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.

  • Review Article
  • Published:

Actin-dependent movement of bacterial pathogens

Key Points

  • Bacteria of the genera Listeria, Rickettsia, Burkholderia, Shigella and Mycobacteria subvert cellular actin dynamics to facilitate their movement within the host cytosol and to infect neighbouring cells while evading host immune surveillance and promoting their intracellular survival. Attaching and effacing (AE) Escherichia coli do not enter host cells but attach intimately to the cell surface, inducing motile actin-rich pedestals, the function of which is unclear.

  • The central role of actin-based motility in bacterial virulence is reflected in the marked attenuation of non-motile mutants of Listeria monocytogenes, Shigella flexneri and Burkholderia pseudomallei. Bacterial factors that control actin assembly are often crucial determinants of virulence and attractive targets for intervention.

  • Analysis of bacterial actin-based motility has revealed novel insights into both pathogen biology and the control of actin assembly in eukaryotic cells. It provides convenient models to study the formation of lamellipodia at the leading edge of motile cells (Listeria and Shigella) and filopodia (Rickettsia).

  • Many of the bacterial factors that are necessary for intracellular motility are expressed at the pole of the bacterium from which the actin-rich tail forms (Listeria ActA, Shigella IcsA and Burkholderia BimA) and are also expressed only to high levels within the host cytosol (Shigella IcsA and Mycobacterium motility factor). In some instances, expression of the bacterial factor has also been shown to be growth-phase dependent (Shigella IcsA).

  • The strategies used by intracellular bacterial pathogens to induce polar actin polymerization, and by AE E. coli to elicit pedestal formation at the plasma membrane, converge on their ability to recruit and activate the cellular Arp2/3 complex. Pathogens that exhibit actin-based movement can be separated into two groups depending on whether they mimic a cellular nucleation-promoting factor (NPF) (Listeria ActA and Rickettsia RickA) or whether they recruit a cellular NPF to the bacterial surface to promote their Arp2/3-mediated motility (Shigella IcsA and AE E. coli Tir).

  • The availability of complete bacterial genome sequences has facilitated identification of new factors that activate actin assembly, including Rickettsia RickA and Burkholderia BimA, and families of homologues of these proteins are emerging in related species that might stimulate actin dynamics in distinct ways.

Abstract

Listeria, Rickettsia, Burkholderia, Shigella and Mycobacterium species subvert cellular actin dynamics to facilitate their movement within the host cytosol and to infect neighbouring cells while evading host immune surveillance and promoting their intracellular survival. 'Attaching and effacing' Escherichia coli do not enter host cells but attach intimately to the cell surface, inducing motile actin-rich pedestals, the function of which is currently unclear. The molecular basis of actin-based motility of these bacterial pathogens reveals novel insights about bacterial pathogenesis and fundamental host-cell pathways.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Interaction of bacterial pathogens with actin.
Figure 2: Mechanisms of intracellular bacterial actin polymerization.
Figure 3: Actin-based motility of Burkholderia pseudomallei
Figure 4: Mechanisms of actin pedestal formation by enteropathogenic Escherichia coli (EPEC) O127:H6 and enterohaemorrhagic E. coli (EHEC) O157:H7.

Similar content being viewed by others

References

  1. Monack, D. M. & Theriot, J. A. Actin-based motility is sufficient for bacterial membrane protrusion formation and host cell uptake. Cell. Microbiol. 3, 633–647 (2001).

    Article  CAS  PubMed  Google Scholar 

  2. Robbins, J. R. et al. Listeria monocytogenes exploits normal host cell processes to spread from cell to cell. J. Cell Biol. 146, 1333–1350 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Perrin, A. J., Jiang, X., Birmingham, C. L., So, N. S. & Brumell, J. H. Recognition of bacteria in the cytosol of mammalian cells by the ubiquitin system. Curr. Biol. 14, 806–811 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Domann, E. et al. A novel bacterial virulence gene in Listeria monocytogenes required for host cell microfilament interaction with homology to the proline-rich region of vinculin. EMBO J. 11, 1981–1990 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Makino, S., Sasakawa, C., Kamata, K., Kurata, T. & Yoshikawa, M. A genetic determinant required for continuous reinfection of adjacent cells on large plasmid in S. flexneri 2a. Cell 46, 551–555 (1986).

