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:

The LC3-conjugation machinery specifies the loading of RNA-binding proteins into extracellular vesicles

Abstract

Traditionally viewed as an autodigestive pathway, autophagy also facilitates cellular secretion; however, the mechanisms underlying these processes remain unclear. Here, we demonstrate that components of the autophagy machinery specify secretion within extracellular vesicles (EVs). Using a proximity-dependent biotinylation proteomics strategy, we identify 200 putative targets of LC3-dependent secretion. This secretome consists of a highly interconnected network enriched in RNA-binding proteins (RBPs) and EV cargoes. Proteomic and RNA profiling of EVs identifies diverse RBPs and small non-coding RNAs requiring the LC3-conjugation machinery for packaging and secretion. Focusing on two RBPs, heterogeneous nuclear ribonucleoprotein K (HNRNPK) and scaffold-attachment factor B (SAFB), we demonstrate that these proteins interact with LC3 and are secreted within EVs enriched with lipidated LC3. Furthermore, their secretion requires the LC3-conjugation machinery, neutral sphingomyelinase 2 (nSMase2) and LC3-dependent recruitment of factor associated with nSMase2 activity (FAN). Hence, the LC3-conjugation pathway controls EV cargo loading and secretion.

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: Identification of proteins secreted via autophagy-dependent pathways using LC3 proximity-dependent biotinylation and quantitative secretomics.
Fig. 2: LC3-II and BirA*–LC3 biotinylated targets are secreted within EVs.
Fig. 3: Endogenous LC3 localizes with endosomes and EV-associated tetraspanins.
Fig. 4: TMT quantitative secretomics identifies EV proteins secreted via the LC3-conjugation machinery.
Fig. 5: LC3-conjugation machinery is required for EV loading and secretion of SAFB and HNRNPK.
Fig. 6: LDELS regulates the small non-coding RNA composition of EVs.
Fig. 7: LDELS requires nSMase2 and FAN.

Similar content being viewed by others

Data availability

The MS proteomics data associated with this study have been deposited to the ProteomeXchange Consortium via the PRIDE80 partner repository with the dataset identifier PXD015479. RNA-seq data have been deposited in GEO: GSE137618. Furthermore, the data and/or reagents that support the findings of this study are available from the corresponding author, J.D., upon reasonable request. Source data for Figs. 17 and Extended Data Figs. 17 are provided online.

References

  1. Kaur, J. & Debnath, J. Autophagy at the crossroads of catabolism and anabolism. Nat. Rev. Mol. Cell Biol. 16, 461–472 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Lock, R., Kenific, C. M., Leidal, A. M., Salas, E. & Debnath, J. Autophagy-dependent production of secreted factors facilitates oncogenic RAS-driven invasion. Cancer Discov. 4, 466–479 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bel, S. et al. Paneth cells secrete lysozyme via secretory autophagy during bacterial infection of the intestine. Science 357, 1047–1052 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. DeSelm, C. J. et al. Autophagy proteins regulate the secretory component of osteoclastic bone resorption. Dev. Cell 21, 966–974 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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.e7 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Murrow, L., Malhotra, R. & Debnath, J. ATG12-ATG3 interacts with Alix to promote basal autophagic flux and late endosome function. Nat. Cell Biol. 17, 300–310 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Dupont, N. et al. Autophagy-based unconventional secretory pathway for extracellular delivery of IL-1beta. EMBO J. 30, 4701–4711 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Duran, J. M., Anjard, C., Stefan, C., Loomis, W. F. & Malhotra, V. Unconventional secretion of Acb1 is mediated by autophagosomes. J. Cell Biol. 188, 527–536 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Manjithaya, R., Anjard, C., Loomis, W. F. & Subramani, S. Unconventional secretion of Pichia pastoris Acb1 is dependent on GRASP protein, peroxisomal functions, and autophagosome formation. J. Cell Biol. 188, 537–546 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Torisu, T. et al. Autophagy regulates endothelial cell processing, maturation and secretion of von Willebrand factor. Nat. Med. 19, 1281–1287 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cadwell, K. & Debnath, J. Beyond self-eating: the control of nonautophagic functions and signaling pathways by autophagy-related proteins. J. Cell Biol. 217, 813–822 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Malhotra, V. Unconventional protein secretion: an evolving mechanism. EMBO J. 32, 1660–1664 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ponpuak, M. et al. Secretory autophagy. Curr. Opin. Cell Biol. 35, 106–116 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Roux, K. J., Kim, D. I., Raida, M. & Burke, B. A promiscuous biotin ligase fusion protein identifies proximal and interacting proteins in mammalian cells. J. Cell Biol. 196, 801–810 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. 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 

