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:

An antibody for analysis of autophagy induction

Abstract

Autophagy is a degradative program that maintains cellular homeostasis. Autophagy defects have been described in numerous diseases. However, analysis of autophagy rates can be challenging, particularly in rare cell populations or in vivo, due to limitations in currently available tools for measuring autophagy induction. Here, we describe a method to monitor autophagy by measuring phosphorylation of the protein ATG16L1. We developed and characterized a monoclonal antibody that can detect phospho-ATG16L1 endogenously in mammalian cells. Importantly, phospho-ATG16L1 is only present on newly forming autophagosomes. Therefore, its levels are not affected by prolonged stress or late-stage autophagy blocks, which can confound autophagy analysis. Moreover, we show that ATG16L1 phosphorylation is a conserved signaling pathway activated by numerous autophagy-inducing stressors. The described antibody is suitable for western blot, immunofluorescence and immunohistochemistry, and measured phospho-ATG16L1 levels directly correspond to autophagy rates. Taken together, this phospho-antibody represents an exciting tool to study autophagy induction.

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: Phosphorylation of ATG16L1 is a conserved indicator of autophagy induction, which is activated by multiple stimuli.
Fig. 2: pATG16L1 can be analyzed by immunofluorescence and is recruited to the expanding autophagosomal membrane.
Fig. 3: pATG16L1 level provides a reliable measurement of autophagy rates independent of late-stage autophagy block and directly reflects autophagic vesicle formation.
Fig. 4: pATG16L1 is compatible with IHC staining of tissue samples to measure autophagic activities in vivo.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Parzych, K. R. & Klionsky, D. J. An overview of autophagy: morphology, mechanism, and regulation. Antioxid. Redox Signal. 20, 460–473 (2014).

    Article  CAS  Google Scholar 

  2. Russell, R. C., Yuan, H. X. & Guan, K. L. Autophagy regulation by nutrient signaling. Cell Res. 24, 42–57 (2014).

    Article  CAS  Google Scholar 

  3. Kuma, A. et al. The role of autophagy during the early neonatal starvation period. Nature 432, 1032–1036 (2004).

    Article  CAS  Google Scholar 

  4. 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  Google Scholar 

  5. Mercer, C. A., Kaliappan, A. & Dennis, P. B. A novel, human Atg13 binding protein, Atg101, interacts with ULK1 and is essential for macroautophagy. Autophagy 5, 649–662 (2009).

    Article  CAS  Google Scholar 

  6. Suzuki, H., Kaizuka, T., Mizushima, N. & Noda, N. N. Structure of the Atg101–Atg13 complex reveals essential roles of Atg101 in autophagy initiation. Nat. Struct. Mol. Biol. 22, 572–580 (2015).

    Article  CAS  Google Scholar 

  7. Hara, T. et al. FIP200, a ULK-interacting protein, is required for autophagosome formation in mammalian cells. J. Cell Biol. 181, 497–510 (2008).

    Article  CAS  Google Scholar 

  8. Ganley, I. G. et al. ULK1.ATG13.FIP200 complex mediates mTOR signaling and is essential for autophagy. J. Biol. Chem. 284, 12297–12305 (2009).

    Article  CAS  Google Scholar 

  9. Hosokawa, N. et al. Nutrient-dependent mTORC1 association with the ULK1–Atg13–FIP200 complex required for autophagy. Mol. Biol. Cell 20, 1981–1991 (2009).

    Article  CAS  Google Scholar 

  10. 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 

  11. 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 

  12. Gammoh, N., Florey, O., Overholtzer, M. & Jiang, X. Interaction between FIP200 and ATG16L1 distinguishes ULK1 complex-dependent and -independent autophagy. Nat. Struct. Mol. Biol. 20, 144–149 (2013).

    Article  CAS  Google Scholar 

  13. Nishimura, T. et al. FIP200 regulates targeting of Atg16L1 to the isolation membrane. EMBO Rep. 14, 284–291 (2013).

    Article  CAS  Google Scholar 

  14. Fujita, N. et al. The Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy. Mol. Biol. Cell 19, 2092–2100 (2008).

    Article  CAS  Google Scholar 

  15. Wu, J. et al. Molecular cloning and characterization of rat LC3A and LC3B—two novel markers of autophagosome. Biochem. Biophys. Res. Commun. 339, 437–442 (2006).

    Article  CAS  Google Scholar 

  16. Kabeya, Y. et al. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 19, 5720–5728 (2000).

    Article  CAS  Google Scholar 

  17. Kimura, S., Fujita, N., Noda, T. & Yoshimori, T. Monitoring autophagy in mammalian cultured cells through the dynamics of LC3. Methods Enzymol. 452, 1–12 (2009).

    Article  CAS  Google Scholar 

  18. Klionsky, D. J. et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 12, 1–222 (2016).

  19. Jain, A. et al. p62/SQSTM1 is a target gene for transcription factor NRF2 and creates a positive feedback loop by inducing antioxidant response element-driven gene transcription. J. Biol. Chem. 285, 22576–22591 (2010).

    Article  CAS  Google Scholar 

  20. Fujita, K. & Srinivasula, S. M. TLR4-mediated autophagy in macrophages is a p62-dependent type of selective autophagy of aggresome-like induced structures (ALIS). Autophagy 7, 552–554 (2011).

    Article  CAS  Google Scholar 

  21. Salminen, A. et al. Emerging role of p62/sequestosome-1 in the pathogenesis of Alzheimer’s disease. Prog. Neurobiol. 96, 87–95 (2012).

    Article  CAS  Google Scholar 

  22. Jiang, X. et al. VPS34 stimulation of p62 phosphorylation for cancer progression. Oncogene 36, 6850–6862 (2017).

    Article  CAS  Google Scholar 

  23. Clausen, T. H. et al. p62/SQSTM1 and ALFY interact to facilitate the formation of p62 bodies/ALIS and their degradation by autophagy. Autophagy 6, 330–344 (2010).

    Article  CAS  Google Scholar 

  24. Kirkin, V., Lamark, T., Johansen, T. & Dikic, I. NBR1 cooperates with p62 in selective autophagy of ubiquitinated targets. Autophagy 5, 732–733 (2009).

    Article  CAS  Google Scholar 

  25. Shi, J. et al. NBR1 is dispensable for PARK2-mediated mitophagy regardless of the presence or absence of SQSTM1. Cell Death Dis. 6, e1943 (2015).

    Article  CAS  Google Scholar 

  26. Yla-Anttila, P., Vihinen, H., Jokitalo, E. & Eskelinen, E. L. Monitoring autophagy by electron microscopy in mammalian cells. Methods Enzymol. 452, 143–164 (2009).

    Article  CAS  Google Scholar 

  27. Roberts, E. A. & Deretic, V. Autophagic proteolysis of long-lived proteins in nonliver cells. Methods Mol. Biol. 445, 111–117 (2008).

    Article  CAS  Google Scholar 

  28. Ueno, T. et al. Autolysosomal membrane-associated betaine homocysteine methyltransferase. Limited degradation fragment of a sequestered cytosolic enzyme monitoring autophagy. J. Biol. Chem. 274, 15222–15229 (1999).

    Article  CAS  Google Scholar 

  29. Warnes, G. Flow cytometric assays for the study of autophagy. Methods 82, 21–28 (2015).

    Article  CAS  Google Scholar 

  30. Guo, S. et al. A rapid and high content assay that measures cyto-ID-stained autophagic compartments and estimates autophagy flux with potential clinical applications. Autophagy 11, 560–572 (2015).

    Article  Google Scholar 

  31. Alsaadi, R. M. et al. ULK1-mediated phosphorylation of ATG16L1 promotes xenophagy, but destabilizes the ATG16L1 Crohn’s mutant. EMBO Rep. 20, e46885 (2019).

    Article  Google Scholar 

  32. Diamanti, M. A. et al. IKKɑ controls ATG16L1 degradation to prevent ER stress during inflammation. J. Exp. Med 214, 423–437 (2017).

    Article  CAS  Google Scholar 

  33. Koyama-Honda, I., Itakura, E., Fujiwara, T. K. & Mizushima, N. Temporal analysis of recruitment of mammalian ATG proteins to the autophagosome formation site. Autophagy 9, 1491–1499 (2013).

    Article  CAS  Google Scholar 

  34. 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  Google Scholar 

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

  36. Sahani, M. H., Itakura, E. & Mizushima, N. Expression of the autophagy substrate SQSTM1/p62 is restored during prolonged starvation depending on transcriptional upregulation and autophagy-derived amino acids. Autophagy 10, 431–441 (2014).

    Article  CAS  Google Scholar 

  37. Mizushima, N., Yamamoto, A., Matsui, M., Yoshimori, T. & Ohsumi, Y. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol. Biol. Cell 15, 1101–1111 (2004).

    Article  CAS  Google Scholar 

  38. Rosenfeldt, M. T., Nixon, C., Liu, E., Mah, L. Y. & Ryan, K. M. Analysis of macroautophagy by immunohistochemistry. Autophagy 8, 963–969 (2012).

    Article  CAS  Google Scholar 

  39. Xi, Y. et al. Knockout of Atg5 delays the maturation and reduces the survival of adult-generated neurons in the hippocampus. Cell Death Dis. 7, e2127 (2016).

    Article  CAS  Google Scholar 

  40. Hoffman, M. A. et al. von Hippel-Lindau protein mutants linked to type 2C VHL disease preserve the ability to downregulate HIF. Hum. Mol. Genet. 10, 1019–1027 (2001).

    Article  CAS  Google Scholar 

  41. Guo, H. et al. Atg5 disassociates the V1V0-ATPase to promote exosome production and tumor metastasis independent of canonical macroautophagy. Dev. Cell 43, 716–730 e717 (2017).

    Article  CAS  Google Scholar 

  42. Mauthe, M. et al. Resveratrol-mediated autophagy requires WIPI-1-regulated LC3 lipidation in the absence of induced phagophore formation. Autophagy 7, 1448–1461 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the uOttawa PALM-Histology Core Facility for processing the IHC tissue samples, the Cell Biology and Image Acquisition Core (CBIA) for assistance in three-dimensional reconstruction and rendering of brain imaging, N. Vernoux for technical assistance in electron microcopy, J.A. Lunde for technical assistance in collecting mouse skeletal muscle samples and members of the Russell laboratory for advice and critical reading of this manuscript. This work was supported by a Canadian Institutes of Health Research Project Grant awarded to R.C.R. (grant no. PJT153034), funding from the Canada Foundation for Innovation to the CBIA core and a Canada Research Chair Tier 2 to M.T.

Author information

Authors and Affiliations

Authors

Contributions

W.T. and R.C.R. wrote the manuscript. W.T. was primarily responsible for data production in all figures except for immunofluorescence and IHC panels in Figs. 2 and 4. R.A. assisted with immunofluorescence experiments and data analysis. Z.G. was responsible for optimizing pATG16L1 antibody protocols for immunofluorescence and IHC. A.K. and D.L. provided the mice used for the study and assisted with IHC experimental planning and tissue staining. M.C. and M.E.T. imaged EM samples. B.L. provided expertise and guidance for EM sample preparation. R.C.R. oversaw manuscript planning and conceived of the study.

Corresponding author

Correspondence to Ryan C. Russell.

Ethics declarations

Competing interest

The authors declare no competing interests.

Additional information

Peer review information Nicole Rusk and Rita Strack were the primary editors on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Supplementary information

Supplementary Information

Supplementary Figs. 1–5 and results.

Reporting Summary

Supplementary Video

Brain hippocampus tissue sections of wild-type and Atg5flox/flox mice were stained with pATG16L1S278 GFP and RFP signals were enhanced with anti-GFP/RFP antibodies. GFP expression is indicative of cells knocked out of Atg5. 3D model of the cell was constructed using the 3D reconstruction function in Imaris based on GRP and RFP signals. N = 2 animals per group. Representative cell from one conditional Atg5 KO mouse sample is shown in the video.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tian, W., Alsaadi, R., Guo, Z. et al. An antibody for analysis of autophagy induction. Nat Methods 17, 232–239 (2020). https://doi.org/10.1038/s41592-019-0661-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-019-0661-y

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