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:

Lipid signalling drives proteolytic rewiring of mitochondria by YME1L

Abstract

Reprogramming of mitochondria provides cells with the metabolic flexibility required to adapt to various developmental transitions such as stem cell activation or immune cell reprogramming, and to respond to environmental challenges such as those encountered under hypoxic conditions or during tumorigenesis1,2,3. Here we show that the i-AAA protease YME1L rewires the proteome of pre-existing mitochondria in response to hypoxia or nutrient starvation. Inhibition of mTORC1 induces a lipid signalling cascade via the phosphatidic acid phosphatase LIPIN1, which decreases phosphatidylethanolamine levels in mitochondrial membranes and promotes proteolysis. YME1L degrades mitochondrial protein translocases, lipid transfer proteins and metabolic enzymes to acutely limit mitochondrial biogenesis and support cell growth. YME1L-mediated mitochondrial reshaping supports the growth of pancreatic ductal adenocarcinoma (PDAC) cells as spheroids or xenografts. Similar changes to the mitochondrial proteome occur in the tumour tissues of patients with PDAC, suggesting that YME1L is relevant to the pathophysiology of these tumours. Our results identify the mTORC1–LIPIN1–YME1L axis as a post-translational regulator of mitochondrial proteostasis at the interface between metabolism and mitochondrial dynamics.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: YME1L-mediated proteolysis is required for spheroid growth.
Fig. 2: mTORC1 acutely regulates YME1L-dependent proteolysis and mitochondrial import.
Fig. 3: mTORC1 regulates mitochondrial PE in a LIPIN1-dependent manner.
Fig. 4: YME1L-mediated proteolysis is required for PDAC cell growth.

Similar content being viewed by others

Data availability

The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the following dataset identifiers: PXD011750 (DDA data—hypoxia/normoxia WT and Yme1l1−/− MEF mitochondria, https://www.ebi.ac.uk/pride/archive/projects/PXD011750) and PXD014405 (MEF MS/MS library and DIA data, https://www.ebi.ac.uk/pride/archive/projects/PXD014405). Transcriptomic data from hypoxia/normoxia WT MEF cells have been deposited to the GEO omnibus (accession number GSE133753). These datasets are presented in Supplementary Tables 16. Uncropped immunoblot images are available in Supplementary Fig. 1 and all Source Data are available online.

References

  1. Porporato, P. E., Filigheddu, N., Pedro, J. M. B., Kroemer, G. & Galluzzi, L. Mitochondrial metabolism and cancer. Cell Res. 28, 265–280 (2018).

    CAS  PubMed  Google Scholar 

  2. Mehta, M. M., Weinberg, S. E. & Chandel, N. S. Mitochondrial control of immunity: beyond ATP. Nat. Rev. Immunol. 17, 608–620 (2017).

    CAS  PubMed  Google Scholar 

  3. Zhang, H., Menzies, K. J. & Auwerx, J. The role of mitochondria in stem cell fate and aging. Development 145, dev143420 (2018).

    PubMed  PubMed Central  Google Scholar 

  4. Mishra, P. & Chan, D. C. Metabolic regulation of mitochondrial dynamics. J. Cell Biol. 212, 379–387 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Mishra, P., Carelli, V., Manfredi, G. & Chan, D. C. Proteolytic cleavage of Opa1 stimulates mitochondrial inner membrane fusion and couples fusion to oxidative phosphorylation. Cell Metab. 19, 630–641 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Wai, T. et al. Imbalanced OPA1 processing and mitochondrial fragmentation cause heart failure in mice. Science 350, aad0116 (2015).

    PubMed  Google Scholar 

  7. Stiburek, L. et al. YME1L controls the accumulation of respiratory chain subunits and is required for apoptotic resistance, cristae morphogenesis, and cell proliferation. Mol. Biol. Cell 23, 1010–1023 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Anand, R. et al. The i-AAA protease YME1L and OMA1 cleave OPA1 to balance mitochondrial fusion and fission. J. Cell Biol. 204, 919–929 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Sprenger, H. G. et al. Loss of the mitochondrial i-AAA protease YME1L leads to ocular dysfunction and spinal axonopathy. EMBO Mol. Med. 11, e9288 (2019).