    Article  CAS  PubMed  Google Scholar 

  6. Bernardini, M. L., Mounier, J., d'Hauteville, H., Coquis-Rondon, M. & Sansonetti, P. J. Identification of icsA, a plasmid locus of Shigella flexneri that governs bacterial intra- and intercellular spread through interaction with F-actin. Proc. Natl Acad. Sci. USA 86, 3867–3871 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sansonetti, P. J., Arondel, J., Fontaine, A., d'Hauteville, H. & Bernardini, M. L. ompB (osmo-regulation) and icsA (cell-to-cell spread) mutants of Shigella flexneri: vaccine candidates and probes to study the pathogenesis of shigellosis. Vaccine 9, 416–422 (1991).

    Article  CAS  PubMed  Google Scholar 

  8. Gouin, E., Welch, M. D. & Cossart, P. Actin-based motility of intracellular pathogens. Curr. Opin. Microbiol. 8, 35–45 (2005).

    Article  CAS  PubMed  Google Scholar 

  9. Kocks, C., Hellio, R., Gounon, P., Ohayon, H. & Cossart, P. Polarized distribution of Listeria monocytogenes surface protein ActA at the site of directional actin assembly. J. Cell Sci. 105, 699–710 (1993).

    Article  CAS  PubMed  Google Scholar 

  10. Goldberg, M. B. & Theriot, J. A. Shigella flexneri surface protein IcsA is sufficient to direct actin-based motility. Proc. Natl Acad. Sci. USA 92, 6572–6576 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Stevens, M. P. et al. Identification of a bacterial factor required for actin-based motility of Burkholderia pseudomallei. Mol. Microbiol. 56, 40–53 (2005). This paper reported the identification of the bacterial factor that is required for B. pseudomallei actin-based motility in host cells. Biochemical studies showed that the factor (BimA) interacts with host-cell actin and can stimulate actin polymerization in vitro in an Arp2/3-independent manner.

    Article  CAS  PubMed  Google Scholar 

  12. Brundage, R. A., Smith, G. A., Camilli, A., Theriot, J. A. & Portnoy, D. A. Expression and phosphorylation of the Listeria monocytogenes ActA protein in mammalian cells. Proc. Natl Acad. Sci. USA 90, 11890–11894 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Magdalena, J. & Goldberg, M. B. Quantification of Shigella IcsA required for bacterial actin polymerization. Cell Motil. Cytoskeleton 51, 187–196 (2002).

    Article  CAS  PubMed  Google Scholar 

  14. Stamm, L. M. et al. Mycobacterium marinum escapes from phagosomes and is propelled by actin-based motility. J. Exp. Med. 198, 1361–1368 (2003). The first study to show that M. marinum escapes from intracellular phagosomes and also spreads from cell to cell. Although the identity of the bacterial actin-based motility factor is still unknown, this study identified host-cell proteins that might be involved in motility by co-localization studies.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Goldberg, M. B., Theriot, J. A. & Sansonetti, P. J. Regulation of surface presentation of IcsA, a Shigella protein essential to intracellular movement and spread, is growth phase dependent. Infect. Immun. 62, 5664–5668 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Welch, M. D., Iwamatsu, A. & Mitchison, T. J. Actin polymerization is induced by Arp2/3 protein complex at the surface of Listeria monocytogenes. Nature 385, 265–269 (1997). Describes the initial purification of the human Arp2/3 complex and its biochemical characterization as an actin-polymerizing factor in L. monocytogenes.

    Article  CAS  PubMed  Google Scholar 

  17. Welch, M. D., Rosenblatt, J., Skoble, J., Portnoy, D. A. & Mitchison, T. J. Interaction of human Arp2/3 complex and the Listeria monocytogenes ActA protein in actin filament nucleation. Science 281, 105–108 (1998).

    Article  CAS  PubMed  Google Scholar 

  18. Kocks, C. et al. L. monocytogenes-induced actin assembly requires the actA gene product, a surface protein. Cell 68, 521–531 (1992).

    Article  CAS  PubMed  Google Scholar 

  19. Mengaud, J., Geoffroy, C. & Cossart, P. Identification of a new operon involved in Listeria monocytogenes virulence: its first gene encodes a protein homologous to bacterial metalloproteases. Infect. Immun. 59, 1043–1049 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Vazquez-Boland, J. A. et al. Nucleotide sequence of the lecithinase operon of Listeria monocytogenes and possible role of lecithinase in cell-to-cell spread. Infect. Immun. 60, 219–230 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kocks, C. et al. The unrelated surface proteins ActA of Listeria monocytogenes and IcsA of Shigella flexneri are sufficient to confer actin-based motility on Listeria innocua and Escherichia coli respectively. Mol. Microbiol. 18, 413–423 (1995).

    Article  CAS  PubMed  Google Scholar 

  22. Smith, G. A., Portnoy, D. A. & Theriot, J. A. Asymmetric distribution of the Listeria monocytogenes ActA protein is required and sufficient to direct actin-based motility. Mol. Microbiol. 17, 945–951 (1995).

    Article  CAS  PubMed  Google Scholar 

  23. Cameron, L. A., Footer, M. J., van Oudenaarden, A. & Theriot, J. A. Motility of ActA protein-coated microspheres driven by actin polymerization. Proc. Natl Acad. Sci. USA 96, 4908–4913 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Boujemaa-Paterski, R. et al. Listeria protein ActA mimics WASP family proteins: it activates filament barbed end branching by Arp2/3 complex. Biochemistry 40, 11390–11404 (2001).

    Article  CAS  PubMed  Google Scholar 

  25. Loisel, T. P., Boujemaa, R., Pantaloni, D. & Carlier, M. F. Reconstitution of actin-based motility of Listeria and Shigella using pure proteins. Nature 401, 613–616 (1999). This study characterized the minimal requirements for motility of both L. monocytogenes and E. coli IcsA in a cell-free system.

    Article  CAS  PubMed  Google Scholar 

  26. Zalevsky, J., Grigorova, I. & Mullins, R. D. Activation of the Arp2/3 complex by the Listeria ActA protein. ActA binds two actin monomers and three subunits of the Arp2/3 complex. J. Biol. Chem. 276, 3468–3475 (2001).

    Article  CAS  PubMed  Google Scholar 

  27. Lommel, S. et al. Actin pedestal formation by enteropathogenic Escherichia coli and intracellular motility of Shigella flexneri are abolished in N-WASP-defective cells. EMBO Rep. 2, 850–857 (2001). This study used N-WASP-deficient cells to prove the essential role of N-WASP in the actin cytoskeletal rearrangements induced by AE E. coli and S. flexneri . It also showed for the first time that N-WASP is dispensable for cellular filopodia formation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lasa, I., David, V., Gouin, E., Marchand, J. B. & Cossart, P. The amino-terminal part of ActA is critical for the actin-based motility of Listeria monocytogenes; the central proline-rich region acts as a stimulator. Mol. Microbiol. 18, 425–436 (1995).

    Article  CAS  PubMed  Google Scholar 

  29. Lasa, I. et al. Identification of two regions in the N-terminal domain of ActA involved in the actin comet tail formation by Listeria monocytogenes. EMBO J. 16, 1531–1540 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Smith, G. A., Theriot, J. A. & Portnoy, D. A. The tandem repeat domain in the Listeria monocytogenes ActA protein controls the rate of actin-based motility, the percentage of moving bacteria, and the localization of vasodilator-stimulated phosphoprotein and profilin. J. Cell Biol. 135, 647–660 (1996).

    Article  CAS  PubMed  Google Scholar 

  31. Golsteyn, R. M., Beckerle, M. C., Koay, T. & Friederich, E. Structural and functional similarities between the human cytoskeletal protein zyxin and the ActA protein of Listeria monocytogenes. J. Cell Sci. 110, 1893–1906 (1997).

    Article  CAS  PubMed  Google Scholar 

  32. Fradelizi, J. et al. ActA and human zyxin harbour Arp2/3-independent actin-polymerization activity. Nature Cell Biol. 3, 699–707 (2001).

    Article  CAS  PubMed  Google Scholar 

  33. Brieher, W. M., Coughlin, M. & Mitchison, T. J. Fascin-mediated propulsion of Listeria monocytogenes independent of frequent nucleation by the Arp2/3 complex. J. Cell Biol. 165, 233–242 (2004). This report showed that L. monocytogenes actin-based motility in cell-free systems can be divided into an Arp2/3-dependent and an Arp2/3-independent phase. The Arp2/3-independent phase (the elongation phase) required the addition of cellular fascin and resulted in tails that consist of parallel bundles similar to those formed by Rickettsia.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Gouin, E. et al. A comparative study of the actin-based motilities of the pathogenic bacteria Listeria monocytogenes, Shigella flexneri and Rickettsia conorii. J. Cell Sci. 112, 1697–1708 (1999).

    Article  CAS  PubMed  Google Scholar 

  35. Heinzen, R. A., Grieshaber, S. S., Van Kirk, L. S. & Devin, C. J. Dynamics of actin-based movement by Rickettsia rickettsii in Vero cells. Infect. Immun. 67, 4201–4207 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Heinzen, R. A., Hayes, S. F., Peacock, M. G. & Hackstadt, T. Directional actin polymerization associated with spotted fever group Rickettsia infection of Vero cells. Infect. Immun. 61, 1926–1935 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Teysseire, N., Chiche-Portiche, C. & Raoult, D. Intracellular movements of Rickettsia conorii and R. typhi based on actin polymerization. Res. Microbiol. 143, 821–829 (1992).

    Article  CAS  PubMed  Google Scholar 

  38. Gouin, E. et al. The RickA protein of Rickettsia conorii activates the Arp2/3 complex. Nature 427, 457–461 (2004). One of two papers published in 2004 reporting the identity of the Rickettsia actin-based motility factor. Characterized the R. conorii RickA protein and demonstrated its expression on intracellular bacteria and its activity in vitro as an NPF. Also reported the conclusive co-localization of Arp2/3 with Rickettsia tails.

    Article  CAS  PubMed  Google Scholar 

  39. Jeng, R. L. et al. A Rickettsia WASP-like protein activates the Arp2/3 complex and mediates actin-based motility. Cell. Microbiol. 6, 761–769 (2004). One of two papers published in 2004 reporting the identity of the Rickettsia actin-based motility factor. This concentrated on characterization of the R. rickettsii RickA protein, describing its similarity in sequence to WASP-family proteins and its ability to promote actin nucleation in vitro.

    Article  CAS  PubMed  Google Scholar 

  40. Simser, J. A., Rahman, M. S., Dreher-Lesnick, S. M. & Azad, A. F. A novel and naturally occurring transposon, ISRpe1 in the Rickettsia peacockii genome disrupting the rickA gene involved in actin-based motility. Mol. Microbiol. 58, 71–79 (2005).

    Article  CAS  PubMed  Google Scholar 

  41. Van Kirk, L. S., Hayes, S. F. & Heinzen, R. A. Ultrastructure of Rickettsia rickettsii actin tails and localization of cytoskeletal proteins. Infect. Immun. 68, 4706–4713 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Harlander, R. S. et al. Effects of ectopically expressed neuronal Wiskott–Aldrich syndrome protein domains on Rickettsia rickettsii actin-based motility. Infect. Immun. 71, 1551–1556 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bear, J. E. et al. Antagonism between Ena/VASP proteins and actin filament capping regulates fibroblast motility. Cell 109, 509–521 (2002).

    Article  CAS  PubMed  Google Scholar 

  44. Friederich, E. et al. Targeting of Listeria monocytogenes ActA protein to the plasma membrane as a tool to dissect both actin-based cell morphogenesis and ActA function. EMBO J. 14, 2731–2744 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Theriot, J. A., Mitchison, T. J., Tilney, L. G. & Portnoy, D. A. The rate of actin-based motility of intracellular Listeria monocytogenes equals the rate of actin polymerization. Nature 357, 257–260 (1992).

    Article  CAS  PubMed  Google Scholar 

  46. McLeod, M. P. et al. Complete genome sequence of Rickettsia typhi and comparison with sequences of other rickettsiae. J. Bacteriol. 186, 5842–5855 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Stevens, M. P. & Galyov, E. E. Exploitation of host cells by Burkholderia pseudomallei. Int. J. Med. Microbiol. 293, 549–555 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Kespichayawattana, W., Rattanachetkul, S., Wanun, T., Utaisincharoen, P. & Sirisinha, S. Burkholderia pseudomallei induces cell fusion and actin-associated membrane protrusion: a possible mechanism for cell-to-cell spreading. Infect. Immun. 68, 5377–5384 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Breitbach, K. et al. Actin-based motility of Burkholderia pseudomallei involves the Arp 2/3 complex, but not N-WASP and Ena/VASP proteins. Cell. Microbiol. 5, 385–393 (2003). A comprehensive study of the cellular factors recruited to B. pseudomallei actin-rich tails in infected cells. Notably, this study reported that Arp2/3 might be required for motility but that N-WASP and members of the Ena/VASP family are dispensable.

    Article  CAS  PubMed  Google Scholar 

  50. Nierman, W. C. et al. Structural flexibility in the Burkholderia mallei genome. Proc. Natl Acad. Sci. USA 101, 14246–14251 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Stevens, J. M. et al. Actin-binding proteins from Burkholderia mallei and B. thailandensis can functionally compensate for the actin-based motility defect of a B. pseudomallei bimA mutant. J. Bacteriol. 187, 7857–7862 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Sansonetti, P. J., Ryter, A., Clerc, P., Maurelli, A. T. & Mounier, J. Multiplication of Shigella flexneri within HeLa cells: lysis of the phagocytic vacuole and plasmid-mediated contact hemolysis. Infect. Immun. 51, 461–469 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Pal, T., Newland, J. W., Tall, B. D., Formal, S. B. & Hale, T. L. Intracellular spread of Shigella flexneri associated with the kcpA locus and a 140-kilodalton protein. Infect. Immun. 57, 477–486 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Allaoui, A., Mounier, J., Prevost, M. C., Sansonetti, P. J. & Parsot, C. icsB: a Shigella flexneri virulence gene necessary for the lysis of protrusions during intercellular spread. Mol. Microbiol. 6, 1605–1616 (1992).

    Article  CAS  PubMed  Google Scholar 

  55. Kadurugamuwa, J. L., Rohde, M., Wehland, J. & Timmis, K. N. Intercellular spread of Shigella flexneri through a monolayer mediated by membranous protrusions and associated with reorganization of the cytoskeletal protein vinculin. Infect. Immun. 59, 3463–3471 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Prevost, M. C. et al. Unipolar reorganization of F-actin layer at bacterial division and bundling of actin filaments by plastin correlate with movement of Shigella flexneri within HeLa cells. Infect. Immun. 60, 4088–4099 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sansonetti, P. J., Mounier, J., Prevost, M. C. & Mege, R. M. Cadherin expression is required for the spread of Shigella flexneri between epithelial cells. Cell 76, 829–839 (1994).

    Article  CAS  PubMed  Google Scholar 

  58. Lett, M. C. et al. virG, a plasmid-coded virulence gene of Shigella flexneri: identification of the VirG protein and determination of the complete coding sequence. J. Bacteriol. 171, 353–359 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Desvaux, M., Parham, N. J. & Henderson, I. R. The autotransporter secretion system. Res. Microbiol. 155, 53–60 (2004).

    Article  CAS  PubMed  Google Scholar 

  60. Egile, C. et al. Activation of the CDC42 effector N-WASP by the Shigella flexneri IcsA protein promotes actin nucleation by Arp2/3 complex and bacterial actin-based motility. J. Cell Biol. 146, 1319–1332 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Suzuki, T., Miki, H., Takenawa, T. & Sasakawa, C. Neural Wiskott–Aldrich syndrome protein is implicated in the actin-based motility of Shigella flexneri. EMBO J. 17, 2767–2776 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Suzuki, T. et al. Neural Wiskott–Aldrich syndrome protein (N-WASP) is the specific ligand for Shigella VirG among the WASP family and determines the host cell type allowing actin-based spreading. Cell. Microbiol. 4, 223–233 (2002).

    Article  CAS  PubMed  Google Scholar 

  63. Moreau, V. et al. A complex of N-WASP and WIP integrates signalling cascades that lead to actin polymerization. Nature Cell Biol. 2, 441–448 (2000).

    Article  CAS  PubMed  Google Scholar 

  64. Suzuki, T. et al. Rho family GTPase Cdc42 is essential for the actin-based motility of Shigella in mammalian cells. J. Exp. Med. 191, 1905–1920 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Mounier, J. et al. Rho family GTPases control entry of Shigella flexneri into epithelial cells but not intracellular motility. J. Cell Sci. 112, 2069–2080 (1999).

    Article  CAS  PubMed  Google Scholar 

  66. Burton, E. A., Oliver, T. N. & Pendergast, A. M. Abl kinases regulate actin comet tail elongation via an N-WASP-dependent pathway. Mol. Cell. Biol. 25, 8834–8843 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Stamm, L. M. et al. Role of the WASP family proteins for Mycobacterium marinum actin tail formation. Proc. Natl Acad. Sci. USA 102, 14837–14842 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Gertler, F. B., Niebuhr, K., Reinhard, M., Wehland, J. & Soriano, P. Mena, a relative of VASP and Drosophila Enabled, is implicated in the control of microfilament dynamics. Cell 87, 227–239 (1996).

    Article  CAS  PubMed  Google Scholar 

  69. Kaper, J. B., Nataro, J. P. & Mobley, H. L. Pathogenic Escherichia coli. Nature Rev. Microbiol. 2, 123–140 (2004).

    Article  CAS  Google Scholar 

  70. Sanger, J. M., Chang, R., Ashton, F., Kaper, J. B. & Sanger, J. W. Novel form of actin-based motility transports bacteria on the surfaces of infected cells. Cell Motil. Cytoskeleton 34, 279–287 (1996).

    Article  CAS  PubMed  Google Scholar 

  71. Shaner, N. C., Sanger, J. W. & Sanger, J. M. Actin and α-actinin dynamics in the adhesion and motility of EPEC and EHEC on host cells. Cell Motil. Cytoskeleton 60, 104–120 (2005).

    Article  CAS  PubMed  Google Scholar 

  72. Kenny, B. et al. Enteropathogenic E. coli (EPEC) transfers its receptor for intimate adherence into mammalian cells. Cell 91, 511–520 (1997). The first demonstration of a bacterium that translocates its own receptor into the eukaryotic host cell. The translocated receptor (Tir) was also shown to be required for actin nucleation at the plasma membrane.

    Article  CAS  PubMed  Google Scholar 

  73. Campellone, K. G. & Leong, J. M. Nck-independent actin assembly is mediated by two phosphorylated tyrosines within enteropathogenic Escherichia coli Tir. Mol. Microbiol. 56, 416–432 (2005).

    Article  CAS  PubMed  Google Scholar 

  74. Liu, H., Magoun, L., Luperchio, S., Schauer, D. B. & Leong, J. M. The Tir-binding region of enterohaemorrhagic Escherichia coli intimin is sufficient to trigger actin condensation after bacterial-induced host cell signalling. Mol. Microbiol. 34, 67–81 (1999).

    Article  PubMed  Google Scholar 

  75. Kenny, B. Phosphorylation of tyrosine 474 of the enteropathogenic Escherichia coli (EPEC) Tir receptor molecule is essential for actin nucleating activity and is preceded by additional host modifications. Mol. Microbiol. 31, 1229–1241 (1999).

    Article  CAS  PubMed  Google Scholar 

  76. Deng, W., Vallance, B. A., Li, Y., Puente, J. L. & Finlay, B. B. Citrobacter rodentium translocated intimin receptor (Tir) is an essential virulence factor needed for actin condensation, intestinal colonization and colonic hyperplasia in mice. Mol. Microbiol. 48, 95–115 (2003).

    Article  CAS  PubMed  Google Scholar 

  77. Campellone, K. G., Giese, A., Tipper, D. J. & Leong, J. M. A tyrosine-phosphorylated 12-amino-acid sequence of enteropathogenic Escherichia coli Tir binds the host adaptor protein Nck and is required for Nck localization to actin pedestals. Mol. Microbiol. 43, 1227–1241 (2002).

    Article  CAS  PubMed  Google Scholar 

  78. Campellone, K. G. et al. Clustering of Nck by a 12-residue Tir phosphopeptide is sufficient to trigger localized actin assembly. J. Cell Biol. 164, 407–416 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Gruenheid, S. et al. Enteropathogenic E. coli Tir binds Nck to initiate actin pedestal formation in host cells. Nature Cell Biol. 3, 856–859 (2001).

    Article  CAS  PubMed  Google Scholar 

  80. Goosney, D. L., DeVinney, R. & Finlay, B. B. Recruitment of cytoskeletal and signaling proteins to enteropathogenic and enterohemorrhagic Escherichia coli pedestals. Infect. Immun. 69, 3315–3322 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Frischknecht, F. et al. Actin-based motility of vaccinia virus mimics receptor tyrosine kinase signalling. Nature 401, 926–929 (1999).

    Article  CAS  PubMed  Google Scholar 

  82. Scaplehorn, N. et al. Grb2 and Nck act cooperatively to promote actin-based motility of vaccinia virus. Curr. Biol. 12, 740–745 (2002).

    Article  CAS  PubMed  Google Scholar 

  83. Campellone, K. G. & Leong, J. M. Tails of two Tirs: actin pedestal formation by enteropathogenic E. coli and enterohemorrhagic E. coli O157:H7. Curr. Opin. Microbiol. 6, 82–90 (2003).

    Article  CAS  PubMed  Google Scholar 

  84. DeVinney, R., Puente, J. L., Gauthier, A., Goosney, D. & Finlay, B. B. Enterohaemorrhagic and enteropathogenic Escherichia coli use a different Tir-based mechanism for pedestal formation. Mol. Microbiol. 41, 1445–1458 (2001).

    Article  CAS  PubMed  Google Scholar 

  85. DeVinney, R. et al. Enterohemorrhagic Escherichia coli O157:H7 produces Tir, which is translocated to the host cell membrane but is not tyrosine phosphorylated. Infect. Immun. 67, 2389–2398 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Paton, A. W., Manning, P. A., Woodrow, M. C. & Paton, J. C. Translocated intimin receptors (Tir) of Shiga-toxigenic Escherichia coli isolates belonging to serogroups O26, O111, and O157 react with sera from patients with hemolytic–uremic syndrome and exhibit marked sequence heterogeneity. Infect. Immun. 66, 5580–5586 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Campellone, K. G., Robbins, D. & Leong, J. M. EspFU is a translocated EHEC effector that interacts with Tir and N-WASP and promotes Nck-independent actin assembly. Dev. Cell 7, 217–228 (2004).

    Article  CAS  PubMed  Google Scholar 

  88. Garmendia, J. et al. TccP is an enterohaemorrhagic Escherichia coli O157:H7 type III effector protein that couples Tir to the actin-cytoskeleton. Cell. Microbiol. 6, 1167–1183 (2004).

    Article  CAS  PubMed  Google Scholar 

  89. Kenny, B. The enterohaemorrhagic Escherichia coli (serotype O157:H7) Tir molecule is not functionally interchangeable for its enteropathogenic E. coli (serotype O127:H6) homologue. Cell. Microbiol. 3, 499–510 (2001).

    Article  CAS  PubMed  Google Scholar 

  90. Vlisidou, I. et al. Role of intimin-Tir interactions and the Tir-cytoskeleton coupling protein (TccP) in the colonization of calves and lambs by Escherichia coli. Infect. Immun. 74, 758–764 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Paavilainen, V. O., Bertling, E., Falck, S. & Lappalainen, P. Regulation of cytoskeletal dynamics by actin-monomer-binding proteins. Trends Cell Biol. 14, 386–394 (2004).

    Article  CAS  PubMed  Google Scholar 

  92. Zigmond, S. H. Formin-induced nucleation of actin filaments. Curr. Opin. Cell Biol. 16, 99–105 (2004).

    Article  CAS  PubMed  Google Scholar 

  93. Quinlan, M. E., Heuser, J. E., Kerkhoff, E. & Mullins, R. D. Drosophila Spire is an actin nucleation factor. Nature 433, 382–388 (2005).

    Article  CAS  PubMed  Google Scholar 

  94. Welch, M. D., DePace, A. H., Verma, S., Iwamatsu, A. & Mitchison, T. J. The human Arp2/3 complex is composed of evolutionarily conserved subunits and is localized to cellular regions of dynamic actin filament assembly. J. Cell Biol. 138, 375–384 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Rohatgi, R., Nollau, P., Ho, H. Y., Kirschner, M. W. & Mayer, B. J. Nck and phosphatidylinositol 4,5-bisphosphate synergistically activate actin polymerization through the N-WASP-Arp2/3 pathway. J. Biol. Chem. 276, 26448–26452 (2001).

    Article  CAS  PubMed  Google Scholar 

  96. Robinson, R. C. et al. Crystal structure of Arp2/3 complex. Science 294, 1679–1684 (2001).

    Article  CAS  PubMed  Google Scholar 

  97. Rottner, K., Lommel, S., Wehland, J. & Stradal, T. E. Pathogen-induced actin filament rearrangement in infectious diseases. J. Pathol. 204, 396–406 (2004).

