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:

Control of plasma membrane lipid homeostasis by the extended synaptotagmins

This article has been updated

Abstract

Acute metabolic changes in plasma membrane (PM) lipids, such as those mediating signalling reactions, are rapidly compensated by homeostatic responses whose molecular basis is poorly understood. Here we show that the extended synaptotagmins (E-Syts), endoplasmic reticulum (ER) proteins that function as PtdIns(4,5)P2- and Ca2+-regulated tethers to the PM, participate in these responses. E-Syts transfer glycerolipids between bilayers in vitro, and this transfer requires Ca2+ and their lipid-harbouring SMP domain. Genome-edited cells lacking E-Syts do not exhibit abnormalities in the major glycerolipids at rest, but exhibit enhanced and sustained accumulation of PM diacylglycerol following PtdIns(4,5)P2 hydrolysis by PLC activation, which can be rescued by expression of E-Syt1, but not by mutant E-Syt1 lacking the SMP domain. The formation of E-Syt-dependent ER–PM tethers in response to stimuli that cleave PtdIns(4,5)P2 and elevate Ca2+ may help reverse accumulation of diacylglycerol in the PM by transferring it to the ER for metabolic recycling.

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: Localization of endogenous E-Syt1.
Figure 2: E-Syt1 is a Ca2+-dependent lipid transfer protein.
Figure 3: Generation of E-Syt1/2 DKO and E-Syt1/2/3 TKO HeLa cells using TALEN and CRISPR.
Figure 4: Rapid isolation and characterization of PM sheets.
Figure 5: Prolonged accumulation of DAG in E-Syt KO cells on phospholipase activation.
Figure 6: SMP-domain-dependent and Ca2+-regulated diacylglycerol transfer by E-Syt1.
Figure 7: PM DAG extraction mediated by E-Syts.

Similar content being viewed by others

Change history

  • 14 April 2016

    In the version of this Article originally published online, '100 mM' in the x axes labels of Fig. 6c should have read '100 μM'. This has been corrected in all versions of the Article.

References

  1. Holthuis, J. C. & Levine, T. P. Lipid traffic: floppy drives and a superhighway. Nat. Rev. Mol. Cell Biol. 6, 209–220 (2005).

    Article  CAS  Google Scholar 

  2. Elbaz, Y. & Schuldiner, M. Staying in touch: the molecular era of organelle contact sites. Trends Biochem. Sci. 36, 616–623 (2011).

    Article  CAS  Google Scholar 

  3. Friedman, J. R. & Voeltz, G. K. The ER in 3D: a multifunctional dynamic membrane network. Trends Cell Biol. 21, 709–717 (2011).

    Article  CAS  Google Scholar 

  4. Toulmay, A. & Prinz, W. A. Lipid transfer and signaling at organelle contact sites: the tip of the iceberg. Curr. Opin. Cell Biol. 23, 458–463 (2011).

    Article  CAS  Google Scholar 

  5. Stefan, C. J. et al. Osh proteins regulate phosphoinositide metabolism at ER-plasma membrane contact sites. Cell 144, 389–401 (2011).

    Article  CAS  Google Scholar 

  6. Mesmin, B. et al. A four-step cycle driven by PI(4)P hydrolysis directs sterol/PI(4)P exchange by the ER-Golgi tether OSBP. Cell 155, 830–843 (2013).

    Article  CAS  Google Scholar 

  7. Prinz, W. A. Bridging the gap: membrane contact sites in signaling, metabolism, and organelle dynamics. J. Cell Biol. 205, 759–769 (2014).

    Article  CAS  Google Scholar 

  8. Giordano, F. et al. PI(4,5)P2-dependent and Ca(2+)-regulated ER-PM interactions mediated by the extended synaptotagmins. Cell 153, 1494–1509 (2013).

    Article  CAS  Google Scholar 

  9. Toulmay, A. & Prinz, W. A. A conserved membrane-binding domain targets proteins to organelle contact sites. J. Cell Sci. 125, 49–58 (2012).

