466:973–977. doi: 10.1038/nature09247.
    1. Moreira-Teixeira L, Mayer-Barber K, Sher A, O’Garra A. Type I interferons in tuberculosis: foe and occasionally friend. J. Exp. Med. 2018;215:1273–1285. doi: 10.1084/jem.20180325.
    1. Bloom CI, et al. Detectable changes in the blood transcriptome are present after two weeks of antituberculosis therapy. PLoS ONE. 2012;7:e46191. doi: 10.1371/journal.pone.0046191.
    1. Cliff JM, et al. Distinct phases of blood gene expression pattern through tuberculosis treatment reflect modulation of the humoral immune response. J. Infect. Dis. 2013;207:18–29. doi: 10.1093/infdis/jis499.
    1. Antonelli LR, et al. Intranasal Poly-IC treatment exacerbates tuberculosis in mice through the pulmonary recruitment of a pathogen-permissive monocyte/macrophage population. J. Clin. Invest. 2010;120:1674–1682. doi: 10.1172/JCI40817.
    1. Dorhoi A, et al. Type I IFN signaling triggers immunopathology in tuberculosis-susceptible mice by modulating lung phagocyte dynamics. Eur. J. Immunol. 2014;44:2380–2393. doi: 10.1002/eji.201344219.
    1. Manca C, et al. Virulence of a Mycobacterium tuberculosis clinical isolate in mice is determined by failure to induce Th1 type immunity and is associated with induction of IFN-alpha /beta. Proc. Natl Acad. Sci. USA. 2001;98:5752–5757. doi: 10.1073/pnas.091096998.
    1. Mayer-Barber KD, et al. Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk. Nature. 2014;511:99–103. doi: 10.1038/nature13489.
    1. McNab FW, et al. TPL-2-ERK1/2 signaling promotes host resistance against intracellular bacterial infection by negative regulation of type I IFN production. J. Immunol. 2013;191:1732–1743. doi: 10.4049/jimmunol.1300146.
    1. Redford PS, et al. Influenza A virus impairs control of Mycobacterium tuberculosis coinfection through a type I interferon receptor-dependent pathway. J. Infect. Dis. 2014;209:270–274. doi: 10.1093/infdis/jit424.
    1. Moreira-Teixeira L, et al. Mouse transcriptome reveals potential signatures of protection and pathogenesis in human tuberculosis. Nat. Immunol. 2020;21:464–476. doi: 10.1038/s41590-020-0610-z.
    1. Eum SY, et al. Neutrophils are the predominant infected phagocytic cells in the airways of patients with active pulmonary TB. Chest. 2010;137:122–128. doi: 10.1378/chest.09-0903.
    1. Gopal R, et al. S100A8/A9 proteins mediate neutrophilic inflammation and lung pathology during tuberculosis. Am. J. Respir. Crit. Care Med. 2013;188:1137–1146. doi: 10.1164/rccm.201304-0803OC.
    1. Mishra BB, et al. Nitric oxide prevents a pathogen-permissive granulocytic inflammation during tuberculosis. Nat. Microbiol. 2017;2:17072. doi: 10.1038/nmicrobiol.2017.72.
    1. Kimmey JM, et al. Unique role for ATG5 in neutrophil-mediated immunopathology during M. tuberculosis infection. Nature. 2015;528:565–569. doi: 10.1038/nature16451.
    1. Casanova JL, Abel L. Genetic dissection of immunity to mycobacteria: the human model. Annu. Rev. Immunol. 2002;20:581–620. doi: 10.1146/annurev.immunol.20.081501.125851.
    1. Flynn JL, Chan J. Immunology of tuberculosis. Annu. Rev. Immunol. 2001;19:93–129. doi: 10.1146/annurev.immunol.19.1.93.
    1. Keane J, et al. Tuberculosis associated with infliximab, a tumor necrosis factor alpha-neutralizing agent. N. Engl. J. Med. 2001;345:1098–1104. doi: 10.1056/NEJMoa011110.
    1. North RJ, Jung YJ. Immunity to tuberculosis. Annu. Rev. Immunol. 2004;22:599–623. doi: 10.1146/annurev.immunol.22.012703.104635.