  16. Nanjappa, V. et al. Plasma Proteome Database as a resource for proteomics research: 2014 update. Nucleic Acids Res. 42, D959–D965 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Szklarczyk, D. et al. STRING v10: protein–protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 43, D447–D452 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Castello, A. et al. Insights into RNA biology from an atlas of mammalian mRNA-binding proteins. Cell 149, 1393–1406 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Thery, C., Amigorena, S., Raposo, G. & Clayton, A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr. Protoc. Cell Biol. 30, 3.22.1–3.22.29 (2006).

  20. Maas, S. L. N., Breakefield, X. O. & Weaver, A. M. Extracellular vesicles: unique intercellular delivery vehicles. Trends Cell Biol. 27, 172–188 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Mathieu, M., Martin-Jaular, L., Lavieu, G. & Thery, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 21, 9–17 (2019).

    Article  CAS  PubMed  Google Scholar 

  22. Pegtel, D. M. & Gould, S. J. Exosomes. Annu. Rev. Biochem. 88, 487–514 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. van Niel, G., D’Angelo, G. & Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 19, 213–228 (2018).

    Article  PubMed  CAS  Google Scholar 

  24. Martell, J. D., Deerinck, T. J., Lam, S. S., Ellisman, M. H. & Ting, A. Y. Electron microscopy using the genetically encoded APEX2 tag in cultured mammalian cells. Nat. Protoc. 12, 1792–1816 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Stenmark, H. et al. Inhibition of rab5 GTPase activity stimulates membrane fusion in endocytosis. EMBO J. 13, 1287–1296 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Trajkovic, K. et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 319, 1244–1247 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Ichimura, Y. et al. A ubiquitin-like system mediates protein lipidation. Nature 408, 488–492 (2000).

    Article  CAS  PubMed  Google Scholar 

  28. Itakura, E., Kishi, C., Inoue, K. & Mizushima, N. Beclin 1 forms two distinct phosphatidylinositol 3-kinase complexes with mammalian Atg14 and UVRAG. Mol. Biol. Cell 19, 5360–5372 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Matsunaga, K. et al. Two Beclin 1-binding proteins, Atg14L and Rubicon, reciprocally regulate autophagy at different stages. Nat. Cell Biol. 11, 385–396 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Ivanov, P., Kedersha, N. & Anderson, P. Stress granules and processing bodies in translational control. Cold Spring Harb. Perspect. Biol. 11, a032813 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Jeppesen, D. K. et al. Reassessment of exosome composition. Cell 177, 428–445.e18 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Stolz, A., Ernst, A. & Dikic, I. Cargo recognition and trafficking in selective autophagy. Nat. Cell Biol. 16, 495–501 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Kishi-Itakura, C., Koyama-Honda, I., Itakura, E. & Mizushima, N. Ultrastructural analysis of autophagosome organization using mammalian autophagy-deficient cells. J. Cell Sci. 127, 4089–4102 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Zhang, H. et al. Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation. Nat. Cell Biol. 20, 332–343 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Villarroya-Beltri, C., Baixauli, F., Gutierrez-Vazquez, C., Sanchez-Madrid, F. & Mittelbrunn, M. Sorting it out: regulation of exosome loading. Semin. Cancer Biol. 28, 3–13 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Dupuis-Sandoval, F., Poirier, M. & Scott, M. S. The emerging landscape of small nucleolar RNAs in cell biology. Wiley Interdiscip. Rev. RNA 6, 381–397 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Adam-Klages, S. et al. FAN, a novel WD-repeat protein, couples the p55 TNF-receptor to neutral sphingomyelinase. Cell 86, 937–947 (1996).

    Article  CAS  PubMed  Google Scholar 

  38. Berg, T. O., Fengsrud, M., Stromhaug, P. E., Berg, T. & Seglen, P. O. Isolation and characterization of rat liver amphisomes. Evidence for fusion of autophagosomes with both early and late endosomes. J. Biol. Chem. 273, 21883–21892 (1998).

