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:

LC3 lipidation is essential for TFEB activation during the lysosomal damage response to kidney injury

An Author Correction to this article was published on 04 October 2022

This article has been updated

Abstract

Sensing and clearance of dysfunctional lysosomes is critical for cellular homeostasis. Here we show that transcription factor EB (TFEB)—a master transcriptional regulator of lysosomal biogenesis and autophagy—is activated during the lysosomal damage response, and its activation is dependent on the function of the ATG conjugation system, which mediates LC3 lipidation. In addition, lysosomal damage triggers LC3 recruitment on lysosomes, where lipidated LC3 interacts with the lysosomal calcium channel TRPML1, facilitating calcium efflux essential for TFEB activation. Furthermore, we demonstrate the presence and importance of this TFEB activation mechanism in kidneys in a mouse model of oxalate nephropathy accompanying lysosomal damage. A proximal tubule-specific TFEB-knockout mouse exhibited progression of kidney injury induced by oxalate crystals. Together, our results reveal unexpected mechanisms of TFEB activation by LC3 lipidation and their physiological relevance during the lysosomal damage response.

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: The lysosomal damage-induced nuclear translocation of TFEB is ATG conjugation system dependent.
Fig. 2: Calcium efflux from lysosomes triggers TFEB nuclear translocation in an ATG conjugation system-dependent manner.
Fig. 3: ATG conjugation system-deficient cells show reduced calcium efflux from lysosomes.
Fig. 4: LC3 localizes on lysosomes during the lysosomal damage response.
Fig. 5: Lipidated LC3 interacts with the TRPML1 channel.
Fig. 6: Lysosomal damage-induced TFEB activation mitigates CaOx nephropathy.

Similar content being viewed by others

Data availability

Data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Change history

References

  1. Papadopoulos, C., Kravic, B. & Meyer, H. Repair or lysophagy: dealing with damaged lysosomes. J. Mol. Biol. https://doi.org/10.1016/j.jmb.2019.08.010 (2019).

  2. Maejima, I. et al. Autophagy sequesters damaged lysosomes to control lysosomal biogenesis and kidney injury. EMBO J. 32, 2336–2347 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hung, Y. H., Chen, L. M., Yang, J. Y. & Yang, W. Y. Spatiotemporally controlled induction of autophagy-mediated lysosome turnover. Nat. Commun. 4, 2111 (2013).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Skowyra, M. L., Schlesinger, P. H., Naismith, T. V. & Hanson, P. I. Triggered recruitment of ESCRT machinery promotes endolysosomal repair. Science https://doi.org/10.1126/science.aar5078 (2018).

  7. Radulovic, M. et al. ESCRT-mediated lysosome repair precedes lysophagy and promotes cell survival. EMBO J. https://doi.org/10.15252/embj.201899753 (2018).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  12. Martina, J. A., Chen, Y., Gucek, M. & Puertollano, R. MTORC1 functions as a transcriptional regulator of autophagy by preventing nuclear transport of TFEB. Autophagy 8, 903–914 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Martina, J. A. & Puertollano, R. Rag GTPases mediate amino acid-dependent recruitment of TFEB and MITF to lysosomes. J. Cell Biol. 200, 475–491 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Paz, I. et al. Galectin-3, a marker for vacuole lysis by invasive pathogens. Cell Microbiol. 12, 530–544 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Chang, C. et al. Bidirectional control of autophagy by BECN1 BARA domain dynamics. Mol. Cell 73, 339–353 (2019).

    Article  CAS  PubMed  Google Scholar 

  18. Puertollano, R., Ferguson, S. M., Brugarolas, J. & Ballabio, A. The complex relationship between TFEB transcription factor phosphorylation and subcellular localization. EMBO J. https://doi.org/10.15252/embj.201798804 (2018).

  19. Li, Y. et al. Protein kinase C controls lysosome biogenesis independently of mTORC1. Nat. Cell Biol. 18, 1065–1077 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sasai, M. et al. Essential role for GABARAP autophagy proteins in interferon-inducible GTPase-mediated host defense. Nat. Immunol. 18, 899–910 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Fujita, N. et al. Recruitment of the autophagic machinery to endosomes during infection is mediated by ubiquitin. J. Cell Biol. 203, 115–128 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  24. Napolitano, G. et al. A substrate-specific mTORC1 pathway underlies Birt–Hogg–Dube syndrome. Nature, https://doi.org/10.1038/s41586-020-2444-0 (2020).

  25. Kilpatrick, B. S., Yates, E., Grimm, C., Schapira, A. H. & Patel, S. Endo-lysosomal TRP mucolipin-1 channels trigger global ER Ca2+ release and Ca2+ influx. J. Cell Sci. 129, 3859–3867 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Cao, Q. et al. BK channels alleviate lysosomal storage diseases by providing positive feedback regulation of lysosomal Ca2+ release. Dev. Cell 33, 427–441 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Shen, D. et al. Lipid storage disorders block lysosomal trafficking by inhibiting a TRP channel and lysosomal calcium release. Nat. Commun. 3, 731 (2012).

    Article  PubMed  Google Scholar 

  28. Grimm, C. Endolysosomal cation channels as therapeutic targets—pharmacology of TRPML channels. Messenger 5, 30–36 (2016).

    Article  Google Scholar 

  29. Mulay, S. R. & Anders, H. J. Crystal nephropathies: mechanisms of crystal-induced kidney injury. Nat. Rev. Nephrol. 13, 226–240 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Mulay, S. R. et al. Calcium oxalate crystals induce renal inflammation by NLRP3-mediated IL-1β secretion. J. Clin. Invest. 123, 236–246 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Knauf, F. et al. NALP3-mediated inflammation is a principal cause of progressive renal failure in oxalate nephropathy. Kidney Int. 84, 895–901 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Mulay, S. R. et al. Cytotoxicity of crystals involves RIPK3–MLKL-mediated necroptosis. Nat. Commun. 7, 10274 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Waikar, S. S. et al. Association of urinary oxalate excretion with the risk of chronic kidney disease progression. JAMA Intern. Med. 179, 542–551 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Kimura, T. et al. Autophagy protects the proximal tubule from degeneration and acute ischemic injury. J. Am. Soc. Nephrol. 22, 902–913 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Mulay, S. R. et al. Mitochondria permeability transition versus necroptosis in oxalate-induced AKI. J. Am. Soc. Nephrol. 30, 1857–1869 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Heckmann, B. L., Boada-Romero, E., Cunha, L. D., Magne, J. & Green, D. R. LC3-associated phagocytosis and inflammation. J. Mol. Biol. 429, 3561–3576 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Fletcher, K. et al. The WD40 domain of ATG16L1 is required for its non-canonical role in lipidation of LC3 at single membranes. EMBO J. 37, e97840 (2018).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Saitoh, T. et al. TWEAK induces NF-κB2 p100 processing and long lasting NF-κB activation. J. Biol. Chem. 278, 36005–36012 (2003).

    Article  CAS  PubMed  Google Scholar 

  41. Morita, S., Kojima, T. & Kitamura, T. Plat-E: an efficient and stable system for transient packaging of retroviruses. Gene Ther. 7, 1063–1066 (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Kobayashi, S. et al. Artificial induction of autophagy around polystyrene beads in nonphagocytic cells. Autophagy 6, 36–45 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Minami, S. et al. Lipophagy maintains energy homeostasis in the kidney proximal tubule during prolonged starvation. Autophagy 13, 1629–1647 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pizzolato, P. Histochemical recognition of calcium oxalate. J. Histochem. Cytochem. 12, 333–336 (1964).

    Article  CAS  PubMed  Google Scholar 

  46. Chaiyarit, S. & Thongboonkerd, V. Changes in mitochondrial proteome of renal tubular cells induced by calcium oxalate monohydrate crystal adhesion and internalization are related to mitochondrial dysfunction. J. Proteome Res. 11, 3269–3280 (2012).