    1. Bryson BD, et al. Heterogeneous GM-CSF signaling in macrophages is associated with control of Mycobacterium tuberculosis. Nat. Commun. 2019;10:2329. doi: 10.1038/s41467-019-10065-8.
    1. Denis M, Ghadirian E. Granulocyte-macrophage colony-stimulating factor restricts growth of tubercle bacilli in human macrophages. Immunol. Lett. 1990;24:203–206. doi: 10.1016/0165-2478(90)90049-V.
    1. Gonzalez-Juarrero M, et al. Disruption of granulocyte macrophage-colony stimulating factor production in the lungs severely affects the ability of mice to control Mycobacterium tuberculosis infection. J. Leukoc. Biol. 2005;77:914–922. doi: 10.1189/jlb.1204723.
    1. Rosen LB, et al. Anti-GM-CSF autoantibodies in patients with cryptococcal meningitis. J. Immunol. 2013;190:3959–3966. doi: 10.4049/jimmunol.1202526.
    1. Rothchild AC, Jayaraman P, Nunes-Alves C, Behar SM. iNKT cell production of GM-CSF controls Mycobacterium tuberculosis. PLoS Pathog. 2014;10:e1003805. doi: 10.1371/journal.ppat.1003805.
    1. Rothchild, A. C. et al. Role of granulocyte-macrophage colony-stimulating factor production by T cells during Mycobacterium tuberculosis infection. MBio8 (2017).
    1. Szeliga J, et al. Granulocyte-macrophage colony stimulating factor-mediated innate responses in. Tuberculosis. Tuberculosis. 2008;88:7–20. doi: 10.1016/j.tube.2007.08.009.
    1. Zumla A, et al. Host-directed therapies for infectious diseases: current status, recent progress, and future prospects. Lancet Infect. Dis. 2016;16:e47–e63. doi: 10.1016/S1473-3099(16)00078-5.
    1. Stanley E, et al. Granulocyte/macrophage colony-stimulating factor-deficient mice show no major perturbation of hematopoiesis but develop a characteristic pulmonary pathology. Proc. Natl Acad. Sci. USA. 1994;91:5592–5596. doi: 10.1073/pnas.91.12.5592.
    1. Dranoff G, et al. Involvement of granulocyte-macrophage colony-stimulating factor in pulmonary homeostasis. Science. 1994;264:713–716. doi: 10.1126/science.8171324.
    1. Singhania A, et al. A modular transcriptional signature identifies phenotypic heterogeneity of human tuberculosis infection. Nat. Commun. 2018;9:2308. doi: 10.1038/s41467-018-04579-w.
    1. Singhania A, et al. Transcriptional profiling unveils type I and II interferon networks in blood and tissues across diseases. Nat. Commun. 2019;10:2887. doi: 10.1038/s41467-019-10601-6.
    1. Gautier EL, et al. Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat. Immunol. 2012;13:1118–1128. doi: 10.1038/ni.2419.
    1. Shibata Y, et al. GM-CSF regulates alveolar macrophage differentiation and innate immunity in the lung through PU.1. Immunity. 2001;15:557–567. doi: 10.1016/S1074-7613(01)00218-7.
    1. Guilliams M, et al. Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J. Exp. Med. 2013;210:1977–1992. doi: 10.1084/jem.20131199.
    1. Segueni N, et al. Controlled Mycobacterium tuberculosis infection in mice under treatment with anti-IL-17A or IL-17F antibodies, in contrast to TNFalpha neutralization. Sci. Rep. 2016;6:36923. doi: 10.1038/srep36923.
    1. Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018;18:134–147. doi: 10.1038/nri.2017.105.
    1. Branzk N, et al. Neutrophils sense microbe size and selectively release neutrophil extracellular traps in response to large pathogens. Nat. Immunol. 2014;15:1017–1025. doi: 10.1038/ni.2987.
    1. Vashakidze S, et al. Retrospective study of clinical and lesion characteristics of patients undergoing surgical treatment for Pulmonary Tuberculosis in Georgia. Int J. Infect. Dis. 2017;56:200–207. doi: 10.1016/j.ijid.2016.12.009.
    1. Marakalala MJ, et al. Inflammatory signaling in human tuberculosis granulomas is spatially organized. Nat. Med. 2016;22:531–538. doi: 10.1038/nm.4073.