    Article  CAS  Google Scholar 

  10. Manford, A. G., Stefan, C. J., Yuan, H. L., Macgurn, J. A. & Emr, S. D. ER-to-plasma membrane tethering proteins regulate cell signaling and ER morphology. Dev. Cell 23, 1129–1140 (2012).

    Article  CAS  Google Scholar 

  11. Fernandez-Busnadiego, R., Saheki, Y. & De Camilli, P. Three-dimensional architecture of extended synaptotagmin-mediated endoplasmic reticulum-plasma membrane contact sites. Proc. Natl Acad. Sci. USA 112, E2004–E2013 (2015).

    Article  CAS  Google Scholar 

  12. Idevall-Hagren, O., Lu, A., Xie, B. & De Camilli, P. Triggered Ca2+ influx is required for extended synaptotagmin 1-induced ER-plasma membrane tethering. EMBO J. 34, 2291–2305 (2015).

    Article  CAS  Google Scholar 

  13. Min, S. W., Chang, W. P. & Sudhof, T. C. E-Syts, a family of membranous Ca2+-sensor proteins with multiple C2 domains. Proc. Natl Acad. Sci. USA 104, 3823–3828 (2007).

    Article  CAS  Google Scholar 

  14. Craxton, M. Genomic analysis of synaptotagmin genes. Genomics 77, 43–49 (2001).

    Article  CAS  Google Scholar 

  15. Craxton, M. Evolutionary genomics of plant genes encoding N-terminal-TM-C2 domain proteins and the similar FAM62 genes and synaptotagmin genes of metazoans. BMC Genomics 8, 259 (2007).

    Article  Google Scholar 

  16. Levy, A., Zheng, J. Y. & Lazarowitz, S. G. Synaptotagmin SYTA forms ER-plasma membrane junctions that are recruited to plasmodesmata for plant virus movement. Curr. Biol. 25, 2018–2025 (2015).

    Article  CAS  Google Scholar 

  17. Perez-Sancho, J. et al. The Arabidopsis synaptotagmin1 is enriched in endoplasmic reticulum-plasma membrane contact sites and confers cellular resistance to mechanical stresses. Plant Physiol. 168, 132–143 (2015).

    Article  CAS  Google Scholar 

  18. Kopec, K. O., Alva, V. & Lupas, A. N. Homology of SMP domains to the TULIP superfamily of lipid-binding proteins provides a structural basis for lipid exchange between ER and mitochondria. Bioinformatics 26, 1927–1931 (2010).

    Article  CAS  Google Scholar 

  19. Kopec, K. O., Alva, V. & Lupas, A. N. Bioinformatics of the TULIP domain superfamily. Biochem. Soc. Trans. 39, 1033–1038 (2011).

    Article  CAS  Google Scholar 

  20. Qiu, X. et al. Crystal structure of cholesteryl ester transfer protein reveals a long tunnel and four bound lipid molecules. Nat. Struct. Mol. Biol. 14, 106–113 (2007).

    Article  Google Scholar 

  21. Oram, J. F., Wolfbauer, G., Vaughan, A. M., Tang, C. & Albers, J. J. Phospholipid transfer protein interacts with and stabilizes ATP-binding cassette transporter A1 and enhances cholesterol efflux from cells. J. Biol. Chem. 278, 52379–52385 (2003).

    Article  CAS  Google Scholar 

  22. Schauder, C. M. et al. Structure of a lipid-bound extended synaptotagmin indicates a role in lipid transfer. Nature 510, 552–555 (2014).

    Article  CAS  Google Scholar 

  23. Kornmann, B. et al. An ER-mitochondria tethering complex revealed by a synthetic biology screen. Science 325, 477–481 (2009).

    Article  CAS  Google Scholar 

  24. AhYoung, A. P. et al. Conserved SMP domains of the ERMES complex bind phospholipids and mediate tether assembly. Proc. Natl Acad. Sci. USA 112, E3179–E3188 (2015).

    Article  CAS  Google Scholar 

  25. Morgan, A. J. & Jacob, R. Ionomycin enhances Ca2+ influx by stimulating store-regulated cation entry and not by a direct action at the plasma membrane. Biochem. J. 300, 665–672 (1994).

    Article  CAS  Google Scholar 

  26. Willars, G. B., Nahorski, S. R. & Challiss, R. A. Differential regulation of muscarinic acetylcholine receptor-sensitive polyphosphoinositide pools and consequences for signaling in human neuroblastoma cells. J. Biol. Chem. 273, 5037–5046 (1998).

    Article  CAS  Google Scholar 

  27. Horowitz, L. F. et al. Phospholipase C in living cells: activation, inhibition, Ca2+ requirement, and regulation of M current. J. Gen. Physiol. 126, 243–262 (2005).

    Article  CAS  Google Scholar 

  28. Suh, B. C., Horowitz, L. F., Hirdes, W., Mackie, K. & Hille, B. Regulation of KCNQ2/KCNQ3 current by G protein cycling: the kinetics of receptor-mediated signaling by Gq. J. Gen. Physiol. 123, 663–683 (2004).

    Article  CAS  Google Scholar 

  29. Cohen, C. M., Kalish, D. I., Jacobson, B. S. & Branton, D. Membrane isolation on polylysine-coated beads. Plasma membrane from HeLa cells. J. Cell Biol. 75, 119–134 (1977).

    Article  CAS  Google Scholar 

  30. Codazzi, F., Teruel, M. N. & Meyer, T. Control of astrocyte Ca(2+) oscillations and waves by oscillating translocation and activation of protein kinase C. Curr. Biol. 11, 1089–1097 (2001).

    Article  CAS  Google Scholar 

  31. Varnai, P. & Balla, T. Visualization of phosphoinositides that bind pleckstrin homology domains: calcium- and agonist-induced dynamic changes and relationship to myo-[3H]inositol-labeled phosphoinositide pools. J. Cell Biol. 143, 501–510 (1998).

    Article  CAS  Google Scholar 

  32. Gallegos, L. L., Kunkel, M. T. & Newton, A. C. Targeting protein kinase C activity reporter to discrete intracellular regions reveals spatiotemporal differences in agonist-dependent signaling. J. Biol. Chem. 281, 30947–30956 (2006).

    Article  CAS  Google Scholar 

  33. Merida, I., Avila-Flores, A. & Merino, E. Diacylglycerol kinases: at the hub of cell signalling. Biochem. J. 409, 1–18 (2008).

    Article  CAS  Google Scholar 

  34. Kim, Y. J., Guzman-Hernandez, M. L., Wisniewski, E. & Balla, T. Phosphatidylinositol-Phosphatidic Acid Exchange by Nir2 at ER-PM contact sites maintains phosphoinositide signaling competence. Dev. Cell 33, 549–561 (2015).

    Article  CAS  Google Scholar 

  35. Whatmore, J., Wiedemann, C., Somerharju, P., Swigart, P. & Cockcroft, S. Resynthesis of phosphatidylinositol in permeabilized neutrophils following phospholipase Cbeta activation: transport of the intermediate, phosphatidic acid, from the plasma membrane to the endoplasmic reticulum for phosphatidylinositol resynthesis is not dependent on soluble lipid carriers or vesicular transport. Biochem. J. 341, 435–444 (1999).

    Article  CAS  Google Scholar 

  36. Chang, C. L. et al. Feedback regulation of receptor-induced Ca2+ signaling mediated by E-Syt1 and Nir2 at endoplasmic reticulum-plasma membrane junctions. Cell Rep. 5, 813–825 (2013).

    Article  Google Scholar 

  37. Chang, C. L. & Liou, J. Phosphatidylinositol 4,5-bisphosphate homeostasis regulated by Nir2 and Nir3 proteins at endoplasmic reticulum-plasma membrane junctions. J. Biol. Chem. 290, 14289–14301 (2015).

    Article  CAS  Google Scholar 

  38. Schapire, A. L. et al. Arabidopsis synaptotagmin 1 is required for the maintenance of plasma membrane integrity and cell viability. Plant Cell 20, 3374–3388 (2008).

    Article  CAS  Google Scholar 

  39. Yamazaki, T., Kawamura, Y., Minami, A. & Uemura, M. Calcium-dependent freezing tolerance in Arabidopsis involves membrane resealing via synaptotagmin SYT1. Plant Cell 20, 3389–3404 (2008).

    Article  CAS  Google Scholar 

  40. Aguilar, P. S., Engel, A. & Walter, P. The plasma membrane proteins Prm1 and Fig1 ascertain fidelity of membrane fusion during yeast mating. Mol. Biol. Cell 18, 547–556 (2007).

    Article  CAS  Google Scholar 

  41. Idevall-Hagren, O., Dickson, E. J., Hille, B., Toomre, D. K. & De Camilli, P. Optogenetic control of phosphoinositide metabolism. Proc. Natl Acad. Sci. USA 109, E2316–E2323 (2012).

    Article  CAS  Google Scholar 

  42. Shibata, Y. et al. The reticulon and DP1/Yop1p proteins form immobile oligomers in the tubular endoplasmic reticulum. J. Biol. Chem. 283, 18892–18904 (2008).

    Article  CAS  Google Scholar 

  43. Sampaio, J. L. et al. Membrane lipidome of an epithelial cell line. Proc. Natl Acad. Sci. USA 108, 1903–1907 (2011).

    Article  CAS  Google Scholar 

  44. Herzog, R. et al. A novel informatics concept for high-throughput shotgun lipidomics based on the molecular fragmentation query language. Genome Biol. 12, R8 (2011).

    Article  CAS  Google Scholar 

  45. Kim, H. et al. Surrogate reporters for enrichment of cells with nuclease-induced mutations. Nat. Methods 8, 941–943 (2011).

    Article  CAS  Google Scholar 

  46. Sanjana, N. E. et al. A transcription activator-like effector toolbox for genome engineering. Nat. Protoc. 7, 171–192 (2012).

    Article  CAS  Google Scholar 

  47. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank H. Shen, S. Ferguson, S. Tomita, R. Dong, W. Hancock-Cerutti, J. Lees and Y. Cai for discussion and/or sharing reagents. We thank F. Wilson, H. Czapla and L. Lucast for technical assistance. This work was supported in part by NIH grants R37NS036251, DK45735 and DA018343 to P.D.C. Y.Saheki was supported by fellowships from the Uehara Memorial Foundation and the Japan Society for the Promotion of Science, and X.B. by a Human Frontier Science Program long-term fellowship.

Author information

Authors and Affiliations

Authors

Contributions

All authors participated in the design of experiments, data analysis and interpretation. Y.Saheki designed and performed all of the genetic manipulations, all of the imaging and biochemical studies and the isolation of plasma membrane sheets. X.B., F.P. and K.M.R. participated in the design of lipid transfer assays that were performed by X.B. C.M.S. and K.M.R. designed SMP domain mutations and performed lipid-binding assays. M.A.S and C.K. performed the lipidomic analysis. Y.Saheki and P.D.C. wrote the manuscript, which was then reviewed by all authors.

Corresponding author

Correspondence to Pietro De Camilli.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 4 Localization of E-Syt1.

(a) Lysates of control and E-Syt1/2 DKO HeLa cells were processed by SDS-PAGE and immunoblotting with the original YU1231 serum (left) or affinity-purified antibodies from YU1231 serum (right). The affinity-purified antibodies were further incubated with aldehyde fixed E-Syt1/2 DKO lysates and used for immunohistochemistry. (b) Confocal images of E-Syt1/2 DKO HeLa cells expressing mRFP-Sec61β. Endogenous localization of E-Syt1 was detected by the affinity-purified antibodies after incubation with aldehyde fixed E-Syt1/2 DKO lysates (see Methods). Note the absence of the E-Syt1 fluorescence signals. Scale bars, 10 μm. (c) TIRF microscopy images of a HeLa cell expressing transfected EGFP-E-Syt1 before and after stimulation with ionomycin (2 μM) in the presence of extracellular Ca2+ at the indicated time. (d) Time-course of ionomycin-induced recruitment of EGFP-E-Syt1 to the PM, as shown in c (mean ± s.e.m., n = 11 cells pooled from 2 independent experiments). Scale bars, 10 μm. Unprocessed original scans of blots are shown in Supplementary Fig. 7.

Supplementary Figure 5 Characterization of the lipid transfer assay.

(a) The self-quenching property of fluorescent lipids was plotted as the ratio of the quenched fluorescence signals ‘F0’ over the fully dequenched fluorescence signal in the presence of detergent ‘F’ (mean ± s.e.m., n = 3 independent experiments) (b) Time-course of normalized fluorescence signals, as assessed by fluorometry, for the mixture of acceptor liposomes with or without PI(4,5)P2 and donor liposomes containing 1% NBD-PE during an incubation with E-Syt1cyto in the presence of 100 μM Ca2+. Note the much slower dequenching of NBD-PE in the absence of PI(4,5)P2 in the acceptor liposomes. (c) Time-course of normalized fluorescence signals, as assessed by fluorometry, for the mixture of liposomes containing 1% NBD-PE either in ER-like or in PM-like liposomes incubated with E-Syt1cyto in the presence of 100 μM Ca2+. Note the comparable dequenching of NBD-PE in the two conditions. (d) (left) Content mixing assay. Time-course of normalized fluorescence signals, as assessed by fluorometer, from the mixture of acceptor and donor liposomes containing self-quenched sulforhodamine B (see Methods), and either E-Syt1cyto or E-Syt1cytoΔSMP, incubated with indicated concentration of Ca2+ in the assay buffer. Note the minimal dequenching of sulforhodamine B that depend neither on the presence nor absence of Ca2+. (right) Lipid transfer assay. Time-course of normalized fluorescence signals, as assessed by fluorometer, from the mixture of acceptor liposomes and donor liposomes containing 1% NBD-PE, and either E-Syt1cyto or E-Syt1cytoΔSMP, incubated with indicated concentration of Ca2+ in the assay buffer. Note the Ca2+- and SMP domain-dependent dequenching of NBD-PE fluorescence signals overtime. (e) Time-course of normalized fluorescence signal, as assessed by fluorometry, for the mixture of acceptor PM-like liposomes and donor ER-like liposomes containing 1% NBD-PE at the indicated ratios and incubated with E-Syt1cyto in the presence of 100 μM Ca2+. (f) Time-course of normalized fluorescence signals, as assessed by fluorometry, from the mixture of acceptor PM-like liposomes and donor ER-like liposomes. Addition of non-labeled PE to acceptor liposomes did not affect the transfer of NBD-PE. For all the liposome-based assays except a, data are from one experiment; the experiments were repeated three times with similar results.

Supplementary Figure 6 Enrichment and characterization of genome-edited E-Syt knockout (KO) cells.

(a) Surveyor nuclease assay for TALEN- and Cas9-mediated cleavage at E-Syt2(left), E-Syt1(middle) and E-Syt3(right) loci in HeLa cells. (b) (top left) Schematics showing the design of the surrogate vector for E-Syt2 KO. (top right) Confocal images of cells expressing the indicated TALEN constructs together with the mCherry-E-Syt2_SMP-EGFP surrogate vector. Scale bars, 100 μm. (bottom left) Schematics showing the design of the surrogate vector for E-Syt1 KO. Protospacer-adjacent motif (PAM) sequences are indicated by yellow boxes; yellow allowheads indicate predicted cleavage sites. (bottom right) Confocal images of cells expressing hCas9 and the indicated guide RNA expression vectors together with the mCherry-E-Syt1(6,7)-EGFP surrogate vector. Scale bars, 10 μm. (c) Surveyor nuclease assay of Cas9-mediated cleavage of E-Syt1 locus. Note approximately 2-fold increase in the cleavage efficiency after FACS. (d) Detection of TALEN-mediated cleavage of E-Syt2 gene by PCR. Asterisks denote clonal cell lines with size changes of the PCR products. (e) Lysates of control HeLa cells (WT) and 8 candidate E-Syt2 KO cell lines were processed by SDS-PAGE and immunoblotting (IB) with indicated antibodies. Arrow indicates endogenous E-Syt2. (f) (top) Nucleotide sequence analysis of the E-Syt2 gene; TALEN-binding sites are highlighted in red, and bold and blue letters indicate TALEN-binding sites and additional nucleotide insertions, respectively. (bottom) Lysates of control HeLa cells (WT) and E-Syt2 KO cell lines transfected with EGFP-E-Syt1 were processed for immunoprecipitation with anti-GFP antibodies. Total lysates (Input) as well as anti-GFP immunoprecipitates were processed by SDS-PAGE and immunoblotted with indicated antibodies. Arrow indicates endogenous E-Syt2. (gh) KO of E-Syt1 and E-Syt3. (g, top) Sequencing analyses of the E-Syt1 gene of two E-Syt1/2 DKO cell lines show one nucleotide insertions. (g, bottom) Lysates of control HeLa cells (WT), E-Syt2 KO cell line and the same cell line treated with guide RNA expressing vectors were processed by SDS-PAGE and immunoblotted with indicated antibodies. (h) Sequencing analysis of the E-Syt3 gene of the E-Syt TKO cell line. Guide RNA-targeting sites and PAM sequences are highlighted in red and green. Unprocessed original scans of blots are shown in Supplementary Fig. 7.

Supplementary Figure 7 PI(4,5)P2 dynamics on muscarinic receptor stimulation is not affected in HeLa cells lacking E-Syts.

(a) (left) Time course of normalized mRFP fluorescence, as assessed by TIRF microscopy, in response to Oxo-M (10 μM) and atropine (50 μM), from WT control HeLa cells, E-Syt1/2 DKO HeLa cells and E-Syt TKO HeLa cells expressing mRFP-PH-PLCδ and M1R. (right) Values of ΔF/F0 corresponding to the indicated time point by arrow. [mean ± s.e.m., n = 11 cells (Ctrl), n = 13 cells (DKO#6-8), n = 12 cells (TKO#5); data are pooled from 2 independent experiments for each condition] Bonferroni’s multiple comparisons test, n.s. = not significant (P > 0.05). (b) Time-course of normalized EGFP, iRFP (left axis) and mCherry (right axis) fluorescence signals in response to Histamine (1 mM), as assessed by TIRF microscopy, from cells expressing endogenously-tagged E-Syt1 as well as iRFP-PH-PLCδ and C1-mCherry. (mean ± s.e.m., n = 13 cells from 3 independent dishes) (c) Time-course of normalized EGFP, iRFP (left axis) and mCherry (right axis) fluorescence signals in response to Oxo-M (10 μM) and atropine (50 μM), as assessed by TIRF microscopy, from cells expressing endogenously-tagged E-Syt1 as well as M1R, iRFP-PH-PLCδ and C1-mCherry. (mean ± s.e.m., n = 25 cells pooled from 4 independent experiments).

Supplementary Figure 8 Enhanced and prolonged increase of DAG in the PM of E-Syt KO cells in response to ionomycin.

(a) (left) Time-course of normalized YFP fluorescence, as assessed by TIRF microscopy, from WT control and E-Syt TKO cells expressing YFP-DBD (a DAG probe), in response to ionomycin (2 μM). (right) Values of ΔF/F0 corresponding to the end of the experiment as indicated by an arrow [mean ± s.e.m., n = 27 cells pooled from 6 independent experiments (Ctrl), n = 19 cells pooled from 3 independent experiments (TKO#5); two-tailed Student’s t-test with unequal variance, P < 0.0001].

Supplementary Figure 9 Dynamics of DAG in E-Syt KO cells.

(a) Time course of normalized EGFP fluorescence, as assessed by TIRF microscopy, in response to Oxo-M (10 μM) and atropine (50 μM), from WT control HeLa cells, E-Syt1/2 DKO HeLa cells and E-Syt TKO HeLa cells expressing C1-EGFP and M1R. [mean ± s.e.m., n = 11 cells (Ctrl), n = 13 cells (DKO#6-8), n = 12 cells (TKO#5); data are pooled from 2 independent experiments for each condition] (b) (left) Time-course of normalized EGFP, iRFP (left axis) and mCherry (right axis) fluorescence signals in response to Oxo-M (10 μM), atropine plus DGKi (50 μM each) and ionomycin (2 μM) in the absence of extracellular Ca2+, as assessed by TIRF microscopy, from cells expressing endogenously-tagged E-Syt1 as well as M1R, iRFP-PH-PLCδ and C1-mCherry. (right) Values of ΔF/F0 corresponding to the end of the experiment as shown here and in Fig. 7d with red arrows [mean ± s.e.m., n = 20 cells (ionomycin with Ca2+), n = 20 cells (ionomycin without Ca2+); data are pooled from 3 independent experiments for each condition; two-tailed Student’s t-test with unequal variance, denotes P = 0.0001]. (c) (left) Time-course of normalized mCherry signal, in response to Oxo-M (10 μM), atropine plus DGKi (50 μM each) and ionomycin (2 μM), as assessed by TIRF microscopy, from control (Ctrl) and E-Syt TKO cells expressing C1-mCherry. Re-expression of EGFP-E-Syt1 in E-Syt TKO cells rescued the accumulation of DAG, as assessed by C1-mCherry, while EGFP-E-Syt1 Cyto that lacks the ER anchor did not. (right) Values of F/F0 corresponding to the end of the experiment as shown with an arrow [mean ± s.e.m., n = 5 cells (Ctrl), n = 7 cells (TKO#5), n = 11 cells (TKO#5 + EGFP-E-Syt1), n = 13 cells (TKO#5 + EGFP-E-Syt1 Cyto)]; data are pooled from 2 independent experiments for each condition; Bonferroni’s multiple comparisons test, denotes P < 0.0001]. NS, not significant (P > 0.05).

Supplementary Figure 10 Unprocessed original scans of blots used for the figures and Supplementary Figures.

Supplementary Table 1 Primary and secondary antibodies used in this study.
Supplementary Table 2 Sequences of primers and oligos used in this study.
Supplementary Table 3 Moles percent of lipids used for the acceptor and donor liposomes in FRET-based lipid transfer experiments.

Supplementary information

Supplementary Information

Supplementary Information (PDF 640 kb)

41556_2016_BFncb3339_MOESM17_ESM.avi

Rapid translocation of endogenously tagged E-Syt1 to the cortical regions of a cell, as monitored by spinning disc confocal microscopy, in response to stimulation with ionomycin (2 μM). (AVI 1043 kb)

41556_2016_BFncb3339_MOESM18_ESM.avi

Rapid translocation of endogenously tagged E-Syt1 to the cortical regions of a cell, as monitored by spinning disc confocal microscopy, in response to thapsigargin (2 μM). (AVI 1340 kb)

Simultaneous TIRF imaging of Nir2-mCherry and endogenously-tagged E-Syt1 (endoEGFP-E-Syt1) in response to the sequential application of Oxo-M (10 μM), atropine plus DGKi (50 μM each) (ATR&DGKi) and ionomycin (2 μM) (Ion.) as indicated.

The increase of fluorescence in the TIRF field is both diffuse and spotted for Nir2 (as this protein is both soluble and partially anchored in the ER via its interaction with VAP36), while it is only spotted for E-Syt1, which is an intrinsic protein of the ER. Dark spots represent ER-PM contacts. (AVI 2390 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Saheki, Y., Bian, X., Schauder, C. et al. Control of plasma membrane lipid homeostasis by the extended synaptotagmins. Nat Cell Biol 18, 504–515 (2016). https://doi.org/10.1038/ncb3339

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3339

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