    Article  CAS  PubMed  Google Scholar 

  39. Fader, C. M., Sanchez, D., Furlan, M. & Colombo, M. I. Induction of autophagy promotes fusion of multivesicular bodies with autophagic vacuoles in k562 cells. Traffic 9, 230–250 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Florey, O., Kim, S. E., Sandoval, C. P., Haynes, C. M. & Overholtzer, M. Autophagy machinery mediates macroendocytic processing and entotic cell death by targeting single membranes. Nat. Cell Biol. 13, 1335–1343 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Heckmann, B. L. et al. LC3-associated endocytosis facilitates beta-amyloid clearance and mitigates neurodegeneration in murine Alzheimer’s disease. Cell 178, 536–551.e14 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Mejlvang, J. et al. Starvation induces rapid degradation of selective autophagy receptors by endosomal microautophagy. J. Cell Biol. 217, 3640–3655 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Puri, C. et al. The RAB11A-positive compartment is a primary platform for autophagosome assembly mediated by WIPI2 recognition of PI3P-RAB11A. Dev. Cell 45, 114–131 e118 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Progida, C. & Bakke, O. Bidirectional traffic between the Golgi and the endosomes - machineries and regulation. J. Cell Sci. 129, 3971–3982 (2016).

    CAS  PubMed  Google Scholar 

  45. Raiborg, C., Wenzel, E. M. & Stenmark, H. ER-endosome contact sites: molecular compositions and functions. EMBO J. 34, 1848–1858 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hubstenberger, A. et al. P-body purification reveals the condensation of repressed mRNA regulons. Mol. Cell 68, 144–157.e5 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. Jain, S. et al. ATPase-modulated stress granules contain a diverse proteome and substructure. Cell 164, 487–498 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Buchan, J. R., Kolaitis, R. M., Taylor, J. P. & Parker, R. Eukaryotic stress granules are cleared by autophagy and Cdc48/VCP function. Cell 153, 1461–1474 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Alberti, S., Mateju, D., Mediani, L. & Carra, S. Granulostasis: protein quality control of RNP granules. Front. Mol. Neurosci. 10, 84 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Ganassi, M. et al. A surveillance function of the HSPB8-BAG3-HSP70 chaperone complex ensures stress granule integrity and dynamism. Mol. Cell 63, 796–810 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Mateju, D. et al. An aberrant phase transition of stress granules triggered by misfolded protein and prevented by chaperone function. EMBO J. 36, 1669–1687 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wang, B. et al. ULK1 and ULK2 regulate stress granule disassembly through phosphorylation and activation of VCP/p97. Mol. Cell 74, 742–757.E8 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Abels, E. R. & Breakefield, X. O. Introduction to extracellular vesicles: biogenesis, RNA cargo selection, content, release, and uptake. Cell. Mol. Neurobiol. 36, 301–312 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Skog, J. et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 10, 1470–1476 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Spinelli, C., Adnani, L., Choi, D. & Rak, J. Extracellular vesicles as conduits of non-coding RNA emission and intercellular transfer in brain tumors. Noncoding RNA 5, 1 (2018).

    PubMed Central  Google Scholar 

  56. Valadi, H. et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9, 654–659 (2007).

    Article  CAS  PubMed  Google Scholar 

  57. Bellingham, S. A., Coleman, B. M. & Hill, A. F. Small RNA deep sequencing reveals a distinct miRNA signature released in exosomes from prion-infected neuronal cells. Nucleic Acids Res. 40, 10937–10949 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Freedman, J. E. et al. Diverse human extracellular RNAs are widely detected in human plasma. Nat. Commun. 7, 11106 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Kaur, S. et al. CD63, MHC class 1, and CD47 identify subsets of extracellular vesicles containing distinct populations of noncoding RNAs. Sci. Rep. 8, 2577 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Lasser, C. et al. Two distinct extracellular RNA signatures released by a single cell type identified by microarray and next-generation sequencing. RNA Biol. 14, 58–72 (2017).