    1. Sakai S, et al. CD4 T cell-derived IFN-gamma plays a minimal role in control of pulmonary Mycobacterium tuberculosis infection and must be actively repressed by PD-1 to prevent lethal disease. PLoS Pathog. 2016;12:e1005667. doi: 10.1371/journal.ppat.1005667.
    1. Cruz A, et al. Pathological role of interleukin 17 in mice subjected to repeated BCG vaccination after infection with Mycobacterium tuberculosis. J. Exp. Med. 2010;207:1609–1616. doi: 10.1084/jem.20100265.
    1. Schmok E, et al. Suppressor of cytokine signaling 3 in macrophages prevents exacerbated interleukin-6-dependent arginase-1 activity and early permissiveness to experimental tuberculosis. Front Immunol. 2017;8:1537. doi: 10.3389/fimmu.2017.01537.
    1. Tobin DM, et al. Host genotype-specific therapies can optimize the inflammatory response to mycobacterial infections. Cell. 2012;148:434–446. doi: 10.1016/j.cell.2011.12.023.
    1. Bloom CI, et al. Transcriptional blood signatures distinguish pulmonary tuberculosis, pulmonary sarcoidosis, pneumonias and lung cancers. PLoS ONE. 2013;8:e70630. doi: 10.1371/journal.pone.0070630.
    1. Abram CL, Roberge GL, Hu Y, Lowell CA. Comparative analysis of the efficiency and specificity of myeloid-Cre deleting strains using ROSA-EYFP reporter mice. J. Immunol. Methods. 2014;408:89–100. doi: 10.1016/j.jim.2014.05.009.
    1. Passegue E, Wagner EF, Weissman IL. JunB deficiency leads to a myeloproliferative disorder arising from hematopoietic stem cells. Cell. 2004;119:431–443. doi: 10.1016/j.cell.2004.10.010.
    1. Greter M, et al. GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells. Immunity. 2012;36:1031–1046. doi: 10.1016/j.immuni.2012.03.027.
    1. King IL, Kroenke MA, Segal BM. GM-CSF-dependent, CD103+ dermal dendritic cells play a critical role in Th effector cell differentiation after subcutaneous immunization. J. Exp. Med. 2010;207:953–961. doi: 10.1084/jem.20091844.
    1. Huang L, Nazarova EV, Tan S, Liu Y, Russell DG. Growth of Mycobacterium tuberculosis in vivo segregates with host macrophage metabolism and ontogeny. J. Exp. Med. 2018;215:1135–1152. doi: 10.1084/jem.20172020.
    1. McNab F, Mayer-Barber K, Sher A, Wack A, O’Garra A. Type I interferons in infectious disease. Nat. Rev. Immunol. 2015;15:87–103. doi: 10.1038/nri3787.
    1. Lowe DM, Redford PS, Wilkinson RJ, O’Garra A, Martineau AR. Neutrophils in tuberculosis: friend or foe? Trends Immunol. 2012;33:14–25. doi: 10.1016/j.it.2011.10.003.
    1. LeVine AM, Reed JA, Kurak KE, Cianciolo E, Whitsett JA. GM-CSF-deficient mice are susceptible to pulmonary group B streptococcal infection. J. Clin. Invest. 1999;103:563–569. doi: 10.1172/JCI5212.
    1. Akk A, Springer LE, Pham CT. Neutrophil extracellular traps enhance early inflammatory response in sendai virus-induced asthma phenotype. Front. Immunol. 2016;7:325. doi: 10.3389/fimmu.2016.00325.
    1. Pillai PS, et al. Mx1 reveals innate pathways to antiviral resistance and lethal influenza disease. Science. 2016;352:463–466. doi: 10.1126/science.aaf3926.
    1. Radermecker C, et al. Locally instructed CXCR4(hi) neutrophils trigger environment-driven allergic asthma through the release of neutrophil extracellular traps. Nat. Immunol. 2019;20:1444–1455. doi: 10.1038/s41590-019-0496-9.
    1. Warnatsch A, Ioannou M, Wang Q, Papayannopoulos V. Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science. 2015;349:316–320. doi: 10.1126/science.aaa8064.
