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.

  • Article
  • Published:

Mammalian Atg8 proteins and the autophagy factor IRGM control mTOR and TFEB at a regulatory node critical for responses to pathogens

An Author Correction to this article was published on 12 August 2020

An Author Correction to this article was published on 12 August 2020

This article has been updated

Abstract

Autophagy is a homeostatic process with multiple functions in mammalian cells. Here, we show that mammalian Atg8 proteins (mAtg8s) and the autophagy regulator IRGM control TFEB, a transcriptional activator of the lysosomal system. IRGM directly interacted with TFEB and promoted the nuclear translocation of TFEB. An mAtg8 partner of IRGM, GABARAP, interacted with TFEB. Deletion of all mAtg8s or GABARAPs affected the global transcriptional response to starvation and downregulated subsets of TFEB targets. IRGM and GABARAPs countered the action of mTOR as a negative regulator of TFEB. This was suppressed by constitutively active RagB, an activator of mTOR. Infection of macrophages with the membrane-permeabilizing microbe Mycobacterium tuberculosis or infection of target cells by HIV elicited TFEB activation in an IRGM-dependent manner. Thus, IRGM and its interactors mAtg8s close a loop between the autophagosomal pathway and the control of lysosomal biogenesis by TFEB, thus ensuring coordinated activation of the two systems that eventually merge during autophagy.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: IRGM controls TFEB nuclear translocation.
Fig. 2: IRGM and TFEB interact.
Fig. 3: IRGM affects mTOR activity and interacts with PPP3CB.
Fig. 4: mAtg8s affect mTOR and the nuclear translocation of TFEB.
Fig. 5: mAtg8s control the transcriptional activity of TFEB.
Fig. 6: Stx17 affects mTOR and regulates TFEB nuclear translocation.
Fig. 7: IRGM affects TFEB nuclear translocation in cells infected with diverse pathogens associated with tuberculosis, AIDS or CD.

Similar content being viewed by others

Data availability

RNA-seq data that support the findings of this study have been deposited in the Gene Expression Omnibus (GEO) under accession code GSE149533. Mass spectrometry proteomics data in this study have been deposited in the MassIVE repository (https://massive.ucsd.edu), with accession number MSV000083251 for Stx17 interactors44 and accession number MSV000085401 for IRGM interactors. All other data supporting the findings of this study are available from the corresponding author upon reasonable request. Source data are provided with this paper.

Change history

References

  1. Mizushima, N., Yoshimori, T. & Ohsumi, Y. The role of Atg proteins in autophagosome formation. Annu. Rev. Cell Dev. Biol. 27, 107–132 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Mizushima, N., Levine, B., Cuervo, A. M. & Klionsky, D. J. Autophagy fights disease through cellular self-digestion. Nature 451, 1069–1075 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Levine, B. & Kroemer, G. Biological functions of autophagy genes: a disease perspective. Cell 176, 11–42 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Singh, S. B., Davis, A. S., Taylor, G. A. & Deretic, V. Human IRGM induces autophagy to eliminate intracellular mycobacteria. Science 313, 1438–1441 (2006).

    Article  CAS  PubMed  Google Scholar 

  5. Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661–678 (2007).

    Article  Google Scholar 

  6. Parkes, M. et al. Sequence variants in the autophagy gene IRGM and multiple other replicating loci contribute to Crohn’s disease susceptibility. Nat. Genet. 39, 830–832 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Deretic, V., Saitoh, T. & Akira, S. Autophagy in infection, inflammation and immunity. Nat. Rev. Immunol. 13, 722–737 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Bekpen, C. et al. The interferon-inducible p47 (IRG) GTPases in vertebrates: loss of the cell autonomous resistance mechanism in the human lineage. Genome Biol. 6, R92 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Bekpen, C. et al. Death and resurrection of the human IRGM gene. PLoS Genet. 5, e1000403 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Mitchell, G. & Isberg, R. R. Innate immunity to intracellular pathogens: balancing microbial elimination and inflammation. Cell Host Microbe 22, 166–175 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. 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  PubMed  Google Scholar 

  12. Singh, S. B. et al. Human IRGM regulates autophagy and cell-autonomous immunity functions through mitochondria. Nat. Cell Biol. 12, 1154–1165 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Chauhan, S., Mandell, M. A. & Deretic, V. IRGM governs the core autophagy machinery to conduct antimicrobial defense. Mol. Cell 58, 507–521 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kumar, S. et al. Mechanism of Stx17 recruitment to autophagosomes via IRGM and mammalian Atg8 proteins. J. Cell Biol. 217, 997–1013 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gregoire, I. P. et al. IRGM is a common target of RNA viruses that subvert the autophagy network. PLoS Pathog. 7, e1002422 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Itakura, E., Kishi-Itakura, C. & Mizushima, N. The hairpin-type tail-anchored SNARE syntaxin 17 targets to autophagosomes for fusion with endosomes/lysosomes. Cell 151, 1256–1269 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Weidberg, H. et al. LC3 and GATE-16/GABARAP subfamilies are both essential yet act differently in autophagosome biogenesis. EMBO J. 29, 1792–1802 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nguyen, T. N. et al. Atg8 family LC3/GABARAP proteins are crucial for autophagosome–lysosome fusion but not autophagosome formation during PINK1/Parkin mitophagy and starvation. J. Cell Biol. 215, 857–874 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Settembre, C. et al. TFEB links autophagy to lysosomal biogenesis. Science 332, 1429–1433 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sardiello, M. et al. A gene network regulating lysosomal biogenesis and function. Science 325, 473–477 (2009).

    Article  CAS  PubMed  Google Scholar 

  21. Puertollano, R., Ferguson, S. M., Brugarolas, J. & Ballabio, A. The complex relationship between TFEB transcription factor phosphorylation and subcellular localization. EMBO J. 37, e98804 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Napolitano, G. & Ballabio, A. TFEB at a glance. J. Cell Sci. 129, 2475–2481 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Brady, O. A., Martina, J. A. & Puertollano, R. Emerging roles for TFEB in the immune response and inflammation. Autophagy 14, 181–189 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Settembre, C. et al. TFEB controls cellular lipid metabolism through a starvation-induced autoregulatory loop. Nat. Cell Biol. 15, 647–658 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Roczniak-Ferguson, A. et al. The transcription factor TFEB links mTORC1 signaling to transcriptional control of lysosome homeostasis. Sci. Signal. 5, ra42 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Settembre, C. et al. A lysosome-to-nucleus signalling mechanism senses and regulates the lysosome via mTOR and TFEB. EMBO J. 31, 1095–1108 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Medina, D. L. et al. Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB. Nat. Cell Biol. 17, 288–299 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Medina, D. L. et al. Transcriptional activation of lysosomal exocytosis promotes cellular clearance. Dev. Cell 21, 421–430 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Nnah, I. C. et al. TFEB-driven endocytosis coordinates MTORC1 signaling and autophagy. Autophagy 15, 151–164 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Mansueto, G. et al. Transcription factor EB controls metabolic flexibility during exercise. Cell Metab. 25, 182–196 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Perera, R. M. et al. Transcriptional control of autophagy–lysosome function drives pancreatic cancer metabolism. Nature 524, 361–365 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Campbell, G. R., Rawat, P., Bruckman, R. S. & Spector, S. A. Human immunodeficiency virus type 1 Nef inhibits autophagy through transcription factor EB sequestration. PLoS Pathog. 11, e1005018 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Gray, M. A. et al. Phagocytosis enhances lysosomal and bactericidal properties by activating the transcription factor TFEB. Curr. Biol. 26, 1955–1964 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Jia, J. et al. Galectin-3 coordinates a cellular system for lysosomal repair and removal. Dev. Cell 52, 69–87 e68 (2020).

    Article  CAS  PubMed  Google Scholar 

  35. Manzanillo, P. S., Shiloh, M. U., Portnoy, D. A. & Cox, J. S. Mycobacterium tuberculosis activates the DNA-dependent cytosolic surveillance pathway within macrophages. Cell Host Microbe 11, 469–480 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Collazo, C. M. et al. Inactivation of LRG-47 and IRG-47 reveals a family of interferon gamma-inducible genes with essential, pathogen-specific roles in resistance to infection. J. Exp. Med. 194, 181–188 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. NCBI. Domain of unknown function (DUF3371). Conserved Domain Database https://www.ncbi.nlm.nih.gov/Structure/cdd/cddsrv.cgi?uid=pfam11851 (2019).

  38. Zoncu, R. et al. mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H+-ATPase. Science 334, 678–683 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Inoki, K., Zhu, T. & Guan, K. L. TSC2 mediates cellular energy response to control cell growth and survival. Cell 115, 577–590 (2003).

    Article  CAS  PubMed  Google Scholar 

  40. Abu-Remaileh, M. et al. Lysosomal metabolomics reveals V-ATPase- and mTOR-dependent regulation of amino acid efflux from lysosomes. Science 358, 807–813 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Behrends, C., Sowa, M. E., Gygi, S. P. & Harper, J. W. Network organization of the human autophagy system. Nature 466, 68–76 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Nezich, C. L., Wang, C., Fogel, A. I. & Youle, R. J. MiT/TFE transcription factors are activated during mitophagy downstream of Parkin and Atg5. J. Cell Biol. 210, 435–450 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gu, Y. et al. Mammalian Atg8 proteins regulate lysosome and autolysosome biogenesis through SNAREs. EMBO J. 38, e101994 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kumar, S. et al. Phosphorylation of syntaxin 17 by TBK1 controls autophagy initiation. Dev. Cell 49, 130–144.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Bar-Peled, L., Schweitzer, L. D., Zoncu, R. & Sabatini, D. M. Ragulator is a GEF for the Rag GTPases that signal amino acid levels to mTORC1. Cell 150, 1196–1208 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Jackson, C. L. & Casanova, J. E. Turning on ARF: the Sec7 family of guanine-nucleotide-exchange factors. Trends Cell. Biol. 10, 60–67 (2000).

    Article  CAS  PubMed  Google Scholar 

  47. Jia, J. et al. Galectins control mTOR in response to endomembrane damage. Mol. Cell 70, 120–135.e8 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Castellano, B. M. et al. Lysosomal cholesterol activates mTORC1 via an SLC38A9–Niemann–Pick C1 signaling complex. Science 355, 1306–1311 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Chauhan, S. et al. TRIMs and galectins globally cooperate and TRIM16 and Galectin-3 Co-direct autophagy in endomembrane damage homeostasis. Dev. Cell 39, 13–27 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Huett, A., McCarroll, S. A., Daly, M. J. & Xavier, R. J. On the level: IRGM gene function is all about expression. Autophagy 5, 96–99 (2009).

    Article  CAS  PubMed  Google Scholar 

  51. Brest, P. et al. A synonymous variant in IRGM alters a binding site for miR-196 and causes deregulation of IRGM-dependent xenophagy in Crohn’s disease. Nat. Genet. 43, 242–245 (2011).

    Article  CAS  PubMed  Google Scholar 

  52. McCarroll, S. A. et al. Deletion polymorphism upstream of IRGM associated with altered IRGM expression and Crohn’s disease. Nat. Genet. 40, 1107–1112 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Carvalho, F. A. et al. Crohn’s disease adherent-invasive Escherichia coli colonize and induce strong gut inflammation in transgenic mice expressing human CEACAM. J. Exp. Med. 206, 2179–2189 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Chowers, M. Y. et al. Optimal infectivity in vitro of human immunodeficiency virus type 1 requires an intact nef gene. J. Virol. 68, 2906–2914 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Huang, R. et al. Deacetylation of nuclear LC3 drives autophagy initiation under starvation. Mol. Cell 57, 456–466 (2015).

    Article  CAS  PubMed  Google Scholar 

  56. Inoki, K., Kim, J. & Guan, K. L. AMPK and mTOR in cellular energy homeostasis and drug targets. Annu. Rev. Pharmacol. Toxicol. 52, 381–400 (2012).

    Article  CAS  PubMed  Google Scholar 

  57. O’Neill, L. A., Kishton, R. J. & Rathmell, J. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 16, 553–565 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Faeste, C. K. et al. Characterisation of potential novel allergens in the fish parasite Anisakis simplex. EuPA Open Proteom. 4, 140–155 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Faeste, C. K. et al. Development of liquid chromatography–tandem mass spectrometry methods for the quantitation of Anisakis simplex proteins in fish. J. Chromatogr. A 1432, 58–72 (2016).

    Article  CAS  PubMed  Google Scholar 

  60. Anonsen, J. H. et al. Novel protein substrates of the phospho-form modification system in Neisseria gonorrhoeae and their connection to O-linked protein glycosylation. Infect. Immun. 80, 22–30 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Olivetta, E. & Federico, M. HIV-1 Nef protects human-monocyte-derived macrophages from HIV-1-induced apoptosis. Exp. Cell Res. 312, 890–900 (2006).

    Article  CAS  PubMed  Google Scholar 

  62. Aiken, C. & Trono, D. Nef stimulates human immunodeficiency virus type 1 proviral DNA synthesis. J. Virol. 69, 5048–5056 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Aiken, C. Pseudotyping human immunodeficiency virus type 1 (HIV-1) by the glycoprotein of vesicular stomatitis virus targets HIV-1 entry to an endocytic pathway and suppresses both the requirement for Nef and the sensitivity to cyclosporin A. J. Virol. 71, 5871–5877 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

V.D. dedicates this work to memory of Beth Levine. We thank R. Puertollano, S. Ferguson and A. Ballabio for the TFEB constructs. This work was supported by NIH grants R37AI042999 and R01AI111935 and center grant P20GM121176 to V.D. T.-E.R and A.J. were supported by grants 262652 and 276070 from the Norwegian Research Council.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: S.K. and V.D. Formal analysis: S.K., A.J., S.W.C., G.P.D.d.S., L.A., M.H.M., R.S.P. and V.D. Investigation and validation: S.K., S.W.C., A.J., M.H.M., M.L., T.-E.R., G.P.D.d.S. and V.D. Resources: V.D., T.-E.R. and M.L. Data curation: L.A. and J.H.A. Writing (original draft): S.K. and V.D. Writing (reviewing and editing): T.-E.R. and M.L. Visualization: S.K., A.J. and V.D. Supervision: V.D. and T.-E.R. Project administration and funding acquisition: V.D.

Corresponding author

Correspondence to Vojo Deretic.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 IRGM affects nuclear translocation of TFEB.

a, confocal microscopy analysis of effects of IRGM KD on TFEB nuclear translocation in response to 2 h starvation. Scale bar 5 µm, (n = 3 biologically independent experiments). b,c, HCM images and quantification to test the effect of IRGM KD on nuclear translocation of TFEB. Cells were permeabilized with Triton. Data, means ± SEM (n = 3 biologically independent experiments) ANOVA, Tukey’s post hoc test; high content microscopy, >500 cells counted per well; minimum number of valid wells 9. Masks; white: algorithm-defined cell boundaries; yellow outline: computer-identified colocalization between TFEB and Hoechst-33342 nuclear stain). Scale bar 10 µm. d,e, HCM images and quantifications to test the effect of IRGM KD on nuclear translocation of TFEB in cells treated with DMSO or pp242. Data, means ± SEM (n = 3 biologically independent experiments) ANOVA, Tukey’s post hoc test; high content microscopy, >500 cells counted per well; minimum number of valid wells. Masks; white: algorithm-defined cell boundaries; yellow outline: computer-identified colocalization between TFEB and Hoechst-33342 nuclear stain). Scale bar 10 µm. Numerical source data for panels b and d are provided in Source Data Extended Data Fig. 1.

Source data

Extended Data Fig. 2 Interactions and localization analyses of IRGM with MiT/TFE family of transcriptional regulators.

a, Confocal microscopy analysis of co-localization between GFP-IRGM and endogenous TFEB. Scale bar 5 µm, (n = 3 biologically independent experiments). b, A screenshot from NCBI showing domain of unknown function (DUF3371) in TFEB. c,d, HCM images and quantifications to analyze the effect of complementation of IRGM KD with GFP-IRGM WT or GFP-IRGM S47N on nuclear translocation of TFEB. Data, means ± SEM (n = 3 biologically independent experiments) ANOVA, Tukey’s post hoc test; HCM, > 500 cells counted per well; minimum number of valid wells 9, 3 independent experiments. Masks; white: algorithm-defined cell boundaries and computer-identified GFP positive cells; blue outline: computer-identified nuclear stain; yellow outline: computer-identified colocalization between TFEB and Hoechst-33342 nuclear stain). Scale bar 10 µm. The masks in gray scale panels are cloned from the merged images. Inset: western blot showing GFP-IRGM expression IRGM KD cells. e, Co-IP analysis of interactions between GFP-MiTF (H isoform) and FLAG-IRGM in 293T cells, (n = 3 biologically independent experiments). f, Co-IP analysis of interactions between GFP-TFE3 and FLAG-IRGM in 293T cells, (n = 3 biologically independent experiments). g,h, Co-IP analysis of interactions between GFP-IRGM WT or GFP-IRGM S47N with MiTF in 293T cells. Data, means ± SEM of normalized intensities (n = 3 biologically independent experiments) paired t-test. Uncropped blots for panels e, f and g and numerical source data for panels c and h are provided in Source Data Extended Data Fig. 2.

Source data

Extended Data Fig. 3 IRGM effects on mTOR and calcineurin and mAtg8s interactions with and effects on TFEB.

a, b, Western blot analysis and quantifications of the effects of IRGM KD on pTFEB (S211) levels in cells treated with pp242. Data, means ± SEM of normalized intensities (n = 3 biologically independent experiments) ANOVA, Tukey’s post hoc test. c-e, western blots analysis of the effects of IRGM on mTOR substrates pS6K and pULK1. Data, means ± SEM of normalized intensities (n = 3 biologically independent experiments) paired t-test. f, HCM image analysis of co-localization between mTOR and LAMP2. Scale bar 10 µm. g,h, HCM analysis of the effects of IRGM KD on LAMP2 puncta. Data, means ± SEM; (n = 3 biologically independent experiments) paired t-test. Scale bar 10 µm. i, HCM analysis of the effect of IRGM expression on cells expressing RagBQ99L and parental 293 T cells on nuclear translocation of TFEB, (n = 3 biologically independent experiments). Scale bar 10 µm. j, confocal microscopy analysis of co-localization between GFP-IRGM and endogenous PPP3CB in HeLa cells (n = 3 biologically independent experiments). Scale bar 5 µm. k,l, HCM analysis of the effect of starvation on colocalization between GFP-IRGM and PPP3CB. Data, means ± SEM (n = 3 biologically independent experiments) paired t-test. Scale bar 10 µm. m, western blot showing PPP3CB KD in HeLa cells (n = 3 biologically independent experiments). n, schematics of LysoIP technique. o, LysoIP to detect indicated proteins on lysosomes (n = 3 biologically independent experiments). p, western blot analysis of the effects of IRGM expression on NFAT mobility shift (n = 3 biologically independent experiments). q, Co-IP analysis of GFP-LC3B and GFP-GABARAP with FLAG-TFEB in 293 T cells. r, GST pull-down analysis of TFEB with WT or LDS mutant of GABARAP. s, HCM images in WT or indicated KO cells in full medium (n = 3 biologically independent experiments). Scale bar 10 µm. Uncropped blots for panels a, c, o, p, q and r and numerical source data for panels b, d, e, g and l are provided in Source Data Extended Data Fig. 3.

Source data

Extended Data Fig. 4 GABARAP and GABARAPL1 but not GABARAPL2 control nuclear translocation of TFEB.

a,b, HCM images and quantifications to test the role of mAtg8s on nuclear translocation of GFP-MiTF in response to autophagy induction (EBSS 2 h). Data, means ± SEM (n = 3 biologically independent experiments) paired t-test. Masks; white: algorithm-defined cell boundaries; blue outline: computer-identified nuclear stain; yellow outline: computer-identified colocalization between TFEB and Hoechst-33342 nuclear stain). Scale bar 10 µm. The masks in gray scale panels are cloned from the merged images, (n = 3 biologically independent experiments). c, HCM image analysis of effects of complementation of HexaKO with GFP-GABARAP on nuclear translocation of TFEB. Scale bar 10 µm. d-g, HCM analysis of the effect of complementation of HexaKO cells with GABARAPL1 or GABARAPL2 on nuclear translocation of TFEB. Data, means ± SEM, ANOVA, Tukey’s post hoc test; HCM, > 500 cells counted per well; minimum number of valid wells 9, (n = 3 biologically independent experiments). Scale bar 10 µm. h, HCM analysis of effect of expression of GABARAP in 293 T cells expressing RagBQ99L or parental 293 T cells on nuclear translocation of TFEB. Masks in c, e, g, h; white: algorithm-defined cell boundaries in GFP positive cells; blue outline: computer-identified nuclear stain; yellow outline: computer-identified co-localization between TFEB and Hoechst-33342 nuclear stain), (n = 3 biologically independent experiments). Scale bar 10 µm. Numerical source data for panels a, d, and f are provided in Source Data Extended Data Fig. 4.

Source data

Extended Data Fig. 5 mAtg8s affect global gene expression.

a, Volcano plot (RNAseq) showing the effect of pan-mAtg8 knockout on differential gene expression (log2 fold change; ratio HeLa HexaKO/HeLaWT). Red points: down-regulated genes in HexaKO cells. Green points: upregulated in HexaKO cells. A subset of genes not identified as TFEB targets are named. Dotted orange line, significance cuttof (p value < 0.05). P values were calculated using Fisher’s exact test adapted for over-dispersed data; edgeR models read counts with negative binomial (NB) distribution (see Methods). (n = 3 biologically independent experiments). b, Heat map representation of genes upregulated or downregulated in HeLaWT vs. HexaKO cells. c, A volcano plot showing RNAseq analysis of HeLaWT vs. ATG3KO cells. P values were calculated using Fisher’s exact test using R package. Named genes are previously identified TFEB targets those were also down-regulated in HexaKO shown in Fig. 5c. (n = 3 biologically independent experiments). d, A volcano plot (RNAseq) listing upregulated and downregulated autophagy-related genes in HeLaWT vs. HexaKO cells. P values were calculated using Fisher’s exact test adapted for over-dispersed data; edgeR models read counts with negative binomial (NB) distribution (see Methods). (n = 3 biologically independent experiments). e, qRT-PCR analysis of p62, ATG9B and ULK1 in HeLaWT vs. HexaKO cells induced for autophagy in EBSS for 2 h; 18 S was used as an internal control, Data, means ± SEM (n = 3 biologically independent experiments). Numerical source data for panel e are provided in Source Data Extended Data Fig. 5.

Source data

Extended Data Fig. 6 mAtg8s affect calcium fluxes and Stx17 affects mTOR and TFEB.

a, A volcano plot showing expression of calcium effectors in HexaKO cells. P values were calculated using Fisher’s exact test adapted for over-dispersed data (see Methods) (n = 3 biologically independent experiments). b,c Flow cytometry using FLUO-3AM to detect intracellular calcium in HeLaWT or HexaKo. Data, means ± SEM; (n = 3 biologically independent experiments) ANOVA, Tukey’s post hoc test. d, Confocal microscopy analysis of the effects of Stx17KO on TFEB localization, (n = 3 biologically independent experiments). Scale bar 5 µm. e-g, confocal microscopy (e) and HCM (f,g) analyses of the effects of Stx17KO on colocalization between TFEB and LAMP2. Scale bar 5 µm (e). Scale bar 10 µm (f). Data, means ± SEM; (n = 3 biologically independent experiments) ANOVA, Tukey’s post hoc test. h, HCM analysis of the effects of Stx17KO on TFEB puncta. Data, means ± SEM; (n = 3 biologically independent experiments) ANOVA, Tukey’s post hoc test. i, Co-IP analysis of interactions between GFP-Stx17 and FLAG-TFEB in 293 T cells (n = 3 biologically independent experiments). j,k, Co-IP analysis of effects of GFP-Stx17 on FLAG-TFEB and IRGM complexes. Data, means ± SEM (n = 3 biologically independent experiments) paired t-test. l,m, MS analysis showing 14-3-3 peptides those interacted with GFP or GFP-Stx17 and GFP-IRGM (n = 3 biologically independent experiments). n-p, Western blot analysis and quantification of the effect of GFP-Stx17 in HeLaWT (full media) or in Stx17KO cells (EBSS 2 h) on mTOR activity. Data, means ± SEM; (n = 3 biologically independent experiments) ANOVA, Tukey’s post hoc test. q-s, Western blot analysis and quantification of pULK1 and pS6K to test the effects of GFP-Stx17 expression in WT 293 T cells and cells expressing RagBQ99L. Data, means ± SEM; (n = 3 biologically independent experiments) ANOVA, Tukey’s post hoc test. t-w, Co-IP analysis of interactions between RagA and FLAG-p18 (t,u) and Raptor and FLAG-RagA (v-w) in Stx17KO or parental HeLa cells. Data, means ± SEM of normalized intensities (n = 3 biologically independent experiments) paired t-test. Uncropped blots for panels i, j, n, q, t and v and numerical source data for panels b, f, h, k, p, o, r, s, u and w are provided in Source Data Extended Data Fig. 6.

Source data

Extended Data Fig. 7 mIR196B affects protective CD variant of IRGM in its role in nuclear translocation of TFEB.

a,b, HCM analysis of the effects of miR196B (shown to downregulate CD protective IRGM variant) and miR20 (control) transfection on TFEB nuclear localization in 293 T cells (c.313 C). HCM (n = 3 biologically independent experiments); >500 primary objects examined per well; minimum number of wells, 9). Masks; white: algorithm-defined cell boundaries; blue: computer-identified nucleus; yellow outline: computer-identified colocalization between TFEB and Hoechst-33342 nuclear stain). Images, a detail from a large database of machine-collected and computer-processed images. Data, means ± SEM; (n = 3 biologically independent experiments) ANOVA, Tukey’s post hoc test. Scale bar 10 µm. c, HCM image analysis of the effects of IRGM KD on AIEC LF82 influenced nuclear translocation of TFEB. K12 was used as control, (n = 3 biologically independent experiments). Scale bar 10 µm. d,e, HC microscopy and quantifications to analyze the effect of HIV infection on TFEB localization in HeLa cells transfected with scramble siRNA or IRGM siRNA. HC microscopy (n = 3 biologically independent experiments; >500 primary objects examined per well; minimum number of wells, 12). Masks; white: algorithm-defined cell boundaries; blue: computer-identified nucleus; yellow outline: computer-identified colocalization between TFEB and Hoechst-33342 nuclear stain). Images, a detail from a large database of machine-collected and computer-processed images. Data, means ± SEM; (n = 3 biologically independent experiments) ANOVA, Tukey’s post hoc test. Scale bar 10 µm. f, The model summarizes the effects of IRGM, Stx17 and mAtg8s/GABARAPs on mTOR inhibition and calcineurin (CN) activation promoting nuclear translocation of TFEB. L, lysosome. Numerical source data for panels a and d are provided in Source Data Extended Data Fig. 7.

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Table 1: RNA-seq analysis (reported as differential gene expression) of HeLa WT and HexaKO cells determining the effects of the deletion of all mAtg8s (HexaKO) on differential gene expression. The cells were incubated in EBSS for 2 h. Previously known TFEB targets are highlighted in yellow. Tab 2 is the same as tab 1, but reporting differential transcript expression (n = 3 biologically independent experiments). Supplementary Table 2: RNA-seq analysis (reported as differential gene expression) of HeLa WT and GABATKO determining the effects of the deletion of all GABARAPs on differential gene expression. The cells were incubated in EBSS for 2 h. Previously known TFEB targets are highlighted in yellow. Tab 2 is the same as tab 1, but reporting differential transcript expression (n = 3 biologically independent experiments). Supplementary Table 3: tab 5 shows RNA-seq analysis (reported as differential gene expression) of HeLa WT and ATG3KO determining the effects of the deletion of ATG3 on differential gene expression. The cells were incubated in EBSS for 2 h. Previously known TFEB targets are highlighted in yellow. Tab 2 is the same as tab 1, but reporting differential transcript expression (n = 3 biologically independent experiments). Supplementary Table 4: mass spectrometry analyses of GFP–IRGM interactors (n = 3 biologically independent experiments).

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Unprocessed blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 6

Unprocessed blots.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed blots.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kumar, S., Jain, A., Choi, S.W. et al. Mammalian Atg8 proteins and the autophagy factor IRGM control mTOR and TFEB at a regulatory node critical for responses to pathogens. Nat Cell Biol 22, 973–985 (2020). https://doi.org/10.1038/s41556-020-0549-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-020-0549-1

This article is cited by

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