Skip to main content

Imaging the Molecular Machines That Power Cell Migration

  • Protocol
  • First Online:
Cell Migration

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1749))

Abstract

Animal cell migration constitutes a complex process involving a multitude of forces generated and maintained by the actin cytoskeleton. Dynamic changes of the cell surface, for instance to effect cell edge protrusion, are at the core of initiating migratory processes, both in tissue culture models and whole animals. Here we sketch different aspects of imaging representative molecular constituents in such actin-driven processes, which power and regulate the polymerisation of actin filaments into bundles and networks, constituting the building blocks of such protrusions. The examples presented illustrate both the diversity of subcellular distributions of distinct molecular components, according to their function, and the complexity of dynamic changes in protrusion size, shape, and/or orientation in 3D. Considering these dynamics helps mechanistically connecting subcellular distributions of molecular machines driving protrusion and migration with their biochemical function.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 79.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 99.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 129.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Krause M, Gautreau A (2014) Steering cell migration: lamellipodium dynamics and the regulation of directional persistence. Nat Rev Mol Cell Biol 15(9):577–590. https://doi.org/10.1038/nrm3861

    Article  CAS  PubMed  Google Scholar 

  2. Ladwein M, Rottner K (2008) On the Rho’d: the regulation of membrane protrusions by Rho-GTPases. FEBS Lett 582(14):2066–2074. https://doi.org/10.1016/j.febslet.2008.04.033

    Article  CAS  PubMed  Google Scholar 

  3. Mattila PK, Lappalainen P (2008) Filopodia: molecular architecture and cellular functions. Nat Rev Mol Cell Biol 9(6):446–454. https://doi.org/10.1038/nrm2406

    Article  CAS  PubMed  Google Scholar 

  4. Small JV, Stradal T, Vignal E, Rottner K (2002) The lamellipodium: where motility begins. Trends Cell Biol 12(3):112–120

    Article  CAS  PubMed  Google Scholar 

  5. Charras G, Paluch E (2008) Blebs lead the way: how to migrate without lamellipodia. Nat Rev Mol Cell Biol 9(9):730–736. https://doi.org/10.1038/nrm2453

    Article  CAS  PubMed  Google Scholar 

  6. Paksa A, Raz E (2015) Zebrafish germ cells: motility and guided migration. Curr Opin Cell Biol 36:80–85. https://doi.org/10.1016/j.ceb.2015.07.007

    Article  CAS  PubMed  Google Scholar 

  7. Paluch EK, Raz E (2013) The role and regulation of blebs in cell migration. Curr Opin Cell Biol 25(5):582–590. https://doi.org/10.1016/j.ceb.2013.05.005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Mishin AS, Belousov VV, Solntsev KM, Lukyanov KA (2015) Novel uses of fluorescent proteins. Curr Opin Chem Biol 27:1–9. https://doi.org/10.1016/j.cbpa.2015.05.002

    Article  CAS  PubMed  Google Scholar 

  9. Rodriguez EA, Campbell RE, Lin JY, Lin MZ, Miyawaki A, Palmer AE, Shu X, Zhang J, Tsien RY (2017) The growing and glowing toolbox of fluorescent and photoactive proteins. Trends Biochem Sci 42(2):111–129. https://doi.org/10.1016/j.tibs.2016.09.010

    Article  CAS  PubMed  Google Scholar 

  10. Demmerle J, Innocent C, North AJ, Ball G, Muller M, Miron E, Matsuda A, Dobbie IM, Markaki Y, Schermelleh L (2017) Strategic and practical guidelines for successful structured illumination microscopy. Nat Protoc 12(5):988–1010. https://doi.org/10.1038/nprot.2017.019

    Article  CAS  PubMed  Google Scholar 

  11. Kanchanawong P, Shtengel G, Pasapera AM, Ramko EB, Davidson MW, Hess HF, Waterman CM (2010) Nanoscale architecture of integrin-based cell adhesions. Nature 468(7323):580–584. https://doi.org/10.1038/nature09621

