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:

Ultrastructure and dynamics of the actin−myosin II cytoskeleton during mitochondrial fission

Abstract

Mitochondrial fission involves the preconstriction of an organelle followed by scission by dynamin-related protein Drp1. Preconstriction is facilitated by actin and non-muscle myosin II through a mechanism that remains unclear, largely due to the unknown cytoskeletal ultrastructure at mitochondrial constrictions. Here, using platinum replica electron microscopy, we show that mitochondria in cells are embedded in an interstitial cytoskeletal network that contains abundant unbranched actin filaments. Both spontaneous and induced mitochondrial constrictions typically associate with a criss-cross array of long actin filaments that comprise part of this interstitial network. Non-muscle myosin II is found adjacent to mitochondria but is not specifically enriched at the constriction sites. During ionomycin-induced mitochondrial fission, F-actin clouds colocalize with mitochondrial constriction sites, whereas dynamic myosin II clouds are present in the vicinity of constrictions. We propose that myosin II promotes mitochondrial constriction by inducing stochastic deformations of the interstitial actin network, which applies pressure on the mitochondrial surface and thus initiates curvature-sensing mechanisms that complete mitochondrial constriction.

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: Ultrastructure of the cell interior in glial cells revealed by PREM.
Fig. 2: CLEM of COS-7 cells.
Fig. 3: Actin filament organization at mitochondrial constriction sites induced in COS-7 cells by expression of the GFP−K38A-Drp1 construct.
Fig. 4: Actin filament organization at mitochondrial constriction sites formed in COS-7 cells coexpressing GFP−K38A-Drp1 and mCherry−INF2-A149D-CAAX.
Fig. 5: Rotenone treatment induces mitochondrial fission and actin reorganization in cells.
Fig. 6: The dynamics of F-actin and NMIIA in the course of ionomycin-induced mitochondrial fission in HeLa cells.
Fig. 7: NMII associated with the interstitial actin network stimulates mitochondrial constrictions.

Similar content being viewed by others

Data availability

Source data for Figs. 5h, 6b and 7b and the statistical results presented in the text have been provided as Supplementary Table 1. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

References

  1. Friedman, J. R. & Nunnari, J. Mitochondrial form and function. Nature 505, 335–343 (2014).

    Article  CAS  Google Scholar 

  2. Mishra, P. & Chan, D. C. Mitochondrial dynamics and inheritance during cell division, development and disease. Nat. Rev. Mol. Cell Biol. 15, 634–646 (2014).

    Article  CAS  Google Scholar 

  3. Wai, T. & Langer, T. Mitochondrial dynamics and metabolic regulation. Trends Endocrinol. Metab. 27, 105–117 (2016).

    Article  CAS  Google Scholar 

  4. Pagliuso, A., Cossart, P. & Stavru, F. The ever-growing complexity of the mitochondrial fission machinery. Cell. Mol. Life Sci. 75, 355–374 (2018).

    Article  CAS  Google Scholar 

  5. Ramachandran, R. Mitochondrial dynamics: the dynamin superfamily and execution by collusion. Semin. Cell Dev. Biol. 76, 201–212 (2018).

    Article  CAS  Google Scholar 

  6. Lee, J. E., Westrate, L. M., Wu, H., Page, C. & Voeltz, G. K. Multiple dynamin family members collaborate to drive mitochondrial division. Nature 540, 139–143 (2016).

    Article  CAS  Google Scholar 

  7. Basu, K. et al. Molecular mechanism of DRP1 assembly studied in vitro by cryo-electron microscopy. PLoS One 12, e0179397 (2017).

    Article  Google Scholar 

  8. Francy, C. A., Alvarez, F. J., Zhou, L., Ramachandran, R. & Mears, J. A. The mechanoenzymatic core of dynamin-related protein 1 comprises the minimal machinery required for membrane constriction. J. Biol. Chem. 290, 11692–11703 (2015).

    Article  CAS  Google Scholar 

  9. Macdonald, P. J. et al. Distinct splice variants of dynamin-related protein 1 differentially utilize mitochondrial fission factor as an effector of cooperative GTPase activity. J. Biol. Chem. 291, 493–507 (2016).

    Article  CAS  Google Scholar 

  10. Korobova, F., Ramabhadran, V. & Higgs, H. N. An actin-dependent step in mitochondrial fission mediated by the ER-associated formin INF2. Science 339, 464–467 (2013).

    Article  CAS  Google Scholar 

  11. Friedman, J. R. et al. ER tubules mark sites of mitochondrial division. Science 334, 358–362 (2011).

    Article  CAS  Google Scholar 

  12. Manor, U. et al. A mitochondria-anchored isoform of the actin-nucleating Spire protein regulates mitochondrial division. eLife 4, e08828 (2015).

    Article  Google Scholar 

  13. DuBoff, B., Gotz, J. & Feany, M. B. Tau promotes neurodegeneration via DRP1 mislocalization in vivo. Neuron 75, 618–632 (2012).

    Article  CAS  Google Scholar 

  14. Korobova, F., Gauvin, T. J. & Higgs, H. N. A role for myosin II in mammalian mitochondrial fission. Curr. Biol. 24, 409–414 (2014).

    Article  CAS  Google Scholar 

  15. Svitkina, T. The actin cytoskeleton and actin-based motility. CSH Perspect. Biol. 10, a018267 (2018).

    Google Scholar 

  16. Li, S. et al. Transient assembly of F-actin on the outer mitochondrial membrane contributes to mitochondrial fission. J. Cell Biol. 208, 109–123 (2015).

    Article  CAS  Google Scholar 

  17. Hatch, A. L., Gurel, P. S. & Higgs, H. N. Novel roles for actin in mitochondrial fission. J. Cell Sci. 127, 4549–4560 (2014).

    Article  Google Scholar 

  18. Cheffings, T. H., Burroughs, N. J. & Balasubramanian, M. K. Actomyosin ring formation and tension generation in eukaryotic cytokinesis. Curr. Biol. 26, 719–737 (2016).

    Article  Google Scholar 

  19. Billington, N., Wang, A., Mao, J., Adelstein, R. S. & Sellers, J. R. Characterization of three full-length human nonmuscle myosin II paralogs. J. Biol. Chem. 288, 33398–33410 (2013).

    Article  CAS  Google Scholar 

  20. Verkhovsky, A. B., Svitkina, T. M. & Borisy, G. G. Myosin II filament assemblies in the active lamella of fibroblasts: their morphogenesis and role in the formation of actin filament bundles. J. Cell Biol. 131, 989–1002 (1995).

    Article  CAS  Google Scholar 

  21. Requejo-Isidro, J. Fluorescence nanoscopy. Methods and applications. J. Chem. Biol. 6, 97–120 (2013).

    Article  Google Scholar 

  22. Svitkina, T. M. Platinum replica electron microscopy: imaging the cytoskeleton globally and locally. Int. J. Biochem. Cell Biol. 86, 37–41 (2017).

    Article  CAS  Google Scholar 

  23. Collins, A., Warrington, A., Taylor, K. A. & Svitkina, T. Structural organization of the actin cytoskeleton at sites of clathrin-mediated endocytosis. Curr. Biol. 21, 1167–1175 (2011).

    Article  CAS  Google Scholar 

  24. Korobova, F. & Svitkina, T. Molecular architecture of synaptic actin cytoskeleton in hippocampal neurons reveals a mechanism of dendritic spine morphogenesis. Mol. Biol. Cell 21, 165–176 (2010).

    Article  CAS  Google Scholar 

  25. Svitkina, T. M. et al. Mechanism of filopodia initiation by reorganization of a dendritic network. J. Cell Biol. 160, 409–421 (2003).

    Article  CAS  Google Scholar 

  26. Svitkina, T. M. & Borisy, G. G. in Cell Biology: A Laboratory Handbook 3rd edn, Vol. 3 (ed. Celis, J.) 277−285 (Elsevier, 2006).

  27. Svitkina, T. Electron microscopic analysis of the leading edge in migrating cells. Methods Cell Biol. 79, 295–319 (2007).

    Article  CAS  Google Scholar 

  28. Svitkina, T. Imaging cytoskeleton components by electron microscopy. Methods Mol. Biol 1365, 99–118 (2016).

    Article  CAS  Google Scholar 

  29. Yang, C. et al. Novel roles of formin mDia2 in lamellipodia and filopodia formation in motile cells. PLoS Biol. 5, e317 (2007).

    Article  Google Scholar 

  30. Heuser, J. The production of ‘cell cortices’ for light and electron microscopy. Traffic 1, 545–552 (2000).

    Article  CAS  Google Scholar 

  31. Sato, F., Asakawa, H., Fukuma, T. & Terada, S. Semi-in situ atomic force microscopy imaging of intracellular neurofilaments under physiological conditions through the ‘sandwich’ method. Microscopy 65, 316–324 (2016).

    Article  CAS  Google Scholar 

  32. Efimova, N. & Svitkina, T. M. Branched actin networks push against each other at adherens junctions to maintain cell-cell adhesion. J. Cell Biol. 217, 1827–1845 (2018).

    Article  CAS  Google Scholar 

  33. Gurel, P. S. et al. Assembly and turnover of short actin filaments by the formin INF2 and profilin. J. Biol. Chem. 290, 22494–22506 (2015).

    Article  CAS  Google Scholar 

  34. Smirnova, E., Griparic, L., Shurland, D. L. & van der Bliek, A. M. Dynamin-related protein Drp1 is required for mitochondrial division in mammalian cells. Mol. Biol. Cell 12, 2245–2256 (2001).

    Article  CAS  Google Scholar 

  35. De Vos, K. J., Allan, V. J., Grierson, A. J. & Sheetz, M. P. Mitochondrial function and actin regulate dynamin-related protein 1-dependent mitochondrial fission. Curr. Biol. 15, 678–683 (2005).

    Article  Google Scholar 

  36. Ji, W. K., Hatch, A. L., Merrill, R. A., Strack, S. & Higgs, H. N. Actin filaments target the oligomeric maturation of the dynamic GTPase Drp1 to mitochondrial fission sites. eLife 4, e11553 (2015).

    Article  Google Scholar 

  37. Ramabhadran, V., Hatch, A. L. & Higgs, H. N. Actin monomers activate inverted formin 2 by competing with its autoinhibitory interaction. J. Biol. Chem. 288, 26847–26855 (2013).

    Article  CAS  Google Scholar 

  38. Barsoum, M. J. et al. Nitric oxide-induced mitochondrial fission is regulated by dynamin-related GTPases in neurons. EMBO J. 25, 3900–3911 (2006).

    Article  CAS  Google Scholar 

  39. Toyama, E. Q. et al. Metabolism. AMP-activated protein kinase mediates mitochondrial fission in response to energy stress. Science 351, 275–281 (2016).

    Article  CAS  Google Scholar 

  40. Sanmartin, C. D. et al. Ryanodine receptor-mediated Ca2+ release underlies iron-induced mitochondrial fission and stimulates mitochondrial Ca(2+) uptake in primary hippocampal neurons. Front. Mol. Neurosci. 7, 13 (2014).

    PubMed  PubMed Central  Google Scholar 

  41. Wales, P. et al. Calcium-mediated actin reset (CaAR) mediates acute cell adaptations. eLife 5, e19850 (2016).

    Article  Google Scholar 

  42. Bao, J., Jana, S. S. & Adelstein, R. S. Vertebrate nonmuscle myosin II isoforms rescue small interfering RNA-induced defects in COS-7 cell cytokinesis. J. Biol. Chem. 280, 19594–19599 (2005).

    Article  CAS  Google Scholar 

  43. Shutova, M. S. et al. Self-sorting of nonmuscle myosins IIA and IIB polarizes the cytoskeleton and modulates cell motility. J. Cell Biol. 216, 2877–2889 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Helle, S. C. J. et al. Mechanical force induces mitochondrial fission. eLife 6, e30292 (2017).

    Article  Google Scholar 

  45. Ban-Ishihara, R., Ishihara, T., Sasaki, N., Mihara, K. & Ishihara, N. Dynamics of nucleoid structure regulated by mitochondrial fission contributes to cristae reformation and release of cytochrome c. Proc. Natl Acad. Sci. USA 110, 11863–11868 (2013).

    Article  CAS  Google Scholar 

  46. Bogenhagen, D. F., Wang, Y., Shen, E. L. & Kobayashi, R. Protein components of mitochondrial DNA nucleoids in higher eukaryotes. Mol. Cell. Proteomics 2, 1205–1216 (2003).

    Article  CAS  Google Scholar 

  47. Garrido, N. et al. Composition and dynamics of human mitochondrial nucleoids. Mol. Biol. Cell 14, 1583–1596 (2003).

    Article  CAS  Google Scholar 

  48. Chakrabarti, R. et al. INF2-mediated actin polymerization at the ER stimulates mitochondrial calcium uptake, inner membrane constriction, and division. J. Cell Biol. 217, 251–268 (2018).

    Article  CAS  Google Scholar 

  49. Cho, B. et al. Constriction of the mitochondrial inner compartment is a priming event for mitochondrial division. Nat. Commun. 8, 15754 (2017).

    Article  Google Scholar 

  50. Wilcox, K. S., Buchhalter, J. & Dichter, M. A. Properties of inhibitory and excitatory synapses between hippocampal neurons in very low density cultures. Synapse 18, 128–151 (1994).

    Article  CAS  Google Scholar 

  51. Efimova, N. et al. βIII spectrin is necessary for formation of the constricted neck of dendritic spines and regulation of synaptic activity in neurons. J. Neurosci. 37, 6442–6459 (2017).

    Article  CAS  Google Scholar 

  52. Svitkina, T. M. & Borisy, G. G. Correlative light and electron microscopy of the cytoskeleton of cultured cells. Methods Enzymol. 298, 570–592 (1998).

    Article  CAS  Google Scholar 

  53. York, A. G. et al. Instant super-resolution imaging in live cells and embryos via analog image processing. Nat. Methods 10, 1122–1126 (2013).

    Article  CAS  Google Scholar 

  54. Strack, S., Wilson, T. J. & Cribbs, J. T. Cyclin-dependent kinases regulate splice-specific targeting of dynamin-related protein 1 to microtubules. J. Cell Biol. 201, 1037–1051 (2013).

    Article  CAS  Google Scholar 

  55. Johnson, H. W. & Schell, M. J. Neuronal IP3 3-kinase is an F-actin-bundling protein: role in dendritic targeting and regulation of spine morphology. Mol. Biol. Cell 20, 5166–5180 (2009).

    Article  CAS  Google Scholar 

  56. Dulyaninova, N. G., House, R. P., Betapudi, V. & Bresnick, A. R. Myosin-IIA heavy-chain phosphorylation regulates the motility of MDA-MB-231 carcinoma cells. Mol. Biol. Cell 18, 3144–3155 (2007).

    Article  CAS  Google Scholar 

  57. Wei, Q. & Adelstein, R. S. Conditional expression of a truncated fragment of nonmuscle myosin II-A alters cell shape but not cytokinesis in HeLa cells. Mol. Biol. Cell 11, 3617–3627 (2000).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank H. N. Higgs for helpful discussions and reagents, A. York and M. Schell for reagents and the members of the Svitkina lab, M. Shutova and N. Efimova, for comments on the manuscript. This work was supported by NIH grant nos. R01 GM 095977 and R01 GM 106000 to T.M.S.

Author information

Authors and Affiliations

Authors

Contributions

C.Y. performed all the experimental work and data analyses. C.Y. and T.S. prepared the figures and wrote the manuscript.

Corresponding author

Correspondence to Tatyana M. Svitkina.

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.

Integrated supplementary information

Supplementary Fig. 1 Additional examples and 3D views of the ultrastructure of the cell interior revealed by PREM in glial cells.

Exposure of the cell interior by saponin-mediated extraction (a-d) or by nitrocellulose-mediated unroofing (e,f). (a, e) Broad windows into the cell interior formed as a result of saponin-mediated extraction (a) or nitrocellulose-mediated unroofing (e) expose membrane organelles associated with the cytoskeleton. Boxed regions in a and e are enlarged in b and f, respectively. (b, f) Enlarged boxed region from a and e, respectively, shown as anaglyph images generated from stereo pairs taken at ±11º. To view 3D organization, use red-cyan anaglyph glasses with red filter over the left eye. These image show unconstructed mitochondria (purple) that interact with putative ER structures (yellow). Cytoskeletal filaments are present in the cytoplasm. They often interact with the ER and sometimes intersect mitochondria. (c, d) PREM anaglyph images corresponding to panels b and c of main Fig. 1. Scale bars: 2 µm (a and e), and 200 nm (b, c-f). The images are representative of n = 6 independent experiments, which gave similar results.

Supplementary Fig. 2 Control experiments for the procedures associated with nitrocellulose-mediated unroofing.

(a) Time lapse imaging of HeLa cells expressing GFP-Ftractin and stained with MitoTracker Deep Red. At t = 0 sec, cell culture medium was replaced with PEM buffer containing 2 µM phalloidin and 10 µM taxol and maintained at room temperature (~22ºC). Imaging was also performed at room temperature. Arrows point to individual mitochondria or F-actin structures to indicate that no apparent changes was induced by the PEM buffer during live cell imaging. The images are representative of n = 2 independent experiments. (b) Confocal images of COS-7 cells expressing Mito-BFP (green) and mCherry-Sec61β (red). Control: cells were directly fixed and imaged. NC-treated: cells were subjected to nitrocellulose-mediated unroofing, fixed, imaged by confocal microscopy, processed for PREM and imaged by electron microscopy. In this sample, some cells were unroofed (bottom row), whereas others remained intact (middle row). The overall organization of Mito-BFP and mCherry-Sec61β is similar in all three cases. Scale bars: 5 µm. The images are representative of n = 3 independent experiments, which gave similar results.

Supplementary Fig. 3 Constitutively active INF2-A149D-CAAX induces abundant actin filaments at the ER surface.

(a-c) Confocal microscopy of a COS-7 cell expressing GFP-INF2-A149D-CAAX (AD-INF2), the ER marker mCherry-Sec61β (Sec61β) and mitochondrial marker Mito-BFP (Mito), and stained with Alexa Fluor 680 phalloidin (F-actin). (a, b) Individual fluorescence channels showing the nearly entire cell (a) and an enlarged boxed region (b). (c) Dual-channel overlays in different combinations of the same region as in b reveal partial colocalization between GFP-INF2-A149D-CAAX, mCherry-Sec61β and F-actin. (d-h) CLEM of mCherry-INF2-A149D-CAAX-expressing COS-7 cell. (d) Fluorescence microscopy of mCherry-INF2-A149D-CAAX in an expressing cell. (e, f) PREM (e) and PREM/fluorescence overlay (f) images of the boxed region in d. (g, h) The INF-A149D-CAAX-positive ER regions are coated with dense actin filament arrays. Both panels show enlargements of the same region outlined by a purple box in f either as PREM/fluorescence overlay (g) or by PREM only (h). Mito, mitochondria. Arrowheads mark actin filaments originating from INF-A149D-CAAX-positive ER and running toward the mitochondrion. See 3D version of panel H in Supplementary Fig. 4 (panel s3). Scale bars: 5 µm (a, d-f), 2 µm (b, c) and 500 nm (g, h). The images are representative of n = 2 independent experiments, which gave similar results.

Supplementary Fig. 4 3D anaglyph versions of the images shown in the main-text figures and Supplementary Fig. 3, as indicated by panel labels.

The images are representative of n = 3 (Fig. 3c, g) or 2 (Figs. 4f, 5b, Supplementary Fig. 3) independent experiments, which gave similar results.

Supplementary Fig. 5 Ionomycin-induced mitochondrial constriction and fission in HeLa cells.

