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:

Elongator stabilizes microtubules to control central spindle asymmetry and polarized trafficking of cell fate determinants

An Author Correction to this article was published on 16 January 2023

This article has been updated

Abstract

Asymmetric cell division gives rise to two daughter cells that inherit different determinants, thereby acquiring different fates. Polarized trafficking of endosomes containing fate determinants recently emerged as an evolutionarily conserved feature of asymmetric cell division to enhance the robustness of asymmetric cell fate determination in flies, fish and mammals. In particular, polarized sorting of signalling endosomes by an asymmetric central spindle contributes to asymmetric cell division in Drosophila melanogaster. However, how central spindle asymmetry arises remains elusive. Here we identify a moonlighting function of the Elongator complex—an established protein acetylase and tRNA methylase involved in the fidelity of protein translation—as a key factor for central spindle asymmetry. Elongator controls spindle asymmetry by stabilizing microtubules differentially on the anterior side of the central spindle. Accordingly, lowering the activity of Elongator on the anterior side using nanobodies mistargets endosomes to the wrong cell. Molecularly, Elongator regulates microtubule dynamics independently of its acetylation and methylation enzymatic activities. Instead, Elongator directly binds to microtubules and increases their polymerization speed while decreasing their catastrophe frequency. Our data establish a non-canonical role of Elongator at the core of cytoskeleton polarity and asymmetric signalling.

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: Elongator controls central spindle asymmetry.
Fig. 2: Elongator controls asymmetric Sara endosome segregation and thereby contributes to Notch signalling.
Fig. 3: Elongator is recruited to the central spindle via its direct interaction with microtubules.
Fig. 4: The enzymatic activity of Elp3 is not required for the effects of Elongator on the central spindle.
Fig. 5: Elongator directly controls microtubule dynamics.
Fig. 6: Asymmetric control of microtubule dynamics by Elongator governs the asymmetric trafficking of Sara endosomes.

Similar content being viewed by others

Data availability

The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE72 partner repository with the dataset identifier PXD036556. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding authors on reasonable request.

Code availability

Custom image processing code specific to this paper have been deposited on our GitHub page in a dedicated repository (https://github.com/deriverylab/2022_Planelles-Herrero). Similarly, our general image registration and wavelet filtering codes can also be found on our GitHub page (https://github.com/deriverylab/GPU_registration and https://github.com/deriverylab/GPU_wavelet_a_trous). Safir denoising code can be found on the Inria website (https://team.inria.fr/serpico/software/nd-safir). All lookup tables applied to images in this paper come from the collection from J. Manton (https://github.com/jdmanton/ImageJ_LUTs).

Change history

References

  1. Kressmann, S. et al. Directional Notch trafficking in Sara endosomes during asymmetric cell division in the spinal cord. Nat. Cell Biol. 17, 333–339 (2015).

    Article  CAS  Google Scholar 

  2. Coumailleau, F., Furthauer, M., Knoblich, J. A. & Gonzalez-Gaitan, M. Directional Delta and Notch trafficking in Sara endosomes during asymmetric cell division. Nature 458, 1051–1055 (2009).

    Article  CAS  Google Scholar 

  3. Montagne, C. & Gonzalez-Gaitan, M. Sara endosomes and the asymmetric division of intestinal stem cells. Development 141, 2014–2023 (2014).

    Article  CAS  Google Scholar 

  4. Derivery, E. et al. Polarized endosome dynamics by spindle asymmetry during asymmetric cell division. Nature 528, 280–285 (2015).

    Article  CAS  Google Scholar 

  5. Kawaguchi, D., Furutachi, S., Kawai, H., Hozumi, K. & Gotoh, Y. Dll1 maintains quiescence of adult neural stem cells and segregates asymmetrically during mitosis. Nat. Commun. https://doi.org/10.1038/ncomms2895 (2013).

  6. Loeffler, D. et al. Asymmetric lysosome inheritance predicts activation of haematopoietic stem cells. Nature https://doi.org/10.1038/s41586-019-1531-6 (2019).

  7. Zhao, X. et al. Polarized endosome dynamics engage cytoplasmic Par-3 that recruits dynein during asymmetric cell division. Sci. Adv. 7, (2021).

  8. Gho, M., Bellaiche, Y. & Schweisguth, F. Revisiting the Drosophila microchaete lineage: a novel intrinsically asymmetric cell division generates a glial cell. Development 126, 3573–3584 (1999).

    Article  CAS  Google Scholar 

  9. Furthauer, M. & Gonzalez-Gaitan, M. Endocytic regulation of notch signalling during development. Traffic 10, 792–802 (2009).

    Article  CAS  Google Scholar 

  10. Loubéry, S. et al. Sara phosphorylation state controls the dispatch of endosomes from the central spindle during asymmetric division. Nat. Commun. https://doi.org/10.1038/ncomms15285 (2017).

  11. Loubéry, S. et al. Uninflatable and Notch control the targeting of Sara endosomes during asymmetric division. Curr. Biol. 24, 2142–2148 (2014).

  12. Glotzer, M. The 3Ms of central spindle assembly: microtubules, motors and MAPs. Nat. Rev. Mol. Cell Biol. 10, 9–20 (2009).

    Article  CAS  Google Scholar 

  13. Buffin, E. & Gho, M. Laser microdissection of sensory organ precursor cells of Drosophila microchaetes. PLoS ONE https://doi.org/10.1371/journal.pone.0009285 (2010).

  14. Krogan, N. J. & Greenblatt, J. F. Characterization of a six-subunit holo-elongator complex required for the regulated expression of a group of genes in Saccharomyces cerevisiae. Mol. Cell. Biol. 21, 8203–8212 (2001).

    Article  CAS  Google Scholar 

  15. Hawkes, N. A. et al. Purification and characterization of the human elongator complex. J. Biol. Chem. 277, 3047–3052 (2002).

    Article  CAS  Google Scholar 

  16. Dauden, M. I. et al. Architecture of the yeast Elongator complex. EMBO Rep. 18, 264–279 (2017).

    Article  CAS  Google Scholar 

  17. Setiaputra, D. T. et al. Molecular architecture of the yeast Elongator complex reveals an unexpected asymmetric subunit arrangement. EMBO Rep. 18, 280–291 (2017).

    Article  CAS  Google Scholar 

  18. Miśkiewicz, K. et al. ELP3 controls active zone morphology by acetylating the ELKS family member bruchpilot. Neuron https://doi.org/10.1016/j.neuron.2011.10.010 (2011).

  19. Creppe, C. et al. Elongator controls the migration and differentiation of cortical neurons through acetylation of α-tubulin. Cell 136, 551–564 (2009).

    Article  CAS  Google Scholar 

  20. Solinger, J. A. et al. The Caenorhabditis elegans elongator complex regulates neuronal α-tubulin acetylation. PLoS Genet. 6, e1000820 (2010).

    Article  Google Scholar 

  21. Bauer, F. et al. Translational control of cell division by Elongator. Cell Rep. 1, 424–433 (2012).

    Article  CAS  Google Scholar 

  22. Chen, C., Tuck, S. & Byström, A. S. Defects in tRNA modification associated with neurological and developmental dysfunctions in Caenorhabditis elegans Elongator mutants. PLoS Genet. 5, e1000561 (2009).

    Article  Google Scholar 

  23. Dauden, M. I. et al. Molecular basis of tRNA recognition by the Elongator complex. Sci. Adv. 5, eaaw2326 (2019).

    Article  CAS  Google Scholar 

  24. Slaugenhaupt, S. A. et al. Tissue-specific expression of a splicing mutation in the IKBKAP gene causes familial dysautonomia. Am. J. Hum. Genet. 68, 598–605 (2001).

    Article  CAS  Google Scholar 

  25. Rapino, F. et al. Codon-specific translation reprogramming promotes resistance to targeted therapy. Nature 558, 605–609 (2018).

    Article  CAS  Google Scholar 

  26. Bento-Abreu, A. et al. Elongator subunit 3 (ELP3) modifies ALS through tRNA modification. Hum. Mol. Genet. 27, 1276–1289 (2018).

    Article  CAS  Google Scholar 

  27. Derivery, E. & Gautreau, A. Generation of branched actin networks: assembly and regulation of the N-WASP and WAVE molecular machines. BioEssays 32, 119–131 (2010).

    Article  CAS  Google Scholar 

  28. Huang, B., Johansson, M. J. O. & Byström, A. S. An early step in wobble uridine tRNA modification requires the Elongator complex. RNA 11, 424–436 (2005).

    Article  CAS  Google Scholar 

  29. Nelissen, H. et al. The elongata mutants identify a functional Elongator complex in plants with a role in cell proliferation during organ growth. Proc. Natl Acad. Sci. USA 102, 7754–7759 (2005).

    Article  CAS  Google Scholar 

  30. Mehlgarten, C. et al. Elongator function in tRNA wobble uridine modification is conserved between yeast and plants. Mol. Microbiol. 76, 1082–1094 (2010).

    Article  CAS  Google Scholar 

  31. Le Borgne, R. & Schweisguth, F. Unequal segregation of Neuralized biases Notch activation during asymmetric cell division. Dev. Cell 5, 139–148 (2003).

    Article  Google Scholar 

  32. Kojic, M. et al. Elp2 mutations perturb the epitranscriptome and lead to a complex neurodevelopmental phenotype. Nat. Commun. 12, 2678 (2021).

    Article  CAS  Google Scholar 

  33. Chang, H. C. et al. Hsc70 is required for endocytosis and clathrin function in Drosophila. J. Cell Biol. 159, 477–487 (2002).

    Article  CAS  Google Scholar 

  34. Bronk, P. et al. Drosophila Hsc70-4 is critical for neurotransmitter exocytosis in vivo. Neuron 30, 475–488 (2001).

    Article  CAS  Google Scholar 

  35. Cheishvili, D. et al. IKAP/Elp1 involvement in cytoskeleton regulation and implication for familial dysautonomia. Hum. Mol. Genet. https://doi.org/10.1093/hmg/ddr036 (2011).

  36. Hunnicutt, B. J., Chaverra, M., George, L. & Lefcort, F. Ikap/elp1 is required in vivo for neurogenesis and neuronal survival, but not for neural crest migration. PLoS ONE 7, e32050 (2012).

  37. George, L. et al. Familial dysautonomia model reveals Ikbkap deletion causes apoptosis of Pax3+ progenitors and peripheral neurons. Proc. Natl Acad. Sci. USA 110, 18698–18703 (2013).

    Article  CAS  Google Scholar 

  38. Jackson, M. Z., Gruner, K. A., Qin, C. & Tourtellotte, W. G. A neuron autonomous role for the familial dysautonomia gene ELP1 in sympathetic and sensory target tissue innervation. Development 141, 2452–2461 (2014).

    Article  CAS  Google Scholar 

  39. Akella, J. S. et al. MEC-17 is an α-tubulin acetyltransferase. Nature 467, 218–222 (2010).

    Article  CAS  Google Scholar 

  40. Wittschieben, B., Fellows, J., Wendy, D., Stillman, D. J. & Svejstrup, J. Q. Overlapping roles for the histone acetyltransferase activities of SAGA and Elongator in vivo. EMBO J. 19, 3060–3068 (2000).

    Article  CAS  Google Scholar 

  41. Walker, J. et al. Role of elongator subunit Elp3 in Drosophila melanogaster larval development and immunity. Genetics 187, 1067–1075 (2011).

    Article  CAS  Google Scholar 

  42. Hawer, H. et al. Roles of Elongator dependent tRNA modification pathways in neurodegeneration and cancer. Genes 10, 2452–2461 (2019).

  43. Björk, G. R., Huang, B., Persson, O. P. & Byström, A. S. A conserved modified wobble nucleoside (mcm5s2U) in lysyl-tRNA is required for viability in yeast. RNA 13, 1245–1255 (2007).

    Article  Google Scholar 

  44. Gell, C. et al. Microtubule dynamics reconstituted in vitro and imaged by single-molecule fluorescence microscopy. Methods Cell. Biol. 95, 221–245 (2010).

    Article  CAS  Google Scholar 

  45. Hein, M. Y. et al. A human interactome in three quantitative dimensions organized by stoichiometries and abundances. Cell https://doi.org/10.1016/j.cell.2015.09.053 (2015).

  46. Holt, C. E., Martin, K. C. & Schuman, E. M. Local translation in neurons: visualization and function. Nat. Struct. Mol. Biol. 26, 557–566 (2019).

    Article  CAS  Google Scholar 

  47. Feng, C. et al. Patronin-mediated minus end growth is required for dendritic microtubule polarity. J. Cell Biol. 218, 2309–2328 (2019).

    Article  CAS  Google Scholar 

  48. Noordstra, I. et al. Control of apico-basal epithelial polarity by the microtubule minus-end-binding protein CAMSAP3 and spectraplakin ACF7. J. Cell Sci. 129, 4278–4288 (2016).

    CAS  Google Scholar 

  49. Abdel-Fattah, W. et al. Phosphorylation of Elp1 by Hrr25 is required for Elongator-dependent tRNA modification in yeast. PLoS Genet. 11, e1004931 (2015).

    Article  Google Scholar 

  50. Mehlgarten, C., Jablonowski, D., Breunig, K. D., Stark, M. J. R. & Schaffrath, R. Elongator function depends on antagonistic regulation by casein kinase Hrr25 and protein phosphatase Sit4. Mol. Microbiol. 73, 869–881 (2009).

    Article  CAS  Google Scholar 

  51. Yang, K., Inoue, A., Lee, Y., Jiang, C. & Lin, F. Loss of Ikbkap/Elp1 in mouse oocytes causes spindle disorganization, developmental defects in preimplantation embryos and impaired female fertility. Sci. Rep. 9, 18875 (2019).

  52. Karpova, N., Bobinnec, Y., Fouix, S., Huitorel, P. & Debec, A. Jupiter, a new Drosophila protein associated with microtubules. Cell Motil. Cytoskeleton 63, 301–312 (2006).

    Article  CAS  Google Scholar 

  53. Emery, G. et al. Asymmetric Rab 11 endosomes regulate delta recycling and specify cell fate in the Drosophila nervous system. Cell 122, 763–773 (2005).

    Article  CAS  Google Scholar 

  54. Bellaiche, Y., Gho, M., Kaltschmidt, J. A., Brand, A. H. & Schweisguth, F. Frizzled regulates localization of cell-fate determinants and mitotic spindle rotation during asymmetric cell division. Nat. Cell Biol. 3, 50–57 (2001).

    Article  CAS  Google Scholar 

  55. Mummery-Widmer, J. L. et al. Genome-wide analysis of Notch signalling in Drosophila by transgenic RNAi. Nature 458, 987–992 (2009).

    Article  CAS  Google Scholar 

  56. Gratz, S. J. et al. Genome engineering of Drosophila with the CRISPR RNA-guided Cas9 nuclease. Genetics 194, 1029–1035 (2013).

    Article  CAS  Google Scholar 

  57. Ponting, C. P. Novel domains and orthologues of eukaryotic transcription elongation factors. Nucleic Acids Res. https://doi.org/10.1093/nar/gkf498 (2002).

  58. Bolukbasi, E. et al. Drosophila poly suggests a novel role for the Elongator complex in insulin receptor-target of rapamycin signalling. Open Biol. https://doi.org/10.1098/rsob.110031 (2012).

  59. Goshima, G. & Vale, R. D. The roles of microtubule-based motor proteins in mitosis: comprehensive RNAi analysis in the Drosophila S2 cell line. J. Cell Biol. 162, 1003–1016 (2003).

    Article  CAS  Google Scholar 

  60. Watson, J. L. et al. High-efficacy subcellular micropatterning of proteins using fibrinogen anchors. J. Cell Biol. https://doi.org/10.1083/jcb.202009063 (2021).

  61. Campbell, J. N. & Slep, K. C. αβ-Tubulin and microtubule-binding assays. Methods Mol. Biol. https://doi.org/10.1007/978-1-61779-252-6_6 (2011).

  62. Lin, T. Y. et al. The Elongator subunit Elp3 is a non-canonical tRNA acetyltransferase. Nat. Commun. 2019 101 10, 625 (2019).

  63. Perkins, D. N., Pappin, D. J. C., Creasy, D. M. & Cottrell, J. S. Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis 20, 3551–3567 (1999).

  64. Keller, A., Nesvizhskii, A. I., Kolker, E. & Aebersold, R. Empirical statistical model to estimate the accuracy of peptide identifications made by MS/MS and database search. Anal. Chem. 74, 5383–5392 (2002).

    Article  CAS  Google Scholar 

  65. Loubery, S. & Gonzalez-Gaitan, M. Monitoring Notch/Delta endosomal trafficking and signaling in Drosophila. Methods Enzym. 534, 301–321 (2014).

    Article  CAS  Google Scholar 

  66. Bell, P. B. Jr. & Safiejko-Mroczka, B. Improved methods for preserving macromolecular structures and visualizing them by fluorescence and scanning electron microscopy. Scanning Microsc. 9, 843–860 (1995).

    CAS  Google Scholar 

  67. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  Google Scholar 

  68. Boulanger, J. et al. Patch-based nonlocal functional for denoising fluorescence microscopy image sequences. IEEE Trans. Med. Imaging https://doi.org/10.1109/TMI.2009.2033991 (2010).

  69. Zala, D. et al. Vesicular glycolysis provides on-board energy for fast axonal transport. Cell https://doi.org/10.1016/j.cell.2012.12.029 (2013).

  70. Holden, S. J. et al. Defining the limits of single-molecule FRET resolution in TIRF microscopy. Biophys. J. 99, 3102–3111 (2010).

    Article  CAS  Google Scholar 

  71. Gardner, M. K., Zanic, M., Gell, C., Bormuth, V. & Howard, J. Depolymerizing kinesins Kip3 and MCAK shape cellular microtubule architecture by differential control of catastrophe. Cell https://doi.org/10.1016/j.cell.2011.10.037 (2011).

  72. Perez-Riverol, Y. et al. The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res. 50, D543–D552 (2022).

    Article  CAS  Google Scholar 

  73. Banerjee, J. J. et al. Meru couples planar cell polarity with apical-basal polarity during asymmetric cell division. eLife 6, e25014 (2017).

Download references

Acknowledgements

This work was supported by the Medical Research Council, as part of United Kingdom Research and Innovation (UK Research and Innovation; grant no. MC_UP_1201/13 to E.D.), the Human Frontier Science Program (Career Development Award grant no. CDA00034/2017 to E.D.) as well as by grants from the SNSF, the ERC (Sara and Morphogen), the NCCR Chemical Biology program, the DIP of the Canton of Geneva and the SystemsX EpiPhysX (SNSF) granted to M.G.G. V.J.P.-H. is supported by an EMBO postdoctoral fellowship. We thank the electronics workshop of the LMB, in particular M. Kyte for his help with the Field Programmable Gate Array hardware required for precise synchronization of our microscope. We also thank the LMB Biophysics and Mass Spectrometry facilities, in particular M. Skehel and F. Begum for performing the mass spectrometry and analysing the mass spectrometry data. We thank I. Diaz (Venki Ramakrishnan laboratory) for sharing tRNA. We thank T. Stevens and J. O’Neill for expert advice on statistics, and F. Karch and P. Verstreken for flies. We thank S. Loubéry for their help with microscopy. We thank the members of the Derivery and Gonzalez-Gaitan laboratories, as well as C. Aumeier, B. Baum, S. Bullock and K. Kruse for critical reading of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

V.J.P.-H. conducted all biochemical experiments as well as all microtubule assays and associated analysis. E.D. and A.B. conducted most fly imaging experiments and associated quantification. A.D. performed additional Elp4 immunostaining and respective imaging. E.D. wrote the codes for automated image processing. C.S. generated the Klp98A–GFP knock-in and conducted genetic interaction experiments. V.J.P.-H., M.G.G. and E.D. designed the project. All authors contributed to writing the paper.

Corresponding authors

Correspondence to Marcos Gonzalez Gaitan or Emmanuel Derivery.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Sebastian Glatt, and the other, anonymous reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Conventions and rationale of the approach that identified Elongator.

(a) Left panel: conventions. We consider a reference frame centred on the central spindle with the y axis parallel to the division plane and the x axis orthogonal to it. The width is defined along the y axis, and the length along the x axis. During division, since the actin ring contracts, the width of the central spindle constricts over time. This constriction is stereotypic between cells and thus can be used to register movies in time a posteriori (methods). Right panel: Average temporal contraction profile of the central spindle length quantified from the Jupiter signal, mean ± SEM (shaded area). Registered time=0 indicates anaphase B onset. n: number of cells. (b) Candidate-based approach that identified Elongator as a key player of central spindle asymmetry. We took the list of all the genes identified by Buffin and Gho13 as being overexpressed in SOP cells, considering only genes with a enrichment of at least a factor two compared to neighbouring cells (green disk, n = 128 genes). This list was cross-referenced with the literature to find proteins within this list that were shown to regulate microtubule dynamics (in flies or other systems, red). We found only one gene matching both criteria, Elp3. Indeed, Elp3 is 2.26-fold overexpressed in SOPs, and Elongator has been suggested to play a role in microtubule dynamics in other systems/species14,15,16, albeit the molecular mechanisms involved was unclear. Note that there are numerous proteins in the Drosophila proteome that affect microtubule dynamics (for example Msps, CLASP, Patronin, Klp10A…), but these were not found specifically overexpressed in SOPs by Buffin and Gho and thus were not further tested here. Conversely, there are numerous proteins that were found specifically overexpressed in SOPs by Buffin and colleagues and that will have an indirect role onto central spindle asymmetry because they control the establishment of the polarity in these cells, which is upstream of central spindle asymmetry (for instance, meru73). But these proteins are not known to directly affect microtubule dynamics and thus were not further tested here. Source numerical data are available in source data.

Source data

Extended Data Fig. 2 Characterization of the antibodies, constructs and RNAi sequenced used in this paper.

(a-b) Characterization of the Elp3 antibody, the Elp3 RNAi sequence and the RNAi-resistant Elp3* construct used in this study. (a) S2 cells stably expressing His-PC-Elp3* (abbreviated HP-Elp3*), or wild-type control (S2) were treated with indicated RNAi before extracts and analysed by Western Blot (WB) against Elp3 (top panels) or acetylated tubulin (bottom panel). His-PC stands for the (His)6 and PC purification tags, respectively. Two different exposures of the western blot are shown to reflect the differences of loadings. Note the band disappearing in the Elp3 WB upon Elp3 RNAi treatment (orange arrowhead). In contrast, the band corresponding to His-PC-Elp3*, shifted upwards because of the PC tag, is not affected by the RNAi treatment. (b) RNAi-treated S2 extracts were subjected to immunoprecipitation using Elp2 antibodies. Note that the band disappearing upon RNAi Elp3 treatment (orange arrowhead) is pulled down by Elp2 antibodies. Altogether, these results demonstrate the specificity of the Elp3 antibody, the Elp3 RNAi sequence and the resistance of the Elp3* construct to the RNAi treatment. n.s.: non-specific band. (c-e) Characterization of the Elp2 (C), Elp4 (D) and Elp5(E) antibodies used in this study. S2 cells were treated with indicated RNAi before extracts were analysed by WB against Elp2 (top panel in c), or Elp4 (top panel in d). Ponceau staining is provided as a loading control (bottom panels in c and d). Note the major band disappearing in the Elp2 (respectively Elp4) WB upon Elp2 (respectively Elp4) RNAi treatment. Similarly, lysates from an identical number of S2 cells stably expressing His-PC-Elp3 and of wild-type control (S2) were processed for immunoprecipitation using anti-PC antibodies followed by Western Blot against PC or endogenous Elp5. Elp5 is specifically enriched upon immunoprecipitation of PC-Elp3, suggesting that it recognizes endogenous Elp5 in enriched fractions. Unprocessed blots are available in source data.

Source data

Extended Data Fig. 3 Several Elongator subunits are needed for its activity on central spindle asymmetry and Sara endosome segregation.

(a) Dividing SOP showing Jupiter–GFP (top row) or Jupiter–GFP with Rainbow RGB LUT (bottom row). Image corresponds to maximum intensity z-projection of the central spindle (SDCM). (b) Enrichment in pIIb. Thick line: median; thin line: quartile; n: number of central spindles analysed. Statistics: Kruskal–Wallis non-parametric ANOVA1 test. P-values of the respective post-hoc tests are indicated. Control and Elp3RNAi datasets are the same as those presented in Fig. 1d, shown here for convenience. (c-d) Elp3 Δ4/Δ4 MARCM clones. (c) Dividing SOP showing Sara endosomes (iDelta20 and mRFP-Pon in the indicated genotype (pIIa cortex; dashed blue). Time in seconds. Note that Sara endosomes are symmetrical in Elp3 mutant (arrowheads). The Elp3Δ4 mutant is a null allele of Elp3 that lacks most of the protein, including the SAM and KAT domains. (d) Quantification of the effects in (c): iDelta20 percentage in pIIa after abscission in SOPs of indicated genotypes (mean ± SEM; Statistics in were performed using a two-sided Welch test (p value indicated); n: SOPs analysed. (e) Dividing SOP showing Sara endosomes (iDelta20) and mRFP-Pon (pIIa cortex; dashed blue). Note the endosomes in the pIIb daughter cell upon Elp1, Elp2 and Elp3 depletion (arrows). (f) Percentage of iDelta20 in pIIa after abscission in SOPs (mean ± SEM; Statistics: one-way ANOVA (P < 0.0001), followed by a Tukey post-hoc test (p value indicated); n: SOPs analysed. The control and Elp3RNAi datasets are the same as those presented in Fig. 2b, shown here for convenience. As expected for stable multiprotein complexes, depletion of the Elongator subunits Elp1, 2 and 3 lead to the same phenotype. However, depletion of Hsc70-4 does not, suggesting that Elongator effect on microtubule dynamics does not require Hsc70-4. Scale bars: 2 µm (a); 5 µm (c,e). Images in this figure are Maximum intensity z-projection of entire cells (SDCM). Source numerical data are available in source data.

Source data

Extended Data Fig. 4 Elp3 depletion induces a short bristle phenotype.

(a) Scanning Electron Microscopy of the notum of representative control, Elp3 RNAi and Elp3 RNAi + Elp3* flies. Dashed lines indicate panier expression region. (b) Microchaete length in the panier region (Mean ± SEM). Statistics: Kruskal–Wallis test (p-value as indicated; n: microchaete number). Note the smaller bristles in the panier region in the Elp3 RNAi genotype. This is rescued by expression of a RNAi-resistant wild-type version of Elp3 (Elp3*). (c) Dividing SOP showing mRFP-Pon after immunolabelling with antibodies against endogenous Elp2 and α-tubulin. Images corresponds to single confocal plane (top panels) or maximum intensity z-projection of two planes (point scanning confocal microscopy). For mRFP-Pon: safir denoising filter (see methods). Top panel displays cell in metaphase and bottom panel a cell in early anaphase. Note that the cell on the bottom panel is at a too early stage of anaphase to display central spindle asymmetry, which gradually builds up during anaphase B (see Fig. 1a). (d,e) Fly nota were stained for tubulin and endogenous Elp2 (d) or Elp4 (e) to image epithelial cells (that is, non-SOP cells). Images corresponds to a maximum intensity z-projection (point scanning confocal microscopy). Both Elp2 and Elp4 antibodies label the mitotic spindle (orange arrow) and central spindle (blue arrow) in epithelial cells. Scale bar: 100 µm (a) and 5 µm (c-e). Source numerical data are available in source data.

Source data

Extended Data Fig. 5 Purification and characterization of the Drosophila Elongator complex.

(a) Stable S2 cell lines expressing His-PC-SNAP tagged version of each of the six Elongator subunits (or a His-PC-SNAP empty vector) were established. The endogenous complex assembling around tagged subunits was then analysed by quantitative Mass Spectrometry. Purification via the Elp3 or Elp4 subunit are the most efficient. (b) Left panel: western blot evaluation of an analytical sucrose gradient (10-30%) after a Protein C affinity step. The full complex migrates as a single unit in the 20% sucrose fractions. Right panel: coomassie-stained gradient SDS–PAGE gel of Elongator complex in the indicated fraction (fraction kept for in vitro analyses). (c) Left panel: chromatogram of the size exclusion (superose 6 increase) step of the purification of His-PC-tagged Elp3 Elongator complex. Right panel: coomassie-stained gradient SDS–PAGE analysis of the main peak of the size exclusion column (fraction used in in vitro analyses). The right panel is identical to Fig. 3d, shown here for convenience. (d) Identification of the subunits of the Elongator complex purified from His-PC-Elp3 by MS. This experiment also demonstrates that the Drosophila homologues of Elp5 and Elp6 are indeed anon-i1 and poly, respectively, previously predicted based on sequence analysis57,58. (e) Coomassie-stained gradient SDS–PAGE gel of Elongator complex purified using a His-PC-SNAP-tagged Elp3 subunit WT or Y531A starting from the same amounts of cells. The Y531A mutation does not affect the assembly of the complex. (f,g) Biochemical characterization of the Drosophila Elongator complex purified in b. (f) Affinity of the direct interaction between Elongator and tRNALys measured by MST (see methods). (g) tRNA modification activity of Elongator (mean ± SD, see methods). Drosophila Elongator behaves similarly to yeast Elongator. n = 3 independent experiments. Statistics: one-way ANOVA followed by a Tukey multiple comparison test (p value indicated). Source numerical data and unprocessed blots are available in source data.

Source data

Extended Data Fig. 6 Elongator subcomplexes do not bind stabilized microtubules.

(a) Coomassie-stained SDS–PAGE gradient gel of a partially purified Elp23 subcomplex (note the absence of Elp1), which copurifies with Hsc70-4. This is a by-product of the sucrose-gradient purification of the Full Elongator complex in the His-PC-SNAP–Elp3 stable cell line (Extended Data Fig. 5b). PC: protein C tag. (b-c) The affinity of the Elp23 + Hsc70-4 subcomplex for microtubules is less than that than of the full complex. (b) The subcomplex shown in (a) was fluorescently labelled with Alexa 488-BG, and the resulting fluorescent complex was incubated with Rhodamine-labelled, GMPCPP-stabilized microtubules (12.5 µM tubulin, 100 nM complex) and imaged by TIRFM. Alternatively, binding to microtubules was assessed by microtubule pelleting assays as in Fig. 3e and compared to the full complex (c). (d) Coomassie-stained SDS–PAGE gradient gel of a purified Elp456 subcomplex tagged with mScarlet on Elp4 and produced in E. coli. (e-f) The Elp456 subcomplex does not bind to microtubules. (e) Microtubule binding of the subcomplex shown in (d) was assessed as in (c) using 1 µM mScarlet–Elp456 subcomplex. The Elp456 subcomplex does not bind quantitatively to microtubules. Note that our purification of the mScarlet–Elp456 subcomplex contains some residual uncleaved GST-mScarlet-Elp4, but that since no mScarlet signal is observed on microtubules, this implies that a potentially dimeric GST-tagged mScarlet–Elp456 also does not bind to microtubules. Alternatively, binding to microtubules was assessed by microtubule pelleting assays and analysed using coomassie staining (f). Altogether these results presented in this figure suggest that the quantitative binding to Microtubules shown in Figs. 3,5 requires the full Elongator complex. Note that this is confirmed by analysis of microtubule dynamics (Fig. 5h,i). Scale bars: 5 μm. Unprocessed blots are available in source data.

Source data

Extended Data Fig. 7 Perturbation of tubulin acetylation by AtatRNAi does not affect central spindle asymmetry and symmetric endosome segregation.

(a-b) Validation of the AtatRNAi construct. (a) Notum of a fly expressing AtatRNAi under the control of the Pnr promoter after immunolabeling using Oregon Green-514 anti-β-tubulin and Atto 647N anti-K40 acetylated-α-tubulin antibodies (SDCM). White line: Pnr expression domain limit. Image corresponds to maximum intensity z-projection of epithelium by SDCM. (b) Quantification of the normalized K40 acetyl-α-tubulin/β-tubulin ratio at the central spindle in conditions shown in a (see methods). Mean±SEM. Statistics: two-sided Mann–Whitney test (p-value<0.0001; n: central spindle number). AtatRNAi leads to a marked decrease of K40-α-tubulin acetylation. (c) Dividing SOP of indicated genotype showing Jupiter–GFP (top row) or Jupiter–GFP with Rainbow RGB LUT applied (bottom row). Anterior/pIIb orientation as determined by the mRFP-Pon signal. Image corresponds to maximum intensity z-projection of the central spindle (SDCM). (d) Quantification of the effects seen in c: pIIb enrichment (see methods) in indicated genotypes. Median±95% confidence interval; n: number of central spindles analysed. Statistics: two-sided Student t-test. The control dataset is the same as the one presented in Fig. 1d, shown here for convenience. Central spindle asymmetry is not affected upon Atat depletion. (e) Dividing SOP showing Sara endosomes (iDelta20) and mRFP-Pon in the indicated genotype (pIIa cortex; dashed blue). Image corresponds to Maximum intensity z-projection of entire cells (SDCM). (f) Quantification of the effects seen in (e): iDelta20 percentage in pIIa after abscission in SOPs of indicated genotypes (mean ± SEM; Statistics: two-sided Student t-test; n: SOPs analysed. The control dataset is the same as the one presented in Fig. 2B, shown here for convenience. Sara endosome asymmetric segregation is not affected upon Atat depletion. (g) Summary of the AtatRNAi and Elp3RNAi phenotypes: Elp3RNAi phenotype cannot be due to a defect of α-tubulin acetylation. Scale bars: 10 µm (a); 2 µm (c); 5 µm (e). Source numerical data are available in source data.

Source data

Extended Data Fig. 8 The enzymatic activity of Elp3 are not required for Elongator effects on the central spindle.

(a) Average central spindle in SOPs after immunolabelling using anti-K40-Acetyl-α-Tubulin antibodies. n: number of spindles analysed. (b) K40-Acetyl-α-Tubulin intensity profile across the spindle (dashed orange in a). While in control SOPs the pIIb peak is ≈25% higher than that of the pIIa, this is abolished in Elp3RNAi mutants. (c) Enrichment in pIIb. Thick line: median; thin line: quartile. n: number of spindles analysed. Statistics: two-sided Mann–Whitney test (P-values indicated). The β-Tubulin datasets are reproduced from Fig. 1g. (d) Acetyl-tubulin/tubulin signal ratio at the central spindle in indicated genotypes (see methods). Mean±SEM. n: number of spindles. Statistics: two-sided, paired Student’s t-test between pIIa/pIIb values for each spindle. Elp3 depletion does not affect central spindle acetylation. (e) Left panel: S2 cells depleted for indicated Elongator complex subunits (or GFP control) were analysed by quantitative Western blot against β-tubulin and K40-Acetyl-α-tubulin. A 2-fold dilution series of the GFP-RNAi control was loaded to demonstrate linearity of the antibodies. Right panel: acetyl-tubulin/tubulin ratio in RNAi-depleted extracts. Depletion of Elongator subunits does not affect tubulin acetylation. (f) Dividing SOP showing Jupiter–GFP with (top row) or without the Rainbow RGB LUT (bottom row). Maximum intensity z-projection of the central spindle (SDCM). Control panel is reproduced from Fig. 4d. (g) Enrichment in pIIb. Thick line: median; thin line: quartile; n: number of central spindles. Statistics: Kruskal–Wallis test. P-values of the respective post-hoc tests indicated. Control, Elp3 RNAi and Elp3 RNAi + Elp3* datasets are reproduced from Fig. 1d. (h) Dividing SOP showing Sara endosomes (iDelta20) and mRFP-Pon (pIIa cortex; dashed blue). Maximum intensity z-projection of entire cells (SDCM). Note the endosomes in the pIIb cell upon Elongator depletion (arrows). Control panel reproduced from Fig. 4f. (i) Percentage of iDelta20 in pIIa after abscission in SOPs (mean ± SEM; Statistics: one-way ANOVA (P < 0.0001), followed by a Tukey post-hoc test (p value indicated); n: number of SOPs. Control, Elp3 RNAi and Elp3 RNAi + Elp3* datasets reproduced from Fig. 2b. Scale bars: 1 µm (a); 2 µm (f); 5 µm (h). Source numerical data and unprocessed blots are available in source data.

Source data

Extended Data Fig. 9 Characterization of the Elongator-induced increase of microtubule growth speed.

(a-e) SNAP-tag-free Elongator behaves the same as SNAP-tagged Elongator (Fig. 5). The effects on microtubule dynamics of a purified full Elongator complex devoid of SNAP tag (Fig. 3d and Extended Data Fig. 5c) were analysed by TIRFM as in Fig. 5. Conditions: 16.5 µM GTP-tubulin, 10% HiLyte 647-tubulin, in the absence (blue data points) or in the presence of 100 nM His-PC-Elongator (green data points). (a,b) Microtubule growth rate at both the minus- (a) and plus-ends (b) under the conditions described above. Data points represent individual growth events quantified from at least two independent experiments. Thick line: median; thin line: quartile. Statistics: two-sided Mann–Whitney test (p-values<0.0001 in both cases; n: microtubule number). (c,d) Cumulative lifetime distribution of microtubules grown from each end under the conditions described above. Elongator increases the microtubule lifetime at both the minus- (c) and the plus-ends (d). Lines: gamma distribution fits. Mean lifetime estimate ±error (that is lifetime at half cumulative distribution, dashed lines): 217 ± 14 s for control (n = 209; blue triangles) and 324 ± 24 s for Elongator (n = 132; green triangles); at the plus end: 163 ± 7 s for control (n = 391; blue circles) and 212 ± 12 s for 25 nM Elongator (n = 237; green circles). Estimate of microtubule catastrophe rate by fitting lifetime histograms by a mono-exponential decay model: Minus end: Control 0.0040 ± 0.0015 sec−1/ Elongator 0.0026 ± 0.0010 sec−1; Plus end: Control 0.0050 ± 0.0022 sec−1, Elongator 0.0037 ± 0.0014 sec−1. (e) Increase of the polymerization speed versus increase of the mean microtubule lifetime (± error) at both ends under the conditions described above. (f) Western blot analysis of in vitro co-sedimentation assays with dynamic microtubules and purified YY531AA Elongator complex (P pellet; S, supernatant). In the presence of microtubules, all the subunits of Elongator identified by immunoblotting are found in the pellet. Source numerical data and unprocessed blots are available in source data.

Source data

Extended Data Fig. 10 Elongator asymmetry controls central spindle asymmetry and thereby polarized trafficking of Sara endosomes.

(a) Percentage of Sara endosomes (iDelta20) in pIIa after abscission in indicated genotypes (mean ± SEM; Statistics: ANOVA1 (P < 0.0001), followed by a Tukey post-hoc test (p value indicated); n: number of SOPs analysed). Control, Elp3 RNAi and Elp3 RNAi + Elp3* datasets are reproduced from Fig. 2b. (b-c) Central spindle asymmetry reversal upon polar targeting of Elongator. (b) SOP expressing Elp3 RNAi + GFP-Elp3*+Elp3RNAi (or control) were processed for K40-Acetyl-α-Tubulin immunolabelling (maximum intensity z-projection, SDCM). (c) Distribution of central spindle asymmetry values in the population of cells described in (b) showing a statistically significant trend for central spindle asymmetry to be reversed. Statistics: Fisher exact test on the proportion of cells with a negative enrichment in pIIb daughter cell (1/26 for Control versus 7/22 for Elp3 RNAi + GFP-Elp3*+GBP-Pon). (d) Percentage of Sara endosome in pIIa cell as a function of the extent of relocalization of Elongator to the anterior cortex in SOP cells expressing Elp3 RNAi + GFP-Elp3*+GBP-Pon (or GFP-Elp3*+GBP-Pon using a different allele of GFP-Elp3*). In this experiment a phenotypic series is generated. The extent of relocalization of Elongator to the anterior cortex positively correlates with the extent of Sara endosomes being targeted to the wrong, pIIb cell. This panel corresponds to unbinned data from panel 6c. Statistics: two-sided Spearman’s correlation coefficient on unbinned data (p value<0.0001). (e) Elongator does not affect the recruitment/activity of Klp98A in SOPs. Sara endosomes (iDelta20) and endogenous Klp98A (GFP knock-in) where imaged in indicated genotypes. Cargo recruitment is a proxy of kinesin activity, thus, the Klp98A–GFP intensity on Sara endosomes can be used as a proxy for kinesin recruitment/activity in Elongator-depleted cells. We specifically looked for a particular situation where, in an Elp3-depleted SOP, two endosomes would be in different cells, but in the same confocal plane, so depth cannot affect fluorescence measurement. Even in this best-case scenario, we could not detect any difference in terms of Klp98A intensity between the two endosomes. (f) Paradigm for the polarized motility of Sara endosomes during SOP division. Scale bars: 5 µm (b, c-left and e), 2 µm (c-right). Source numerical data are available in source data.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2.

Reporting Summary

Peer Review File

Supplementary Video 1

Dynamics of central spindle symmetry breaking in control cells. Time-lapse video of a dividing control SOP showing Jupiter–GFP/mRFP-PonLD (left) and Jupiter–GFP with the Rainbow RGB LUT for signal density (right). Spinning disk confocal imaging (images are maximum intensity z-projections of the entire SOP cell). Registered time is indicated in seconds. Registered time = 0 indicates anaphase B onset. Note that the central spindle becomes asymmetric during anaphase, with an enrichment of microtubule density on the anterior (Pon-positive) side of the spindle. Scale bar, 5 µm.

Supplementary Video 2

Central spindle asymmetry is abolished in Elp3-depleted SOPs. Time-lapse video of a dividing Elp3RNAi SOP showing Jupiter–GFP/mRFP-PonLD (left) and Jupiter–GFP with the Rainbow RGB LUT for signal density (right). Spinning disk confocal imaging (images are maximum intensity z-projections of the entire SOP cell). Registered time is indicated in seconds. Registered time = 0 indicates anaphase B onset. Note that the central spindle never becomes asymmetric. Scale bar, 5 µm.

Supplementary Video 3

Asymmetric Sara endosome segregation is abolished in Elp3 mutants. Dividing SOP showing Sara endosomes (iDelta20) and mRFP-Pon in the indicated genotype. Images are maximum intensity z-projections of entire cells (SDCM). Elapsed time is indicated in seconds. Note that Sara endosomes are asymmetrical in controls but symmetrical in Elp3 mutants. Scale bar: 5 µm.

Supplementary Video 4

Elongator directly stabilizes microtubules. Biotinylated, rhodamine-labelled, GMPCPP-stabilized seeds (red) were anchored on a glass surface via fibrinogen–NeutrAvidin, 12.5 µM GTP-tubulin (10% labelled with HiLyte 647-fluorescent GTP–tubulin, cyan) was then added in the presence or absence of 25 nM (His)6-PC-SNAP–Elongator and microtubule dynamics were observed by TIRFM (top). Time is indicated in seconds. Note that the SNAP-tagged Elongator is not labelled with any BG-conjugated dye here. Control in the absence of Elongator was performed with the same buffer used for the experiment in the presence of Elongator. Kymographs of the microtubules displayed in the top panels (orange lines; bottom). Note that microtubules grow faster and undergo less catastrophe in the presence of Elongator. Scale bar, 10 µm.

Source data

Source Data Fig. 1

Statistical source data for Fig. 1.

Source Data Fig. 2

Statistical source data for Fig. 2.

Source Data Fig. 3

Statistical source data for Fig. 3.

Source Data Fig. 3

Unprocessed western blots for Fig. 3.

Source Data Fig. 4

Statistical source data for Fig. 4.

Source Data Fig. 4

Unprocessed gel for Fig. 4.

Source Data Fig. 5

Statistical source data for Fig. 5.

Source Data Fig. 6

Statistical source data for Fig. 6.

Source Data Extended Data Fig. 1

Statistical source data for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Unprocessed western blots for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Statistical source data for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Statistical source data for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Statistical source data for Extended Data Fig. 5.

Source Data Extended Data Fig. 5

Unprocessed western blots and gels for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Unprocessed western blots and gels for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Statistical source data for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Statistical source data for Extended Data Fig. 8.

Source Data Extended Data Fig. 8

Unprocessed western blots for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Statistical source data for Extended Data Fig. 9.

Source Data Extended Data Fig. 9

Unprocessed western blots for Extended Data Fig. 9.

Source Data Extended Data Fig. 10

Statistical source data for Extended Data Fig. 10.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) 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

Planelles-Herrero, V.J., Bittleston, A., Seum, C. et al. Elongator stabilizes microtubules to control central spindle asymmetry and polarized trafficking of cell fate determinants. Nat Cell Biol 24, 1606–1616 (2022). https://doi.org/10.1038/s41556-022-01020-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-01020-9

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