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:

Antimicrobial mechanisms of phagocytes and bacterial evasion strategies

Key Points

  • The innate immune response constitutes the first line of defence against pathogenic microorganisms.

  • Phagocytes, such as macrophages and neutrophils, engulf microorganisms into a vacuole or phagosome that is gradually converted into an effective microbicidal organelle.

  • Such remodelling of the phagocytic vacuole, a process known as maturation, occurs by coordinated fission and fusion events involving multiple subcompartments of the endocytic pathway.

  • Phagosome maturation is directed by small GTPases, requires SNARE (soluble NSF-attachment protein receptor) proteins and involves extensive lipid remodelling of the vacuolar membrane.

  • A number of intracellular pathogens have evolved strategies to arrest or divert phagosome maturation, or can escape the confines of the phagocytic vacuole.

  • We discuss examples of bacterial effectors that are injected into the host cells and cause disruption of the maturation pathway, often by co-opting the host cell machinery, to generate a niche conducive to bacterial survival and replication.

Abstract

Professional phagocytes have a vast and sophisticated arsenal of microbicidal features. They are capable of ingesting and destroying invading organisms, and can present microbial antigens on their surface, eliciting acquired immune responses. To survive this hostile response, certain bacterial species have developed evasive strategies that often involve the secretion of effectors to co-opt the cellular machinery of the host. In this Review, we present an overview of the antimicrobial defences of the host cell, with emphasis on macrophages, for which phagocytosis has been studied most extensively. In addition, using Mycobacterium tuberculosis, Listeria monocytogenes, Legionella pneumophila and Coxiella burnetii as examples, we describe some of the evasive strategies used by bacteria.

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

Figure 1: Stages of phagosomal maturation.
Figure 2: The microbicidal arsenal of phagocytes versus the defensive mechanisms of the microorganism.
Figure 3: Strategies used by professional intracellular bacterial pathogens to modulate phagosome maturation.

Similar content being viewed by others

References

  1. Roitt, I. M. Essential Immunology (Blackwell Science, Oxford, 1994).

    Google Scholar 

  2. Ghazizadeh, S., Bolen, J. B. & Fleit, H. B. Physical and functional association of Src-related protein tyrosine kinases with FcγRII in monocytic THP-1 cells. J. Biol. Chem. 269, 8878–8884 (1994).

    CAS  PubMed  Google Scholar 

  3. Daëron, M. Fc receptor biology. Annu. Rev. Immunol. 15, 203–234 (1997).

    Article  PubMed  Google Scholar 

  4. Caron, E. & Hall, A. Identification of two distinct mechanisms of phagocytosis controlled by different Rho GTPases. Science 282, 1717–1721 (1998).

    Article  CAS  PubMed  Google Scholar 

  5. Patel, J. C., Hall, A. & Caron, E. Vav regulates activation of Rac but not Cdc42 during FcγR-mediated phagocytosis. Mol. Biol. Cell 13, 1215–1226 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hall, A. B. et al. Requirements for Vav guanine nucleotide exchange factors and Rho GTPases in FcγR- and complement-mediated phagocytosis. Immunity 24, 305–316 (2006). This article refutes the prevailing view concerning Rho GTPases during phagocytosis. It also shows that Rac regulates actin polymerization during both Fcγ- and complement-mediated phagocytosis and that Rho is implicated in both modes of uptake, but at a step distinct from actin polymerization.

    Article  CAS  PubMed  Google Scholar 

  7. Coppolino, M. G. et al. Evidence for a molecular complex consisting of Fyb/SLAP, SLP-76, Nck, VASP and WASP that links the actin cytoskeleton to Fcγ receptor signalling during phagocytosis. J. Cell Sci. 114, 4307–4318 (2001).

    CAS  PubMed  Google Scholar 

  8. May, R. C. et al. Involvement of the Arp2/3 complex in phagocytosis mediated by FcγR or CR3. Nature Cell Biol. 2, 246–248 (2000).

    Article  CAS  PubMed  Google Scholar 

  9. Colucci-Guyon, E. et al. A role for mammalian diaphanous-related formins in complement receptor (CR3)-mediated phagocytosis in macrophages. Curr. Biol. 15, 2007–2012 (2005).

    Article  CAS  PubMed  Google Scholar 

  10. Araki, N., Johnson, M. T. & Swanson, J. A. A role for phosphoinositide 3-kinase in the completion of macropinocytosis and phagocytosis by macrophages. J. Cell Biol. 135, 1249–1260 (1996).

    Article  CAS  PubMed  Google Scholar 

  11. Cox, D. et al. Myosin X is a downstream effector of PI(3)K during phagocytosis. Nature Cell Biol. 4, 469–477 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Lennartz, M. R. et al. Phospholipase A2 inhibition results in sequestration of plasma membrane into electronlucent vesicles during IgG-mediated phagocytosis. J. Cell Sci. 110, 2041–2052 (1997).

    CAS  PubMed  Google Scholar 

  13. Kusner, D. J., Hall, C. F. & Jackson, S. Fcγ receptor-mediated activation of phospholipase D regulates macrophage phagocytosis of IgG-opsonized particles. J. Immunol. 162, 2266–2274 (1999).

    CAS  PubMed  Google Scholar 

  14. Holevinsky, K. O. & Nelson, D. J. Membrane capacitance changes associated with particle uptake during phagocytosis in macrophages. Biophys. J. 75, 2577–2586 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bajno, L. et al. Focal exocytosis of VAMP3-containing vesicles at sites of phagosome formation. J. Cell Biol. 149, 697–706 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Braun, V. et al. TI–VAMP/VAMP7 is required for optimal phagocytosis of opsonised particles in macrophages. EMBO J. 23, 4166–4176 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Czibener, C. et al. Ca2+ and synaptotagmin VII-dependent delivery of lysosomal membrane to nascent phagosomes. J. Cell Biol. 174, 997–1007 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Huynh, K. K. et al. Fusion, fission, and secretion during phagocytosis. Physiology (Bethesda) 22, 366–372 (2007).

    CAS  Google Scholar 

  19. Cox, D. et al. A Rab11-containing rapidly recycling compartment in macrophages that promotes phagocytosis. Proc. Natl Acad. Sci. USA 97, 680–685 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Niedergang, F. et al. ADP ribosylation factor 6 is activated and controls membrane delivery during phagocytosis in macrophages. J. Cell Biol. 161, 1143–1150 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Desjardins, M. et al. Biogenesis of phagolysosomes proceeds through a sequential series of interactions with the endocytic apparatus. J. Cell Biol. 124, 677–688 (1994).

    Article  CAS  PubMed  Google Scholar 

  22. Mayorga, L. S., Bertini, F. & Stahl, P. D. Fusion of newly formed phagosomes with endosomes in intact cells and in a cell-free system. J. Biol. Chem. 266, 6511–6517 (1991).

    CAS  PubMed  Google Scholar 

  23. Desjardins, M. et al. Maturation of phagosomes is accompanied by changes in their fusion properties and size-selective acquisition of solute materials from endosomes. J. Cell Sci. 110, 2303–2314 (1997).

    CAS  PubMed  Google Scholar 

  24. Mukherjee, S., Ghosh, R. N. & Maxfield, F. R. Endocytosis. Physiol. Rev. 77, 759–803 (1997).

    Article  CAS  PubMed  Google Scholar 

  25. Bucci, C. et al. The small GTPase rab5 functions as a regulatory factor in the early endocytic pathway. Cell 70, 715–728 (1992).

    Article  CAS  PubMed  Google Scholar 

  26. Kitano, M. et al. Imaging of Rab5 activity identifies essential regulators for phagosome maturation. Nature 453, 241–245 (2008). This article shows that GAPVD1 is the essential GEF for Rab5A activation during the phagocytosis of apoptotic cells and that its phagosomal delivery is mediated by MAPRE1 (microtubule-associated protein RP/EB family member 1) on microtubules.

    Article  CAS  PubMed  Google Scholar 

  27. Vieira, O. V. et al. Distinct roles of class I and class III phosphatidylinositol 3-kinases in phagosome formation and maturation. J. Cell Biol. 155, 19–25 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Gaullier, J. M. et al. FYVE fingers bind PtdIns(3)P. Nature 394, 432–433 (1998).

    Article  CAS  PubMed  Google Scholar 

  29. Kanai, F. et al. The PX domains of p47phox and p40phox bind to lipid products of PI(3)K. Nature Cell Biol. 3, 675–678 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Lawe, D. C. et al. The FYVE domain of early endosome antigen 1 is required for both phosphatidylinositol 3-phosphate and Rab5 binding. Critical role of this dual interaction for endosomal localization. J. Biol. Chem. 275, 3699–3705 (2000).

    Article  CAS  PubMed  Google Scholar 

  31. Callaghan, J. et al. Direct interaction of EEA1 with Rab5b. Eur. J. Biochem. 265, 361–366 (1999).

    Article  CAS  PubMed  Google Scholar 

  32. McBride, H. M. et al. Oligomeric complexes link Rab5 effectors with NSF and drive membrane fusion via interactions between EEA1 and syntaxin 13. Cell 98, 377–386 (1999).

    Article  CAS  PubMed  Google Scholar 

  33. Mills, I. G., Urbé, S. & Clague, M. J. Relationships between EEA1 binding partners and their role in endosome fusion. J. Cell Sci. 114, 1959–1965 (2001).

    CAS  PubMed  Google Scholar 

  34. Botelho, R. J. et al. Role of COPI in phagosome maturation. J. Biol. Chem. 275, 15717–15727 (2000).

    Article  CAS  PubMed  Google Scholar 

  35. Leiva, N. et al. Reconstitution of recycling from the phagosomal compartment in streptolysin O-permeabilized macrophages: role of Rab11. Exp. Cell Res. 312, 1843–1855 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Damiani, M. T. et al. Rab coupling protein associates with phagosomes and regulates recycling from the phagosomal compartment. Traffic 5, 785–797 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Lindsay, A. J. et al. Rab coupling protein (RCP), a novel Rab4 and Rab11 effector protein. J. Biol. Chem. 277, 12190–12199 (2002).

    Article  CAS  PubMed  Google Scholar 

  38. Soldati, T. & Schliwa, M. Powering membrane traffic in endocytosis and recycling. Nature Rev. Mol. Cell Biol. 7, 897–908 (2006).

    Article  CAS  Google Scholar 

  39. Gokool, S., Tattersall, D. & Seaman, M. N. J. EHD1 interacts with retromer to stabilize SNX1 tubules and facilitate endosome-to-Golgi retrieval. Traffic 8, 1873–1886 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Traer, C. J. et al. SNX4 coordinates endosomal sorting of TfnR with dynein-mediated transport into the endocytic recycling compartment. Nature Cell Biol. 9, 1370–1380 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Lee, W. L. et al. Role of ubiquitin and proteasomes in phagosome maturation. Mol. Biol. Cell 16, 2077–2090 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Muzioł, T. et al. Structural basis for budding by the ESCRT-III factor CHMP3. Dev. Cell 10, 821–830 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Hanson, P. I. et al. Plasma membrane deformation by circular arrays of ESCRT-III protein filaments. J. Cell Biol. 180, 389–402 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Whitley, P. et al. Identification of mammalian Vps24p as an effector of phosphatidylinositol 3,5-bisphosphate-dependent endosome compartmentalization. J. Biol. Chem. 278, 38786–38795 (2003).