    PubMed  Google Scholar 

  10. Jiang, L. et al. Reductive carboxylation supports redox homeostasis during anchorage-independent growth. Nature 532, 255–258 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Mizushima, N. et al. A protein conjugation system essential for autophagy. Nature 395, 395–398 (1998).

    ADS  CAS  PubMed  Google Scholar 

  12. Rainbolt, T. K., Lebeau, J., Puchades, C. & Wiseman, R. L. Reciprocal degradation of YME1L and OMA1 adapts mitochondrial proteolytic activity during stress. Cell Rep. 14, 2041–2049 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. González, A. & Hall, M. N. Nutrient sensing and TOR signaling in yeast and mammals. EMBO J. 36, 397–408 (2017).

    PubMed  PubMed Central  Google Scholar 

  14. Saxton, R. A. & Sabatini, D. M. mTOR signaling in growth, metabolism, and disease. Cell 169, 361–371 (2017).

    CAS  PubMed  Google Scholar 

  15. Brugarolas, J. et al. Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex. Genes Dev. 18, 2893–2904 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Huang, J. & Manning, B. D. The TSC1-TSC2 complex: a molecular switchboard controlling cell growth. Biochem. J. 412, 179–190 (2008).

    CAS  PubMed  Google Scholar 

  17. Cunningham, J. T. et al. mTOR controls mitochondrial oxidative function through a YY1–PGC-1α transcriptional complex. Nature 450, 736–740 (2007).

    ADS  CAS  PubMed  Google Scholar 

  18. Morita, M. et al. mTORC1 controls mitochondrial activity and biogenesis through 4E-BP-dependent translational regulation. Cell Metab. 18, 698–711 (2013).

    CAS  PubMed  Google Scholar 

  19. Laplante, M. & Sabatini, D. M. An emerging role of mTOR in lipid biosynthesis. Curr. Biol. 19, R1046–R1052 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Aaltonen, M. J. et al. MICOS and phospholipid transfer by Ups2-Mdm35 organize membrane lipid synthesis in mitochondria. J. Cell Biol. 213, 525–534 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Tasseva, G. et al. Phosphatidylethanolamine deficiency in mammalian mitochondria impairs oxidative phosphorylation and alters mitochondrial morphology. J. Biol. Chem. 288, 4158–4173 (2013).

    CAS  PubMed  Google Scholar 

  22. Wai, T. et al. The membrane scaffold SLP2 anchors a proteolytic hub in mitochondria containing PARL and the i-AAA protease YME1L. EMBO Rep. 17, 1844–1856 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Eaton, J. M., Mullins, G. R., Brindley, D. N. & Harris, T. E. Phosphorylation of lipin 1 and charge on the phosphatidic acid head group control its phosphatidic acid phosphatase activity and membrane association. J. Biol. Chem. 288, 9933–9945 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Huffman, T. A., Mothe-Satney, I. & Lawrence, J. C., Jr. Insulin-stimulated phosphorylation of lipin mediated by the mammalian target of rapamycin. Proc. Natl Acad. Sci. USA 99, 1047–1052 (2002).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Peterson, T. R. et al. mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway. Cell 146, 408–420 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Craddock, C. P., Adams, N., Bryant, F. M., Kurup, S. & Eastmond, P. J. PHOSPHATIDIC ACID PHOSPHOHYDROLASE regulates phosphatidylcholine biosynthesis in Arabidopsis by phosphatidic acid-mediated activation of CTP:PHOSPHOCHOLINE CYTIDYLYLTRANSFERASE activity. Plant Cell 27, 1251–1264 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Jacquemyn, J., Cascalho, A. & Goodchild, R. E. The ins and outs of endoplasmic reticulum-controlled lipid biosynthesis. EMBO Rep. 18, 1905–1921 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhang, P. & Reue, K. Lipin proteins and glycerolipid metabolism: Roles at the ER membrane and beyond. Biochim. Biophys. Acta Biomembr. 1859, 1583–1595 (2017).

    CAS  PubMed  Google Scholar 

  29. Vaziri-Gohar, A., Zarei, M., Brody, J. R. & Winter, J. M. Metabolic dependencies in pancreatic cancer. Front. Oncol. 8, 617 (2018).