    Article  CAS  PubMed  Google Scholar 

  47. Takahashi, A. et al. Autophagy guards against cisplatin-induced acute kidney injury. Am. J. Pathol. 180, 517–525 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Lazarou (Monash University) for providing us with ATG8s-deficient HeLa cells. S.N. is supported by AMED-PRIME (grant no. 17gm6110003h0001), JSPS KAKENHI (grant no. 17H05064), the Senri Life Science Foundation, Takeda Science Foundation, Nakajima Foundation, MSD Life Science Foundation, Astellas Foundation for Research on Metabolic Disorders and Mochida Memorial Foundation for Medical and Pharmaceutical Research. T.Y. is supported by JST CREST (grant no. JPMJCR17H6), AMED (grant no. JP19gm5010001), Takeda Science Foundation, JSPS A3 Foresight Program and an HFSP research grant. This work was also supported by grants from the Italian Telethon Foundation (grant no. TGM16CB6), MIUR FIRB (grant no. RBAP11Z3YA; A.B.), European Research Council (advanced investigator grant no. 694282; LYSOSOMICS; A.B.), Associazione Italiana per la Ricerca sul Cancro (A.I.R.C.) (A.B.), US National Institutes of Health (grant no. R01-NS078072; A.B.), the Huffington foundation (A.B.), European Regional Development Fund—POR Campania FESR 2014/2020 (A.B.), MIUR PRIN (grant no. 2017YF9FBS; G.N.), University of Naples ‘Federico II’ STAR L1 2018 (G.N.) and Associazione Italiana per la Ricerca sul Cancro MFAG 2019, grant no. 23538 (G.N.).

Author information

Authors and Affiliations

Authors

Contributions

S.N., S.M., G.N., A.B., Y.T. and T.Y. wrote the manuscript with input from the other authors. S.N. and T.Y. supervised the project. S.N., S.S. and T.M. generated the stable cell lines and performed calcium imaging. S.N., S.S., S.M., A.K., M.M., T.S., S.A., A.E., G.N. and A.Tokumura conducted western blotting and immunohistochemistry using cell culture. S.N, S.M., S.S. and T.N.-H. obtained and analysed the transmission electron microscopy images. S.M., J.N., Y.T. and A.Takahashi conducted the mouse experiments. K.Y., M.H., M.Y., Y. Okada, T.N., Y.T., Y.I., H.X. and M.K. analysed data and provided intellectual support. S.N., M.S., Y. Oe, T. F. and S.T. generated the KO cells and plasmid constructs. A.B. generated the TFEB construct and TFEB flox mice.

Corresponding authors

Correspondence to Shuhei Nakamura or Tamotsu Yoshimori.

Ethics declarations

Competing interests

S.N. and T.Y. have applied for a patent for the assay related to this work (patent application no. 2020-63351). T.Y. is founder of AutoPhagyGO. A.B. is a co-founder of CASMA Therapeutics, Inc.

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 TFEB is essential for lysosomal homeostasis after the injury.

(a) Representative immunoblots confirming efficient reduction of ATG16L1 and TFEB protein levels by indicated siRNA knockdown (representative of 3 independent experiments). Actin was used as a loading control. (b) Representative immunoblots confirming absence of TFEB protein in established TFEB-KO cells (representative of 3 independent experiments). (c) LLOMe treatment and subsequent chase did not affect the cell viability of WT and TFEB-KO HeLa cells. Cells were treated with 1 mM LLOMe for 1 h and then subjected to cell viability assay at the indicated time point after LLOMe washout. Bars represent mean ± s.d. (n = 3 biologically independent experiments). (d) Transmission electron microscopy (TEM) observation of ultrastructural changes of lysosomes in WT and TFEB-KO cells after LLOMe treatment (representative of 2 independent experiments). Cells were treated with 1 mM LLOMe for 1 h and then fixed at the indicated time point after LLOMe washout. Lysosomes showed enlarged and disorganized morphology soon after LLOMe treatment. After 3 h wash-off by DMEM, the morphology of lysosomes appeared to be normal round shape in WT cells, while it showed still distorted shape in TFEB-KO cells. Scale bars, 500 nm. Uncropped blots and statistical source data are provided in Source Data Extended Data Fig. 1.

Source data

Extended Data Fig. 2 Lysosomal damage, but not starvation, induces TFEB nuclear translocation in an ATG conjugation system-dependent manner.

(a, b) Representative immunoblots showing TFEB, phospho S6K, total S6K, phospho GSKαβ, total GSKβ, LC3 (a) and actin in WT and several KO HeLa cells (3 independent experiments for both a and b). TFEB dephosphorylation (downshift) was observed in all KO cells under EBSS starvation (b), whereas TFEB dephosphorylation was partly impaired in ATG7-KO and ATG16L1-KO cells under LLOMe treatment (a). Samples were collected after EBSS starvation (4 h), or LLOMe treatment (3 h washout after 1 h LLOMe treatment). (c, d) Representative immunoblots showing TFEB expression in WT and ATG3-KO (c) or ATG5-KO (d) HeLa cells under control conditions, EBSS starvation (4 h), or LLOMe treatment (3 h washout after 1 h LLOMe treatment) (n = 3 independent experiments for both c and d). TFEB dephosphorylation (downshift) induced by LLOMe but not EBSS was impaired in ATG3-KO and ATG5-KO cells. (e) Representative immunoblots showing LC3 in WT and several ATG conjugation-deficient HeLa cells which we generated in this study treated with or without Bafilomycin A (3 independent experiments). Note that lipidated LC3-II form was not present in these ATG conjugation-deficient cells. Uncropped blots are provided in Source Data Extended Data Fig. 2.

Source data

Extended Data Fig. 3 Requirement of Atg8 paralogues for TFEB nuclear localization during lysosomal damage.

(a) MEF cell lines stably expressing TFEB::mNeonGreen (green) were treated with or without LLOMe (3 h washout after 1 h LLOMe treatment). (b) Quantification of nuclear/cytoplasmic ratio of TFEB::mNeonGreen under each condition. More than 100 cells were analysed per condition by cell profiler and the experiments were repeated three times. Bars represent mean ± s.d. (n = 3 biologically independent experiments, one-way ANOVA with Tukey’s test, *P = 0.0166) Scale bars, 50 μm. Statistical source data are provided in Source Data Extended Data Fig. 3.

Source data

Extended Data Fig. 4 Transfection of Effectene-coated beads induces TFEB nuclear localization in an ATG conjugation system-dependent manner.

(a) Representative immunofluorescence images showing TFEB::mNeonGreen and Gal-3 counterstained by DAPI. Transfection of Effectene-coated beads damaged endosomes or lysosomes, whereas transfection with PEI-coated beads did not. Note that cells harbouring the damage visualized by Gal-3 exhibited TFEB nuclear translocation (open arrowheads). DIC images showed that transfected beads were internalized in cells. (b) Ratio of nuclear to cytoplasmic TFEB::mNeonGreen in WT and ATG7-KO cells transfected with either Effectene- or PEI-coated beads. More than 100 cells were analysed per condition by cell profiler and the experiments were repeated three times. Bars represent mean ± s.d. (n = 3 biologically independent experiments, one-way ANOVA with Tukey’s test, **P = 0.001 for WT PEI versus effectene, P = 0.9711 for ATG7-KO PEI versus effectene) Scale bars, 50 μm. Statistical source data are provided in Source Data Extended Data Fig. 4.

Source data

Extended Data Fig. 5 Calcium efflux from lysosomes triggers TFEB nuclear translocation in an ATG conjugation system-dependent manner.

(a) Representative immunoblots showing TFEB expression in WT and ATG3-KO HeLa cells treated with DMSO (control) or ML-SA1 for 1 h (3 independent experiments). In ATG3-KO cells, TFEB activation (downshift) was defective after ML-SA1 treatment. (b) The Ca2+ chelator BAPTA-AM reduced TFEB::mNeonGreen nuclear localization induced by ML-SA1. (c) Ratio of nuclear to cytoplasmic TFEB::mNeonGreen in WT HeLa cells treated with indicated drugs. Bars represent mean ± s.d. (n = 3 biologically independent experiments, one-way ANOVA with Tukey’s test, *P = 0.0435 for DMSO control versus ML-SA1, P = 0.216 for BAPTA control versus ML-SA1). (d) Representative images showing that knockdown of PPP3CB did not abolish TFEB::mNeonGreen nuclear translocation after LLOMe treatment (3 h after 1 h LLOMe treatment) (3 independent experiments). (e) Representative blots for TFEB and PPP3CB (3 independent experiments). Knockdown of PPP3CB did not interfere with TFEB dephosphorylation. (f) Representative blots for TFEB and PPP3CB (3 independent experiments). Double knockdown of PPP3CB and PPP3CA did not largely affect TFEB status by LLOMe treatment. (g) qRT-PCR confirmed PPP3CA transcripts were significantly reduced by PPP3CA knockdown. Two different siRNA (#1 and #2) for PPP3CA were used. Bars represent mean ± s.d. (n = 3 biologically independent experiments, one-way ANOVA with Tukey’s test, ****P < 0.0001 for siLuc versus siPPP3CA#1 or #2). Scale bars, 50 μm (b and d). Uncropped blots and statistical source data are provided in Source Data Extended Data Fig. 5.

Source data

Extended Data Fig. 6 TRPML1 interacts with ATG8s.

(a) Co-immunoprecipitation experiments revealed that TRPML1::HA interacted with endogenous LC3A, LC3B, GABARAP, GABARAPL1 and GABARAPL2 (representative of 3 independent experiments). (b) Co-immunoprecipitation experiments revealed that TRPML1::HA interacted with 3x FLAG::LC3C (representative of 3 independent experiments). (c) TRPML1::HA overexpression increased LC3 dots. Most of LC3 dots(magenta) were colocalized on lysosomes in TRPML1::HA overexpressed cells. (d) Quantification of numbers of LC3 dots per cells with HA or TRPML1::HA overexpressed cells. Bars represent mean ± s.d. (n = 3 biologically independent experiments, two-tailed unpaired t-test, *P = 0.0262). (e) Representative immunoblots confirming efficient reduction of TSG101 and ALIX protein levels by indicated siRNA knockdown (3 independent experiments). Scale bars, 50 μm (c). Uncropped blots and statistical source data are provided in Source Data Extended Data Fig. 6.

Source data

Extended Data Fig. 7 CaOx crystals cause lysosomal damage in mouse kidneys.

(a) Representative images of cultured PTECs stably expressing GFP-galectin 3 under the indicated conditions (n = 3 animals in each group). (b) Levels of plasma UN and creatinine after oxalate administration (n = 6 animals for 0 h, n = 3 animals for 2, 6, 12, 24 and 48 h). (c) Representative images of Pizzolato staining on the kidney sections of mice 24 h after oxalate administration. CaOx crystals are shown within the PTECs (arrows) as well as in the tubular lumen. (d) Quantification of positive area after Pizzolato staining is shown (n = 2 animals for 12 h; n = 3 animals for 0, 2, 6, 24 and 48 h). (ej) WT mice were assessed 24 h after vehicle or oxalate injection (75 mg/kg) (n = 3 animals in each group). Representative images of PAS staining (e), immunostaining for LAMP1 (f), Galectin-3 and LAMP1 (g), Galectin-3 and LRP2/MEGALIN (h, left), Galectin-3 and THP (h, middle), Galectin-3 and AQP2 (h, right), and TFEB (j) on the kidney sections. Kidney sections were counterstained with hematoxylin (f) or DAPI (g and h). (i) Electron micrographs of PTECs. Arrowheads indicate elongating isolation membrane along the surface of the lysosome (n = 3 animals). Mt, mitochondria; asterisk: lysosome. Original magnification: ×100 (c, left); ×200 (e, left; f, left; j, left); ×400 (c, middle; e, middle; j, right);×1000 (c, right; e, right; f, right). Scale bars, 20 μm (a, g and h), 50 μm (c, e, f and j), 500 nm (i). Uncropped blots and statistical source data are provided in Source Data Extended Data Fig. 7.

Source data

Extended Data Fig. 8 Lysosomal damage induces TFEB nuclear translocation in an ATG5-dependent manner in PTECs.

(a) Representative immunofluorescence images of mNeonGreen-tagged TFEB in ATG5-negative or ATG5-positive PTECs counterstained with DAPI (blue). Non-treated control samples and samples 1 h after 1 mM LLOMe treatment (1 h) are shown (3 independent experiments). (b, c) Representative images of PAS staining (a) and electron micrographs (b) of the kidneys in control and PTEC-specific Tfeb-deficient mice [n = 6 animals (a) and n = 3 animals (b) in each group]. Mt, mitochondria; asterisk, lysosome. Scale bars, 20 μm (a), 50 μm (b), 500 nm (c).

Extended Data Fig. 9 The severity of kidney injury is comparable between control and TFEB-KO mice at 6 h after oxalate administration.