    Article  PubMed  Google Scholar 

  61. Lefebvre, F. A. et al. Comparative transcriptomic analysis of human and Drosophila extracellular vesicles. Sci. Rep. 6, 27680 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Rimer, J. M. et al. Long-range function of secreted small nucleolar RNAs that direct 2ʹ-O-methylation. J. Biol. Chem. 293, 13284–13296 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Shurtleff, M. J. et al. Broad role for YBX1 in defining the small noncoding RNA composition of exosomes. Proc. Natl Acad. Sci. USA 114, E8987–E8995 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Hong, E. et al. Unravelling the RNA-binding properties of SAFB proteins in breast cancer cells. Biomed. Res. Int. 2015, 395816 (2015).

    PubMed  PubMed Central  Google Scholar 

  65. Falaleeva, M. & Stamm, S. Processing of snoRNAs as a new source of regulatory non-coding RNAs: snoRNA fragments form a new class of functional RNAs. Bioessays 35, 46–54 (2013).

    Article  CAS  PubMed  Google Scholar 

  66. Holley, C. L. et al. Cytosolic accumulation of small nucleolar RNAs (snoRNAs) is dynamically regulated by NADPH oxidase. J. Biol. Chem. 290, 11741–11748 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Li, M. W. et al. Nuclear export factor 3 regulates localization of small nucleolar RNAs. J. Biol. Chem. 292, 20228–20239 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Pear, W. S., Scott, M. L. & Nolan, G. P. Generation of high-titer, helper-free retroviruses by transient transfection. Methods Mol. Med. 7, 41–57 (1997).

    CAS  PubMed  Google Scholar 

  69. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    Article  CAS  PubMed  Google Scholar 

  70. Tyanova, S. et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 13, 731–740 (2016).

    Article  CAS  PubMed  Google Scholar 

  71. Kuksa, P. P. et al. DASHR 2.0: integrated database of human small non-coding RNA genes and mature products. Bioinformatics 35, 1033–1039 (2019).

    Article  CAS  PubMed  Google Scholar 

  72. Mi, H., Muruganujan, A., Ebert, D., Huang, X. & Thomas, P. D. PANTHER version 14: more genomes, a new PANTHER GO-slim and improvements in enrichment analysis tools. Nucleic Acids Res. 47, D419–D426 (2019).

    Article  CAS  PubMed  Google Scholar 

  73. Szklarczyk, D. et al. STRINGv11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47, D607–D613 (2019).

    Article  CAS  PubMed  Google Scholar 

  74. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Hulsen, T., de Vlieg, J. & Alkema, W. BioVenn - a web application for the comparison and visualization of biological lists using area-proportional Venn diagrams. BMC Genomics 9, 488 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Costes, S. V. et al. Automatic and quantitative measurement of protein–protein colocalization in live cells. Biophys. J. 86, 3993–4003 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Hayashi, S. & McMahon, A. P. Efficient recombination in diverse tissues by a tamoxifen-inducible form of Cre: a tool for temporally regulated gene activation/inactivation in the mouse. Dev. Biol. 244, 305–318 (2002).

    Article  CAS  PubMed  Google Scholar 

  79. Albuquerque, C., Joseph, D. J., Choudhury, P. & MacDermott, A. B. Dissection, plating, and maintenance of cortical astrocyte cultures. Cold Spring Harb. Protoc. https://doi.org/10.1101/pdb.prot5273 (2009).

    Google Scholar 

  80. Vizcaino, J. A. et al. 2016 update of the PRIDE database and its related tools. Nucleic Acids Res. 44, 11033 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Krummel (UCSF) for generously providing reagents, members of the Debnath laboratory for helpful discussions and R. Perera (UCSF) for critical reading of the manuscript. We also thank S. Kilinc and A. Goga (UCSF) for assistance with EV concentration and size analysis, and A. Navickas for advice on RNA-seq library preparation. Grant support includes the NIH (CA201849, CA126792, CA201849 and CA213775 to J.D.; R00CA194077 and R01CA240984 to H.G.; K08CA184116 to A.P.W.; and AG057462 to J.D. and E.J.H.), the DOD BCRP (W81XWH-11-1-0130 to J.D.), the Samuel Waxman Cancer Research Foundation (to J.D.), a UCSF QB3 Calico Longevity Fellowship (to J.D. and A.M.L.) and a Dale Frey Breakthrough Award from the Damon Runyon Cancer Research Foundation (DFS 14-15 to A.P.W.). Fellowship support includes a Banting Postdoctoral Fellowship from the Government of Canada (201409BPF-335868) and a Cancer Research Society Scholarship for Next Generation of Scientists to A.M.L., NSF Graduate Student Fellowships (1650113 to T.S. and 1144247 to J.G.), a Canadian Institutes of Health Research Post-doctoral Fellowship to F.K., NRSA awards from the NCI (F31CA217015 to T. Marsh, F30CA224693 to J.Y.L.), a UCSF IRACDA Postdoctoral Fellowship (K12GM081266) to T. Monkkonen and an NCI T32 training grant (T32CA108462) to A.V.

Author information

Authors and Affiliations

Authors

Contributions

J.D. and A.M.L. conceived the study and designed the experiments. A.M.L., H.H.H., A.P.W. and J.D. designed and optimized the quantitative proteomics workflows. A.M.L., J.D. and H.G. designed, performed and analysed the extracellular and intracellular RNA-seq experiments. A.M.L., T.S., J.Y., F.K., J.Y.L., T. Monkkonen and A.V. performed biochemical and cell biological experiments. H.H.H. and A.P.W. performed MS and analysed the resulting liquid chromatography (LC)–MS/MS data with A.M.L. A.M.L. and J.G. performed bioinformatics analysis of the BirA*–LC3-labelled secretome. A.M.L., T. Marsh, Y.-H.H. and E.J.H. performed mouse experiments and primary astrocyte isolation. D.Z. and L.Y. performed APEX staining and TEM. A.M.L. and J.D. analysed the biochemical and cell biological data. A.M.L. and J.D. wrote the paper, with input from all other authors.

Corresponding author

Correspondence to Jayanta Debnath.

Ethics declarations

Competing interests

J.D. is a Scientific Advisory Board Member for Vescor Therapeutics, LLC.

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 Functional validation of the BirA*-LC3 recombinant probe.

a, Cells stably expressing myc-BirA*-LC3, myc-BirA* or vector control were incubated in either full (F) or serum free media (S) for 4h in the absence or presence of 50 µM chloroquine (CQ) for the last 1h. Cells were lysed and subject to immunoblotting for indicated proteins (n=2 biologically independent experiments). b, Representative images of cells stably expressing GFP-LC3 and myc-BirA*-LC3 or myc-BirA* and immunostained with anti-myc antibody (n=3 biologically independent samples). c, Representative images of cells stably expressing myc-BirA*-LC3 or myc-BirA* and co-immunostained with anti-LC3 (green) and anti-myc (magenta) antibody (n=3 biologically independent samples). d, Biotinylation blots reproduced from Fig. 1b with accompanying Ponceau S stained membranes of whole cell lysate (WCL) and conditioned media (CM) (n=3 biologically independent experiments). e, Schematic of experiment to test for intracellular versus extracellular origin of BirA* and BirA*-LC3-mediated biotinylated targets isolated from CM. f, Representative Strep-HRP blot for biotinylated proteins in the precipitated CM from myc-BirA*-LC3 or myc-BirA* cells co-incubated (Co) or post-incubated (Post) with 50 µM biotin for 24h and negative control (Neg). CM was probed to validate expression and secretion of the myc-tagged recombinant proteins (n=2 biologically independent experiments). Unprocessed blots available in Source Data Extended Data Fig. 1.

Source data

Extended Data Fig. 2 BirA*-LC3B-labelled secretome is enriched in RBPs.

a, Volcano plot of BirAf*-LC3-labeled secretome quantified by mass spectrometry. SILAC labelled biotin-tagged proteins plotted according to -log10 p-values as determined by two-tailed t-test and log2 fold enrichment (BirA*-LC3/BirA*) (n=3 biologically independent samples). Grey horizontal dotted line: significance cut-off with p-value of 0.05. Log2 fold change reflects LC3-BirA* to BirA* alone ratio. Grey vertical dotted line: 2-fold enriched and de-enriched cut-off. Pink: significantly enriched proteins relative to BirA* alone. Red: Class I enriched proteins represented in heat map in Fig. 1. Inset: Expanded view of significantly enriched proteins. b, Venn diagram showing overlap of secretory autophagy candidates (Class I and II hits) with the LC3B intracellular interactome defined in Behrends et al. 2010. c, Venn diagram showing the overlap of secretory autophagy candidates (Class I and II hits) with the entire ATG8 intracellular interactome defined in Behrends et al. 2010. d, Ranked list of proteins with greatest connectivity to secretory autophagy candidates as determined by the Enrichr gene enrichment analysis tool (n=3 biologically independent samples; 200 enriched proteins in Class I + II datasets). Statistical significance calculated by one-way Fisher’s exact test and adjusted using the Benjamini–Hochberg method. LC3 family members highlighted in red. e, Network map of autophagy-dependent secretion candidates. Class I and II secretory autophagy candidates mapped to zero-order protein interaction network using Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and proteins associated with extracellular exosomes or with RNA-binding functions coloured in red and blue, respectively. f, Pie chart plotting percentage of Class I and II secretory autophagy candidates assigned to Gene Ontology (GO) term extracellular exosome by PANTHER. g, Venn diagram showing overlap of class I and II secretory autophagy candidates with the mRNA binding proteins from Castello et al. 2012. Data available in Source Data Extended Data Fig. 2.

Source data

Extended Data Fig. 3 Endogenous LC3-II is secreted within EVs isolated from cultured cells and murine plasma.

a, Whole cell lysate (WCL) and extracellular vesicle lysates (EVs) from murine RAW264.7 macrophages treated with 100 ng/ml LPS for 24h and 20 μM nigericin for 1 h, murine B16F10 melanoma cells, and murine LLC1 cells were immunoblotted for LC3, SAFB, HNRNPK and extracellular vesicle marker proteins (n=2 biologically independent experiments). b, Workflow employed to obtain plasma and tissue from CAG-CreER;Atg12flox/flox mice in which Atg12 was systemically deleted via tamoxifen treatment. c, Extracellular vesicles (EVs) isolated from the plasma of naïve wild-type mice (CAG-CreER or Atg12flox/flox) and mice in which Atg12 was systemically deleted by 4-OHT treatment (CAG-CreER; Atg12flox/flox) were lysed and immunoblotted for LC3 and the indicated extracellular vesicle marker proteins (n=2 biologically independent experiments). d, Whole cell lysates (WCL) derived from the renal tissue of mice in Panel c were immunoblotted for LC3 and the indicated marker proteins (n=2 biologically independent experiments). e, Workflow employed to obtain CM from murine astrocytes (CAG-CreER;Atg5flox/flox) in which Atg5 was deleted ex vivo via 4-OHT treatment. f, Extracellular vesicles (EVs) isolated from the conditioned media of naive wild-type (Atg5flox/flox) primary astrocytes and astrocyte cultures in which Atg5 was deleted ex vivo (CAG-CreER; Atg5flox/flox) by 4-OHT treatment were lysed and immunoblotted for LC3 and CD9 (n=2 biologically independent experiments). g, Whole cell lysates (WCL) primary astrocyte cultures in Panel d were immunoblotted for LC3 and CD9 (n=2 biologically independent experiments). h, Representative fluorescence micrographs from wild-type, ATG7-/- and ATG14-/- HEK293T cells transfected with mCherry-Rab5Q79L (yellow). Cells were immunostained for endogenous LC3 (green) and CD63 (magenta) (n=3 biologically independent samples). Scale bar=10μm. Unprocessed blots available in Source Data Extended Data Fig. 3.

Source data

Extended Data Fig. 4 Components of stress granules and P-bodies secreted in EVs through mechanisms requiring the LC3-conjugation machinery.

a, Proportion of BirA*-LC3B- labelled secretome (Class I, II candidates) detected in the total extracellular vesicle (EV) proteome defined by TMT quantitative mass spectrometry. b, Venn diagram showing overlap of EV components requiring ATG7 and ATG12 for secretion with the fixed and unfixed stress granule proteome from Jain et al., 2016. c, Venn diagram showing the overlap of EV components requiring ATG7 and ATG12 for secretion with the P-body proteome from Hubstenberger et al., 2017. d, Representative fluorescence micrographs from wild-type HEK293T cells transfected with mCherry-Rab5Q79L (blue) and immunostained for endogenous LC3 (green) and SAFB or HNRNPK (magenta) (n=3 biologically independent samples). Scale bar=10μm. e, Whole cell (WCL) and EV lysates harvested from equal numbers of cells stably expressing non-targeting (NT) or ATG3 shRNA were immunoblotted for indicated proteins (n=3 biologically independent samples). f, Quantification of indicated protein levels in EVs from cells stably expressing shRNAs targeting ATG3 relative to non-targeting shRNA (mean ±s.e.m.; n=3 biologically independent samples). g, Quantification of Lactate Dehydrogenase (LDH) in EV-depleted conditioned media from wild-type (WT) HEK293T cells treated 100 μM Etoposide (Etop) for 24h or WT and ATG knockout cells serum starved for 24h (mean ±s.e.m.; n=3 biologically independent experiments). h, Cell death in wild-type (WT) and ATG knockout cells (KO) after 24h in full serum media (FM) or serum starved media (SS) quantified using Calcein-AM and ethidium bromide staining (mean ±s.e.m.; n=3 biologically independent experiments). i, Whole cell and EV lysates from wild-type cells grown in EV-depleted full serum media (FM) or EV-depleted FM with 100 nM Rapamycin (Rap) for 24h. Immunoblots probed against the indicated proteins (n=2 biologically independent experiments). j, Quantification of the relative levels of indicated proteins in EVs from Rap-treated cells in Panel d (line=mean; n=2 biologically independent experiments). Data and unprocessed blots available in Source Data Extended Data Fig. 4.

Source data

Extended Data Fig. 5 LC3-conjugation machinery controls EV-mediated secretion of diverse RBPs.

a, EVs from WT and ATG deficient cells normalized for protein concentration and immunoblotted to detect endogenous LC3A, LC3B, LC3C, GABARAP (GR), GABARAPL1 (GRL1), GABARAPL2 (GRL2), and indicated marker proteins (n=2 biologically independent experiments). b, Whole cell (WCL) and EV lysates from WT and ATG7-/- cells were normalized for protein concentration and immunoblotted for indicated proteins (n=3 biologically independent experiments). c, HEK293T cells co-transfected with FLAG-tagged G3BP1, LARP1 or SF3A1, and myc-tagged LC3B, GABARAP (GR), LC3C respectively, or myc-BirA* were lysed, immunoprecipitated (IP) with anti-myc antibody and immunoblotted (WB) with indicated antibodies (n=3 biologically independent experiments). d, Diagram mapping the domains and primary LC3-interaction region (LIR) in SAFB. e, Volcano plot of mRNA and long non-coding RNA (large RNA) detected in EVs from WT and ATG7-/- cells. Results plotted according to -log10 p-values as determined by DESeq2 and log2 fold enrichment (n=3 biologically independent samples; WT/ATG7-/-). Grey dots: RNAs not enriched in EVs from WT or ATG7-/- cells identified with a p-value >0.05 and/or log2 fold change between -0.5 and 0.5 (-0.5<log2FC<0.5). Black dots: Large RNAs enriched in EVs from WT cells or ATG7-/- cells. f, Volcano plot of mRNA and long non-coding RNA (large RNA) detected in EVs from WT and ATG12-/- cells. Results plotted according to -log10 p-values as determined by DESeq2 and log2 fold enrichment (n=3 biologically independent samples; WT/ATG12-/-). Grey dots: RNAs not enriched in EVs from WT or ATG12-/- cells identified with a p-value >0.05 and/or log2 fold change between -0.5 and 0.5 (-0.5<log2FC<0.5). Black dots: Large RNAs enriched in EVs from WT or ATG12-/- cells. g, Venn diagram showing the overlap of mRNA and long non-coding RNAs (large RNAs) enriched in EVs from WT relative to ATG7-/- cells and EVs from WT relative to ATG12-/- cells. Data and unprocessed blots available in Source Data Extended Data Fig. 5.

Source data

Extended Data Fig. 6 LC3 delivery into ILVs of Rab5Q79L endosomes requires CHMP4b and nSMase2, but is independent of other ESCRT machinery components.

a, Representative fluorescence micrographs from wild-type HEK293T cells co-transfected with mCherry-Rab5Q79L (magenta) and non-targeting (NT) control siRNA or siRNAs that deplete ATG7, ALIX, TSG101, VPS4a/b, CHMP3, CHMP4b and nSMase2. Cells were immunostained for endogenous LC3 (green) (n=2 biologically independent experiments). Scale bar=10μm. b, Scatter plot of the proportion of mCherry-Rab5Q79L endosomes that overlap with LC3 in immuno-stained cells in Panel a (mean ± s.e.m.; n=23 biologically independent samples). Statistical significance calculated by one-way ANOVA coupled with Fisher’s least significant difference test. c, Lysates from cells in Panel a were immunoblotted with antibodies the various siRNA targets and GAPDH as a loading control. Representative blots are shown (n=2 biologically independent experiments). Non-specific bands are indicated with an asterisk (*). d, Quantitative PCR (QPCR) measurement of nSMase2 mRNA in HEK293T cells transfected with siRNAs targeting nSMase2, nSMase2 (nSM2) relative to non-targeting siRNA (NT) control cells (line=mean; n=1, 2 technical replicates). Data and unprocessed blots available in Source Data Extended Data Fig. 6.

Source data

Extended Data Fig. 7 LC3-dependent EV loading and secretion (LDELS) requires FAN and nSMase2.

a, Whole cell lysate harvested from equal numbers of HEK293Ts stably expressing non-targeting (NT) shRNA or shRNAs that deplete ATG7 or nSMase2(nSM2) immunoblotted for indicated proteins (n=2 biologically independent experiments). b, Quantitative PCR (QPCR) for nSMase2 mRNA in HEK293Ts stably expressing shRNAs targeting ATG7, nSMase2 (nSM2) relative to non-targeting shRNA (NT) control cells (line=mean; n=1, 2 technical replicates). c, Whole cell lysates from HEK293Ts stably expressing non-targeting shRNA (NT) or shRNAs that deplete ATG7 or FAN were immunoblotted with antibodies for the indicated proteins (n=2 biologically independent experiments). d, HEK293Ts were EBSS starved for the indicated times, treated with DMSO or 50nM Bafilomycin A1 (Baf A1) for 1 h prior to lysis, lysed and immunoblotted for FAN and the indicated proteins (n=2 biologically independent experiments). e, HEK293Ts expressing non-targeting (NT) shRNA or shRNA that depletes FAN were starved in EBSS for 4h, treated with DMSO or 50nM Bafilomycin A1 (Baf A1) for 1h prior to lysis, lysed and immunoblotted for the indicated proteins (n=2 biologically independent experiments). f, Representative fluorescence micrographs from wild-type cells co-transfected with mCherry-Rab5Q79L (magenta) and non-targeting (NT) control siRNA or siRNAs that deplete FAN. Cells were immunostained for endogenous LC3 (green)(n=2 biologically experiments). Scale bar=10μm. g, Lysates from cells in Panel f were immunoblotted with antibodies against FAN and GAPDH as a loading control (n=2 biologically independent experiments). h, Scatter plot of the proportion of mCherry-Rab5Q79L endosomes overlapping with LC3 in immuno-stained cells in Panel f (mean ±s.e.m.; n=22 biologically independent samples). Statistical significance calculated by unpaired two-tailed t-test. i, Whole cell lysate from HEK293Ts analysed in Fig. 7j that were co-expressing non-targeting (NT) shRNA or shRNA that depletes endogenous FAN along with FLAG-tagged wild-type FAN (WT) or mutant FAN (F199A) were immunoblotted for the indicated proteins (n=2 biologically independent experiments). j, Proposed model for LC3-dependent EV loading and secretion (LDELS) in comparison to classical autophagy. Data and unprocessed blots available in Source Data Extended Data Fig. 7.

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Tables 1–3.

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. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed blots.

Source Data Extended Data Fig. 1

Unprocessed blots.

Source Data Extended Data Fig. 2

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. 4

Unprocessed blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed blots.

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.

Source Data Extended Data Fig. 7

Unprocessed blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Leidal, A.M., Huang, H.H., Marsh, T. et al. The LC3-conjugation machinery specifies the loading of RNA-binding proteins into extracellular vesicles. Nat Cell Biol 22, 187–199 (2020). https://doi.org/10.1038/s41556-019-0450-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0450-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