(a) Dynamics of mitochondria, the ER and F-actin in a HeLa cell expressing Mito-BFP (white), GFP-Ftractin (green) and mCherry-NMIIA (red) after addition of ionomycin at time 0:00 (min:sec). Ionomycin treatment leads to accumulation of F-actin that largely overlaps with the ER, whereas mitochondria are mostly located in the voids of the network. However, sites of mitochondrial constriction and fission (arrows and arrowheads) overlap with both F-actin and ER signals. (b) Dynamics of mitochondria, F-actin and NMIIA in a HeLa cell expressing Mito-BFP (white), GFP-Ftractin (green) and mCherry-NMIIA (red) after addition of ionomycin at time 0:00 (min:sec). Initiation of mitochondrial constriction correlates with the appearance of F-actin and, occasionally, NMIIA clouds intersecting the constriction (arrows and arrowheads). Subsequently, F-actin remains at the constriction sites until mitochondrial fission, whereas NMIIA mostly fluctuates in the mitochondrion vicinity. (c) A 3D PREM image of a constricted mitochondrion (purple) in an unroofed HeLa cell treated with ionomycin for 1.5 min. Membranous structures likely representing the ER are shaded in yellow. Actin filaments apparently originating from the ER intersect the mitochondrion close to the constriction site. Scale bars: 1 µm (a, b) and 200 nm (c). The images are representative of n = 2 independent experiments, which gave similar results.

Supplementary Fig. 6 Ionomycin-induced mitochondrial constriction and fission in COS-7 cells.

(a) F-actin assembly in COS-7 cells after ionomycin treatment. COS-7 cell expressing mCherry-Ftractin is shown before (top) and after (middle) ionomycin treatment. Merged image (bottom) of these two time points shows reddish color in the cell center indicating F-actin assembly in the cytoplasm after ionomycin treatment. (b) Time-lapse sequence of a mitochondrial fission event in COS-7 cell expressing Mito-BFP (white), mCherry-Ftractin (red) and GFP-NMIIB (green) after addition of ionomycin at time 0:00 (min:sec). Initiation of mitochondrial constriction (arrow, t=0:21) correlates with the appearance of F-actin and NMIIB clouds intersecting the constriction (arrows and arrowheads). Subsequently, F-actin remains at the constriction sites until mitochondrial fission, whereas NMIIA mostly fluctuates in the mitochondrion vicinity. (c) Immunogold PREM of NMIIB localization at mitochondrial constriction in an unroofed COS-7 cell treated with ionomycin for 5 min. Immunogold particles (18 nm) marked by yellow dots localize at comparable densities at the constrictions site and in the surrounding network. Mitochondrion is shaded in purple. Scale bars: 10 µm (a), 1 µm (b) and 200 nm (c). The images are representative of n = 2 independent experiments, which gave similar results.

Supplementary Information

Supplementary Information

Supplementary Figures 1–6, Supplementary Table title/legend, Supplementary Video titles/legends.

Reporting Summary

Supplementary Video 1

Three-dimensional organization of actin filaments at the mitochondrial constriction site in a COS-7 cell expressing GFP−K38A-Drp1.

Supplementary Video 2

Rotenone treatment induces actin assembly in the cytoplasm in association with mitochondria.

Supplementary Video 3

Ionomycin treatment leads to F-actin assembly in association with the ER.

Supplementary Video 4

Dynamics of mitochondria, F-actin and NMIIA in HeLa cells (example 1).

Supplementary Video 5

Dynamics of mitochondria, F-actin and NMIIA in HeLa cells (example 2).

Supplementary Video 6

Dynamics of mitochondria, F-actin and NMIIB in COS-7 cells.

Supplementary Video 7

3D organization of actin filaments and immunogold-labelled NMIIA at the mitochondrial constriction site in a HeLa cell treated with ionomycin for 2 min.

Supplementary Table 1

Statistics source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, C., Svitkina, T.M. Ultrastructure and dynamics of the actin−myosin II cytoskeleton during mitochondrial fission. Nat Cell Biol 21, 603–613 (2019). https://doi.org/10.1038/s41556-019-0313-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0313-6

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