(af) Images of Pizzolato staining (a), quantification of positive area (b), PAS staining (c), PAS injury score (d), levels of plasma UN and creatinine (e), mRNA expression levels of kidney injury marker genes (f), TUNEL staining (g), and numbers of TUNEL-positive cells (h) are shown. TfebF/F and TfebF/F;KAP-Cre mice were subjected to oxalate injection (75 mg/kg) and verified after 6 h (a–f, n = 3 animals in each group) and 48 h (g and h, TfebF/F, n = 15 animals; TfebF/F;KAP-Cre, n = 9 animals). two-tailed unpaired t-test, P = * 0.0285 (h). Bars: means ± s.d. Scale bars, 50 μm (a, c and g). Statistical source data are provided in Source Data Extended Data Fig. 9.

Source data

Extended Data Fig. 10 Mitochondrial morphological changes during the lysosomal damage response.

(a) TfebF/F and TfebF/F;KAP-Cre mice were subjected to oxalate injection (75 mg/kg) and verified after 48 h (3 animals in each group). Representative images of electron microscopy are shown. Mt, mitochondria. (b) Transmission electron microscopy (TEM) observation of ultrastructural changes of mitochondria in WT and TFEB-KO HeLa cells after LLOMe treatment (representative of 2 independent experiments). Cells were treated with 1 mM LLOMe for 1 h and then fixed for TEM imaging at the indicated time point after LLOMe washout. Fragmented mitochondria was observed soon after LLOMe treatment (0 h) both in WT and TFEB- KO cells. The fragmentation seems to be recovered after 3h LLOMe washout. In TFEB -KO cells, the fragmented mitochondria was observed even in control cells. (c) Representative images showing mitochondrial morphological changes immunostained by TOMM20 antibody (green) during the lysosomal damage (3 independent experiments). Mitochondrial fragmentation occurred after 1 h LLOMe treatment, while interestingly, during the continuous LLOMe treatment for 3 h mitochondrial morphology recovered to normal. These morphological changes occurred in WT, FIP200-KO, ATG7-KO and TFEB-KO cells, indicating that these changes depend neither on the function of autophagy nor TFEB. (d) Representative immunoblots showing the change of complex III core subunit 1 protein level in indicated cells during lysosomal damage response (3 independent experiments). Mitophagy was not induced by 1 mM LLOMe treatment for 1 h or 3 h. (e) Wild type mice were subjected to oxalate injection (75 mg/kg) and verified after 48 h (n = 3 animals in each group). Representative images of electron microscopic analysis revealed that contents of lysosome-like structures were observed within the tubular lumen, indicating the presence of lysosomal exocytosis. N, nucleus; TL, tubular lumen; arrows, contents of lysosome-like structures. Scale bars, 1 μm (a and e), 500 nm (b), 50 μm (c). Uncropped blots are provided in Source Data Extended Data Fig. 10.

Source data

Supplementary information

Reporting Summary

Supplementary Video 1

Effect of 50 μM ML-SA1 on the cytosolic Ca2+ levels of Fura-2-loaded WT HeLa cells. Images were acquired every 3 s for 15 min and played back at a rate of 30 frames s−1. Warmer colours represent the Fura-2 fluorescence ratio, which is proportional to the Ca2+ concentration. Biologically independent experiments were repeated three times with similar results.

Supplementary Video 2

Fura-2 calcium imaging in ATG7-KO HeLa cells. Effect of 50 μM ML-SA1 on the cytosolic Ca2+ levels of Fura-2-loaded ATG7-KO HeLa cells. Images were acquired every 3 s for 15 min and played back at rate of 30 frames s−1. Warmer colours represent the Fura-2 fluorescence ratio, which is proportional to the Ca2+ concentration. Biologically independent experiments were repeated three times with similar results.

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

Statistical source data

Source Data Extended Data Fig. 1

Statistical source data

Source Data Extended Data Fig. 1

Unprocessed western blots

Source Data Extended Data Fig. 2

Unprocessed western blots

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. 5

Unprocessed western blots

Source Data Extended Data Fig. 6

Statistical source data

Source Data Extended Data Fig. 6

Unprocessed western blots

Source Data Extended Data Fig. 7

Statistical source data

Source Data Extended Data Fig. 9

Statistical source data

Source Data Extended Data Fig. 10

Unprocessed western blots

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nakamura, S., Shigeyama, S., Minami, S. et al. LC3 lipidation is essential for TFEB activation during the lysosomal damage response to kidney injury. Nat Cell Biol 22, 1252–1263 (2020). https://doi.org/10.1038/s41556-020-00583-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-020-00583-9

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