    1. Guimaraes-Costa AB, Rochael NC, Oliveira F, Echevarria-Lima J, Saraiva EM. Neutrophil extracellular traps reprogram IL-4/GM-CSF-induced monocyte differentiation to anti-inflammatory macrophages. Front. Immunol. 2017;8:523. doi: 10.3389/fimmu.2017.00523.
    1. Ramos-Kichik V, et al. Neutrophil extracellular traps are induced by Mycobacterium tuberculosis. Tuberculosis. 2009;89:29–37. doi: 10.1016/j.tube.2008.09.009.
    1. Su R, et al. Mycobacterium tuberculosis infection induces low-density granulocyte generation by promoting neutrophil extracellular trap formation via ROS pathway. Front. Microbiol. 2019;10:1468. doi: 10.3389/fmicb.2019.01468.
    1. Ong CW, et al. Neutrophil-derived MMP-8 drives AMPK-dependent matrix destruction in human pulmonary tuberculosis. PLoS Pathog. 2015;11:e1004917. doi: 10.1371/journal.ppat.1004917.
    1. de Melo MGM, et al. Imbalance of NET and alpha-1-antitrypsin in tuberculosis patients is related with hyper inflammation and severe lung tissue damage. Front. Immunol. 2018;9:3147. doi: 10.3389/fimmu.2018.03147.
    1. Schechter MC, et al. Neutrophil extracellular trap (NET) levels in human plasma are associated with active TB. PLoS ONE. 2017;12:e0182587. doi: 10.1371/journal.pone.0182587.
    1. Ravimohan, S., Kornfeld, H., Weissman, D. & Bisson, G. P. Tuberculosis and lung damage: from epidemiology to pathophysiology. Eur. Respir. Rev.27, 170077 (2018).
    1. Garcia-Romo GS, et al. Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci. Transl. Med. 2011;3:73ra20. doi: 10.1126/scitranslmed.3001201.
    1. Gul E, et al. Type I IFN-related NETosis in ataxia telangiectasia and Artemis deficiency. J. Allergy Clin. Immunol. 2018;142:246–257. doi: 10.1016/j.jaci.2017.10.030.
    1. Mayer-Barber KD, et al. Innate and adaptive interferons suppress IL-1alpha and IL-1beta production by distinct pulmonary myeloid subsets during Mycobacterium tuberculosis infection. Immunity. 2011;35:1023–1034. doi: 10.1016/j.immuni.2011.12.002.
    1. McNab FW, et al. Type I IFN induces IL-10 production in an IL-27-independent manner and blocks responsiveness to IFN-gamma for production of IL-12 and bacterial killing in Mycobacterium tuberculosis-infected macrophages. J. Immunol. 2014;193:3600–3612. doi: 10.4049/jimmunol.1401088.
    1. Kamphuis E, Junt T, Waibler Z, Forster R, Kalinke U. Type I interferons directly regulate lymphocyte recirculation and cause transient blood lymphopenia. Blood. 2006;108:3253–3261. doi: 10.1182/blood-2006-06-027599.
    1. Le Bon A, et al. Cutting edge: enhancement of antibody responses through direct stimulation of B and T cells by type I IFN. J. Immunol. 2006;176:2074–2078. doi: 10.4049/jimmunol.176.4.2074.
    1. Bankhead P, et al. QuPath: open source software for digital pathology image analysis. Sci. Rep. 2017;7:16878. doi: 10.1038/s41598-017-17204-5.
    1. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinforma. 2011;12:323. doi: 10.1186/1471-2105-12-323.
    1. Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. doi: 10.1093/bioinformatics/bts635.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8.
    1. Team, R.D.C. R Development Core Team. R: A language and environment for statistical computing. (R Foundation for Statistical Computing, Vienna, Austria, 2008).
    1. Murtagh F, Legendre P. Ward’s hierarchical agglomerative clustering method: which algorithms implement Ward’s criterion? J. Classification. 2014;31:274–295. doi: 10.1007/s00357-014-9161-z.
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Newman AM, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods. 2015;12:453–457. doi: 10.1038/nmeth.3337.
    1. Yaari G, Bolen CR, Thakar J, Kleinstein SH. Quantitative set analysis for gene expression: a method to quantify gene set differential expression including gene-gene correlations. Nucleic Acids Res. 2013;41:e170. doi: 10.1093/nar/gkt660.

Source: PubMed

3
订阅