Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Unique role for ATG5 in neutrophil-mediated immunopathology during M. tuberculosis infection

Abstract

Mycobacterium tuberculosis, a major global health threat, replicates in macrophages in part by inhibiting phagosome–lysosome fusion, until interferon-γ (IFNγ) activates the macrophage to traffic M. tuberculosis to the lysosome. How IFNγ elicits this effect is unknown, but many studies suggest a role for macroautophagy (herein termed autophagy), a process by which cytoplasmic contents are targeted for lysosomal degradation1. The involvement of autophagy has been defined based on studies in cultured cells where M. tuberculosis co-localizes with autophagy factors ATG5, ATG12, ATG16L1, p62, NDP52, BECN1 and LC3 (refs 2, 3, 4, 5, 6), stimulation of autophagy increases bacterial killing6,7,8, and inhibition of autophagy increases bacterial survival1,2,4,6,7. Notably, these studies reveal modest (~1.5–3-fold change) effects on M. tuberculosis replication. By contrast, mice lacking ATG5 in monocyte-derived cells and neutrophils (polymorponuclear cells, PMNs) succumb to M. tuberculosis within 30 days4,9, an extremely severe phenotype similar to mice lacking IFNγ signalling10,11. Importantly, ATG5 is the only autophagy factor that has been studied during M. tuberculosis infection in vivo and autophagy-independent functions of ATG5 have been described12,13,14,15,16,17,18. For this reason, we used a genetic approach to elucidate the role for multiple autophagy-related genes and the requirement for autophagy in resistance to M. tuberculosis infection in vivo. Here we show that, contrary to expectation, autophagic capacity does not correlate with the outcome of M. tuberculosis infection. Instead, ATG5 plays a unique role in protection against M. tuberculosis by preventing PMN-mediated immunopathology. Furthermore, while Atg5 is dispensable in alveolar macrophages during M. tuberculosis infection, loss of Atg5 in PMNs can sensitize mice to M. tuberculosis. These findings shift our understanding of the role of ATG5 during M. tuberculosis infection, reveal new outcomes of ATG5 activity, and shed light on early events in innate immunity that are required to regulate disease pathology and bacterial replication.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: ATG5, in contrast to other autophagy factors, is essential to control M. tuberculosis infection.
Figure 2: Loss of Atg5 in LysM+ cells leads to earlier and more severe lung inflammation during M. tuberculosis infection.
Figure 3: Depletion of PMNs allows for survival of Atg5fl/fl-Lysm-cre mice during M. tuberculosis infection.
Figure 4: Loss of Atg5 in PMNs, but not alveolar macrophages or dendritic cells, can cause susceptibility to M. tuberculosis.

Similar content being viewed by others

References

  1. Deretic, V. Autophagy in tuberculosis. Cold Spring Harb. Persp. Med. 4, a018481 (2014)

    Article  Google Scholar 

  2. Dutta, R. K., Kathania, M., Raje, M. & Majumdar, S. IL-6 inhibits IFN-γ induced autophagy in Mycobacterium tuberculosis H37Rv infected macrophages. Int. J. Biochem. Cell Biol. 44, 942–954 (2012)

    Article  CAS  Google Scholar 

  3. Juárez, E. et al. NOD2 enhances the innate response of alveolar macrophages to Mycobacterium tuberculosis in humans. Eur. J. Immunol. 42, 880–889 (2012)

    Article  Google Scholar 

  4. Watson, R. O., Manzanillo, P. S. & Cox, J. S. Extracellular M. tuberculosis DNA targets bacteria for autophagy by activating the host DNA-sensing pathway. Cell 150, 803–815 (2012)

    Article  CAS  Google Scholar 

  5. Seto, S., Tsujimura, K., Horii, T. & Koide, Y. Autophagy adaptor protein p62/SQSTM1 and autophagy-related gene Atg5 mediate autophagosome formation in response to Mycobacterium tuberculosis infection in dendritic cells. PLoS ONE 8, e86017 (2013)

    Article  ADS  Google Scholar 

  6. Sakowski, E. T. et al. Ubiquilin 1 promotes IFN-γ-induced xenophagy of Mycobacterium tuberculosis . PLoS Pathog. 11, e1005076 (2015)

    Article  Google Scholar 

  7. Gutierrez, M. G. et al. Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell 119, 753–766 (2004)

    Article  CAS  Google Scholar 

  8. Wang, J. et al. MicroRNA-155 promotes autophagy to eliminate intracellular mycobacteria by targeting Rheb. PLoS Pathog. 9, e1003697 (2013)

    Article  Google Scholar 

  9. Castillo, E. F. et al. Autophagy protects against active tuberculosis by suppressing bacterial burden and inflammation. Proc. Natl Acad. Sci. USA 109, E3168–E3176 (2012)

    Article  CAS  Google Scholar 

  10. Cooper, A. M. et al. Disseminated tuberculosis in interferon gamma gene-disrupted mice. J. Exp. Med. 178, 2243–2247 (1993)

    Article  CAS  Google Scholar 

  11. Nandi, B. & Behar, S. M. Regulation of neutrophils by interferon-γ limits lung inflammation during tuberculosis infection. J. Exp. Med. 208, 2251–2262 (2011)

    Article  CAS  Google Scholar 

  12. Choi, J. et al. The parasitophorous vacuole membrane of Toxoplasma gondii is targeted for disruption by ubiquitin-like conjugation systems of autophagy. Immunity 40, 924–935 (2014)

    Article  CAS  Google Scholar 

  13. Hwang, S. et al. Nondegradative role of Atg5-Atg12/Atg16L1 autophagy protein complex in antiviral activity of interferon gamma. Cell Host Microbe 11, 397–409 (2012)

    Article  CAS  Google Scholar 

  14. Zhao, Z. et al. Autophagosome-independent essential function for the autophagy protein Atg5 in cellular immunity to intracellular pathogens. Cell Host Microbe 4, 458–469 (2008)

    Article  CAS  Google Scholar 

  15. Martinez, J. et al. Molecular characterization of LC3-associated phagocytosis reveals distinct roles for Rubicon, NOX2 and autophagy proteins. Nature Cell Biol. 17, 893–906 (2015)

    Article  CAS  Google Scholar 

  16. Jounai, N. et al. The Atg5 Atg12 conjugate associates with innate antiviral immune responses. Proc. Natl Acad. Sci. USA 104, 14050–14055 (2007)

    Article  ADS  CAS  Google Scholar 

  17. Yousefi, S. et al. Calpain-mediated cleavage of Atg5 switches autophagy to apoptosis. Nature Cell Biol. 8, 1124–1132 (2006)

    Article  CAS  Google Scholar 

  18. Maskey, D. et al. ATG5 is induced by DNA-damaging agents and promotes mitotic catastrophe independent of autophagy. Nature Commun. 4, 2130 (2013)

    Article  ADS  Google Scholar 

  19. Jakubzick, C. et al. Lymph-migrating, tissue-derived dendritic cells are minor constituents within steady-state lymph nodes. J. Exp. Med. 205, 2839–2850 (2008)

    Article  CAS  Google Scholar 

  20. Abram, C. L., Roberge, G. L., Hu, Y. & Lowell, C. A. Comparative analysis of the efficiency and specificity of myeloid-Cre deleting strains using ROSA-EYFP reporter mice. J. Immunol. Methods 408, 89–100 (2014)

    Article  CAS  Google Scholar 

  21. Flynn, J. L. & Chan, J. Immunology of tuberculosis. Annu. Rev. Immunol. 19, 93–129 (2001)

    Article  CAS  Google Scholar 

  22. Jayaswal, S. et al. Identification of host-dependent survival factors for intracellular Mycobacterium tuberculosis through an siRNA Screen. PLoS Pathog. 6, e1000839 (2010)

    Article  Google Scholar 

  23. Jung, C. H. et al. ULK-Atg13–FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol. Biol. Cell 20, 1992–2003 (2009)

    Article  CAS  Google Scholar 

  24. Mariño, G. et al. Autophagy is essential for mouse sense of balance. J. Clin. Invest. 120, 2331–2344 (2010)

    Article  Google Scholar 

  25. Lin, H. H. et al. Dynamic involvement of ATG5 in cellular stress responses. Cell Death Dis. 5, e1478 (2014)

    Article  CAS  Google Scholar 

  26. Cadwell, K. et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature 456, 259–263 (2008)

    Article  ADS  CAS  Google Scholar 

  27. Abdel Fattah, E., Bhattacharya, A., Herron, A., Safdar, Z. & Eissa, N. T. Critical role for IL-18 in spontaneous lung inflammation caused by autophagy deficiency. J. Immunol. 194, 5407–5416 (2015)

    Article  CAS  Google Scholar 

  28. Kanayama, M., He, Y.-W. & Shinohara, M. L. The lung is protected from spontaneous inflammation by autophagy in myeloid cells. J. Immunol. 194, 5465–5471 (2015)

    Article  CAS  Google Scholar 

  29. Saitoh, T. et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1β production. Nature 456, 264–268 (2008)

    Article  ADS  CAS  Google Scholar 

  30. Leemans, J. C. et al. Depletion of alveolar macrophages exerts protective effects in pulmonary tuberculosis in mice. J. Immunol. 166, 4604–4611 (2001)

    Article  CAS  Google Scholar 

  31. Jia, W. & He, Y.-W. Temporal regulation of intracellular organelle homeostasis in T lymphocytes by autophagy. J. Immunol. 186, 5313–5322 (2011)

    Article  CAS  Google Scholar 

  32. Malhotra, R., Warne, J. P., Salas, E., Xu, A. W. & Debnath, J. Loss of Atg12, but not Atg5, in pro-opiomelanocortin neurons exacerbates diet-induced obesity. Autophagy 11, 145–154 (2015)

    PubMed  PubMed Central  Google Scholar 

  33. Komatsu, M. et al. Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell 131, 1149–1163 (2007)

    Article  CAS  Google Scholar 

  34. Lee, E.-J. & Tournier, C. The requirement of uncoordinated 51-like kinase 1 (ULK1) and ULK2 in the regulation of autophagy. Autophagy 7, 689–695 (2011)

    Article  CAS  Google Scholar 

  35. McAlpine, F., Williamson, L. E., Tooze, S. A. & Chan, E. Y. W. Regulation of nutrient-sensitive autophagy by uncoordinated 51-like kinases 1 and 2. Autophagy 9, 361–373 (2013)

    Article  CAS  Google Scholar 

  36. Fleming, T. J., Fleming, M. L. & Malek, T. R. Selective expression of Ly-6G on myeloid lineage cells in mouse bone marrow. RB6-8C5 mAb to granulocyte-differentiation antigen (Gr-1) detects members of the Ly-6 family. J. Immunol. 151, 2399–2408 (1993)

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

C.L.S. is supported by a Beckman Young Investigator Award from the Arnold and Mabel Beckman Foundation. J.M.K. is supported by a National Science Foundation Graduate Research Fellowship DGE-1143954 and the NIGMS Cell and Molecular Biology Training Grant GM007067. J.P.H. is supported by a National Science Foundation Graduate Research Fellowship DGE-1143954. H.W.V., S.P. and A.K. are supported by U19 AI109725. We would like to acknowledge D. Kreamalmeyer for assistance with the mouse colonies and T. Malek and L. D. Sibley for providing the 1A8 hybridoma.

Author information

Authors and Affiliations

Authors

Contributions

J.M.K. designed and performed experiments, analysed data, and wrote the manuscript. J.P.H. and L.A.W. performed experiments. S.P., A.K. and J.D. generated mouse strains. H.W.V. provided all mouse strains, analysed data and wrote the manuscript. C.L.S. designed experiments, analysed data and wrote the manuscript.

Corresponding author

Correspondence to Christina L. Stallings.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Survival of mice with defects in autophagy genes other than Atg5.

Per cent survival of mice following infection with 100 colony-forming units (c.f.u.) of aerosolized M. tuberculosis. a, Survival of C57Bl/6 (open squares), Ulk1−/− (blue triangles), Ulk2−/− (inverted pink triangles), Atg4b−/− (red diamonds), and p62−/− (green circles) mice. b, Survival of Atg14lfl/fl-Lysm-cre (purple diamonds), Atg12fl/fl-Lysm-cre (red inverted triangles), Atg16l1fl/fl-Lysm-cre (green triangles), Atg7fl/fl-Lysm-cre (pink diamonds), Atg3fl/fl-Lysm-cre (brown circles) and corresponding floxed control mice. Floxed control mice are shown in open shapes, LysM–Cre-expressing mice are shown in closed shapes. c, Survival of C57Bl/6 (open squares), Atg16l1HM1 (open circles). Samples represent biological replicates. See Supplementary Fig. 6 for sample sizes.

Source data

Extended Data Figure 2 Analysis of autophagy in bronchoalveolar macrophages.

Western blot analysis of p62, LC3, and actin levels in ex vivo macrophages isolated from bronchoalveolar lavages of uninfected mice. For gel source data, see Supplementary Fig. 1.

Extended Data Figure 3 Atg5fl/fl bone-marrow-derived macrophages are hypomorphic for ATG5.

Western blot analysis of ATG5 (ATG5–ATG12 conjugate, 56 kDa) and actin in uninfected-bone-marrow-derived macrophages. For gel source data, see Supplementary Fig. 1.

Extended Data Figure 4 Loss of Atg5 or Atg16l1 in LysM+ cells does not lead to increased c.f.u. at 2 w.p.i. log pulmonary c.f.u. at 2 w.p.i.

Samples represent biological replicates; error bars represent mean ± s.e.m. See Supplementary Fig. 7 for sample sizes and results from all statistical comparisons.

Source data

Extended Data Figure 5 Cytokine levels in uninfected lungs.

Concentration of cytokines in lungs (homogenized in 1 ml PBS plus 0.05% Tween 80) from uninfected mice. Levels of IFNγ, IL-6, MIP-1α, IL-17, and G-CSF were below the limit of detection. C57Bl/6 (grey solid bars), Atg5fl/fl (blue striped bars), Atg5fl/fl-Lysm-cre (blue solid bars), Atg16l1fl/fl (green striped bars), Atg16l1fl/fl-Lysm-cre (green solid bars). Statistical differences were determined by one-way ANOVA and Bonferonni’s multiple comparison test. n.s., not significant. Samples represent biological replicates; error bars represent mean ± s.e.m. See Supplementary Fig. 8 for sample sizes and results from all statistical comparisons.

Source data

Extended Data Figure 6 Number of inflammatory cells in lungs of mice at 2 and 3 w.p.i. (related to Fig. 2).

a, Gating strategy for analysis of inflammatory cells in lungs at 2 and 3 w.p.i. Single lung cells were gated based on CD11b, CD11c, Ly6G, Ly6C and autofluorescence (auto). The parental gate is shown above each contour plot. Representative data are shown from an Atg5fl/fl mouse at 2 w.p.i. b, c, C57Bl/6 (grey solid bars), Atg5fl/fl (blue striped bars), Atg5fl/fl-Lysm-cre (blue solid bars), Atg16l1fl/fl (green striped bars), Atg16l1fl/fl-Lysm-cre (green solid bars). Mean number of alveolar macrophages, PMNs, recruited macrophages, and inflammatory monocytes in lungs at 2 w.p.i. (b) and 3 w.p.i. (c). d, e, Flow cytometry data presented in b and c and in Fig. 2 are the compilation of results from five experiments. In some experiments, different amounts of lung were collected for analysis, making it difficult to compare the average number of each cell type between strains, unless the data are normalized (as done in Fig. 2c, d—percentage of total cells). Therefore, to compare the raw number of cells detected in each cell population, each mouse analysed at 2 w.p.i. (d) and 3 w.p.i. (e) has been graphed individually. Each line represents a different mouse, with dots indicating the number of total cells, alveolar macrophages, PMNs, recruited macrophages and inflammatory monocytes. Statistical differences were determined by one-way ANOVA and Bonferonni’s multiple comparison test (b, c); *P < 0.05; n.s., not significant. Samples represent biological replicates; error bars represent mean ± s.e.m. See Supplementary Fig. 9 for sample sizes and results from all statistical comparisons.

Source data

Extended Data Figure 7 Number of inflammatory cells in lungs of mice at 3 w.p.i. (related to Fig. 4).

Number of alveolar macrophages, PMNs, recruited macrophages, and inflammatory monocytes in lungs at 3 w.p.i. C57Bl/6 (grey solid bars), Atg5fl/fl (blue striped bars), Atg5fl/fl-Lysm-cre (blue solid bars), ‘healthy’ Atg5fl/fl-MRP8-cre (purple striped bars), and ‘susceptible’ Atg5fl/fl-MRP8-cre (purple solid bars). Statistical differences were determined by one-way ANOVA and Bonferonni’s multiple comparison test; *P < 0.05; n.s., not significant. Samples represent biological replicates; error bars represent mean ± s.e.m. See Supplementary Fig. 10 for sample sizes and results from all statistical comparisons.

Source data

Extended Data Figure 8 Analysis of autophagy in bone marrow PMNs.

Western blot analysis of p62, LC3, and actin in bone marrow PMNs from uninfected mice. Each lane represents an individual mouse. Two replicates of the Atg5fl/fl-Lysm-cre and Atg16l1fl/fl-Lysm-cre mice are shown. For gel source data, see Supplementary Fig. 1.

Supplementary information

Supplementary Figure 1

This file contains source gel data showing uncropped western blot images and ladder markers. Boxes indicate how each image was cropped for final figures. (PDF 651 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kimmey, J., Huynh, J., Weiss, L. et al. Unique role for ATG5 in neutrophil-mediated immunopathology during M. tuberculosis infection. Nature 528, 565–569 (2015). https://doi.org/10.1038/nature16451

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature16451

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing