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.

ATG14 promotes membrane tethering and fusion of autophagosomes to endolysosomes

Abstract

Autophagy, an important catabolic pathway implicated in a broad spectrum of human diseases, begins by forming double membrane autophagosomes that engulf cytosolic cargo and ends by fusing autophagosomes with lysosomes for degradation1,2. Membrane fusion activity is required for early biogenesis of autophagosomes and late degradation in lysosomes3,4,5,6,7. However, the key regulatory mechanisms of autophagic membrane tethering and fusion remain largely unknown. Here we report that ATG14 (also known as beclin-1-associated autophagy-related key regulator (Barkor) or ATG14L), an essential autophagy-specific regulator of the class III phosphatidylinositol 3-kinase complex8,9,10,11, promotes membrane tethering of protein-free liposomes, and enhances hemifusion and full fusion of proteoliposomes reconstituted with the target (t)-SNAREs (soluble N-ethylmaleimide-sensitive factor attachment protein receptors) syntaxin 17 (STX17) and SNAP29, and the vesicle (v)-SNARE VAMP8 (vesicle-associated membrane protein 8). ATG14 binds to the SNARE core domain of STX17 through its coiled-coil domain, and stabilizes the STX17–SNAP29 binary t-SNARE complex on autophagosomes. The STX17 binding, membrane tethering and fusion-enhancing activities of ATG14 require its homo-oligomerization by cysteine repeats. In ATG14 homo-oligomerization-defective cells, autophagosomes still efficiently form but their fusion with endolysosomes is blocked. Recombinant ATG14 homo-oligomerization mutants also completely lose their ability to promote membrane tethering and to enhance SNARE-mediated fusion in vitro. Taken together, our data suggest an autophagy-specific membrane fusion mechanism in which oligomeric ATG14 directly binds to STX17–SNAP29 binary t-SNARE complex on autophagosomes and primes it for VAMP8 interaction to promote autophagosome–endolysosome fusion.

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

Figure 1: ATG14 interacts with STX17–SNAP29 on mature autophagosomes.
Figure 2: ATG14 promotes membrane tethering and enhances autophagic SNARE-mediated fusion.
Figure 3: ATG14 homo-oligomerization is required for autophagic SNARE binding.
Figure 4: ATG14 homo-oligomerization is required for autophagosomal fusion with endolysosomes in vivo and in vitro.

Accession codes

Primary accessions

Protein Data Bank

Data deposits

Coordinates and structure factor amplitudes have been deposited in the RCSB Protein Data Bank (http://www.rcsb.org) under accession code 4WY4.

References

  1. Levine, B. & Klionsky, D. J. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev. Cell 6, 463–477 (2004)

    Article  CAS  Google Scholar 

  2. Huang, W. P. & Klionsky, D. J. Autophagy in yeast: a review of the molecular machinery. Cell Struct. Funct. 27, 409–420 (2002)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  4. Nair, U. et al. SNARE proteins are required for macroautophagy. Cell 146, 290–302 (2011)

    Article  CAS  Google Scholar 

  5. Ragusa, M. J., Stanley, R. E. & Hurley, J. H. Architecture of the Atg17 complex as a scaffold for autophagosome biogenesis. Cell 151, 1501–1512 (2012)

    Article  CAS  Google Scholar 

  6. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  8. Sun, Q. et al. Identification of Barkor as a mammalian autophagy-specific factor for Beclin 1 and class III phosphatidylinositol 3-kinase. Proc. Natl Acad. Sci. USA 105, 19211–19216 (2008)

    Article  ADS  CAS  Google Scholar 

  9. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  11. Zhong, Y. et al. Distinct regulation of autophagic activity by Atg14L and Rubicon associated with Beclin 1-phosphatidylinositol-3-kinase complex. Nature Cell Biol. 11, 468–476 (2009)

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  13. Diao, J. et al. A single vesicle–vesicle fusion assay for in vitro studies of SNAREs and accessory proteins. Nature Protocols 7, 921–934 (2012)

    Article  CAS  Google Scholar 

  14. Diao, J. et al. Native alpha-synuclein induces clustering of synaptic-vesicle mimics via binding to phospholipids and synaptobrevin-2/VAMP2. eLife 2, e00592 (2013)

    Article  Google Scholar 

  15. Fan, W., Nassiri, A. & Zhong, Q. Autophagosome targeting and membrane curvature sensing by Barkor/Atg14(L). Proc. Natl Acad. Sci. USA 108, 7769–7774 (2011)

    Article  ADS  CAS  Google Scholar 

  16. Bigay, J., Casella, J. F., Drin, G., Mesmin, B. & Antonny, B. ArfGAP1 responds to membrane curvature through the folding of a lipid packing sensor motif. EMBO J. 24, 2244–2253 (2005)

    Article  CAS  Google Scholar 

  17. Farsad, K. et al. Generation of high curvature membranes mediated by direct endophilin bilayer interactions. J. Cell Biol. 155, 193–200 (2001)

    Article  CAS  Google Scholar 

  18. Kyoung, M. et al. In vitro system capable of differentiating fast Ca2+-triggered content mixing from lipid exchange for mechanistic studies of neurotransmitter release. Proc. Natl Acad. Sci. USA 108, E304–E313 (2011)

    Article  CAS  Google Scholar 

  19. Matsunaga, K. et al. Autophagy requires endoplasmic reticulum targeting of the PI3-kinase complex via Atg14L. J. Cell Biol. 190, 511–521 (2010)

    Article  CAS  Google Scholar 

  20. 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 

  21. Nair, U., Thumm, M., Klionsky, D. J. & Krick, R. GFP-Atg8 protease protection as a tool to monitor autophagosome biogenesis. Autophagy 7, 1546–1550 (2011)

    Article  CAS  Google Scholar 

  22. 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 

  23. Takats, S. et al. Interaction of the HOPS complex with Syntaxin 17 mediates autophagosome clearance in Drosophila. Mol. Biol. Cell 25, 1338–1354 (2014)

    Article  Google Scholar 

  24. Nakatogawa, H., Ichimura, Y. & Ohsumi, Y. Atg8, a ubiquitin-like protein required for autophagosome formation, mediates membrane tethering and hemifusion. Cell 130, 165–178 (2007)

    Article  CAS  Google Scholar 

  25. Chen, D. et al. A mammalian autophagosome maturation mechanism mediated by TECPR1 and the Atg12-Atg5 conjugate. Mol. Cell 45, 629–641 (2012)

    Article  CAS  Google Scholar 

  26. Su, Z., Ishitsuka, Y., Ha, T. & Shin, Y. K. The SNARE complex from yeast is partially unstructured on the membrane. Structure 16, 1138–1146 (2008)

    Article  CAS  Google Scholar 

  27. Sun, Q. et al. The RUN domain of rubicon is important for hVps34 binding, lipid kinase inhibition, and autophagy suppression. J. Biol. Chem. 286, 185–191 (2011)

    Article  CAS  Google Scholar 

  28. Studier, F. W. Protein production by auto-induction in high-density shaking cultures. Protein Expr. Purif. 41, 207–234 (2005)

    Article  CAS  Google Scholar 

  29. Kabsch, W. Automatic processing of rotation diffraction data from crystals of initially unknown symmetry and cell constants. J. Appl. Crystallogr. 26, 795–800 (1993)

    Article  CAS  Google Scholar 

  30. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007)

    Article  CAS  Google Scholar 

  31. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D 60, 2126–2132 (2004)

    Article  Google Scholar 

  32. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D 66, 213–221 (2010)

    Article  CAS  Google Scholar 

  33. Sutton, R. B., Fasshauer, D., Jahn, R. & Brunger, A. T. Crystal structure of a SNARE complex involved in synaptic exocytosis at 2.4 A resolution. Nature 395, 347–353 (1998)