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Schermelleh L, Heintzmann R, Leonhardt H (2010) A guide to super-resolution fluorescence microscopy. J Cell Biol 190(2):165–175. https://doi.org/10.1083/jcb.201002018

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Block J, Breitsprecher D, Kuhn S, Winterhoff M, Kage F, Geffers R, Duwe P, Rohn JL, Baum B, Brakebusch C, Geyer M, Stradal TE, Faix J, Rottner K (2012) FMNL2 drives actin-based protrusion and migration downstream of Cdc42. Curr Biol 22(11):1005–1012. https://doi.org/10.1016/j.cub.2012.03.064

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kage F, Winterhoff M, Dimchev V, Mueller J, Thalheim T, Freise A, Bruhmann S, Kollasser J, Block J, Dimchev G, Geyer M, Schnittler HJ, Brakebusch C, Stradal TE, Carlier MF, Sixt M, Kas J, Faix J, Rottner K (2017) FMNL formins boost lamellipodial force generation. Nat Commun 8:14832. https://doi.org/10.1038/ncomms14832

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Krause M, Dent EW, Bear JE, Loureiro JJ, Gertler FB (2003) Ena/VASP proteins: regulators of the actin cytoskeleton and cell migration. Annu Rev Cell Dev Biol 19:541–564. https://doi.org/10.1146/annurev.cellbio.19.050103.103356

    Article  CAS  PubMed  Google Scholar 

  16. Rottner K, Behrendt B, Small JV, Wehland J (1999) VASP dynamics during lamellipodia protrusion. Nat Cell Biol 1(5):321–322. https://doi.org/10.1038/13040

    Article  CAS  PubMed  Google Scholar 

  17. Breitsprecher D, Kiesewetter AK, Linkner J, Vinzenz M, Stradal TE, Small JV, Curth U, Dickinson RB, Faix J (2011) Molecular mechanism of Ena/VASP-mediated actin-filament elongation. EMBO J 30(3):456–467. https://doi.org/10.1038/emboj.2010.348

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Hansen SD, Mullins RD (2010) VASP is a processive actin polymerase that requires monomeric actin for barbed end association. J Cell Biol 191(3):571–584. https://doi.org/10.1083/jcb.201003014

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Mejillano MR, Kojima S, Applewhite DA, Gertler FB, Svitkina TM, Borisy GG (2004) Lamellipodial versus filopodial mode of the actin nanomachinery: pivotal role of the filament barbed end. Cell 118(3):363–373. https://doi.org/10.1016/j.cell.2004.07.019

    Article  CAS  PubMed  Google Scholar 

  20. Svitkina TM, Bulanova EA, Chaga OY, Vignjevic DM, Kojima S, Vasiliev JM, Borisy GG (2003) Mechanism of filopodia initiation by reorganization of a dendritic network. J Cell Biol 160(3):409–421. https://doi.org/10.1083/jcb.200210174

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Edwards M, Zwolak A, Schafer DA, Sept D, Dominguez R, Cooper JA (2014) Capping protein regulators fine-tune actin assembly dynamics. Nat Rev Mol Cell Biol 15(10):677–689. https://doi.org/10.1038/nrm3869

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Iwasa JH, Mullins RD (2007) Spatial and temporal relationships between actin-filament nucleation, capping, and disassembly. Curr Biol 17(5):395–406. https://doi.org/10.1016/j.cub.2007.02.012

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lai FP, Szczodrak M, Block J, Faix J, Breitsprecher D, Mannherz HG, Stradal TE, Dunn GA, Small JV, Rottner K (2008) Arp2/3 complex interactions and actin network turnover in lamellipodia. EMBO J 27(7):982–992. https://doi.org/10.1038/emboj.2008.34

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Alekhina O, Burstein E, Billadeau DD (2017) Cellular functions of WASP family proteins at a glance. J Cell Sci 130(14):2235–2241. https://doi.org/10.1242/jcs.199570

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bisi S, Disanza A, Malinverno C, Frittoli E, Palamidessi A, Scita G (2013) Membrane and actin dynamics interplay at lamellipodia leading edge. Curr Opin Cell Biol 25(5):565–573. https://doi.org/10.1016/j.ceb.2013.04.001

