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:

Recycling of autophagosomal components from autolysosomes by the recycler complex

Abstract

Autolysosomes contain components from autophagosomes and lysosomes. The contents inside the autolysosomal lumen are degraded during autophagy, while the fate of autophagosomal components on the autolysosomal membrane remains unknown. Here we report that the autophagosomal membrane components are not degraded, but recycled from autolysosomes through a process coined in this study as autophagosomal components recycling (ACR). We further identified a multiprotein complex composed of SNX4, SNX5 and SNX17 essential for ACR, which we termed ‘recycler’. In this, SNX4 and SNX5 form a heterodimer that recognizes autophagosomal membrane proteins and is required for generating membrane curvature on autolysosomes, both via their BAR domains, to mediate the cargo sorting process. SNX17 interacts with both the dynein–dynactin complex and the SNX4–SNX5 dimer to facilitate the retrieval of autophagosomal membrane components. Our discovery of ACR and identification of the recycler reveal an important retrieval and recycling pathway on autolysosomes.

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

Fig. 1: STX17 is retrieved from autolysosomes.
Fig. 2: SNX4 is required for STX17 recycling from autolysosomes.
Fig. 3: SNX5 is required for STX17 recycling from autolysosomes.
Fig. 4: SNX5 cooperates with SNX4 to mediate STX17 sorting from autolysosomes.
Fig. 5: SNX17 interacts with both the STX17–SNX4–SNX5 and dynein–dynactin complexes to facilitate STX17 retrieval.
Fig. 6: SNX4–SNX5–SNX17 forms the recycler complex.
Fig. 7: ATG9A is retrieved from autolysosomes by recycler.
Fig. 8: Inhibited autophagic flux in recycler-deficient cells is rescued by SNX4, but not SNX4 Y252A, cells.

Similar content being viewed by others

Data availability

Source data are provided with this paper. The authors declare that all relevant data supporting the findings of this study are available within the paper and its Supplementary Information files. The proteomics data have been deposited in the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD031183.

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. Bento, C. F. et al. Mammalian autophagy: how does it work? Annu. Rev. Biochem. 85, 685–713 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Yu, L., Chen, Y. & Tooze, S. A. Autophagy pathway: cellular and molecular mechanisms. Autophagy 14, 207–215 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Dikic, I. & Elazar, Z. Mechanism and medical implications of mammalian autophagy. Nat. Rev. Mol. Cell Biol. 19, 349–364 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Zhao, Y. G. & Zhang, H. Core autophagy genes and human diseases. Curr. Opin. Cell Biol. 61, 117–125 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Mizushima, N. The ATG conjugation systems in autophagy. Curr. Opin. Cell Biol. 63, 1–10 (2019).

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Poillet-Perez, L. & White, E. Role of tumor and host autophagy in cancer metabolism. Genes Dev. 33, 610–619 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Yang, Y. & Klionsky, D. J. Autophagy and disease: unanswered questions. Cell Death Differ. 27, 858–871 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Gomez-Sanchez, R., Tooze, S. A. & Reggiori, F. Membrane supply and remodeling during autophagosome biogenesis. Curr. Opin. Cell Biol. 71, 112–119 (2021).

    Article  CAS  PubMed  Google Scholar 

  11. Axe, E. L. et al. Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum. J. Cell Biol. 182, 685–701 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Hayashi-Nishino, M. et al. A subdomain of the endoplasmic reticulum forms a cradle for autophagosome formation. Nat. Cell Biol. 11, 1433–1437 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Yla-Anttila, P., Vihinen, H., Jokitalo, E. & Eskelinen, E. L. 3D tomography reveals connections between the phagophore and endoplasmic reticulum. Autophagy 5, 1180–1185 (2009).

    Article  PubMed  Google Scholar 

  14. Zoppino, F. C., Militello, R. D., Slavin, I., Alvarez, C. & Colombo, M. I. Autophagosome formation depends on the small GTPase Rab1 and functional ER exit sites. Traffic 11, 1246–1261 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. Graef, M., Friedman, J. R., Graham, C., Babu, M. & Nunnari, J. ER exit sites are physical and functional core autophagosome biogenesis components. Mol. Biol. Cell 24, 2918–2931 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hailey, D. W. et al. Mitochondria supply membranes for autophagosome biogenesis during starvation. Cell 141, 656–667 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ravikumar, B., Moreau, K., Jahreiss, L., Puri, C. & Rubinsztein, D. C. Plasma membrane contributes to the formation of pre-autophagosomal structures. Nat. Cell Biol. 12, 747–757 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Moreau, K., Ravikumar, B., Renna, M., Puri, C. & Rubinsztein, D. C. Autophagosome precursor maturation requires homotypic fusion. Cell 146, 303–317 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Young, A. R. et al. Starvation and ULK1-dependent cycling of mammalian Atg9 between the TGN and endosomes. J. Cell Sci. 119, 3888–3900 (2006).

    Article  CAS  PubMed  Google Scholar 

  20. Geng, J., Nair, U., Yasumura-Yorimitsu, K. & Klionsky, D. J. Post-Golgi Sec proteins are required for autophagy in Saccharomyces cerevisiae. Mol. Biol. Cell 21, 2257–2269 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Guo, Y. et al. AP1 is essential for generation of autophagosomes from the trans-Golgi network. J. Cell Sci. 125, 1706–1715 (2012).

    CAS  PubMed  Google Scholar 

  22. Longatti, A. et al. TBC1D14 regulates autophagosome formation via Rab11- and ULK1-positive recycling endosomes. J. Cell Biol. 197, 659–675 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ge, L., Melville, D., Zhang, M. & Schekman, R. The ER–Golgi intermediate compartment is a key membrane source for the LC3 lipidation step of autophagosome biogenesis. eLife 2, e00947 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Ge, L., Zhang, M. & Schekman, R. Phosphatidylinositol 3-kinase and COPII generate LC3 lipidation vesicles from the ER–Golgi intermediate compartment. eLife 3, e04135 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Ge, L. et al. Remodeling of ER-exit sites initiates a membrane supply pathway for autophagosome biogenesis. EMBO Rep. 18, 1586–1603 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Yu, L. et al. Termination of autophagy and reformation of lysosomes regulated by mTOR. Nature 465, 942–946 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Rong, Y. et al. Spinster is required for autophagic lysosome reformation and mTOR reactivation following starvation. Proc. Natl Acad. Sci. USA 108, 7826–7831 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Rong, Y. et al. Clathrin and phosphatidylinositol-4,5-bisphosphate regulate autophagic lysosome reformation. Nat. Cell Biol. 14, 924–934 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Du, W. et al. Kinesin 1 drives autolysosome tubulation. Dev. Cell 37, 326–336 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. 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 

  31. Takats, S. et al. Autophagosomal Syntaxin17-dependent lysosomal degradation maintains neuronal function in Drosophila. J. Cell Biol. 201, 531–539 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Tsuboyama, K. et al. The ATG conjugation systems are important for degradation of the inner autophagosomal membrane. Science 354, 1036–1041 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Uematsu, M., Nishimura, T., Sakamaki, Y., Yamamoto, H. & Mizushima, N. Accumulation of undegraded autophagosomes by expression of dominant-negative STX17 (syntaxin 17) mutants. Autophagy 13, 1452–1464 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Traer, C. J. et al. SNX4 coordinates endosomal sorting of TfnR with dynein-mediated transport into the endocytic recycling compartment. Nat. Cell Biol. 9, 1370–1380 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. van Weering, J. R. et al. Molecular basis for SNX-BAR-mediated assembly of distinct endosomal sorting tubules. EMBO J. 31, 4466–4480 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Anton, Z. et al. A heterodimeric SNX4–SNX7 SNX-BAR autophagy complex coordinates ATG9A trafficking for efficient autophagosome assembly. J. Cell Sci. 133, jcs246306 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Arighi, C. N., Hartnell, L. M., Aguilar, R. C., Haft, C. R. & Bonifacino, J. S. Role of the mammalian retromer in sorting of the cation-independent mannose 6-phosphate receptor. J. Cell Biol. 165, 123–133 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wassmer, T. et al. A loss-of-function screen reveals SNX5 and SNX6 as potential components of the mammalian retromer. J. Cell Sci. 120, 45–54 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. Cullen, P. J. Endosomal sorting and signalling: an emerging role for sorting nexins. Nat. Rev. Mol. Cell Biol. 9, 574–582 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Wassmer, T. et al. The retromer coat complex coordinates endosomal sorting and dynein-mediated transport, with carrier recognition by the trans-Golgi network. Dev. Cell 17, 110–122 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Cullen, P. J. & Steinberg, F. To degrade or not to degrade: mechanisms and significance of endocytic recycling. Nat. Rev. Mol. Cell Biol. 19, 679–696 (2018).

    Article  CAS  PubMed  Google Scholar 

  42. Chen, K. E., Healy, M. D. & Collins, B. M. Towards a molecular understanding of endosomal trafficking by Retromer and Retriever. Traffic 20, 465–478 (2019).

    Article  CAS  PubMed  Google Scholar 

  43. Peter, B. J. et al. BAR domains as sensors of membrane curvature: the amphiphysin BAR structure. Science 303, 495–499 (2004).

    Article  CAS  PubMed  Google Scholar 

  44. McMahon, H. T. & Gallop, J. L. Membrane curvature and mechanisms of dynamic cell membrane remodelling. Nature 438, 590–596 (2005).

    Article  CAS  PubMed  Google Scholar 

  45. Suetsugu, S., Toyooka, K. & Senju, Y. Subcellular membrane curvature mediated by the BAR domain superfamily proteins. Semin. Cell Dev. Biol. 21, 340–349 (2010).

    Article  CAS  PubMed  Google Scholar 

  46. van Kerkhof, P. et al. Sorting nexin 17 facilitates LRP recycling in the early endosome. EMBO J. 24, 2851–2861 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Steinberg, F., Heesom, K. J., Bass, M. D. & Cullen, P. J. SNX17 protects integrins from degradation by sorting between lysosomal and recycling pathways. J. Cell Biol. 197, 219–230 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. McNally, K. E. et al. Retriever is a multiprotein complex for retromer-independent endosomal cargo recycling. Nat. Cell Biol. 19, 1214–1225 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Engelender, S. et al. Huntingtin-associated protein 1 (HAP1) interacts with the p150Glued subunit of dynactin. Hum. Mol. Genet. 6, 2205–2212 (1997).

    Article  CAS  PubMed  Google Scholar 

  50. Hoogenraad, C. C. et al. Mammalian Golgi-associated Bicaudal-D2 functions in the dynein–dynactin pathway by interacting with these complexes. EMBO J. 20, 4041–4054 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Holleran, E. A. et al. beta III spectrin binds to the Arp1 subunit of dynactin. J. Biol. Chem. 276, 36598–36605 (2001).

    Article  CAS  PubMed  Google Scholar 

  52. Jordens, I. et al. The Rab7 effector protein RILP controls lysosomal transport by inducing the recruitment of dynein–dynactin motors. Curr. Biol. 11, 1680–1685 (2001).

    Article  CAS  PubMed  Google Scholar 

  53. Matanis, T. et al. Bicaudal-D regulates COPI-independent Golgi–ER transport by recruiting the dynein–dynactin motor complex. Nat. Cell Biol. 4, 986–992 (2002).

    Article  CAS  PubMed  Google Scholar 

  54. Hoogenraad, C. C. et al. Bicaudal D induces selective dynein-mediated microtubule minus end-directed transport. EMBO J. 22, 6004–6015 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Gauthier, L. R. et al. Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell 118, 127–138 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Johansson, M. et al. Activation of endosomal dynein motors by stepwise assembly of Rab7-RILP-p150Glued, ORP1L, and the receptor betalll spectrin. J. Cell Biol. 176, 459–471 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Hong, Z. et al. The retromer component SNX6 interacts with dynactin p150Glued and mediates endosome-to-TGN transport. Cell Res. 19, 1334–1349 (2009).

    Article  CAS  PubMed  Google Scholar 

  58. Matsui, T. et al. Autophagosomal YKT6 is required for fusion with lysosomes independently of syntaxin 17. J. Cell Biol. 217, 2633–2645 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Yamamoto, H. et al. Atg9 vesicles are an important membrane source during early steps of autophagosome formation. J. Cell Biol. 198, 219–233 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Orsi, A. et al. Dynamic and transient interactions of Atg9 with autophagosomes, but not membrane integration, are required for autophagy. Mol. Biol. Cell 23, 1860–1873 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Reggiori, F. & Tooze, S. A. Autophagy regulation through Atg9 traffic. J. Cell Biol. 198, 151–153 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Klionsky, D. J., Elazar, Z., Seglen, P. O. & Rubinsztein, D. C. Does bafilomycin A1 block the fusion of autophagosomes with lysosomes? Autophagy 4, 849–850 (2008).

    Article  CAS  PubMed  Google Scholar 

  63. Rojas, R., Kametaka, S., Haft, C. R. & Bonifacino, J. S. Interchangeable but essential functions of SNX1 and SNX2 in the association of retromer with endosomes and the trafficking of mannose 6-phosphate receptors. Mol. Cell. Biol. 27, 1112–1124 (2007).

    Article  CAS  PubMed  Google Scholar 

  64. Weeratunga, S., Paul, B. & Collins, B. M. Recognising the signals for endosomal trafficking. Curr. Opin. Cell Biol. 65, 17–27 (2020).

    Article  CAS  PubMed  Google Scholar 

  65. Kvainickas, A. et al. Cargo-selective SNX-BAR proteins mediate retromer trimer independent retrograde transport. J. Cell Biol. 216, 3677–3693 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Simonetti, B., Danson, C. M., Heesom, K. J. & Cullen, P. J. Sequence-dependent cargo recognition by SNX-BARs mediates retromer-independent transport of CI-MPR. J. Cell Biol. 216, 3695–3712 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Elwell, C. A. et al. Chlamydia interfere with an interaction between the mannose-6-phosphate receptor and sorting nexins to counteract host restriction. eLife 6, e22709 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Simonetti, B. et al. Molecular identification of a BAR domain-containing coat complex for endosomal recycling of transmembrane proteins. Nat. Cell Biol. 21, 1219–1233 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Griffin, C. T., Trejo, J. & Magnuson, T. Genetic evidence for a mammalian retromer complex containing sorting nexins 1 and 2. Proc. Natl Acad. Sci. USA 102, 15173–15177 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ma, M., Burd, C. G. & Chi, R. J. Distinct complexes of yeast Snx4 family SNX-BARs mediate retrograde trafficking of Snc1 and Atg27. Traffic 18, 134–144 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Suzuki, S. W. & Emr, S. D. Membrane protein recycling from the vacuole/lysosome membrane. J. Cell Biol. 217, 1623–1632 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Ravussin, A., Brech, A., Tooze, S. A. & Stenmark, H. The phosphatidylinositol 3-phosphate-binding protein SNX4 controls ATG9A recycling and autophagy. J. Cell Sci. 134, jcs250670 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hamasaki, M. et al. Autophagosomes form at ER–mitochondria contact sites. Nature 495, 389–393 (2013).

    Article  CAS  PubMed  Google Scholar 

  74. Kumar, S. et al. Phosphorylation of syntaxin 17 by TBK1 controls autophagy initiation. Dev. Cell 49, 130–144 e136 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. 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 

  76. De Tito, S., Hervas, J. H., van Vliet, A. R. & Tooze, S. A. The Golgi as an assembly line to the autophagosome. Trends Biochem. Sci 45, 484–496 (2020).

    Article  PubMed  CAS  Google Scholar 

  77. Merino-Trigo, A. et al. Sorting nexin 5 is localized to a subdomain of the early endosomes and is recruited to the plasma membrane following EGF stimulation. J. Cell Sci. 117, 6413–6424 (2004).

    Article  CAS  PubMed  Google Scholar 

  78. Leprince, C. et al. Sorting nexin 4 and amphiphysin 2, a new partnership between endocytosis and intracellular trafficking. J. Cell Sci. 116, 1937–1948 (2003).

    Article  CAS  PubMed  Google Scholar 

  79. Hung, V. et al. Spatially resolved proteomic mapping in living cells with the engineered peroxidase APEX2. Nat. Protoc. 11, 456–475 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are deeply grateful to L. Yu (Tsinghua University), Q. Zhong (Shanghai Jiao Tong University), W. Liu (Zhejiang University), Q. Sun (Zhejiang University) and L. Ge (Tsinghua University) for helpful suggestions on this study. We thank J. Liu (Institute of Genetics and Developmental Biology, Chinese Academy of Sciences) for gifting plasmids. The work was supported by grants from NSFC 91854116 and 31771529 (to Y.R.) and the Junior Thousand Talents Program of China (to Y.R.). The work was partially supported by the Fundamental Research Funds for the Central Universities 5003510089 (to Y.R.).

Author information

Authors and Affiliations

Authors

Contributions

Y.R., C.Z. and Z.W. conceived and designed the experiments. C.Z., Z.W., H.Q. and Y.W. performed the biological and biochemical experiments. W.D. carried out the in vitro experiments. C.Z., Z.W., W.D., H.Q., Y.W. and Y.R. analysed the data and wrote the manuscript with the help of all authors.

Corresponding author

Correspondence to Yueguang Rong.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Noboru Mizushima and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 GFP-STX17-TM fails to get to lysosomes in FIP200 knock-out cells and ATG9A knock-out cells.

a and c, The delivery of STX17 to autolysosomes was blocked in FIP200-KO cells. Wild-type or FIP200-KO MEF cells stably expressing GFP-STX17-TM were starved with EBSS for 2 hours in a or treated with bafilomycin A1 (100 nM) for another 3 hours after 2 hours EBSS starvation in c. Cells were stained with antibodies against GFP and LAMP1. Scale bar, 5 μm. Inset scale bar, 2 μm. b and d, Images from a and c were analyzed, data are means ± s.d. (n = 3, 50 cells from 3 independent experiments were quantified). Unpaired two-tailed t-test. e and g, The delivery of STX17 to autolysosomes was blocked in ATG9-KO cells. Wild-type or ATG9-KO Hela cells stably expressing GFP-STX17-TM were starved with EBSS for 2 hours in e or treated with bafilomycin A1 (100 nM) for another 3 hours after 2 hours EBSS starvation in g. Then cells were stained with antibodies against GFP and LAMP1. Scale bar, 5 μm. Inset scale bar, 2 μm. f and h, Images e and g were analyzed. Data are means ± s.d. (n = 3, 50 cells from 3 independent experiments were quantified). Unpaired two-tailed t-test. Source numerical data are available in source data.

Source data

Extended Data Fig. 2 The full length STX17 fails to get to lysosomes in FIP200 knock-out cells and ATG9A knock-out cells.

a and c, The delivery of STX17 to autolysosomes is blocked in FIP200-KO cells. Wild-type or FIP200-KO MEF cells stably expressing Flag-STX17 were starved with EBSS for 2 hours in a or treated with bafilomycin A1 (100 nM) for another 3 hours after 2 hours EBSS starvation in c. Cells were stained with antibodies against Flag and LAMP1. Scale bar, 5 μm. Inset scale bar, 2 μm. b and d, Images from a and c were analyzed, data are means ± s.d. (n = 3, 50 cells from 3 independent experiments were quantified). Unpaired two-tailed t-test. e and g, The delivery of STX17 to autolysosomes is blocked in ATG9A-KO cells. Wild-type or ATG9-KO Hela cells stably expressing Flag-STX17 were starved with EBSS for 2 hours in e or treated with bafilomycin A1 (100 nM) for another 3 hours after 2 hours EBSS starvation in g. Then cells were stained with antibodies against Flag and LAMP1. Scale bar, 5 μm. Inset scale bar, 2 μm. f and h, Images from e and g are analyzed. Data are means ± s.d. (n = 3, 50 cells from 3 independent experiments were quantified). Unpaired two-tailed t-test. Source numerical data are available in source data.

Source data

Extended Data Fig. 3 STX17 is recycled from autolysosomes.

a, STX17 is recycled from autolysosomes. U2OS cells stably expressing GFP-STX17 TM, BFP-LC3 were starved for 2 h with EBSS and stained with LysoTracker. Time-lapse images were taken. Selected frames were shown as indicated time points. Scale bar, 1 μm. b-c, The recycling events and recycling frequency on autolysosomes were analyzed. Data are presented as mean values ± s.e.m., (n = 41 biologically independent experiments). d-f, Hela cells, HepG2 cells and COS-7 cells were transfected with GFP-STX17-TM. Twenty-four hours after transfection, cells stained with LysoTracker were starved and time-lapse images were taken. Selected frames were shown as indicated time points. Scale bar, 500 nm. Source numerical data are available in source data.

Source data

Extended Data Fig. 4 ALR genes are not required for STX17 recycling from autolysosomes.

a, ALR genes depletion has no effect on STX17 recycling from autolysosomes. U2OS cells stably expressing Flag-STX17 were transfected with indicated siRNAs. Forty-eight hours after transfection, cells were starved with EBSS for indicated hours and stained with antibodies against Flag and LAMP1. Scale bar, 5 μm. Inset scale bar, 2 μm. b, Quantification of STX17 positive autolysosomes in a. Data are means ± s.e.m. (n = 3, 50 cells from 3 independent experiments were quantified). Unpaired two-tailed t-test. Source numerical data are available in source data.

Source data

Extended Data Fig. 5 Bafilomycin A1 leads to STX17 entrance into autolysosomes.

a, The relative fluorescent intensity of STX17 and LysoTracker on autolysosomes and newly generated STX17 vesicles. Selected images from Fig. 1g were analyzed. Inset scale bar, 2 μm. b, U2OS cells stably expressing GFP-STX17-TM and LAMP1-mCherry were starved with EBSS for 2 h, then cells were treated with or without bafilomycin A1 for another 6 h. Scale bar, 5 μm. Inset scale bar, 2 μm. c, Percentage of cells with STX17 inside autolysosomes. Images in b were analyzed. Data are means ± s.d. (n = 3, 50 cells from 3 independent experiments were quantified), unpaired two-tailed t-test. d, The relative fluorescent intensity of STX17 and LAMP1 on autolysosomes. Selected images from b were analyzed. Inset scale bar, 2 μm. Source numerical data are available in source data.

Source data

Extended Data Fig. 6 The localization of sorting nexins.

The localization of sorting nexins. U2OS cells stably expressing GFP-STX17-TM and LAMP1-CFP were transfected with mCherry-SNX fusion proteins. Twenty-four hours after transfection, cells were starved with EBSS for 2 hours and images were taken. Scale bar, 5 μm. Inset scale bar, 2 μm.

Extended Data Fig. 7 The effect of sorting nexins on STX17 recycling from autolysosomes.

The effect of SNXs on STX17 recycling from autolysosomes. U2OS cells stably expressing Flag-STX17 were transfected with the indicated siRNAs. Forty-eight hours after transfection, cells were starved with EBSS for 5 hours and stained with antibodies against Flag and LAMP1. Scale bar, 5 μm. Inset scale bar, 2 μm.

Extended Data Fig. 8 KIBRA, SNX7, and SNX30 are not required for STX17 recycling from autolysosomes.

a-c, U2OS cells stably expressing Flag-STX17 were transfected with indicated siRNAs. Forty-eight hours after transfection, cells were subjected to immunoblot with antibodies against SNX4, KIBRA and SNX30. d, Representative mRNA level for the knockdown efficiency of SNX7. Data are presented as mean values ± s.d. e, The depletion of KIBRA, SNX7, and SNX30 has no effect on STX17 recycling from autolysosomes. U2OS cells stably expressing Flag-STX17 were transfected with non-targeting siRNA (NC) or siRNAs against KIBRA, SNX7, and SNX30. Forty-eight hours after transfection, cells were starved with EBSS for the indicated hours. Scale bar, 5 μm. Inset scale bar, 2 μm. Source numerical data and unprocessed blots are available in source data.

Source data

Extended Data Fig. 9 Retromer is not required for STX17 recycling from autolysosomes.

a-c, U2OS cells stably expressing Flag-STX17 were transfected with indicated siRNAs. Forty-eight hours after transfection, cells were subjected to immunoblot with antibodies against SNX5, VPS35 and SNX6. d, Depletion of VPS35 and SNX6 has no effect on STX17 recycling from autolysosomes. U2OS cells stably expressing Flag-STX17 were transfected with non-targeting siRNA (NC) or siRNAs against VPS35 and SNX6. Forty-eight hours after transfection, cells were starved with EBSS for the indicated hours. Scale bar, 5 μm. Inset scale bar, 2 μm. Source unprocessed blots are available in source data.

Source data

Extended Data Fig. 10 SNX4, SNX5 and SNX17 are required for STX17 recycling from autolysosomes.

a, Depletion of SNX4, SNX5 and SNX17 causes STX17 recycling defect. U2OS cells stably expressing Flag-STX17 were transfected with the indicated siRNAs. Forty-eight hours after transfection, cells were starved with or without EBSS for indicated duration and stained with antibodies against Flag, LAMP1 and LC3. Scale bar, 5 μm. Inset scale bar, 2 μm. b, Quantification of the number of STX17-positive autolysosomes. Images in a were analyzed. Data are means ± s.d. (n = 3, 50 cells from 3 independent experiments were quantified). Unpaired two-tailed t-test. c, U2OS cells stably expressing Flag-STX17 were transfected with the indicated siRNAs. Forty-eight hours after transfection, cells were subjected to immunoblot with antibodies against SNX4, SNX5 and SNX17. * indicates a non-specific band. Source numerical data and unprocessed blots are available in source data.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–21 and figure legend.

Reporting Summary

Supplementary Video 1

STX17 is retrieved from autolysosomes.

Supplementary Tables

Supplementary Tables 1–3.

Supplementary Data

Statistical source data for Additional Supplementary Figures.

Supplementary Data

Unprocessed western blots for additional supplementary figures.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 9

Unprocessed western blots.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, C., Wu, Z., Du, W. et al. Recycling of autophagosomal components from autolysosomes by the recycler complex. Nat Cell Biol 24, 497–512 (2022). https://doi.org/10.1038/s41556-022-00861-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-00861-8

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