    Article  CAS  PubMed  Google Scholar 

  45. Bucci, C. et al. Rab7: a key to lysosome biogenesis. Mol. Biol. Cell 11, 467–480 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Harrison, R. E. et al. Phagosomes fuse with late endosomes and/or lysosomes by extension of membrane protrusions along microtubules: role of Rab7 and RILP. Mol. Cell Biol. 23, 6494–6506 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Rink, J. et al. Rab conversion as a mechanism of progression from early to late endosomes. Cell 122, 735–749 (2005). Rink and colleagues used advanced quantitative live-cell imaging to show that early-to-late endosomal maturation requires the gradual exchange of Rab5A for Rab7A (Rab conversion) on the same organelle.

    Article  CAS  PubMed  Google Scholar 

  48. Peterson, M. R. & Emr, S. D. The class C Vps complex functions at multiple stages of the vacuolar transport pathway. Traffic 2, 476–486 (2001).

    Article  CAS  PubMed  Google Scholar 

  49. Poupon, V. et al. The role of mVps18p in clustering, fusion, and intracellular localization of late endocytic organelles. Mol. Biol. Cell 14, 4015–4027 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Jordens, I. et al. The Rab7 effector protein RILP controls lysosomal transport by inducing the recruitment of dynein–dynactin motors. Curr. Biol. 11, 1680–1685 (2001).

    Article  CAS  PubMed  Google Scholar 

  51. Antonin, W. et al. A SNARE complex mediating fusion of late endosomes defines conserved properties of SNARE structure and function. EMBO J. 19, 6453–6464 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wade, N. et al. Syntaxin 7 complexes with mouse Vps10p tail interactor 1b, syntaxin 6, vesicle-associated membrane protein (VAMP)8, and VAMP7 in b16 melanoma cells. J. Biol. Chem. 276, 19820–19827 (2001).

    Article  CAS  PubMed  Google Scholar 

  53. Vieira, O. V. et al. Modulation of Rab5 and Rab7 recruitment to phagosomes by phosphatidylinositol 3-kinase. Mol. Cell. Biol. 23, 2501–2514 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Odorizzi, G. The multiple personalities of Alix. J. Cell Sci. 119, 3025–3032 (2006).

    Article  CAS  PubMed  Google Scholar 

  55. Kobayashi, T. et al. A lipid associated with the antiphospholipid syndrome regulates endosome structure and function. Nature 392, 193–197 (1998).

    Article  CAS  PubMed  Google Scholar 

  56. Gillooly, D. J. et al. Localization of phosphatidylinositol 3-phosphate in yeast and mammalian cells. EMBO J. 19, 4577–4588 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Griffiths, G. et al. The mannose 6-phosphate receptor and the biogenesis of lysosomes. Cell 52, 329–341 (1988).

    Article  CAS  PubMed  Google Scholar 

  58. Beyenbach, K. W. & Wieczorek, H. The V-type H+ ATPase: molecular structure and function, physiological roles and regulation. J. Exp. Biol. 209, 577–589 (2006).

    Article  CAS  PubMed  Google Scholar 

  59. Huynh, K. K. & Grinstein, S. Regulation of vacuolar pH and its modulation by some microbial species. Microbiol. Mol. Biol. Rev. 71, 452–462 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. van Deurs, B., Holm, P. K. & Sandvig, K. Inhibition of the vacuolar H+-ATPase with bafilomycin reduces delivery of internalized molecules from mature multivesicular endosomes to lysosomes in HEp-2 cells. Eur. J. Cell Biol. 69, 343–350 (1996).

    CAS  PubMed  Google Scholar 

  61. Gordon, A. H., Hart, P. D. & Young, M. R. Ammonia inhibits phagosome–lysosome fusion in macrophages. Nature 286, 79–80 (1980).

    Article  CAS  PubMed  Google Scholar 

  62. Aniento, F. et al. An endosomal βCOP is involved in the pH-dependent formation of transport vesicles destined for late endosomes. J. Cell Biol. 133, 29–41 (1996).

    Article  CAS  PubMed  Google Scholar 

  63. Hurtado-Lorenzo, A. et al. V-ATPase interacts with ARNO and Arf6 in early endosomes and regulates the protein degradative pathway. Nature Cell Biol. 8, 124–136 (2006). The authors found that the intraluminal domains of two 'a' subunits of the V-ATPase function as low pH sensors that undergo a conformational change in response to acidification and bind to ARF6 and its GEF, cytohesin 2, to regulate the endocytic degradative pathway.

    Article  CAS  PubMed  Google Scholar 

  64. Babior, B. M. NADPH oxidase. Curr. Opin. Immunol. 16, 42–47 (2004).

    Article  CAS  PubMed  Google Scholar 

  65. Heyworth, P. G., Cross, A. R. & Curnutte, J. T. Chronic granulomatous disease. Curr. Opin. Immunol. 15, 578–584 (2003).

    Article  CAS  PubMed  Google Scholar 

  66. Ambruso, D. R. et al. Human neutrophil immunodeficiency syndrome is associated with an inhibitory Rac2 mutation. Proc. Natl Acad. Sci. USA 97, 4654–4659 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Zhao, X., Carnevale, K. A. & Cathcart, M. K. Human monocytes use Rac1, not Rac2, in the NADPH oxidase complex. J. Biol. Chem. 278, 40788–40792 (2003).