    PubMed  PubMed Central  Google Scholar 

  30. Son, J. et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature 496, 101–105 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Saita, S. et al. PARL partitions the lipid transfer protein STARD7 between the cytosol and mitochondria. EMBO J. 37, e97909 (2018).

    PubMed  PubMed Central  Google Scholar 

  32. Hartmann, B. et al. Homozygous YME1L1 mutation causes mitochondriopathy with optic atrophy and mitochondrial network fragmentation. eLife 5, e16078 (2016).

    PubMed  PubMed Central  Google Scholar 

  33. Baker, M. J. et al. Stress-induced OMA1 activation and autocatalytic turnover regulate OPA1-dependent mitochondrial dynamics. EMBO J. 33, 578–593 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Cybulski, N., Zinzalla, V. & Hall, M. N. Inducible raptor and rictor knockout mouse embryonic fibroblasts. Methods Mol. Biol. 821, 267–278 (2012).

    CAS  PubMed  Google Scholar 

  35. Robitaille, A. M. et al. Quantitative phosphoproteomics reveal mTORC1 activates de novo pyrimidine synthesis. Science 339, 1320–1323 (2013).

    ADS  CAS  PubMed  Google Scholar 

  36. Borowicz, S. et al. The soft agar colony formation assay. J. Vis. Exp. e51998 (2014).

  37. Fuhrer, T., Heer, D., Begemann, B. & Zamboni, N. High-throughput, accurate mass metabolome profiling of cellular extracts by flow injection-time-of-flight mass spectrometry. Anal. Chem. 83, 7074–7080 (2011).

    CAS  PubMed  Google Scholar 

  38. Rappsilber, J., Ishihama, Y. & Mann, M. Stop and go extraction tips for matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS sample pretreatment in proteomics. Anal. Chem. 75, 663–670 (2003).

    CAS  PubMed  Google Scholar 

  39. 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).

    CAS  PubMed  Google Scholar 

  40. Cox, J. et al. Andromeda: a peptide search engine integrated into the MaxQuant environment. J. Proteome Res. 10, 1794–1805 (2011).

    ADS  CAS  PubMed  Google Scholar 

  41. Cox, J. & Mann, M. 1D and 2D annotation enrichment: a statistical method integrating quantitative proteomics with complementary high-throughput data. BMC Bioinformatics 13 (Suppl. 16), S12 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Nolte, H., MacVicar, T. D., Tellkamp, F. & Krüger, M. Instant Clue: a software suite for interactive data visualization and analysis. Sci. Rep. 8, 12648 (2018).

    ADS  PubMed  PubMed Central  Google Scholar 

  43. Tatsuta, T. Quantitative analysis of glycerophospholipids in mitochondria by mass spectrometry. Methods Mol. Biol. 1567, 79–103 (2017).

    CAS  PubMed  Google Scholar 

  44. Bligh, E. G. & Dyer, W. J. A rapid method of total lipid extraction and purification. Can. J. Biochem. Physiol. 8, 911–917 (1959).

    Google Scholar 

  45. Schwarz, D. et al. Preparative scale expression of membrane proteins in Escherichia coli-based continuous exchange cell-free systems. Nat. Protocols 2, 2945–2957 (2007).

    CAS  PubMed  Google Scholar 

  46. Potting, C. et al. TRIAP1/PRELI complexes prevent apoptosis by mediating intramitochondrial transport of phosphatidic acid. Cell Metab. 18, 287–295 (2013).

    CAS  PubMed  Google Scholar 

  47. Titov, D. V. et al. Complementation of mitochondrial electron transport chain by manipulation of the NAD+/NADH ratio. Science 352, 231–235 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. Scheuner, D. et al. Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol. Cell 7, 1165–1176 (2001).

    CAS  PubMed  Google Scholar 

  49. Thoreen, C. C. et al. A unifying model for mTORC1-mediated regulation of mRNA translation. Nature 485, 109–113 (2012).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D. Ehrentraut for expert technical assistance; L. Raatz and M. Heitmann for assisting with xenograft analysis and tissue protein extraction; and A. Wilbrand-Hennes and U. Cullmann of the CECAD proteomics facility for technical assistance. RNA sequencing was performed at the Cologne Center for Genomics and Data Analysis in the MPI Biology of Ageing bioinformatics core facility. We thank J. Altmüller, M. Franitza, F. Metge and J. Boucas for technical and bioinformatic assistance. This work was supported by grants from the Deutsche Forschungsgemeinschaft (La918/15-1; SFB1218/A1), the German-Israel-Project (DIP; RA1028/10-1) and the Max-Planck-Society to T.L.; EMBO and Alexander von Humboldt fellowships to T.M. (GA-2013-609409, ALTF 1220-2014); and a fellowship of the Japan Society for the Promotion of Science (JSPS) for research abroad, The Osamu Hayaishi Memorial Scholarship for Study Abroad and grants from the Uehara Memorial Foundation to Y.O. We are grateful for the following gifts: Tsc2/ MEFs from C. Demetriades and D. Kwiatkowski, Atg5−/− MEFs from K. Winklhofer, WT (S/S) and EIF2αS51A knock-in MEFs from R. Kaufman, 4E-BP DKO, iRaptor and iRictor MEFs from H. McBride and pUC57-LbNOX from V. Mootha (Addgene plasmid # 75285).

Author information

Authors and Affiliations

Authors

Contributions

The study was conceived and designed by T.M., Y.O. and T.L.; T.M. performed the analysis in hypoxia and spheroids; Y.O. performed lipid analysis and in vitro reconstitution experiments; proteomic experiments were performed and evaluated by T.M., B.L., F.C.M., H.N. and M.K.; metabolomic experiments were performed by T.M. and N.Z.; T.T. performed lipid transfer assays; H.-G.S. supported data interpretation; Y.Z., J.L. and C.B. provided support for clinical materials and performed the xenograft assay; T.M., M.H. and J.R. performed and evaluated import and labelling experiments; R.S., M.P., S.H. and J.C.B. assisted with in vivo studies and supported data interpretation; T.M., Y.O. and T.L. wrote the manuscript, which was edited by all authors.

Corresponding author

Correspondence to Thomas Langer.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Peer review information Nature thanks Thomas Becker, Markus Ralser and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Fig. 1 Characterization of YME1L-dependent spheroid growth.

a, b, Spheroid surface area (a) and ATP levels (b) of HEK293 cell lines after 7 days of culture (a, n = 5 independent experiments; b, n = 3 independent experiments; # is number of spheroids per condition). c, d, Spheroid surface area (c) and ATP content (d) of WT and YME1L/ HeLa cells after 7 days (n = 3 independent experiments; # is number of spheroids per condition). e, Quantification of the NAD/NADH ratio in WT and YME1L/ HEK293 cells cultured in normoxia (21% O2) or hypoxia (0.5% O2) for 24 h (n = 3 independent experiments). f, Spheroid surface area of WT and YME1L/ HEK293 cells and of YME1L/ HEK293 cells expressing Lb-NOX (NADH oxidase from Lactobacillus brevis)47 after 7 days (n = 1 experiment; # is number of spheroids per condition). The expression of Lb-NOX was determined by SDS–PAGE and immunoblotting of control and Lb-NOX-expressing WT and YME1L/− HEK293 cells in normoxia (21% O2) or hypoxia (0.5% O2) for 24 h. g, The dependency of WT and YME1L/ HEK293 cells on glucose oxidation (left) and fatty acid oxidation (right) was monitored using the XF Fuel Flex Assay. Dependencies are calculated as a percentage of combined glucose, fatty acid and glutamine oxidation (n = 4 independent experiments). hj, Quantification of cellular metabolites from HEK293 cells (h, i) and MEFs (j). Metabolite abundance was normalized to those of WT cells (n = 5 independent experiments). Mean ± s.e.m.; one-way ANOVA with Dunnett’s multiple comparisons test (a, b); two tailed t-test (c, d, g); two-way ANOVA with Sidak’s multiple comparisons test (e).

Source data

Extended Data Fig. 2 HIF1α drives YME1L-dependent proteolysis and occurs independently of mitophagy.

a, SDS–PAGE and immunoblot analysis of WT and YME1L/ HEK293 cells expressing YME1L or YME1L(E543Q) cultured for 7 days as monolayers (M) or spheroids (S) (n = 1 experiment). b, Representative immunoblot of WT and YME1L/ HEK293 cells and YME1L/ HEK293 cells expressing YME1L or YME1L(E543Q) cultured in normoxia (21% O2) or hypoxia (0.5% O2) for 16 h (n = 4 independent experiments). c, SDS–PAGE and immunoblot analysis of the indicated cell lines treated with control (Scr) or Yme1l siRNA and incubated in normoxia (21% O2) or hypoxia (0.5% O2) for 16 h (n = 1 experiment). d, SDS–PAGE and immunoblot analysis of WT and YME1L/ HEK293 cells treated with Co(II)Cl2 (200 µM) for 24 h or DFP (1 mM) for 24 h. Quantification of PRELID1 and TIMM23 protein levels is shown (n = 3 independent experiments; fc, fold change). e, Quantified PRELID1 and TIMM17A protein levels in HeLa cells transfected with Gfp or Hif1α esiRNA cultured in normoxia or hypoxia for 24 h (example blot shown in Fig. 1 f; n = 3 independent experiments; fc, fold change). f, SDS–PAGE and immunoblot analysis of WT and Atg5/ MEFs cultured in hypoxia (0.5% O2) for the indicated time. Quantification of PRELID1 and TIMM23 protein levels is shown (n = 3 independent experiments; fc, fold change). Mean ± s.e.m.; two-way ANOVA with Sidak’s multiple comparisons test (e).

Source data

Extended Data Fig. 3 YME1L reshapes the mitochondrial proteome independently of OMA1.

a, Box plot analysis of the log2 ratio distribution comparing hypoxia and normoxia in WT and Yme1l/ MEFs of total proteins and mitochondrial proteins. P values calculated by Wilcoxon sum-rank test. Centre lines denote medians, box limits denote 25th and 75th percentiles; whiskers denote maxima and minima (1.5 times the interquartile range). Data located outside the maxima or minima were denoted as outliers and removed. b, Volcano plot representation of proteins determined by quantitative mass spectrometry after isolation of mitochondria from WT and Yme1l/ MEFs cultivated under normoxic conditions (dataset as in Fig. 1g, n = 5 independent experiments, two-tailed t-test). Filled plots indicate proteins that differ significantly between Yme1l/ and WT MEFs at a permutation-based estimated FDR < 0.05. Among these, mitochondrial proteins (according to Gene Ontology Cellular Component) are highlighted in blue. Mitochondrial proteins enriched in Yme1l/ compared to WT are putative YME1L substrates (class I or II). c, d, Volcano plots of mitochondrial protein changes in hypoxia versus normoxia from WT (c) or Yme1l/ (d) MEFs (dataset as in Fig. 1g, n = 5 independent experiments, two-tailed t-test). Class I YME1L substrates are highlighted in red. e, Z-score of log2-transformed LFQ intensities of class I YME1L substrates in WT, Oma1/ and Oma1/Yme1l/ MEFs treated as in Fig. 1g (n = 5 independent experiments). f, SDS–PAGE and immunoblot analysis of WT, Yme1l/ and Oma1/ MEFs cultured in normoxia (21% O2) or hypoxia (0.5% O2) for 24 h. # denotes nonspecific cross-reaction (n = 1 experiment). g, Spheroid surface area of the indicated HEK293 cell lines after 7 days (means from 16 spheroids shown, n = 1 experiment).

Source data

Extended Data Fig. 4 mTORC1 acutely regulates YME1L-dependent proteolysis and mitochondrial import.

ac, SDS–PAGE and immunoblot analysis of WT and YME1L−/− HEK293 cells cultured in hypoxia (0.5% O2) (a), glutamine-depleted medium (b) or in the presence of 400 nM Torin1 (c) for the indicated times (representative immunoblots from n = 3 independent experiments, quantification shown in Fig. 2b). d, Immunofluorescence of WT and YME1L/ HeLa cells treated with Torin1 (400 nM) for 4 h. Cells were immunostained with TIMM17A- and ATP5β-specific antibodies. Quantification of mean fluorescence intensity per cell is shown (TIMM17A mean from three independent experiments, two-way ANOVA with Sidak multiple comparisons test; ATP5β mean from two independent experiments). # is number of cells per condition; au, arbitrary unit; scale bar, 10 µm. e, Immunoblot of WT, Raptor and Rictor knockout (KO) MEFs. Quantified PRELID1 protein levels are shown (Raptor, n = 4 independent experiments; Rictor, n = 3 independent experiments; mean ± s.e.m.; two-tailed t-test). fc, fold change. # denotes nonspecific cross-reaction. f, HEK293 cells were cultured in glutamine-depleted medium (Starve) for 16 h and then cultured in non-essential amino acid (NEAA)-containing medium for the indicated time. Cell lysates were analysed by SDS–PAGE and immunoblotting (n = 1 experiment). g, HEK293 cells were cultured in serum-depleted medium for 16 h and treated with insulin (100 nM) for the indicated time. Cell lysates were analysed by SDS–PAGE and immunoblotting (n = 1 experiment). h, i, [35S]-SOD2 (h) and [35S]-LIAS (i) were imported into mitochondria isolated from WT and YME1L−/− HEK293 cells treated with or without 400 nM Torin1 for 18 h. Import was stopped after 1.5, 3 and 4.5 min in the presence or absence of membrane potential (∆ψ). Mitochondria were analysed by SDS–PAGE and autoradiography (representative blots from n = 2 (h) and n = 3 (i) independent experiments). Arrows indicate the mature form of SOD2. j, [35S]-LIAS and [35S]-SOD2 were imported into mitochondria isolated from WT and YME1L−/− HEK293 cells incubated in normoxia (21% O2) or hypoxia (0.5% O2) for 18 h. Import was stopped after 5, 10 and 15 min. Mitochondria were analysed by SDS–PAGE and levels of imported [35S]-LIAS and [35S]-SOD2 were observed by autoradiography (n = 1 experiment).

Source data

Extended Data Fig. 5 mTORC1 regulates YME1L-mediated proteolysis in a post-translational manner.

a, b, YME1L degradation of substrates does not depend on eIF2α phosphorylation and integrated stress response (ISR) activation. SDS–PAGE and immunoblot analysis of WT (S/S) and EIF2αS51A knock-in (A/A) MEFs, which cannot activate the ISR48, treated with Torin1 for 4 h (a; n = 3 independent experiments, mean ± s.e.m.) or cultured in normoxia (21% O2) or hypoxia (0.5% O2) for 24 h (b; n = 3 independent experiments). c, d, YME1L degradation of substrates does not depend on mTORC1 regulation of translation initiator factor 4 binding proteins (4E-BPs). SDS–PAGE and immunoblot analysis of WT and Eif4ebp1/Eif4ebp2/ (4E-BP DKO) MEFs treated with Torin1 for 4 h (c; n = 4 independent experiments, mean ± s.e.m.) or cultured in normoxia (21% O2) or hypoxia (0.5% O2) for 16 h (d; n = 3 independent experiments). e, f, We observed a slight reduction in newly synthesized TIMM17A in Torin1-treated WT HEK293 cells, consistent with previous reports49. After treatment with DMSO or Torin1 for 2 h, cells were incubated in labelling medium containing [35S]-methionine for the indicated time before lysis and immunoprecipitation with an antibody targeting TIMM17A. Input (5% of total) and immunoprecipitates (IP) were analysed by SDS–PAGE and autoradiography. [35S]-TIMM17A (indicated by an arrow) was quantified (f). Total TIMM17A protein level was determined by immunoblotting (n = 3 independent experiments, mean ± s.e.m.). au, arbitrary unit. gi, TIMM17A synthesis and the majority of global translation was restored in Torin1-treated MEFs that lack 4E-BP proteins. This confirms that the reduced synthesis of TIMM17A upon mTORC1 inhibition reflects 4E-BP-dependent attenuation of translation49. After treatment with DMSO or Torin1 for 2 h, cells were incubated in labelling medium containing [35S]-methionine for 60 min before lysis and immunoprecipitation with an antibody targeting TIMM17A. TIMM17A levels (h) were analysed and quantified as in f (n = 3 independent experiments, mean ± s.e.m.). Total protein synthesis was determined by the intensity of all bands in input lanes (i) (n = 2 independent experiments). The synthesis rate of TIMM17A was quantified in WT and Eif4ebp1/Eif4ebp2/ (4E-BP DKO) MEFs. au, arbitrary unit. jl, Post-translational degradation of YME1L substrates monitored by [35S]-methionine pulse-chase experiment in HEK293 cells. After labelling for 1 h in [35S]-methionine containing medium, cells were incubated for 4 h and 6 h in radioactive-free medium in the presence and absence of Torin1. [35S]-labelled TIMM17A and STARD7 were immunoprecipitated and their levels determined by SDS–PAGE and autoradiography. Quantification of the fraction of [35S]-TIMM17A (k) and [35S]-STARD7 (l) remaining after 4 and 6 h chase is shown. Torin1 treatment accelerated TIMM17A and STARD7 degradation (n = 3 independent experiments for 4 h, n = 4 independent experiments for 6 h; mean ± s.e.m.; two tailed t-test).

Source data

Extended Data Fig. 6 Decreased mitochondrial PE promotes YME1L-mediated proteolysis.

a, b, Phospholipid analysis of mitochondrial fractions from HeLa cells treated with Torin1 for 4 h (n = 3 independent experiments); a, relative distribution; b, absolute abundance. c, Acyl chain composition of mitochondrial PE from HeLa cells treated with Torin1 for 4 h (n = 3 independent experiments). d, Phospholipid analysis of mitochondrial fractions from WT, PRELID3B/ and PRELID3B/ HeLa cells expressing PRELID3B–Flag or PRELID3B(T57K)–Flag (n = 3 independent experiments). e, Quantification of protein levels from Fig. 2h (n = 4 independent experiments). f, g, Immunoblot and mitochondrial phospholipid analysis of scrambled control (Scr) or Prelid3b siRNA-transfected HeLa cells. Quantification of indicated protein levels is shown (immunoblot, n = 6 independent experiments; phospholipid analysis, n = 3 independent experiments). h, NBD-PS transfer by PRELID3B (black circles), PRELID3B(T57K) (red circles) or without addition of protein (white circles). PRELID3B(T57K) contains a mutation in the PS-binding site, which abolishes PS transfer but does not interfere with the assembly of PRELID3B into lipid transfer complexes. Average of n = 3 independent experiments. i, SDS–PAGE analysis of recombinant PRELID3B–TRIAP1 complexes and the T57K variant (40 pmol) by CBB staining (n = 1 experiment). j, Quantification of protein levels from Fig. 2i (n = 5 independent experiments). k, l, Phospholipid analysis of the mitochondrial fraction from HeLa cells treated with scrambled control (Scr) or Pisd siRNA (n = 3 independent experiments) (k), WT and Tsc2−/− MEFs (n = 3 independent experiments) (l). Mean ± s.e.m.; two tailed t-test (a, b, f, g, jl), one-way ANOVA with Dunnett’s multiple comparisons test (d, e). fc, fold change.

Source data

Extended Data Fig. 7 PE regulates YME1L-mediated proteolysis.

a, b, HeLa cells pretreated with ethanolamine (Etn, 200 µM) for 24 h were treated with Torin1 for 4 h. Phospholipids in mitochondrial fractions were analysed by mass spectrometry (a) (n = 3 independent experiments). Cell lysates were analysed by immunoblotting and indicated protein levels were quantified (n = 5 independent experiments) (b). c, d, HEK293 cells were treated with Torin1 and/or 100 µM LPE for 4 h. Phospholipids in mitochondrial fractions were analysed by mass spectrometry (c) (n = 3 independent experiments). Cell lysates were analysed by immunoblotting and indicated protein levels were quantified (n = 4–5 independent experiments) (d). e, f, HeLa cells transfected with scrambled control (Scr) or Pisd siRNA were treated with 100 µM LPE for 24 h. Cell lysates were analysed by immunoblotting and indicated protein levels were quantified (n = 5 independent experiments) (e). PE levels in mitochondrial fractions were determined by mass spectrometry (n = 3 independent experiments) (f). g, h, YME1L (WT or E543Q) and OPA1ΔC (g) or TIMM17A (h) reconstituted in liposomes was incubated in the presence or absence of ATP at 37 °C for the indicated times. Samples were analysed by immunoblotting (representative data from n = 3 independent experiments). i, j, YME1L (WT or E543Q) and OPA1ΔC (i) or TIMM17A (j) reconstituted in liposomes containing different amounts of PE were incubated in the presence of ATP at 37 °C for the indicated times. Samples were analysed by immunoblotting (representative data from n = 4 independent experiments). Mean ± s.e.m.; two-way ANOVA with Tukey’s multiple comparisons test (ad), with Holm-Sidak’s multiple comparisons test (e, f). fc, fold change.

Source data

Extended Data Fig. 8 Depletion of LIPIN1 preserves YME1L-mediated proteolysis after inhibition of mTORC1.

a, HeLa cells transfected with the indicated esiRNAs were treated with Torin1 for 4 h before immunoblotting. Representative immunoblot and quantification of TIMM17A protein levels are shown (n = 3 independent experiments; fold change (fc) from DMSO control). b, c, Immunoblot analysis of HeLa cells transfected with Gfp or Lipin1 esiRNA (b) or scrambled control (Scr) or Lipin1 siRNA (c) treated with Torin1 for 4 h. Quantification of PRELID1 and TIMM17A protein levels is shown (n = 7 independent experiments (b), n = 6 independent experiments (c)). d, Immunoblot analysis of MEFs transfected with Gfp or Lipin1 esiRNA and treated with Torin1 for 4 h. Quantification of TIMM17A protein levels is shown (n = 5 independent experiments). e, Immunoblot analysis of HeLa cells transfected with the indicated esiRNAs and treated with Torin1 for 4 h (representative data from six independent experiments). f, The mTORC1–LIPIN1–YME1L regulatory axis. Upon inhibition of mTORC1, dephosphorylation of LIPIN1 stimulates its PA-phosphatase activity and leads to a decrease in PA. The depletion of PA inhibits CCTα and therefore limits the supply of PS to mitochondria. Reduced PS transfer to the inner membrane limits accumulation of PE, stimulating YME1L-mediated proteolysis. Mean ± s.e.m.; two-way ANOVA with Tukey’s multiple comparisons test (bd).

Source data

Extended Data Fig. 9 YME1L-mediated proteolysis is enhanced in PDAC and supports the growth of PDAC cells.

a, b, SDS–PAGE and immunoblot analysis of pancreatic tissue from patients with PDAC (a) and liver tissue from patients with HCC (b). T, tumour tissue; NT, corresponding adjacent non-tumour tissue. Quantification of indicated protein levels is shown below (n = 6 independent patient samples). YME1L substrate proteins are highlighted in red, other mitochondrial proteins shown in dark grey. The immunoblots from two other patients are shown in Fig. 4a, b. c, Cellular ATP levels in spheroids of the indicated cell lines cultured for 6 days (n = 3 independent experiments for HPAF-II, HepG2 and Huh7, n = 4 independent experiments for PANC1; # is number of spheroids; fc, fold change). d, e, Immunoblot analysis of spheroids from HPAF-II (d) and Huh7 (e) cells treated with Gfp or Lipin1 esiRNA, quantified in Fig. 4e, f. n = 1 immunoblot. f, Representative immunoblot of HPAF-II cells expressing shGfp and shYme1l (n = 3 independent samples). g, Weights of xenograft tumours derived from HPAF-II cells treated with Gfp or Yme1l shRNA 28 days after injection (Gfp n = 7 mice; Yme1l n = 8 mice). Mean ± s.e.m.; two tailed unpaired t-test (ac, g).

Source data

Extended Data Table 1 Class I and class II YME1L substrates

Supplementary information

Supplementary Figure

Supplementary Figure 1 contains all electrophoresis (gels) source data (PDF).

Reporting Summary

Supplementary Tables

This file contains supplementary tables 1-12.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

MacVicar, T., Ohba, Y., Nolte, H. et al. Lipid signalling drives proteolytic rewiring of mitochondria by YME1L. Nature 575, 361–365 (2019). https://doi.org/10.1038/s41586-019-1738-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1738-6

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: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research