    Article  ADS  CAS  Google Scholar 

  34. Strop, P., Kaiser, S. E., Vrljic, M. & Brunger, A. T. The structure of the yeast plasma membrane SNARE complex reveals destabilizing water-filled cavities. J. Biol. Chem. 283, 1113–1119 (2008)

    Article  CAS  Google Scholar 

  35. Zwilling, D. et al. Early endosomal SNAREs form a structurally conserved SNARE complex and fuse liposomes with multiple topologies. EMBO J. 26, 9–18 (2007)

    Article  CAS  Google Scholar 

  36. Antonin, W. et al. A SNARE complex mediating fusion of late endosomes defines conserved properties of SNARE structure and function. EMBO J. 19, 6453–6464 (2000)

    Article  CAS  Google Scholar 

  37. Diao, J. et al. Synaptic proteins promote calcium-triggered fast transition from point contact to full fusion. eLife 1, e00109 (2012)

    Article  Google Scholar 

  38. Rocco, C. J., Dennison, K. L., Klenchin, V. A., Rayment, I. & Escalante-Semerena, J. C. Construction and use of new cloning vectors for the rapid isolation of recombinant proteins from Escherichia coli. Plasmid 59, 231–237 (2008)

    Article  CAS  Google Scholar 

  39. Miroux, B. & Walker, J. E. Over-production of proteins in Escherichia coli: mutant hosts that allow synthesis of some membrane proteins and globular proteins at high levels. J. Mol. Biol. 260, 289–298 (1996)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Q. Sun, W. Fan, M. Padolina and R. Bellerose for technical assistance, Y. Cheng and S. Wu for help in the cryo-electron microscopy experiments, A. Liang at OCS Microscopy Core of New York University Langone Medical Center for electron microscope analysis, the Northeastern Collaborative Access Team (supported by National Institutes of Health (NIH) P41 GM103403) at the Advanced Photon Source for X-ray data collection, and B. Levine and R. Sumpter for reading the manuscript. The work was supported by grants to Q.Z. from the Welch Foundation (I-1864), the Cancer Prevention & Research Institute of Texas (RP140320), an American Cancer Society Research Scholar Grant (RSG-11-274-01-CCG) and NIH R01 (CA133228), and NIH R01 (R37-MH63105) to A.T.B. The work was partly supported by China Scholarship Council to R.L. This work was also supported by the National Cancer Institute of the NIH under award number 5P30CA142543.

Author information

Authors and Affiliations

Authors

Contributions

R.L., Y.R., J.Z., L.M.W. and J.L. performed the biological and biochemical experiments characterizing ATG14 function. J.D., Y.L. and R.A.P. performed the in vitro membrane tethering and fusion experiments; M.Z. and Q.Z. determined the crystal structure of the autophagic SNARE complex and performed the cryo-electron microscopy experiments; S.V. performed the SEC–MALS experiments, J.D., R.L., A.T.B. and Q.Z. conceived the project, designed the experiments, analysed the data and wrote the manuscript with the help of all authors.

Corresponding author

Correspondence to Qing Zhong.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Interaction among ATG14, STX17 and beclin 1.

a, Coomassie-stained SDS gel of recombinantly expressed and purified full-length Flag-tagged STX17, SNAP29 and VAMP8. b, Overexpression of Myc-ATG14 stabilizes the STX17–SNAP29 binary t-SNARE complex in a co-immunoprecipitation assay. c, ATG14 interacts with the SNARE core domain of STX17 in a co-immunoprecipitation assay. d, The CCD domain of ATG14 interacts with STX17 in an co-immunoprecipitation assay. e, Fractionation of beclin 1, ATG14, STX17 with or without chloroquine treatment by Superdex 200. f, STX17 does not associate with beclin 1. Flag-tagged STX17 was co-transfected with either Myc-ATG14 or Myc-beclin 1 in HEK293T cells. ATG14 but not beclin 1 co-immunoprecipitated with STX17. The immunoprecipitation efficiency (immunoprecipitation/input) was normalized by the ratio of immunoprecipitated ATG14 or beclin 1 versus their inputs. g, STX17 interacts with ATG14 in a complex that is distinct from the beclin 1/PI3KC3 complex. Cell lysates from U2OS cells stably expressing Flag–beclin 1 or ATG14 were immunoprecipitated with anti-Flag M2 beads, and endogenous beclin 1, ATG14 and STX17 were detected in the immunoprecipitates.

Extended Data Figure 2 Co-localization of ATG14 and STX17 on autophagosomes upon chloroquine or bafilomycin A1 treatments.

a, b, GFP–ATG14 was co-transfected with STX17–Flag in chloroquine- (a) or bafilomycin-A1-treated (b) U2OS cells, and detected by GFP fluorescence and anti-Flag antibody in immunostaining. Endogenous LC3, LAMP1 or Atg16 were detected by anti-LC3, LAMP2 or Atg16 antibodies in immunostaining (n = 20). Scale bars, 5 μm.

Extended Data Figure 3 Characterization of ATG14-mediated membrane tethering.

a, Scheme of the single-vesicle/liposome-tethering assay13,14. DiD-labelled liposomes were attached to the imaging surface through the interaction between biotin/NeutrAvidin; surface binding was assessed by red laser excitation (633 nm). Tethered DiI-labelled liposomes were detected by green laser excitation (532 nm). This assay was used in Figs 2a and 4e and Extended Data Figs 3c–h, 5d and 5g. b, Scheme of the FRET-based single-vesicle/liposome lipid-mixing assay13. Tethered DiI-labelled liposomes were excited by illumination with a green laser (532 nm). Detection of emission in both green and red spectral regions was performed simultaneously by using a dichroic beam-splitter. The total number of tethered DiI-labelled liposomes were counted in the green fluorescence channel, and FRET to the DiD acceptor dyes was observed in the red fluorescence channel. This assay was only used in Fig. 2b. The field of view is 45 μm × 90 μm. See Methods for more details. c, The BATS domain deletion mutant of ATG14 (900 nM) does not promote liposome tethering. Top panels: quantitation. Bottom panels: representative fluorescence images of tethered liposomes (n = 15). d, The BATS domain alone does not promote liposome tethering. Top panels: quantitation. Bottom panels: representative fluorescence images of tethered liposomes (n = 15). e, Purified recombinant ADP-ribosylation factor GTPase-activating protein 1 (ARFGAP1, 870 nM) does not promote liposome tethering. Top panels: quantitation. Bottom panels: representative fluorescence images of tethered liposomes (n = 15). f, Purified recombinant endophilin 1/Bif 1 (2 μM) does not promote liposome tethering. Top panels: quantitation. Bottom panels: representative fluorescence images of tethered liposomes (n = 15). g, Incorporation of 2% PI3P into small liposomes (with a 50 nm diameter) failed to enhance the liposome-tethering activity by ATG14. Top panels: quantitation. Bottom panels: representative fluorescence images of tethered liposomes (n = 15). h, Incorporation of 2% PI3P into large liposomes (with a 400 nm diameter) enhances the liposome-tethering activity by ATG14. Top panels: quantitation. Bottom panels: representative fluorescence images of tethered liposomes (n = 15). Results in ch are presented as the mean (± s.d.) of random imaging locations (n = 15) in the same sample channel.

Extended Data Figure 4 Structure of the autophagic SNARE complex.

a, Boundaries of SNARE domains in VAMP8, STX17 and SNAP29. b, Purification of the autophagic SNARE complex. The four SNARE core domains of VAMP8, STX17 and SNAP29 (with two SNARE core domains) were cloned, expressed in E. coli and co-purified (see Methods). The four fragments form a complex that appears as a single peak by SEC. The SDS–PAGE gel of the indicated four eluted fractions shows individual bands corresponding to the four SNARE core domains. Note that VAMP8 (10–74), STX17 (164–277) and SNAP29 (194–258) migrate at approximately the same position. The leftmost lane is a protein ladder with the molecular masses labelled. c, Comparison of SNARE structures. Shown are cartoon representation and surface charge distribution of the autophagic, neuronal, yeast, early endosomal and endosomal SNARE complex structures in two different orientations. The structures were placed such that the carboxy termini of the SNARE complexes are facing to the right. The surface charge distribution was generated using default ‘vacuum electrostatics’ in PyMOL for qualitative comparison only. Positive and negative charges are marked in blue and red colours respectively.

Extended Data Figure 5 Determining the specificity of ATG14 to autophagic SNAREs-mediated membrane fusion and characterizing recombinant ATG14 homo-oligomerization.

a, Scheme of the ensemble content-mixing assay using autophagic SNARE-reconstituted proteoliposomes (v- and t-proteoliposomes). b, Purified recombinant Flag–STX17, His-VAMP2, His-SNAP25 and His-Syntaxin1 were incubated with IgG Sepharose associated with recombinant ZZ–Flag–ATG14, and the ATG14 binding proteins were cleaved by TEV protease and detected by Coomassie blue staining (upper panel). ZZ–Flag–ATG14 bound to IgG Sepahrose was detected by western blotting in the bottom panel. ATG14 binds to STX17 but not to neuronal SNAREs. c, ATG14 had no detectable effect on promoting ensemble lipid-mixing of proteoliposomes reconstituted with neuronal SNAREs (n = 3). d, The membrane-tethering activity of ATG14 CCD deletion mutant is largely intact. Shown is the mean number of tethered vesicles (± s.d.) (n = 15) in the same sample channel. e, The ATG14 CCD deletion mutant fails to enhance ensemble lipid mixing of proteoliposomes reconstituted with autophagic SNAREs (n = 3). f, The oligomeric states of recombinant ATG14 were determined by SEC–MALS. g, Membrane-tethering activities of ATG14 monomer and dimer measured by the single protein-free vesicle/liposome membrane-tethering assay. Shown are the mean numbers of tethered vesicles (± s.d.) (n = 15) in the same sample channel. Representative images are shown in the bottom panels (n = 15). h, Mapping ATG14 oligomerization sites to its N terminus. Flag-tagged full-length ATG14 or an ATG14 truncating mutant (lacking the first 70 residues) were transfected into HEK293T cells and treated with cross-linking agent DSS (0, 0.1, 0.2, 0.4 mM) for 30 min, then subjected to SDS–PAGE analysis. ATG14 was probed by anti-Flag antibody. i, ATG14 CCD deletion mutant still forms oligomer in the DSS cross-linking assay. j, Purified recombinant ATG14 (36 nM) was boiled in non-reducing SDS sample buffer with 12.5 mM TCEP (lane 2), 25 mM DTT (lane 3) or 2.5% β-mercaptoethanol (lane 4), or mock-treated (lane 1), and the samples were loaded on non-reducing SDS–PAGE and probed for anti-Flag antibody for ATG14. k, Flag–ATG14 was transfected into HEK293T cells and treated with rapamycin (500 nM for 12 h) and/or cross-linking agent DSS (0.2 μM) for 30 min, then subjected to SDS–PAGE analysis. ATG14 was probed by anti-Flag antibody.

Extended Data Figure 6 ATG14 homo-oligomerization is not essential for beclin 1 interaction and PI3KC3 activation.

a, Interaction between Flag-tagged WT ATG14 and mutants with HA-tagged beclin 1 observed in a co-immunoprecipitation assay. b, Purification of Flag-tagged PI3KC3 complex subunits from insect cells. c, Graph showing p150 but not beclin 1 stimulated Vps34 lipid kinase activity using in vitro TLC kinase assays. Shown are the mean intensity values of radioactive PI3P spots (± s.d.) (n = 3). AU, arbitrary unit. d, ATG14 enhanced the lipid kinase activity of Vps34–p150–beclin 1. Shown are the mean intensity values of radioactive PI3P spots (± s.d.) (n = 3). e, The requirement of beclin 1 and p150 to the promotion effect of ATG14. Shown are the mean intensity values of radioactive PI3P spots (± s.d.) (n = 3). f, Purification of ATG14 WT and mutants from insect cells. Two doses of each recombinant protein were loaded. g, Effect of ATG14 WT and mutants on Vps34(V)–p150(P)–beclin 1(B) lipid kinase activity. Shown are the mean intensity values of radioactive PI3P spots (± s.d.) (n = 3).

Extended Data Figure 7 Autophagosome targeting of ATG14 homo-oligomerization-deficient mutants.

GFP, GFP–ATG14 WT, GFP–ATG14 C43/46A, GFP–ATG14 C55/58A and GFP–ATG14 4C4A were co-transfected with STX17–Flag in U2OS cells and detected by immunostaining. Endogenous LC3 was detected by anti-LC3 antibody in immunostaining (n = 20). Scale bars, 5 μm.

Extended Data Figure 8 Characterization of the subcellular localization of ATG14 HOD mutants.

a, U2OS cells transfected with GFP–ATG14 WT or ATG14 4C4A and mCherry-Sec61β (ER marker) were treated with EBSS for 2 h and co-stained with endogenous Tom20 (mitochondria marker). Both GFP–ATG14 and GFP–ATG14 4C4A were found at the junction of ER (Sec61β) and mitochondria (Tom20) (n = 20). Scale bars, 5 μm. b, Accumulation of endogenous LC3 and GFP–STX17, but not mCherry-Atg16, in ATG14 4C4A mutant but not in ATG14 WT or knockdown (KD) cells (n = 20). Scale bars, 5 μm. c, ATG14 knockdown U2OS cells stably complemented with ATG14 WT or 4C4A mutant ATG14 were transfected with GFP–ATG16, and endogenous LC3 staining and ATG16 fluorescence were imaged. LC3 but not ATG16 puncta were accumulated in ATG14 4C4A cells (n = 20). Scale bars, 5 μm. d, ATG14 knockdown U2OS cells stably complemented with ATG14 WT or 4C4A mutant ATG14 were transfected with GFP–STX17, and endogenous ATG16 staining and STX17 fluorescence were imaged. STX17 but not ATG16 puncta were accumulated in ATG14 4C4A cells (n = 20). Scale bars, 5 μm.

Extended Data Figure 9 ATG14 homo-oligomerization is required for autophagosome maturation.

a, mRFP–GFP–LC3, with WT or mutant ATG14, were expressed in U2OS cells, and LC3 puncta (overlapped green and red fluorescence) in ATG14-expressing cells were imaged (n = 20). Scale bars, 5 μm. Yellow LC3 signals represent phagophores or autophagosomes before fusion. Red-only LC3 signals represent acidified mature autophagosomes. Un, untreated. Rap, rapamycin. The corresponding quantitative statistical analysis is shown in Fig. 4c. b, Stable expression of Flag–ATG14 or Flag–ATG14 4C4A in ATG14 knockdown U2OS cells. c, Protease protection assay in U2OS ATG14 WT cells treated with chloroquine (2 h). U2OS ATG14 WT cells were infected with lentivirus expressing GFP–LC3 for 24 h and treated with chloroquine 2 h before harvest. The autophagosome-enriched fraction from these cells was isolated by centrifugation, and incubated with trypsin (10 μg ml−1) with and without 0.4% Triton X-100 for 25 min at 30 °C. GFP or GFP–LC3 were detected by western blotting.

Extended Data Table 1 Statistics of X-ray data collection and refinement for the autophagic SNARE complex

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Diao, J., Liu, R., Rong, Y. et al. ATG14 promotes membrane tethering and fusion of autophagosomes to endolysosomes. Nature 520, 563–566 (2015). https://doi.org/10.1038/nature14147

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14147

This article is cited by

Comments

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

Search

Quick links

Nature Briefing

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

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