    Article  CAS  PubMed  Google Scholar 

  68. Quinn, M. T. & Gauss, K. A. Structure and regulation of the neutrophil respiratory burst oxidase: comparison with nonphagocyte oxidases. J. Leukoc. Biol. 76, 760–781 (2004).

    Article  CAS  PubMed  Google Scholar 

  69. Minakami, R. & Sumimotoa, H. Phagocytosis-coupled activation of the superoxide-producing phagocyte oxidase, a member of the NADPH oxidase (nox) family. Int. J. Hematol. 84, 193–198 (2006).

    Article  CAS  PubMed  Google Scholar 

  70. Shepherd, V. L. The role of the respiratory burst of phagocytes in host defense. Semin. Respir. Infect. 1, 99–106 (1986).

    CAS  PubMed  Google Scholar 

  71. Reeves, E. P. et al. Killing activity of neutrophils is mediated through activation of proteases by K+ flux. Nature 416, 291–297 (2002).

    Article  CAS  PubMed  Google Scholar 

  72. Reeves, E. P. et al. Reassessment of the microbicidal activity of reactive oxygen species and hypochlorous acid with reference to the phagocytic vacuole of the neutrophil granulocyte. J. Med. Microbiol. 52, 643–651 (2003).

    Article  CAS  PubMed  Google Scholar 

  73. DeCoursey, T. E. During the respiratory burst, do phagocytes need proton channels or potassium channels, or both? Sci. STKE 2004, pe21 (2004).

    PubMed  Google Scholar 

  74. Fang, F. C. Antimicrobial reactive oxygen and nitrogen species: concepts and controversies. Nature Rev. Microbiol. 2, 820–832 (2004).

    Article  CAS  Google Scholar 

  75. Stuehr, D. J. Mammalian nitric oxide synthases. Biochim. Biophys. Acta 1411, 217–230 (1999).

    Article  CAS  PubMed  Google Scholar 

  76. Webb, J. L. et al. Macrophage nitric oxide synthase associates with cortical actin but is not recruited to phagosomes. Infect. Immun. 69, 6391–6400 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. De Groote, M. A., Fang, F. G. Nitric oxide and Infection (Kluwer Academic–Plenum, New York, 1999).

    Google Scholar 

  78. Borregaard, N. et al. Granules and secretory vesicles of the human neutrophil. Clin. Exp. Immunol. 101 (Suppl. 1), 6–9 (1995).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Masson, P. L., Heremans, J. F. & Schonne, E. Lactoferrin, an iron-binding protein in neutrophilic leukocytes. J. Exp. Med. 130, 643–658 (1969).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Cellier, M. F., Courville, P. & Campion, C. Nramp1 phagocyte intracellular metal withdrawal defense. Microbes Infect. 9, 1662–1670 (2007).

    Article  CAS  PubMed  Google Scholar 

  81. Lehrer, R. I., Lichtenstein, A. K. & Ganz, T. Defensins: antimicrobial and cytotoxic peptides of mammalian cells. Annu. Rev. Immunol. 11, 105–128 (1993).

    Article  CAS  PubMed  Google Scholar 

  82. Zanetti, M. The role of cathelicidins in the innate host defenses of mammals. Curr. Issues Mol. Biol. 7, 179–196 (2005).

    CAS  PubMed  Google Scholar 

  83. Pillay, C. S., Elliott, E. & Dennison, C. Endolysosomal proteolysis and its regulation. Biochem. J. 363, 417–429 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Claus, V. et al. Lysosomal enzyme trafficking between phagosomes, endosomes, and lysosomes in J774 macrophages. Enrichment of cathepsin H in early endosomes. J. Biol. Chem. 273, 9842–9851 (1998).

    Article  CAS  PubMed  Google Scholar 

  85. Garrity-Ryan, L. et al. The arginine finger domain of ExoT contributes to actin cytoskeleton disruption and inhibition of internalization of Pseudomonas aeruginosa by epithelial cells and macrophages. Infect. Immun. 68, 7100–7113 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Grosdent, N. et al. Role of Yops and adhesins in resistance of Yersinia enterocolitica to phagocytosis. Infect. Immun. 70, 4165–4176 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Rooijakkers, S. H. M. et al. Immune evasion by a staphylococcal complement inhibitor that acts on C3 convertases. Nature Immunol. 6, 920–927 (2005).

    Article  CAS  Google Scholar 

  88. Hong, Y. Q. & Ghebrehiwet, B. Effect of Pseudomonas aeruginosa elastase and alkaline protease on serum complement and isolated components C1q and C3. Clin. Immunol. Immunopathol. 62, 133–138 (1992).

    Article  CAS  PubMed  Google Scholar 

  89. Prasadarao, N. V. et al. A novel interaction of outer membrane protein A with C4b binding protein mediates serum resistance of Escherichia coli K1. J. Immunol. 169, 6352–6360 (2002).

    Article  CAS  PubMed  Google Scholar 

  90. Davis, J. M., Rasmussen, S. B. & O'Brien, A. D. Cytotoxic necrotizing factor type 1 production by uropathogenic Escherichia coli modulates polymorphonuclear leukocyte function. Infect. Immun. 73, 5301–5310 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Vandal, O. H. et al. A membrane protein preserves intrabacterial pH in intraphagosomal Mycobacterium tuberculosis. Nature Med. 14, 849–854 (2008).

    Article  CAS  PubMed  Google Scholar 

  92. Park, Y. K. et al. Internal pH crisis, lysine decarboxylase and the acid tolerance response of Salmonella typhimurium. Mol. Microbiol. 20, 605–611 (1996).

    Article  CAS  PubMed  Google Scholar 

  93. Schmidtchen, A. et al. Proteinases of common pathogenic bacteria degrade and inactivate the antibacterial peptide LL-37. Mol. Microbiol. 46, 157–168 (2002).

    Article  CAS  PubMed  Google Scholar 

  94. Peschel, A. et al. Staphylococcus aureus resistance to human defensins and evasion of neutrophil killing via the novel virulence factor MprF is based on modification of membrane lipids with L-lysine. J. Exp. Med. 193, 1067–1076 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Trent, M. S. et al. An inner membrane enzyme in Salmonella and Escherichia coli that transfers 4-amino-4-deoxy-L-arabinose to lipid A: induction on polymyxin-resistant mutants and role of a novel lipid-linked donor. J. Biol. Chem. 276, 43122–43131 (2001).

    Article  CAS  PubMed  Google Scholar 

  96. St John, G. et al. Peptide methionine sulfoxide reductase from Escherichia coli and Mycobacterium tuberculosis protects bacteria against oxidative damage from reactive nitrogen intermediates. Proc. Natl Acad. Sci. USA 98, 9901–9906 (2001).

    Article  CAS  PubMed  Google Scholar 

  97. Ng, V. H. et al. Role of KatG catalase-peroxidase in mycobacterial pathogenesis: countering the phagocyte oxidative burst. Mol. Microbiol. 52, 1291–1302 (2004).

    Article  CAS  PubMed  Google Scholar 

  98. Mott, J., Rikihisa, Y. & Tsunawaki, S. Effects of Anaplasma phagocytophila on NADPH oxidase components in human neutrophils and HL-60 cells. Infect. Immun. 70, 1359–1366 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Davis, A. S. et al. Mechanism of inducible nitric oxide synthase exclusion from mycobacterial phagosomes. PLoS Pathog. 3, e186 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Luo, M., Fadeev, E. A. & Groves, J. T. Mycobactin-mediated iron acquisition within macrophages. Nature Chem. Biol. 1, 149–153 (2005).

    Article  CAS  Google Scholar 

  101. Velayudhan, J. et al. The role of ferritins in the physiology of Salmonella enterica sv. Typhimurium: a unique role for ferritin B in iron–sulphur cluster repair and virulence. Mol. Microbiol. 63, 1495–1507 (2007).

    Article  CAS  PubMed  Google Scholar 

  102. Robey, M. & Cianciotto, N. P. Legionella pneumophila feoAB promotes ferrous iron uptake and intracellular infection. Infect. Immun. 70, 5659–5669 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Raivio, T. L. Envelope stress responses and Gram-negative bacterial pathogenesis. Mol. Microbiol. 56, 1119–1128 (2005).

    Article  CAS  PubMed  Google Scholar 

  104. Pethe, K. et al. Isolation of Mycobacterium tuberculosis mutants defective in the arrest of phagosome maturation. Proc. Natl Acad. Sci. USA 101, 13642–13647 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Wel, N. V. D. et al. M. tuberculosis and M. leprae translocate from the phagolysosome to the cytosol in myeloid cells. Cell 129, 1287–1298 (2007).

    Article  CAS  PubMed  Google Scholar 

  106. Gan, H. et al. Mycobacterium tuberculosis blocks crosslinking of annexin-1 and apoptotic envelope formation on infected macrophages to maintain virulence. Nature Immunol. 9, 1189–1197 (2008).

    Article  CAS  Google Scholar 

  107. Rao, V. et al. Mycobacterium tuberculosis controls host innate immune activation through cyclopropane modification of a glycolipid effector molecule. J. Exp. Med. 201, 535–543 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Simeone, R., Bottai, D. & Brosch, R. ESX/type VII secretion systems and their role in host–pathogen interaction. Curr. Opin. Microbiol. 12, 4–10 (2009).

    Article  CAS  PubMed  Google Scholar 

  109. Schlesinger, L. S. et al. Phagocytosis of Mycobacterium tuberculosis is mediated by human monocyte complement receptors and complement component C3. J. Immunol. 144, 2771–2780 (1990).

    CAS  PubMed  Google Scholar 

  110. Fratti, R. A. et al. Role of phosphatidylinositol 3-kinase and Rab5 effectors in phagosomal biogenesis and mycobacterial phagosome maturation arrest. J. Cell Biol. 154, 631–644 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Fratti, R. A. et al. Mycobacterium tuberculosis glycosylated phosphatidylinositol causes phagosome maturation arrest. Proc. Natl Acad. Sci. USA 100, 5437–5442 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Vergne, I., Chua, J. & Deretic, V. Tuberculosis toxin blocking phagosome maturation inhibits a novel Ca2+/calmodulin–PI3K hVPS34 cascade. J. Exp. Med. 198, 653–659 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Vergne, I. et al. Mechanism of phagolysosome biogenesis block by viable Mycobacterium tuberculosis. Proc. Natl Acad. Sci. USA 102, 4033–4038 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Beatty, W. L. et al. Trafficking and release of mycobacterial lipids from infected macrophages. Traffic 1, 235–247 (2000).

    Article  CAS  PubMed  Google Scholar 

  115. Malik, Z. A. et al. Cutting edge: Mycobacterium tuberculosis blocks Ca2+ signaling and phagosome maturation in human macrophages via specific inhibition of sphingosine kinase. J. Immunol. 170, 2811–2815 (2003).

    Article  CAS  PubMed  Google Scholar 

  116. Thompson, C. R. et al. Sphingosine kinase 1 (SK1) is recruited to nascent phagosomes in human macrophages: inhibition of SK1 translocation by Mycobacterium tuberculosis. J. Immunol. 174, 3551–3561 (2005).

    Article  CAS  PubMed  Google Scholar 

  117. Randhawa, A. K., Ziltener, H. J. & Stokes, R. W. CD43 controls the intracellular growth of Mycobacterium tuberculosis through the induction of TNF-α-mediated apoptosis. Cell. Microbiol. 10, 2105–2117 (2008).

    Article  CAS  PubMed  Google Scholar 

  118. Bonecini-Almeida, M. G. et al. Induction of in vitro human macrophage anti-Mycobacterium tuberculosis activity: requirement for IFN-γ and primed lymphocytes. J. Immunol. 160, 4490–4499 (1998).

    CAS  PubMed  Google Scholar 

  119. Gutierrez, M. G. et al. Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell 119, 753–766 (2004). This is the first study to show that IFNγ-treated macrophages are better able to eradicate intracellular M. tuberculosis through increased autophagy.

    Article  CAS  PubMed  Google Scholar 

  120. Delgado, M. A. et al. Toll-like receptors control autophagy. EMBO J. 27, 1110–1121 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Singh, S. B. et al. Human IRGM induces autophagy to eliminate intracellular mycobacteria. Science 313, 1438–1441 (2006).

    Article  CAS  PubMed  Google Scholar 

  122. Fremond, C. M. et al. IL-1 receptor-mediated signal is an essential component of MyD88-dependent innate response to Mycobacterium tuberculosis infection. J. Immunol. 179, 1178–1189 (2007).

    Article  CAS  PubMed  Google Scholar 

  123. Master, S. S. et al. Mycobacterium tuberculosis prevents inflammasome activation. Cell Host Microbe 3, 224–232 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Hamon, M., Bierne, H. & Cossart, P. Listeria monocytogenes: a multifaceted model. Nature Rev. Microbiol. 4, 423–434 (2006).

    Article  CAS  Google Scholar 

  125. Gaillard, J. L. et al. Entry of L. monocytogenes into cells is mediated by internalin, a repeat protein reminiscent of surface antigens from Gram-positive cocci. Cell 65, 1127–1141 (1991).

    Article  CAS  PubMed  Google Scholar 

  126. Dramsi, S. et al. Entry of Listeria monocytogenes into hepatocytes requires expression of InIB, a surface protein of the internalin multigene family. Mol. Microbiol. 16, 251–261 (1995).

    Article  CAS  PubMed  Google Scholar 

  127. Mengaud, J. et al. E-cadherin is the receptor for internalin, a surface protein required for entry of L. monocytogenes into epithelial cells. Cell 84, 923–932 (1996).

    Article  CAS  PubMed  Google Scholar 

  128. Shen, Y. et al. InIB-dependent internalization of Listeria is mediated by the Met receptor tyrosine kinase. Cell 103, 501–510 (2000).

    Article  CAS  PubMed  Google Scholar 

  129. Braun, L., Ghebrehiwet, B. & Cossart, P. gC1q-R/p32, a C1q-binding protein, is a receptor for the InlB invasion protein of Listeria monocytogenes. EMBO J. 19, 1458–1466 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Dunne, D. W. et al. The type I macrophage scavenger receptor binds to Gram-positive bacteria and recognizes lipoteichoic acid. Proc. Natl Acad. Sci. USA 91, 1863–1867 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Alvarez-Dominguez, C., Carrasco-Marin, E. & Leyva-Cobian, F. Role of complement component C1q in phagocytosis of Listeria monocytogenes by murine macrophage-like cell lines. Infect. Immun. 61, 3664–3672 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Drevets, D. A. & Campbell, P. A. Roles of complement and complement receptor type 3 in phagocytosis of Listeria monocytogenes by inflammatory mouse peritoneal macrophages. Infect. Immun. 59, 2645–2652 (1991).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Beauregard, K. E. et al. pH-dependent perforation of macrophage phagosomes by listeriolysin O from Listeria monocytogenes. J. Exp. Med. 186, 1159–1163 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Singh, R., Jamieson, A. & Cresswell, P. GILT is a critical host factor for Listeria monocytogenes infection. Nature 455, 1244–1247 (2008). This investigation shows that a host phagosome- specific protein is required for activation of the listeriolysin toxin and highlights the level to which L. monocytogenes has adapted to its host.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Henry, R. et al. Cytolysin-dependent delay of vacuole maturation in macrophages infected with Listeria monocytogenes. Cell. Microbiol. 8, 107–119 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Shaughnessy, L. M. et al. Membrane perforations inhibit lysosome fusion by altering pH and calcium in Listeria monocytogenes vacuoles. Cell. Microbiol. 8, 781–792 (2006). This manuscript reports how L. monocytogenes rapidly induces a delay in phagosome maturation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Smith, G. A. et al. The two distinct phospholipases C of Listeria monocytogenes have overlapping roles in escape from a vacuole and cell-to-cell spread. Infect. Immun. 63, 4231–4237 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Tilney, L. G. & Portnoy, D. A. Actin filaments and the growth, movement, and spread of the intracellular bacterial parasite, Listeria monocytogenes. J. Cell Biol. 109, 1597–1608 (1989).

    Article  CAS  PubMed  Google Scholar 

  139. Eylert, E. et al. Carbon metabolism of Listeria monocytogenes growing inside macrophages. Mol. Microbiol. 69, 1008–1017 (2008).

    Article  CAS  PubMed  Google Scholar 

  140. Lambrechts, A. et al. Listeria comet tails: the actin-based motility machinery at work. Trends Cell Biol. 18, 220–227 (2008).

    Article  CAS  PubMed  Google Scholar 

  141. Portnoy, D. A., Auerbuch, V. & Glomski, I. J. The cell biology of Listeria monocytogenes infection: the intersection of bacterial pathogenesis and cell-mediated immunity. J. Cell Biol. 158, 409–414 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Birmingham, C. L. et al. Listeriolysin O allows Listeria monocytogenes replication in macrophage vacuoles. Nature 451, 350–354 (2008). This study revealed a novel intracellular fate for L. monocytogenes that was previously unappreciated.

    Article  CAS  PubMed  Google Scholar 

  143. Borella, P. et al. Water ecology of Legionella and protozoan: environmental and public health perspectives. Biotechnol. Annu. Rev. 11, 355–380 (2005).

    Article  CAS  PubMed  Google Scholar 

  144. Brüggemann, H., Cazalet, C. & Buchrieser, C. Adaptation of Legionella pneumophila to the host environment: role of protein secretion, effectors and eukaryotic-like proteins. Curr. Opin. Microbiol. 9, 86–94 (2006).

    Article  CAS  PubMed  Google Scholar 

  145. Bellinger-Kawahara, C. & Horwitz, M. A. Complement component C3 fixes selectively to the major outer membrane protein (MOMP) of Legionella pneumophila and mediates phagocytosis of liposome–MOMP complexes by human monocytes. J. Exp. Med. 172, 1201–1210 (1990).

    Article  CAS  PubMed  Google Scholar 

  146. Payne, N. R. & Horwitz, M. A. Phagocytosis of Legionella pneumophila is mediated by human monocyte complement receptors. J. Exp. Med. 166, 1377–1389 (1987).

    Article  CAS  PubMed  Google Scholar 

  147. Clemens, D. L., Lee, B. Y. & Horwitz, M. A. Deviant expression of Rab5 on phagosomes containing the intracellular pathogens Mycobacterium tuberculosis and Legionella pneumophila is associated with altered phagosomal fate. Infect. Immun. 68, 2671–2684 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Joshi, A. D., Sturgill-Koszycki, S. & Swanson, M. S. Evidence that Dot-dependent and -independent factors isolate the Legionella pneumophila phagosome from the endocytic network in mouse macrophages. Cell. Microbiol. 3, 99–114 (2001).

    Article  CAS  PubMed  Google Scholar 

  149. Robinson, C. G. & Roy, C. R. Attachment and fusion of endoplasmic reticulum with vacuoles containing Legionella pneumophila. Cell. Microbiol. 8, 793–805 (2006).

    Article  CAS  PubMed  Google Scholar 

  150. Murata, T. et al. The Legionella pneumophila effector protein DrrA is a Rab1 guanine nucleotide-exchange factor. Nature Cell Biol. 8, 971–977 (2006).

    Article  CAS  PubMed  Google Scholar 

  151. Machner, M. P. & Isberg, R. R. Targeting of host Rab GTPase function by the intravacuolar pathogen Legionella pneumophila. Dev. Cell 11, 47–56 (2006).

    Article  CAS  PubMed  Google Scholar 

  152. Brombacher, E. et al. Rab1 guanine nucleotide exchange factor SidM is a major phosphatidylinositol 4-phosphate-binding effector protein of Legionella pneumophila. J. Biol. Chem. 284, 4846–4856 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Ingmundson, A. et al. Legionella pneumophila proteins that regulate Rab1 membrane cycling. Nature 450, 365–369 (2007).

    Article  CAS  PubMed  Google Scholar 

  154. Machner, M. P. & Isberg, R. R. A bifunctional bacterial protein links GDI displacement to Rab1 activation. Science 318, 974–977 (2007).

    Article  CAS  PubMed  Google Scholar 

  155. Nagai, H. et al. A bacterial guanine nucleotide exchange factor activates ARF on Legionella phagosomes. Science 295, 679–682 (2002). This is the first study to show that an Icm–Dot-secreted factor from L. pneumophila targets host vesicle trafficking.

    Google Scholar 

  156. Pan, X. et al. Ankyrin repeat proteins comprise a diverse family of bacterial type IV effectors. Science 320, 1651–1654 (2008). The authors show that C. burnetii secretes effectors into host cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Sauer, J. et al. Specificity of Legionella pneumophila and Coxiella burnetii vacuoles and versatility of Legionella pneumophila revealed by coinfection. Infect. Immun. 73, 4494–4504 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Sturgill-Koszycki, S. & Swanson, M. S. Legionella pneumophila replication vacuoles mature into acidic, endocytic organelles. J. Exp. Med. 192, 1261–1272 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Wieland, H., Goetz, F. & Neumeister, B. Phagosomal acidification is not a prerequisite for intracellular multiplication of Legionella pneumophila in human monocytes. J. Infect. Dis. 189, 1610–1614 (2004).

    Article  PubMed  Google Scholar 

  160. Swanson, M. S., Fernandez-Moreira, E. & Fernandez-Moreia, E. A microbial strategy to multiply in macrophages: the pregnant pause. Traffic 3, 170–177 (2002).

    Article  CAS  PubMed  Google Scholar 

  161. Amer, A. O. & Swanson, M. S. Autophagy is an immediate macrophage response to Legionella pneumophila. Cell. Microbiol. 7, 765–778 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Isberg, R. R., O'Connor, T. J. & Heidtman, M. The Legionella pneumophila replication vacuole: making a cosy niche inside host cells. Nature Rev. Microbiol. 7, 13–24 (2009).

    Article  CAS  Google Scholar 

  163. Albert-Weissenberger, C., Cazalet, C. & Buchrieser, C. Legionella pneumophila — a human pathogen that co-evolved with fresh water protozoa. Cell. Mol. Life Sci. 64, 432–448 (2007).

    Article  CAS  PubMed  Google Scholar 

  164. Voth, D. E. & Heinzen, R. A. Lounging in a lysosome: the intracellular lifestyle of Coxiella burnetii. Cell. Microbiol. 9, 829–840 (2007).

    Article  CAS  PubMed  Google Scholar 

  165. Heinzen, R. A. et al. Differential interaction with endocytic and exocytic pathways distinguish parasitophorous vacuoles of Coxiella burnetii and Chlamydia trachomatis. Infect. Immun. 64, 796–809 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Capo, C. et al. Subversion of monocyte functions by Coxiella burnetii: impairment of the cross-talk between αvβ3 integrin and CR3. J. Immunol. 163, 6078–6085 (1999).

    CAS  PubMed  Google Scholar 

  167. Meconi, S. et al. Activation of protein tyrosine kinases by Coxiella burnetii: role in actin cytoskeleton reorganization and bacterial phagocytosis. Infect. Immun. 69, 2520–2526 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Meconi, S. et al. Coxiella burnetii induces reorganization of the actin cytoskeleton in human monocytes. Infect. Immun. 66, 5527–5533 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Berón, W. et al. Coxiella burnetii localizes in a Rab7-labeled compartment with autophagic characteristics. Infect. Immun. 70, 5816–5821 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Gutierrez, M. G. et al. Autophagy induction favours the generation and maturation of the Coxiella-replicative vacuoles. Cell. Microbiol. 7, 981–993 (2005).

    Article  CAS  PubMed  Google Scholar 

  171. Romano, P. S. et al. The autophagic pathway is actively modulated by phase II Coxiella burnetii to efficiently replicate in the host cell. Cell. Microbiol. 9, 891–909 (2007).

    Article  CAS  PubMed  Google Scholar 

  172. Maurin, M. et al. Phagolysosomes of Coxiella burnetii-infected cell lines maintain an acidic pH during persistent infection. Infect. Immun. 60, 5013–5016 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Hackstadt, T. & Williams, J. C. Biochemical stratagem for obligate parasitism of eukaryotic cells by Coxiella burnetii. Proc. Natl Acad. Sci. USA 78, 3240–3244 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Mertens, K. et al. Constitutive SOS expression and damage-inducible AddAB-mediated recombinational repair systems for Coxiella burnetii as potential adaptations for survival within macrophages. Mol. Microbiol. 69, 1411–1426 (2008).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Work in the authors' laboratory is supported by the Canadian Cystic Fibrosis Foundation, the Heart and Stroke Foundation of Ontario and the Canadian Institutes of Health Research (CIHR). R.S.F. is supported by a CIHR Fellowship and G.C. is supported by the National Council of Science and Technology of Mexico (CONACYT).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sergio Grinstein.

Related links

Related links

DATABASES

Entrez Genome Project

Coxiella burnetii

Legionella pneumophila

Listeria monocytogenes

Mycobacterium tuberculosis

Glossary

Endocytic pathway

The route followed inside the cell by vesicles derived from the plasma membrane by endocytosis, including their membrane-associated cargo and trapped fluid. Vesicles or vacuoles derived from the plasma membrane undergo fusion and fission events that deliver some of their components to lysosomes for degradation, whereas others are recycled.

Phosphoinositide

An inositol phospholipid that can be phosphorylated separately or at all possible combinations of the D-3, D-4 and D-5 positions of the inositol ring.

SNARE protein

A member of the soluble N-ethylmaleimide sensitive factor attachment protein receptor family that mediates docking and fusion of cellular membranes. Cognate SNARE pairs on the vesicular and target membranes intertwine to form a SNARE pin that brings the membranes into close apposition, driving their fusion.

Multivesicular body

(MVB). A defined stage in the transit between early endosomes and late endosomes or lysosomes. MVBs are characterized by a limiting membrane that encloses internal vesicles rich in lysobisphosphatidic acid, CD63 and phosphatidylinositol-3-phosphate. Proteins destined for degradation are sorted to internal vesicles of MVBs.

Azurophil or primary granule

A specialized neutrophil granule, also called a peroxidase-positive granule, that resembles lysosomes, in that it contains degradative enzymes, such as β-glucuronidase, cathepsins, elastase, lysozyme and myeloperoxidase, as well as antimicrobial peptides, such as defensins.

Specific or secondary granule

A specialized neutrophil granule, also called a peroxidase-negative granule, that exists as a heterogeneous continuum of granules with varying amounts of lactoferrin, collagenase, heparanase, lysozyme and antimicrobial cathelicidins.

Autophagy

A complex cellular process by which intracellular components, including entire organelles, are sequestered in double-membrane vesicles or vacuoles called autophagosomes that eventually fuse with lysosomes, bringing about the degradation of their contents.

SarI–COPII-coated secretory vesicle

A vesicle derived from the endoplasmic reticulum (ER) through coating with the coatomer protein complex II (COPII) protein complex, a process initiated at specialized ER exit sites by the GTPase SarI.

Type IV secretion system

A macromolecular apparatus used by bacteria to secrete effector molecules. This secretion system is ancestrally related to bacterial DNA conjugation systems, and is often expressed by pathogenic bacteria, which contributes to their virulence.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Flannagan, R., Cosío, G. & Grinstein, S. Antimicrobial mechanisms of phagocytes and bacterial evasion strategies. Nat Rev Microbiol 7, 355–366 (2009). https://doi.org/10.1038/nrmicro2128

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrmicro2128

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