    Article  CAS  PubMed  Google Scholar 

  98. Chakraborty, T. et al. A focal adhesion factor directly linking intracellularly motile Listeria monocytogenes and Listeria ivanovii to the actin-based cytoskeleton of mammalian cells. EMBO J. 14, 1314–1321 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Gouin, E., Dehoux, P., Mengaud, J., Kocks, C. & Cossart, P. iactA of Listeria ivanovii, although distantly related to Listeria monocytogenes actA, restores actin tail formation in an L. monocytogenes actA mutant. Infect. Immun. 63, 2729–2737 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors gratefully acknowledge the support of the Biotechnology and Biological Sciences Research Council and the Department for the Environment, Food and Rural Affairs.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mark P. Stevens.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Related links

Related links

DATABASES

Entrez Genome Project

Burkholderia pseudomallei

Citrobacter rodentium

EHEC O157:H7

Listeria innocua

Listeria monocytogenes

Mycobacterium marinum

Rickettsia conorii

Rickettsia prowazekii

Rickettsia rickettsii

Rickettsia sibirica

Rickettsia typhi

Shigella boydii

Shigella dysenteriae

Shigella flexneri

Streptococcus pneumoniae

FURTHER INFORMATION

The Institute for Animal Health

Glossary

Lamellipodia

Sheet-like protrusions of the plasma membrane associated with exploratory cell movements and cell motility. Composed of orthogonal, highly crosslinked actin arrays. The Rho GTPase Rac has a crucial role in lamellipodia formation through activation of NPFs.

Filopodia

Protrusions of the plasma membrane with finger-like morphology reaching 200 μm in length. Composed of dynamic bundles of actin filaments. Associated with rapid membrane remodelling, transient matrix adhesions and cell motility. Cdc42 has a crucial role in initiating filopodia formation.

Facultative intracellular pathogen

A pathogen that can replicate either within the host cell or as a cell-free unit.

Wiskott–Aldrich syndrome protein

(WASP). The prototypic member of a family of proteins that stimulate the low intrinsic nucleation activity of the Arp2/3 complex.

Src homology-3 (SH3) domain

A small module of 50 amino-acid residues found in proteins that interact with proline-rich motifs.

Focal adhesion sites

Adhesions by which cells attach to the underlying substrate. Many structural, cytoskeletal and signalling proteins are concentrated in these structures.

Stress fibres

Contractile filaments involved in the maintenance of cell shape that support the motile apparatus of the cell. At least one end is anchored by a focal adhesion site.

Central and acidic (CA) domain

Forms part of a larger domain called a WCA domain. This region of WASP-family proteins stimulates the Arp2/3 complex.

WASP homology-2 (WH2) domain

WASP homology-2 domain motifs are composed of approximately 35 amino acids and are conserved in cellular proteins that recruit actin monomers.

CAAX motif

A motif that is post-translationally modified by farnesylation at the cysteine residue and that results in localization to the inner leaflet of the plasma membrane.

Type III secretion system

A transmembrane-spanning 'needle complex' involved in the secretion of two classes of proteins, translocators and effectors.

Type V secreted protein

A protein translocated across the outer membrane of Gram-negative bacteria through a transmembrane pore formed by a self-encoded β-barrel structure.

Pleckstrin homology domain

These domains share little sequence conservation but all have a common fold involved in lipid binding and targeting of proteins to the plasma membrane.

CRIB domain

Cdc42/Rac interactive binding (CRIB) domains bind Cdc42 and other Rho-family GTPases.

WAVE

WASP-family verprolin homologous (WAVE)/Scar proteins are members of the WASP family of Arp2/3 activators that lack the domains required for Rho GTPase binding. As such, they are thought to be regulated in a different manner to other WASP-family members.

Adaptor proteins

Proteins involved in signalling cascades that lack any intrinsic enzymatic activities. They mediate protein–protein interactions to form diverse protein complexes.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Stevens, J., Galyov, E. & Stevens, M. Actin-dependent movement of bacterial pathogens. Nat Rev Microbiol 4, 91–101 (2006). https://doi.org/10.1038/nrmicro1320

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrmicro1320

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