    Article  CAS  PubMed  Google Scholar 

  26. Innocenti M, Zucconi A, Disanza A, Frittoli E, Areces LB, Steffen A, Stradal TE, Di Fiore PP, Carlier MF, Scita G (2004) Abi1 is essential for the formation and activation of a WAVE2 signalling complex. Nat Cell Biol 6(4):319–327. https://doi.org/10.1038/ncb1105ncb1105[pii]

    Article  CAS  PubMed  Google Scholar 

  27. Kunda P, Craig G, Dominguez V, Baum B (2003) Abi, Sra1, and Kette control the stability and localization of SCAR/WAVE to regulate the formation of actin-based protrusions. Curr Biol 13(21):1867–1875

    Article  CAS  PubMed  Google Scholar 

  28. Steffen A, Rottner K, Ehinger J, Innocenti M, Scita G, Wehland J, Stradal TE (2004) Sra-1 and Nap1 link Rac to actin assembly driving lamellipodia formation. EMBO J 23(4):749–759. https://doi.org/10.1038/sj.emboj.7600084

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Stradal TE, Scita G (2006) Protein complexes regulating Arp2/3-mediated actin assembly. Curr Opin Cell Biol 18(1):4–10. https://doi.org/10.1016/j.ceb.2005.12.003

    Article  CAS  PubMed  Google Scholar 

  30. Stradal T, Courtney KD, Rottner K, Hahne P, Small JV, Pendergast AM (2001) The Abl interactor proteins localize to sites of actin polymerization at the tips of lamellipodia and filopodia. Curr Biol 11(11):891–895

    Article  CAS  PubMed  Google Scholar 

  31. Gertler FB, Niebuhr K, Reinhard M, Wehland J, Soriano P (1996) Mena, a relative of VASP and Drosophila enabled, is implicated in the control of microfilament dynamics. Cell 87(2):227–239

    Article  CAS  PubMed  Google Scholar 

  32. Haffner C, Jarchau T, Reinhard M, Hoppe J, Lohmann SM, Walter U (1995) Molecular cloning, structural analysis and functional expression of the proline-rich focal adhesion and microfilament-associated protein VASP. EMBO J 14(1):19–27

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Rottner K, Hall A, Small JV (1999) Interplay between Rac and Rho in the control of substrate contact dynamics. Curr Biol 9(12):640–648

    Article  CAS  PubMed  Google Scholar 

  34. Ballestrem C, Wehrle-Haller B, Imhof BA (1998) Actin dynamics in living mammalian cells. J Cell Sci 111(Pt 12):1649–1658

    Article  CAS  PubMed  Google Scholar 

  35. Hall A (1998) Rho GTPases and the actin cytoskeleton. Science 279(5350):509–514

    Article  CAS  PubMed  Google Scholar 

  36. Nobes CD, Hall A (1995) Rho, rac, and cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia. Cell 81(1):53–62. doi:0092-8674(95)90370-4[pii]

    Article  CAS  PubMed  Google Scholar 

  37. Koestler SA, Steffen A, Nemethova M, Winterhoff M, Luo N, Holleboom JM, Krupp J, Jacob S, Vinzenz M, Schur F, Schluter K, Gunning PW, Winkler C, Schmeiser C, Faix J, Stradal TE, Small JV, Rottner K (2013) Arp2/3 complex is essential for actin network treadmilling as well as for targeting of capping protein and cofilin. Mol Biol Cell 24(18):2861–2875. https://doi.org/10.1091/mbc.E12-12-0857

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Steffen A, Koestler SA, Rottner K (2014) Requirements for and consequences of Rac-dependent protrusion. Eur J Cell Biol 93(5–6):184–193. https://doi.org/10.1016/j.ejcb.2014.01.008

    Article  CAS  PubMed  Google Scholar 

  39. Avnur Z, Geiger B (1981) The removal of extracellular fibronectin from areas of cell-substrate contact. Cell 25(1):121–132

    Article  CAS  PubMed  Google Scholar 

  40. Cailleteau L, Estrach S, Thyss R, Boyer L, Doye A, Domange B, Johnsson N, Rubinstein E, Boucheix C, Ebrahimian T, Silvestre JS, Lemichez E, Meneguzzi G, Mettouchi A (2010) alpha2beta1 integrin controls association of Rac with the membrane and triggers quiescence of endothelial cells. J Cell Sci 123(Pt 14):2491–2501. https://doi.org/10.1242/jcs.058875

    Article  CAS  PubMed  Google Scholar 

  41. Mettouchi A, Klein S, Guo W, Lopez-Lago M, Lemichez E, Westwick JK, Giancotti FG (2001) Integrin-specific activation of Rac controls progression through the G(1) phase of the cell cycle. Mol Cell 8(1):115–127

    Article  CAS  PubMed  Google Scholar 

  42. Price LS, Leng J, Schwartz MA, Bokoch GM (1998) Activation of Rac and Cdc42 by integrins mediates cell spreading. Mol Biol Cell 9(7):1863–1871

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Rottner K, Kaverina IN, Stradal TEB (2006) Cytoskeleton proteins. In: Celis JE (ed) Cell biology, a laboratory handbook, vol. 3, 3rd edn. Elsevier, London, pp 111–119

    Google Scholar 

  44. Doyon JB, Zeitler B, Cheng J, Cheng AT, Cherone JM, Santiago Y, Lee AH, Vo TD, Doyon Y, Miller JC, Paschon DE, Zhang L, Rebar EJ, Gregory PD, Urnov FD, Drubin DG (2011) Rapid and efficient clathrin-mediated endocytosis revealed in genome-edited mammalian cells. Nat Cell Biol 13(3):331–337. https://doi.org/10.1038/ncb2175

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hille F, Charpentier E (2016) CRISPR-Cas: biology, mechanisms and relevance. Philos Trans R Soc Lond Ser B Biol Sci 371(1707). https://doi.org/10.1098/rstb.2015.0496

  46. Swedlow JR, Porter IM, Posch M, Swift S (2010) In vivo imaging of mammalian cells. In: Goldman RD, Swedlow JR, Spector DL (eds) Live cell imaging: a laboratory manual, 2nd edn. Cold Spring Harbor Laboratory Press, New York, NY, pp 317–332

    Google Scholar 

  47. Poulter NS, Pitkeathly WT, Smith PJ, Rappoport JZ (2015) The physical basis of total internal reflection fluorescence (TIRF) microscopy and its cellular applications. Methods Mol Biol 1251:1–23. https://doi.org/10.1007/978-1-4939-2080-8_1

    Article  CAS  PubMed  Google Scholar 

  48. Geese M, Schluter K, Rothkegel M, Jockusch BM, Wehland J, Sechi AS (2000) Accumulation of profilin II at the surface of Listeria is concomitant with the onset of motility and correlates with bacterial speed. J Cell Sci 113(Pt 8):1415–1426

    Article  CAS  PubMed  Google Scholar 

  49. Nejedla M, Li Z, Masser AE, Biancospino M, Spiess M, Mackowiak SD, Friedlander MR, Karlsson R (2017) A fluorophore fusion construct of human profilin I with non-compromised poly(L-proline) binding capacity suitable for imaging. J Mol Biol 429(7):964–976. https://doi.org/10.1016/j.jmb.2017.01.004

    Article  CAS  PubMed  Google Scholar 

  50. Okreglak V, Drubin DG (2007) Cofilin recruitment and function during actin-mediated endocytosis dictated by actin nucleotide state. J Cell Biol 178(7):1251–1264. https://doi.org/10.1083/jcb.200703092

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sakurai-Yageta M, Recchi C, Le Dez G, Sibarita JB, Daviet L, Camonis J, D’Souza-Schorey C, Chavrier P (2008) The interaction of IQGAP1 with the exocyst complex is required for tumor cell invasion downstream of Cdc42 and RhoA. J Cell Biol 181(6):985–998. https://doi.org/10.1083/jcb.200709076

    Article  PubMed  PubMed Central  Google Scholar 

  52. Steffen A, Le Dez G, Poincloux R, Recchi C, Nassoy P, Rottner K, Galli T, Chavrier P (2008) MT1-MMP-dependent invasion is regulated by TI-VAMP/VAMP7. Curr Biol 18(12):926–931. https://doi.org/10.1016/j.cub.2008.05.044

    Article  CAS  PubMed  Google Scholar 

  53. Rottner K, Krause M, Gimona M, Small JV, Wehland J (2001) Zyxin is not colocalized with vasodilator-stimulated phosphoprotein (VASP) at lamellipodial tips and exhibits different dynamics to vinculin, paxillin, and VASP in focal adhesions. Mol Biol Cell 12(10):3103–3113

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Dimchev G, Steffen A, Kage F, Dimchev V, Pernier J, Carlier MF, Rottner K (2017) Efficiency of lamellipodia protrusion is determined by the extent of cytosolic actin assembly. Mol Biol Cell 28(10):1311–1325. https://doi.org/10.1091/mbc.E16-05-0334

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Schafer DA, Welch MD, Machesky LM, Bridgman PC, Meyer SM, Cooper JA (1998) Visualization and molecular analysis of actin assembly in living cells. J Cell Biol 143(7):1919–1930

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Derivery E, Sousa C, Gautier JJ, Lombard B, Loew D, Gautreau A (2009) The Arp2/3 activator WASH controls the fission of endosomes through a large multiprotein complex. Dev Cell 17(5):712–723. https://doi.org/10.1016/j.devcel.2009.09.010

    Article  CAS  PubMed  Google Scholar 

  57. Gomez TS, Billadeau DD (2009) A FAM21-containing WASH complex regulates retromer-dependent sorting. Dev Cell 17(5):699–711. https://doi.org/10.1016/j.devcel.2009.09.009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Rottner K, Hanisch J, Campellone KG (2010) WASH, WHAMM and JMY: regulation of Arp2/3 complex and beyond. Trends Cell Biol 20(11):650–661. https://doi.org/10.1016/j.tcb.2010.08.014

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported in part by the German Research Foundation (DFG) and by the Helmholtz Centre for Infection Research (HZI, Braunschweig, Germany). We would like to thank Dr. Yu-Li Wang (Pittsburgh, USA) for insightful discussions on fluorescence imaging.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Klemens Rottner .

Editor information

Editors and Affiliations

1 Electronic Supplementary Materials

Movie 1

EGFP-VASP accumulates in protruding lamellipodia. Time-lapse epifluorescence and phase-contrast microscopy reveals VASP to be enriched at the lamellipodial front as long as the edge protrudes forward, while it disappears from the focal plane in local phases of upward and rearward ruffling and/or retraction. EGFP-VASP also accumulates in protruding filopodia and focal adhesions. Time is displayed in minutes and seconds (AVI 2021 kb)

Movie 2

Filopodia can emerge from ruffling lamellipodia. Time-lapse fluorescence and phase-contrast microscopy of B16-F1 cell transfected with EGFP-VASP, which is enriched at lamellipodia rims, filopodia tips, and focal adhesions. Upper panel shows an overview of the cell, grey box in phase-contrast movie marks the zoom region that is shown in higher magnification below. Note that when the lamellipodium starts to ruffle, the VASP signal transiently vanishes from the front because of leaving the focal plane, but comes back as soon as the rearward folded lamellipodium returns into the focal plane. At this point, the ruffling edge of the rearward-folded lamellipodium develops and protrudes a filopodium in rearward direction of migration. Simultaneously, a new lamellipodium protrudes from the front, which accumulates EGFP-VASP signal, and the cycle of protrusion and ruffling and/or retraction starts from the beginning. All these activities highlight the complexity of the dynamics of molecules operating in actin filament regulation in cell edge protrusions. Time is shown in minutes and seconds (AVI 1608 kb)

Movie 3

FMNL2-EGFP localizes to the tip regions of lamellipodia and filopodia. Time-lapse fluorescence and phase-contrast microscopy of B16-F1 cell migrating on laminin and transiently transfected with FMNL2 tagged with EGFP at its C-terminus (FMNL2-EGFP). Note that formin accumulation is restricted to protruding lamellipodia or filopodia. It is absent from retracting regions of the cell. Time is given in minutes and seconds (AVI 3037 kb)

Movie 4

Full length EGFP-FMNL2 displays cytosolic localization. B16-F1 cell transiently transfected with EGFP-tagged full length FMNL2. Time-lapse epifluorescence imaging reveals a mostly cytosolic localization of the fusion protein due to inhibition of myristoylation by N-terminal EGFP-tagging [13]. Note the stark contrast with C-terminal tagging displayed in Movie 3. Time is given in minutes and seconds (AVI 635 kb)

Movie 5

EGFP-CP localizes to the lamellipodium and endosomes. The movie shows a NIH3T3 cell expressing EGFP-CP. Fluorescence (left) and phase-contrast (right) time-lapse microscopy of the whole cell (top panels) and details of the regions (bottom panels) as indicated at the top. The cell was co-transfected with myc-Rac1-L61, leading to constitutive treadmilling of lamellipodia at the periphery as well as at the ventral side. CP is localized to the lamellipodium, as well as ruffles and endosomes. It is difficult in these conditions to differentiate small, ventral ruffles from endosomes, except that those structures appearing at the periphery are likely ruffles since the fluorescent accumulation of CP is accompanied by fuzzy dark-grey structures in phase contrast, more likely corresponding to ruffles. Endosomes, instead, appear as white, grey or dark grey round vesicular structures (see top right panel). Note that co-expression of appropriate marker proteins would facilitate identifying structures like endosomes and ruffles, or distinguishing them from one another. Note that upon ruffling, CP seems to enrich at the extreme periphery; however, this can be accounted for by the transient, local thickening of the cell periphery, as revealed in corresponding phase-contrast time lapse images, and not in this case by a real restriction to the lamellipodium tip, as shown with other components such as Abi1 (Fig. 5 and Movie 6). Time is given in minutes and seconds (AVI 7317 kb)

Movie 6

EGFP-Abi1 exclusively accumulates at the tips of protruding lamellipodia. The movie shows a NIH3T3 cell expressing EGFP-Abi1. Fluorescence (left) and phase-contrast (right) time-lapse microscopy of the whole cell (top panels) and details of the regions as indicated at the top in bottom panels. The cell was co-transfected with myc-Rac1-L61, leading to constitutive treadmilling of lamellipodia at the periphery as well as at the ventral side. Note that EGFP-Abi1 is localized at the extreme tip of the periphery. As shown in the details at the bottom, the lamellipodium is frequently lifting up, which can be appreciated by a fuzzy periphery in phase contrast. The EGFP-Abi1 signal is disappearing correspondingly, due to out of focus localization. Protrusive ventral lamellipodia can be seen in both fluorescence as well as phase-contrast channels. Time is given in minutes and seconds (AVI 6078 kb)

Movie 7

Control NIH3T3 cell expressing EGFP. Fluorescence (left) and phase-contrast (right) time-lapse microscopy of the whole cell (top panel) and details of the regions as indicated (bottom panel) are shown. Time is given in minutes and seconds. Note that when the periphery of the cell is ruffling, the cytosolic EGFP signal seems to correspond to a distinct “localization” at the periphery, however, this is just due to the increased thickness of the cell as illustrated also by phase-contrast microscopy. In images using the latter, thicker and/or denser regions of the cell appear darker, whereas very dense material appears as dark spots, and fatty or liquid-containing vesicles such as macropinosomes constitute white structures (AVI 4714 kb)

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer Science+Business Media, LLC

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Steffen, A., Kage, F., Rottner, K. (2018). Imaging the Molecular Machines That Power Cell Migration. In: Gautreau, A. (eds) Cell Migration. Methods in Molecular Biology, vol 1749. Humana, New York, NY. https://doi.org/10.1007/978-1-4939-7701-7_19

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-7701-7_19

  • Published:

  • Publisher Name: Humana, New York, NY

  • Print ISBN: 978-1-4939-7700-0

  • Online ISBN: 978-1-4939-7701-7

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics