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:

Modulation of innate immunity by Toxoplasma gondii virulence effectors

Key Points

  • The parasite Toxoplasma gondii is extremely widespread in animals and is a common cause of food- and water-borne infection in people. Although most infections are benign, they can have severe consequences in immunocompromised patients and following congenital infection.

  • T. gondii is regarded as a model intracellular parasite for which forward- and reverse-genetics tools are available. In combination with the mouse model of toxoplasmosis (including the many genetic knockout and transgenic mouse lines that are available), these tools for genetic manipulation of the parasite have enabled researchers to explore the molecular determinants of T. gondii pathogenesis and host defence.

  • Forward-genetics crosses conducted in T. gondii, using strains of different genotypes and virulences in mice, revealed that acute virulence is largely mediated by a family of effector proteins that are secreted into the host cell cytoplasm during parasite invasion. These proteins are derived from a secretory organelle called the rhoptry and, hence, are called ROP effectors.

  • ROPs include a family of serine/threonine kinases that affect host targets and have important roles in infection in the mouse. Among these, ROP18 phosphorylates immunity-related GTPases, thus promoting parasite survival in activated macrophages, whereas ROP16 phosphorylates signal transducer and activator of transcription 3 (STAT3) and STAT6 and, hence, alters host gene transcription. Curiously, the activity of ROP18 is mediated by another family member called ROP5, which is a pseudokinase.

  • Although a limited subset of ROP kinases can largely explain the virulence of T. gondii in the mouse, their role in other hosts has not been established. The genome encodes more than 40 ROPs, and these different proteins might have distinct roles during infection in the wide range of hosts infected by T. gondii. Understanding these patterns might help in the prevention and treatment of human infections.

Abstract

Toxoplasma gondii is a common parasite of animals and humans and can cause serious opportunistic infections. However, the majority of infections are asymptomatic, possibly because the organism has co-evolved with its many vertebrate hosts and has developed multiple strategies to persist asymptomatically for the lifetime of the host. Over the past two decades, infection studies in the mouse, combined with forward-genetics approaches aimed at unravelling the molecular basis of infection, have revealed that T. gondii virulence is mediated, in part, by secretion of effector proteins into the host cell during invasion. Here, we review recent advances that illustrate how these virulence factors disarm innate immunity and promote survival of the parasite.

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: The complex life cycle of Toxoplasma gondii
Figure 2: Innate immune responses to Toxoplasma gondii during infection.
Figure 3: Structure and function of rhoptry protein 2 family members.
Figure 4: The role of Toxoplasma gondii virulence factors in modulating immune signalling in the host.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

  1. Dubey, J. P. Toxoplasmosis of Animals and Humans (CRC Press, 2010).

    Google Scholar 

  2. Dubey, J. P. & Frenkel, J. F. Cyst-induced toxoplasmosis in cats. J. Protozool. 19, 155–177 (1972).

    Article  CAS  PubMed  Google Scholar 

  3. Su, C. et al. Recent expansion of Toxoplasma through enhanced oral transmission. Science 299, 414–416 (2003).

    Article  CAS  PubMed  Google Scholar 

  4. Jones, J. L. & Dubey, J. P. Foodborne toxoplasmosis. Clin. Infect. Dis. 55, 845–851 (2012).

    Article  PubMed  Google Scholar 

  5. Moura, L. et al. Waterborne toxoplasmosis, Brazil, from field to gene. Emerg. Infect. Dis. 12, 326–329 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Joynson, D. H. & Wreghitt, T. J. Toxoplasmosis: A Comprehensive Clinical Guide. (Cambridge Univ. Press, 2001).

    Book  Google Scholar 

  7. Boothroyd, J. C. & Grigg, M. E. Population biology of Toxoplasma gondii and its relevance to human infection: do different strains cause different disease? Curr. Opin. Microbiol. 5, 438–442 (2002).

    Article  PubMed  Google Scholar 

  8. Jamieson, S. E. et al. Genetic and epigenetic factors at COL2A1 and ABCA4 influence clinical outcome in congenital toxoplasmosis. PLoS ONE 3, e2285 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. McLeod, R. et al. Prematurity and severity are associated with Toxoplasma gondii alleles (NCCCTS, 1981–2009). Clin. Infect. Dis. 54, 1595–1605 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Suzuki, Y. et al. Evidence for genetic regulation of susceptibility to toxoplasmic encephalitis in AIDS patients. J. Infect. Dis. 173, 265–268 (1996).

    Article  CAS  PubMed  Google Scholar 

  11. Mead, P. S. et al. Food-related illness and death in the United States. Emerg. Infect. Dis. 5, 607–625 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Buzoni-Gatel, D., Schulthess, J., Menard, L. C. & Kasper, L. H. Mucosal defenses against orally acquired protozoan parasites, emphasis on Toxoplasma gondii infections. Cell. Microbiol. 8, 535–544 (2006).

    Article  CAS  PubMed  Google Scholar 

  13. Daher, W. & Soldati-Favre, D. Mechanisms controlling glideosome function in apicomplexans. Curr. Opin. Microbiol. 12, 408–414 (2009).

    Article  CAS  PubMed  Google Scholar 

  14. Sibley, L. D. How apicomplexan parasites move in and out of cells. Curr. Opin. Biotechnol. 21, 592–598 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Sibley, L. D. Invasion and intracellular survival by protozoan parasites. Immunol. Rev. 240, 72–91 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Tait, E. D. & Hunter, C. A. Advances in understanding immunity to Toxoplasma gondii. Mem. Inst. Oswaldo Cruz 104, 201–210 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Montoya, J. G. & Liesenfeld, O. Toxoplasmosis. Lancet 363, 1965–1976 (2004).

    Article  CAS  PubMed  Google Scholar 

  18. Ajioka, J. W. & Soldati, D. Toxoplasma: Molecular and Cellular Biology. (Horizon Biosciences, 2007).

    Google Scholar 

  19. Kim, K. & Weiss, L. M. Toxoplasma gondii: the model apicomplexan. Int. J. Parasitol. 34, 423–432 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Nicolle, C. & Manceaux, L. H. Sur une infection a corp de Leishman (ou organismes voisins) du gondi. C.R. Scéances Acad. Sci. III 147, 763–766 (1908) (in French).

    Google Scholar 

  21. Howe, D. K. & Sibley, L. D. Toxoplasma gondii comprises three clonal lineages: correlation of parasite genotype with human disease. J. Infect. Dis. 172, 1561–1566 (1995).

    Article  CAS  PubMed  Google Scholar 

  22. Boyle, J. P. et al. Just one cross appears capable of dramatically altering the population biology of a eukaryotic pathogen like Toxoplasma gondii. Proc. Natl Acad. Sci. USA 103, 10514–10519 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sibley, L. D. & Ajioka, J. W. Population structure of Toxoplasma gondii: clonal expansion driven by infrequent recombination and selective sweeps. Ann. Rev. Microbiol. 62, 329–351 (2008).

    Article  CAS  Google Scholar 

  24. Su, C. L. et al. Globally diverse Toxoplasma gondii isolates comprise six major clades originating from a small number of distinct ancestral lineages. Proc. Natl Acad. Sci. USA 109, 5844–5849 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Munoz, M., Liesenfeld, O. & Heimesaat, M. M. Immunology of Toxoplasma gondii. Immunol. Rev. 240, 269–285 (2011).

    Article  CAS  PubMed  Google Scholar 

  26. Sibley, L. D. & Boothroyd, J. C. Virulent strains of Toxoplasma gondii comprise a single clonal lineage. Nature 359, 82–85 (1992).

    Article  CAS  PubMed  Google Scholar 

  27. Radke, J. R. et al. Defining the cell cycle for the tachyzoite stage of Toxoplasma gondii. Mol. Biochem. Parasitol. 115, 165–175 (2001).

    Article  CAS  PubMed  Google Scholar 

  28. Barragan, A. & Sibley, L. D. Transepithelial migration of Toxoplasma gondii is linked to parasite motility and virulence. J. Exp. Med. 195, 1625–1633 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Liesenfeld, O. Oral infection of C57BL/6 mice with Toxoplasma gondii: a new model of inflammatory bowel disease? J. Infect. Dis. 185, S96–101 (2002).

    Article  PubMed  Google Scholar 

  30. Suzuki, Y. & Joh, K. Effect of the strain of Toxoplasma gondii on the development of toxoplasmic encephalitis in mice treated with antibody to interferon-gamma. Parasitol. Res. 80, 125–130 (1994).

    Article  CAS  PubMed  Google Scholar 

  31. Khan, I. A., Matsuura, T. & Kasper, L. H. Interleukin-12 enhances murine survival against acute toxoplasmosis. Infect. Immun. 62, 1639–1642 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Johnson, L. L. SCID mouse models of acute and relapsing chronic Toxoplasma gondii infections. Infect. Immun. 60, 3719–3724 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Gazzinelli, R. T., Hieny, S., Wynn, T. A., Wolf, S. & Sher, A. Interleukin 12 is required for the T-lymphocyte-independent induction of interferon γ by an intracellular parasite and induces resistance in T-cell-deficient hosts. Proc. Natl Acad. Sci. USA 90, 6115–6119 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hunter, C. A., Subauste, C. S., Van Cleave, V. H. & Remington, J. S. Production of gamma interferon by natural killer cells from Toxoplasma gondii-infected SCID mice: regulation by interleukin-10, interleukin-12, and tumor necrosis factor alpha. Infect. Immun. 62, 2818–2824 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Denkers, E. Y. et al. Perforin-mediated cytolysis plays a limited role in host resistance to Toxoplasma gondii. J. Immunol. 159, 1903–1908 (1997).

    CAS  PubMed  Google Scholar 

  36. Robben, P. M. et al. Production of IL-12 by macrophages infected with Toxoplasma gondii depends on the parasite genotype. J. Immunol. 172, 3686–3694 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Reis e Sousa, C. et al. In vivo microbial stimulation induces rapid CD40 ligand-independent production of interleukin 12 by dendritic cells and their redistribution to T cell areas. J. Exp. Med. 186, 1819–1829 (1997).

    CAS  PubMed  Google Scholar 

  38. Mashayekhi, M. et al. CD8α+ dendritic cells are the critical source of interleukin-12 that controls acute infection by Toxoplasma gondii tachyzoites. Immunity 35, 249–259 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bierly, A. L., Shufesky, W. J., Sukhumavasi, W., Morelli, A. E. & Denkers, E. Y. Dendritic cells expressing plasmacytoid marker PDCA-1 are Trojan horses during Toxoplasma gondii infection. J. Immunol. 181, 8485–8491 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Pepper, M. et al. Plasmacytoid dendritic cells are activated by Toxoplasma gondii to present antigen and produce cytokines. J. Immunol. 180, 6229–6236 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Bliss, S. K., Butcher, B. A. & Denkers, E. Y. Rapid recruitment of neutrophils containing prestored IL-12 during microbial infection. J. Immunol. 165, 4515–4521 (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Goldszmid, R. S. et al. NK cell-derived interf eron-γ orchestrates cellular dynamics and the differentiation of monocytes into dendritic cells at the site of infection. Immunity 36, 1047–1059 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Dunay, I. R. et al. Gr1+ inflammatory monocytes are required for mucosal resistance to the pathogen Toxoplasma gondii. Immunity 29, 306–317 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Robben, P. R., LaRegina, M., Kuziel, W. A. & Sibley, L. D. Recruitment of Gr-1+ monocytes is essential for control of acute toxoplasmosis. J. Exp. Med. 201, 1761–1769 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Scanga, C. A. et al. Cutting edge: MyD88 is required for resistance to Toxoplasma gondii infection and regulates parasite-induced IL-12 production by dendritic cells. J. Immunol. 168, 5997–6001 (2002).

    Article  CAS  PubMed  Google Scholar 

  46. Debierre-Grockiego, F. et al. Activation of TLR2 and TLR4 by glycosylphosphatidylinositols derived from Toxoplasma gondii. J. Immunol. 179, 1129–1137 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Yarovinsky, F. et al. TLR11 activation of dendritic cells by a protozoan profilin-like protein. Science 308, 1626–1629 (2005).

    Article  CAS  PubMed  Google Scholar 

  48. Gazzinelli, R. T. & Denkers, E. Y. Protozoan encounters with Toll-like receptor signalling pathways: implications for host parasitism. Nature Rev. Immunol. 6, 895–906 (2006).

    Article  CAS  Google Scholar 

  49. Melo, M. B. et al. UNC93B1 mediates host resistance to infection with Toxoplasma gondii. PLoS Pathog. 6, e1001071 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Pifer, R., Benson, A., Sturge, C. R. & Yarovinsky, F. UNC93B1 is essential for TLR11 activation and IL-12-dependent host resistance to Toxoplasma gondii. J. Biol. Chem. 286, 3307–3314 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Suzuki, Y., Orellana, M. A., Schreiber, R. D. & Remington, J. S. Interferon-γ: the major mediator of resistance against Toxoplasma gondii. Science 240, 516–518 (1988).

    Article  CAS  PubMed  Google Scholar 

  52. Yap, G. S. & Sher, A. Effector cells of both nonhemopoietic and hemopoietic origin are required for interferon (IFN)-γ- and tumor necrosis factor (TNF)- α-dependent host resistance to the intracellular pathogen, Toxoplasma gondii. J. Exp. Med. 189, 1083–1092 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Pfefferkorn, E. R. Interferon-γ blocks the growth of Toxoplasma gondii in human fibroblasts by inducing the host to degrade tryptophan. Proc. Natl Acad. Sci. USA 81, 908–912 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Dimier, I. H. & Bout, D. T. Interferon-γ-activated primary enterocytes inhibit Toxoplasma gondii replication: a role for intracellular iron. Immunology 94, 488–495 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Murray, H. W., Rubin, B. Y., Carriero, S. M., Harris, A. M. & Jaffee, E. A. Human mononuclear phagocyte antiprotozoal mechanisms: oxygen-dependent versus oxygen-independent activity against intracellular Toxoplasma gondii. J. Immunol. 134, 1982–1988 (1985).

    CAS  PubMed  Google Scholar 

  56. Adams, L. B., Hibbs, J. B., Taintor, R. R. & Krahenbuhl, J. L. Microbiostatic effect of murine activated macrophages for Toxoplasma gondii. Role for synthesis of inorganic nitrogen oxides from L-Arginine. J. Immunol. 144, 2725–2729 (1990).

    CAS  PubMed  Google Scholar 

  57. Scharton-Kersten, T. M., Yap, G., Magram, J. & Sher, A. Inducible nitric oxide is essential for host control of persistent but not acute infection with the intracellular pathogen Toxoplasma gondii. J. Exp. Med. 185, 1261–1273 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Taylor, G. A. & Feng, C. G. & Sher, A. p47 GTPases: regulators of immunity to intracellular pathogens. Nature Rev. Immunol. 4, 100–109 (2004).

    Article  CAS  Google Scholar 

  59. Taylor, G. A., Feng, C. G. & Sher, A. Control of IFN-γ-mediated host resistance to intracellular pathogens by immunity-related GTPases (p47 GTPases). Microb. Infect. 9, 1644–1651 (2007).

    Article  CAS  Google Scholar 

  60. Howard, J. C., Hunn, J. P. & Steinfeldt, T. The IRG protein-based resistance mechanism in mice and its relation to virulence in Toxoplasma gondii. Curr. Opin. Microbiol. 14, 414–421 (2011).

    Article  CAS  PubMed  Google Scholar 

  61. Yamamoto, M. et al. A cluster of Interferon-γ-inducible p65 GTPases plays a critical role in host defense against Toxoplasma gondii. Immunity 37, 302–313 (2012).

    Article  CAS  PubMed  Google Scholar 

  62. Shenoy, A. R. et al. Emerging themes in IFN-γ-induced macrophage immunity by the p47 and p65 GTPase families. Immunobiol. 212, 771–784 (2008).

    Article  CAS  Google Scholar 

  63. Kim, B. H. et al. A family of IFN-γ-inducible 65-kD GTPases protects against bacterial infection. Science 332, 717–721 (2011).

    Article  CAS  PubMed  Google Scholar 

  64. MacMicking, J. D. Interferon-inducible effector mechanisms in cell-autonomous immunity. Nature Rev. Immunol. 12, 367–382 (2012).

    Article  CAS  Google Scholar 

  65. Blanchard, N. & Shastri, N. Topological journey of parasite-derived antigens for presentation by MHC class I molecules. Trends Immunol. 31, 414–421 (2010).

    Article  CAS  PubMed  Google Scholar 

  66. Lüder, C. G. K., Lang, T., Beuerle, B. & Gross, U. Down-regulation of MHC class II molecules and inability to up-regulate class I molecules in murine macrophages after infection with Toxoplasma gondii. Clin. Exp. Immunol. 112, 308–316 (1998).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Tait, E. D. et al. Virulence of Toxoplasma gondii is associated with distinct dendritic cell responses and reduced numbers of activated CD8+ T cells. J. Immunol. 185, 1502–1512 (2010).

    Article  CAS  PubMed  Google Scholar 

  68. Blanchard, N. et al. Immunodominant, protective response to the parasite Toxoplasma gondii requires antigen processing in the endoplasmic reticulum. Nature Immunol. 9, 937–944 (2008).

    Article  CAS  Google Scholar 

  69. Frickel, E. M. et al. Parasite stage-specific recognition of endogenous Toxoplasma gondii-derived CD8+ T cell epitopes. J. Infect. Dis. 198, 1625–1633 (2008).

    Article  CAS  PubMed  Google Scholar 

  70. Lekutis, C., Ferguson, D. J., Grigg, M. E., Camps, M. & Boothroyd, J. C. Surface antigens of Toxoplasma gondii: variations on a theme. Int. J. Parasitol. 31, 1285–1292 (2001).

    Article  CAS  PubMed  Google Scholar 

  71. Blader, I., Manger, I. D. & Boothroyd, J. C. Microarray analysis reveals previously unknown changes in Toxoplasma gondii infected human cells. J. Biol. Chem. 276, 24223–24231 (2001).

    Article  CAS  PubMed  Google Scholar 

  72. Lüder, C. G., Walter, W., Beuerle, B., Maeurer, M. J. & Gross, U. Toxoplasma gondii down-regulates MHC class II gene expression and antigen presentation by murine macrophages via interference with nuclear translocation of STAT1α. Eur. J. Immunol. 31, 1475–1484 (2001).

    Article  PubMed  Google Scholar 

  73. Butcher, B. A., Kim, L., Johnson, P. F. & Denkers, E. Y. Toxoplasma gondii tachyzoites inhibit proinflammatory cytokine induction in infected macrophages by preventing nuclear translocation of the transcription factor NF-κB. J. Immunol. 167, 2193–2201 (2001).

    Article  CAS  PubMed  Google Scholar 

  74. Shapira, S. et al. Initiation and termination of NF-κB signalling by the intracellular protozoan parasite Toxoplasma gondii. J. Cell Sci. 118, 3501–3508 (2005).

    Article  CAS  PubMed  Google Scholar 

  75. Shapira, S., Speirs, K., Gerstein, A., Caamano, J. & Hunter, C. A. Suppression of NF-κB activation by infection with Toxoplasma gondii. J. Infect. Dis. 185 (Suppl. 1), S66–S72 (2002).

    Article  CAS  PubMed  Google Scholar 

  76. Kim, S. K., Fouts, A. E. & Boothroyd, J. C. Toxoplasma gondii dysregulates IFN-γ inducible gene expression in human fiboblasts: insights from a genome-wide transcriptional profiling. J. Immunol. 178, 5154–5165 (2007).

    Article  CAS  PubMed  Google Scholar 

  77. Lang, C. et al. Impaired chromatin remodelling at STAT1-regulated promoters leads to global unresponsiveness of Toxoplasma gondii-infected macrophages to IFN-γ. PLoS Pathog. 8, e1002483 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Stutz, A., Kessler, H., Kaschel, M. E., Meissner, M. & Dalpke, A. H. Cell invasion and strain dependent induction of suppressor of cytokine signalling-1 by Toxoplasma gondii. Immunobiology 217, 28–36 (2012).

    Article  CAS  PubMed  Google Scholar 

  79. Zimmermann, S., Murray, P. J., Heeg, K. & Dalpke, A. H. Induction of suppressor of cytokine signalling-1 by Toxoplasma gondii contributes to immune evasion in macrophages by blocking IFN-γ signalling. J. Immunol. 176, 1840–1847 (2006).

    Article  CAS  PubMed  Google Scholar 

  80. Whitmarsh, R. J. et al. A critical role for SOCS3 in innate resistance to Toxoplasma gondii. Cell Host Microbe 10, 224–236 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Butcher, B. A. et al. IL-10-independent STAT3 activation by Toxoplasma gondii mediates suppression of IL-12 and TNF-α in host macrophages. J. Immunol. 174, 3148–3152 (2005).

    Article  CAS  PubMed  Google Scholar 

  82. Andrade, R. M., Wessendarp, M., Gubbels, J. M., Striepen, B. & Subaste, C. S. CD40 induces macrophage anti-Toxoplasma gondii activity by triggering autophagy-dependent fusion of pathogen-containing vacuoles and lysosomes. J. Clin. Invest. 116, 2366–2377 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Reichmann, G. et al. The CD40/CD40 ligand interaction is required for resistance in toxoplasmic encephalitis. Infect. Immun. 68, 1312–1318 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Sibley, L. D., Adams, L. B., Fukutomi, Y. & Krahenbuhl, J. L. Tumor necrosis factor-α triggers antitoxoplasmal activity of IFN-γ primed macrophages. J. Immunol. 147, 2340–2345 (1991).

    CAS  PubMed  Google Scholar 

  85. Denkers, E. Y., Butcher, B. A., Del Rio, L. & Kim, L. Manipulations of mitogen-activated protein kinase/nuclear factor-κB-signalling cascades during intracellular Toxoplasma gondii infection. Immunol. Rev. 201, 191–205 (2004).

    Article  CAS  PubMed  Google Scholar 

  86. Mason, N. J., Artis, D. & Hunter, C. A. New lessons from old pathogens: what parasitic infections have taught us about the role of nuclear factor-κB in the regulation of immunity. Immunol. Rev. 201, 48–56 (2004).

    Article  CAS  PubMed  Google Scholar 

  87. Pfefferkorn, E. R. & Kasper, L. H. Toxoplasma gondii: genetic crosses reveal phenotypic suppression of hydroxyurea resistance by fluorodeoxyuridine resistance. Exp. Parasitol. 55, 207–218 (1983).

    Article  CAS  PubMed  Google Scholar 

  88. Sibley, L. D. et al. Forward genetics in Toxoplasma gondii reveals a family of rhoptry kinases that mediates pathogenesis. Eukaryot. Cell 8, 1085–1093 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Håkansson, S., Charron, A. J. & Sibley, L. D. Toxoplasma evacuoles: a two-step process of secretion and fusion forms the parasitophorous vacuole. EMBO J. 20, 3132–3144 (2001).

    Article  PubMed  PubMed Central  Google Scholar 

  90. Taylor, S. et al. A secreted serine-threonine kinase determines virulence in the eukaryotic pathogen Toxoplasma gondii. Science 314, 1776–1780 (2006). A forward-genetics analysis of acute virulence in type I T. gondii strains, revealing that ROP18 is primarily responsible for the differences between type I and avirulent type III strains.

    Article  CAS  PubMed  Google Scholar 

  91. Saeij, J. P. J. et al. Polymorphic secreted kinases are key virulence factors in toxoplasmosis. Science 314, 1780–1783 (2006). A forward-genetics analysis of the differences in mouse infectivity between type II and type III T. gondii strains; this analysis identifies ROP18 , along with several other loci.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Khan, A., Taylor, S., Ajioka, J. W., Rosenthal, B. M. & Sibley, L. D. Selection at a single locus leads to widespread expansion of Toxoplasma gondii lineages that are virulence in mice. PLoS Genet. 5, e1000404 (2009). A study of different T. gondii populations in South America finds that many of these strains share the acute virulence of type I strains and express virulent type I variants of ROP18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Saeij, J. P. J. et al. Toxoplasma co-opts host gene expression by injection of a polymorphic kinase homologue. Nature 445, 324–327 (2007). A forward-genetics investigation that leads to the discovery of ROP16, a polymorphic kinase that affects host gene expression.

    Article  CAS  PubMed  Google Scholar 

  94. Reese, M. L., Zeiner, G. M., Saeij, J. P., Boothroyd, J. C. & Boyle, J. P. Polymorphic family of injected pseudokinases is paramount in Toxoplasma virulence. Proc. Natl Acad. Sci. USA 108, 9625–9630 (2011). A forward-genetics analysis indicating that ROP5 contributes substantially to the differences between intermediate type II and avirulent type III T. gondii strains in mice.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Behnke, M. S. et al. Virulence differences in Toxoplasma mediated by amplification of a family of polymorphic pseuodokinases. Proc. Natl Acad. Sci. USA 108, 9631–9636 (2011). A forward-genetics study demonstrating that ROP5 is responsible for most of the differences in acute virulence between highly virulent type I and intermediate type II strains in mice.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Hanks, S. K. & Hunter, T. Protein kinases 6. The eukaryotic protein kinase superfamily: kinase (catalytic) domain structure and classification. FASEB J. 9, 576–596 (1995).

    Article  CAS  PubMed  Google Scholar 

  97. Boudeau, J., Miranda-Saavedra, D., Barton, G. J. & Alessi, D. R. Emerging roles of pseudokinases. Trends Cell Biol. 16, 443–452 (2006).

    Article  CAS  PubMed  Google Scholar 

  98. Peixoto, L. et al. Integrative genomics approaches highlight a family of parasite-specific kinases that regulate host responses. Cell Host Microbe 8, 208–218 (2010). A bioinformatic analysis that provides a complete summary of the 'kinome' in T. gondii and demonstrates the divergent and unique nature of the ROP kinase family.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. El Hajj, H. et al. The ROP2 family of Toxoplasma gondii rhoptry proteins: proteomic and genomic characterization and molecular modeling. Proteomics 6, 5773–5784 (2006).

    Article  CAS  PubMed  Google Scholar 

  100. Labesse, G. et al. ROP2 from Toxoplasma gondii: a virulence factor with a protein-kinase fold and no enzymatic activity. Structure 17, 139–146 (2009).

    Article  CAS  PubMed  Google Scholar 

  101. Qiu, W. et al. Novel structural and regulatory features of rhoptry secretory kinases in Toxoplasma gondii. EMBO J. 28, 969–979 (2008).

    Article  CAS  Google Scholar 

  102. Reese, M. L. & Boothroyd, J. C. A conserved non-canonical motif in the pseudoactive site of the ROP5 pseudokinase domain mediates its effect on Toxoplasma virulence. J. Biol. Chem. 286, 29366–29375 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Reese, M. L. & Boothroyd, J. C. A helical membrane-binding domain targets the Toxoplasma ROP2 family to the parasitophorous vacuole. Traffic 10, 1458–1470 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. El Hajj, H. et al. ROP18 is a rhoptry kinase controlling the intracellular proliferation of Toxoplasma gondii. PLoS Pathog. 3, e14 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Fentress, S. J., Steinfeldt, T., Howard, J. C. & Sibley, L. D. The arginine-rich N-terminal domain of ROP18 is necessary for vacuole targeting and virulence of Toxopalsma gondii. Cell. Microbiol. 20 Aug 2012 (doi:10.1111/cmi.12022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Yamamoto, M. et al. A single polymorphic amino acid on Toxoplasma gondii kinase ROP16 determines the direct and strain-specific activation of Stat3. J. Exp. Med. 206, 2747–2760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Ong, Y. C., Reese, M. L. & Boothroyd, J. C. Toxoplasma rhoptry protein 16 (ROP16) subverts host function by direct tyrosine phosphorylation of STAT6. J. Biol. Chem. 285, 28731–28740 (2010). References 106 and 107 demonstrate that the molecular targets of ROP16 include STAT3 and STAT6, and that these transcription factors are directly phosphorylated on key tyrosine residues, resulting in their prolonged activation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Butcher, B. A. et al. Toxoplasma gondii rhoptry kinase ROP16 activates STAT3 and STAT6 resulting in cytokine inhibition and arginase-1-dependent growth control. PLoS Pathog. 7, e1002236 (2011). This work shows the complex signalling that is affected by ROP16, and that this results in the inhibition of cytokine responses and can lead to the induction of parasite control.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Jensen, K. D. et al. Toxoplasma polymorphic effectors determine macrophage polarization and intestinal inflammation. Cell Host Microbe 9, 472–483 (2011). This article reports that the combination of ROP16 and GRA15 variants determines the polarization of macrophage phenotypes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Denkers, E. Y., Bzik, D. J., Fox, B. A. & Butcher, B. A. An inside job: hacking into Janus kinase/signal transducer and activator of transcription signalling cascades by the intracellular protozoan Toxoplasma gondii. Infect. Immun. 80, 476–482 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. El Kasmi, K. C. et al. Toll-like receptor-induced arginase 1 in macrophages thwarts effective immunity against intracellular pathogens. Nature Immunol. 9, 1399–1406 (2008).

    Article  CAS  Google Scholar 

  112. Rosowski, E. E. et al. Strain-specific activation of the NF-κB pathway by GRA15, a novel Toxoplasma gondii dense granule protein. J. Exp. Med. 208, 195–212 (2011). Forward-genetics mapping identifies a dense-granule protein as the factor that is primarily responsible for strain-dependent differences in NF-κB activation by T. gondii.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Kim, L. et al. Toxoplasma gondii genotype determines MyD88-dependent signalling in infected macrophages. J. Immunol. 177, 2584–2591 (2006).

    Article  CAS  PubMed  Google Scholar 

  114. Goerdt, S. et al. Alternative versus classical activation of macrophages. Pathobiology 67, 222–226 (1999).

    Article  CAS  PubMed  Google Scholar 

  115. Bekpen, C. et al. The interferon-inducible p47 (IRG) GTPases in vertebrates: loss of the cell autonomous resistance mechanism in the human lineage. Genome Biol. 6, R92 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Khaminets, A. et al. Coordinated loading of IRG resistance GTPases on to the Toxoplasma gondii parasitophorous vacuole. Cell. Microbiol. 12, 939–961 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Martens, S. et al. Disruption of Toxoplasma gondii parasitophorous vacuoles by the mouse p47-resistance GTPases. Plos Pathog. 1, e24 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Zhao, Z. et al. Autophagosome-independent essential function for the autophagy protein Atg5 in cellular immunity to intracellular pathogens. Cell Host Microbe 4, 458–469 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Zhao, Y. et al. Virulent Toxoplasma gondii evade immunity-related GTPas-mediated parasite vacuole disruption within primed macrophages. J. Immunol. 182, 3775–3781 (2009). Together with reference 116, this work demonstrates the correlation between virulent strains of T. gondii and the avoidance of IRG recruitment to the PVM.

    Article  CAS  PubMed  Google Scholar 

  120. Fentress, S. J. et al. Phosphorylation of immunity-related GTPases by a parasite secretory kinase promotes macrophage survival and virulence. Cell Host Microbe 16, 484–495 (2010). A study showing that ROP18 phosphorylates IRG proteins in activated macrophages, thus protecting the parasite against clearance.

    Article  CAS  Google Scholar 

  121. Steinfeldt, T. et al. Phosphorylation of mouse immunity-related GTPase (IRG) resistance proteins is an evasion strategy for virulent Toxoplasma gondii. PLoS Biol. 8, e1000576 (2010). The specific sites in Irga6 that are phosphorylated by ROP18 are shown to disrupt GTPase activity and prevent oligomerization of this IRG.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Martens, S. et al. Mechanisms regulating the positioning of mouse p47 resistance GTPases LRG-47 and IIGP1 on cellular membranes: retargeting to plasma membrane induced by phagocytosis. J. Immunol. 173, 2594–2606 (2004).

    Article  CAS  PubMed  Google Scholar 

  123. Yamamoto, M. et al. ATF6β is a host cellular target of the Toxoplasma gondii virulence factor ROP18. J. Exp. Med. 208, 1533–1546 (2011). Using a yeast two-hybrid screen, these authors find that ROP18 phosphorylates ATF6β.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Fleckenstein, M. C. et al. A Toxoplasma gondii pseudokinase inhibits host IRG resistance proteins. PLoS Biol. 10, e1001358 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Niedelman, W. et al. The rhoptry proteins ROP18 and ROP5 mediate Toxoplasma gondii evasion of the murine, but not the human, interferon-gamma response. PLoS Pathog. 8, e1002784 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Behnke, M. S. et al. The polymorphic pseudokinase ROP5 controls virulence in Toxoplasma gondii by regulating the active kinase ROP18. PLoS Pathog. (in the press).

  127. Zeqiraj, E. & van Aalten, D. M. Pseudokinases-remnants of evolution or key allosteric regulators? Curr. Opin. Struct. Biol. 20, 772–781 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Gavrilescu, L. C. & Denkers, E. Y. IFN-γ overproduction and high level apoptosis are associated with high but not low virulence Toxoplasma gondii infection. J. Immunol. 167, 902–909 (2001).

    Article  CAS  PubMed  Google Scholar 

  129. Mordue, D. G., Monroy, F., La Regina, M., Dinarello, C. A. & Sibley, L. D. Acute toxoplasmosis leads to lethal overproduction of Th1 cytokines. J. Immunol. 167, 4574–4584 (2001).

    Article  CAS  PubMed  Google Scholar 

  130. Frenkel, J. K. Host, strain and treatment variation as factors in the pathogenesis of toxoplasmosis. Am. J. Trop. Med. Hyg. 2, 390–415 (1953).

    Article  CAS  PubMed  Google Scholar 

  131. Dubey, J. P. & Frenkel, J. K. Toxoplasmosis of rats: a review, with considerations of their value as an animal model and their possible role in epidemiology. Vet. Parasitol. 77, 1–32 (1998).

    Article  CAS  PubMed  Google Scholar 

  132. Dubey, J. P. Toxoplasma gondii infections in chickens (Gallus domesticus): prevalence, clinical disease, diagnosis, and public health significance. Zoonoses Public Health 57, 60–73 (2008).

    Article  Google Scholar 

  133. Backert, S. & Meyer, T. F. Type IV secretion systems and their effectors in bacterial pathogenesis. Curr. Opin. Microbiol. 9, 207–217 (2006).

    Article  CAS  PubMed  Google Scholar 

  134. Cascales, E. & Christie, P. J. The versatile bacterial type IV secretion systems. Nature Rev. Microbiol. 1, 137–149 (2003).

    Article  CAS  Google Scholar 

  135. Erhardt, M., Namba, K. & Hughes, K. T. Bacterial nanomachines: the flagellum and type III injectisome. Cold Spring Harb. Perspect. Biol. 2, a000299 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Koshy, A. A. et al. Toxoplasma co-opts host cells it does not invade. PLoS Pathog. 8, e1002825 (2012). An article describing T. gondii -mediated injection of ROP proteins into cells that the parasite does not directly infect; this activity potentially alters the behaviour of uninfected cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Boothroyd, J. C. & Dubremetz, J. F. Kiss and spit: the dual roles of Toxoplasma rhoptries. Nature Rev. Microbiol. 6, 79–88 (2008).

    Article  CAS  Google Scholar 

  138. Carruthers, V. B. & Tomley, F. M. Microneme proteins in apicomplexans. Subcell. Biochem. 47, 33–45 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  139. Bradley, P. J. et al. Proteomic analysis of rhoptry organelles reveals many novel constituents for host-parasite interactions in T. gondii. J. Biol. Chem. 280, 34245–34258 (2005).

    Article  CAS  PubMed  Google Scholar 

  140. Besteiro, S., Dubremetz, J. F. & Lebrun, M. The moving junction of apicomplexan parasites: a key structure for invasion. Cell. Microbiol. 13, 797–805 (2011).

    Article  CAS  PubMed  Google Scholar 

  141. Shen, B. & Sibley, L. D. The moving junction, a key portal to host cell invasion by apicomplexan parasites. Curr. Opin. Microbiol. 15, 449–455 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Bradley, P. J. & Sibley, L. D. Rhoptries: an arsenal of secreted virulence factors. Curr. Opin. Microbiol. 10, 582–587 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Sibley, L. D. Development of forward genetics in Toxoplasma gondii. Int. J. Parasitol. 39, 915–924 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Khan, A. et al. Composite genome map and recombination parameters derived from three archetypal lineages of Toxoplasma gondii. Nucleic Acids Res. 33, 2980–2992 (2005). Genetic linkage maps of T. gondii are constructed from three pairwise crosses between strain types I, II and III, and enable the discovery of genes controlling important traits such as virulence and host gene transcription.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Churchill, G. A. & Doerge, R. W. Empirical threshold values for quantitative trait mapping. Genetics 138, 963–971 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Barragan, A. & Sibley, L. D. Migration of Toxoplasma gondii across biological barriers. Trends Microbiol. 11, 426–430 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Work in the authors' laboratories is supported by the US National Institutes of Health (L.D.S. and C.A.H.) and the State of Pennsylvania, USA (C.A.H.). The authors regret not being able to cite all of the appropriate primary literature owing to space limitations.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information Table S1

Major mediators of resistance to T. gondii as defined by genetic knockouts in the mouse. (PDF 315 kb)

Supplementary information Table S1

Host pathways altered by T. gondii infection (PDF 306 kb)

Related links

Related links

FURTHER INFORMATION

Protein Data Bank

Glossary

Apicomplexans

Protozoans of the phylum Apicomplexa that are known for their apical complex consisting of a specialized microtubule-organizing centre, called the conoid, and secretory organelles involved in host cell invasion. Toxoplasma spp. belong to the tissue cyst-forming coccidian group of apicomplexans.

Oocysts

The diploid stages of parasite development, resulting from the fusion of Toxoplasma gondii gametes. Oocysts are shed in cat faeces and thus contaminate the environment, giving rise to infection by accidental ingestion.

Tachyzoites

The rapidly replicating intracellular forms of many tissue cyst-forming coccidians, such as Toxoplasma gondii. The name is derived from the Greek tachys, meaning fast.

Bradyzoites

The slow-growing forms of certain parasites; these forms reside within long-lived tissue cysts that are associated with chronic infection. The bradyzoite is one of the parasite stages that is specialized for transmission if ingested. The name is derived from the Greek bradys, meaning slow.

Micronemes

Organelles that are discharged in response to elevated cytoplasmic calcium levels. Microneme proteins mediate parasite adhesion to substrates and host cells.

Toll-like receptors

(TLRs). Pattern recognition receptors that mediate immune responses by detecting pathogen-associated molecular patterns.

Glycosylphosphatidylinositol-anchored proteins

Proteins with a common glycolipid anchor that is covalently attached to the carboxyl terminus as a post-translational modification.

Inducible nitric oxide synthase

(iNOS). A soluble enzyme that produces nitric oxide from l-arginine. This enzyme is upregulated by interferon-γ and tumour necrosis factor, and constitutes a major antimicrobial activity of macrophages. It is encoded by NOS2.

Immunity-related GTPases

(IRGs). A family of 45–47kDa GTPases that are strongly upregulated by interferon-γ and contribute to resistance to intracellular pathogens. These GTPases are ubiquitous in the mouse, but are more rare in other vertebrates and largely absent in humans.

p67 guanylate-binding proteins

(GBPs). A family of 65 kDa GTPases that are upregulated by interferon-γ and contribute to resistance to intracellular pathogens. These GTPases are widely distributed in vertebrates, indicating that they might have an important general role in resistance to intracellular pathogens.

Major histocompatibility complex

A cell surface molecule that allows recognition of epitopes from foreign or self antigens through presentation of these antigens to T cells.

Signal transducer and activator of transcription 1

(STAT1). One of a family of transcription factors that are activated by Janus kinases (JAKs) and regulate gene expression by binding to nuclear promoters. STATs have a role in development and the immune system.

Nuclear factor-κB

(NF-κB). A complex that controls DNA transcription in response to various signalling inputs and has a key role in regulating innate and adaptive immune responses.

Suppressor of cytokine signalling

(SOCS). A family of intracellular proteins that regulate cytokine signalling by either direct inhibition of receptors or increased degradation of signalling proteins.

Tumour necrosis factor

(TNF). A cytokine involved in inflammation, tumour suppression and host defence.

Pseudokinase

A protein that contains a conserved protein kinase fold but lacks key residues in the nucleotide-binding pocket, resulting in an inability to transfer phosphate to a donor substrate. Although these proteins are not catalytically active, they serve regulatory or scaffolding roles.

Quantitative trait locus

(QTL). A genetic region (or locus) that contributes to a quantitative trait (a trait that varies by degree). Such traits are typically polygenic, with multiple QTLs contributing to the phenotype.

Mitogen-activated protein kinases

(MAPKs). Serine/threonine kinases that are involved in cellular signalling in response to a diverse array of stimuli.

Centimorgan

A genetic unit used for establishing genetic linkage based on recombination frequency in the progeny of a genetic cross. 1 centimorgan is equal to the distance over which the average proportion of recombination events in a single generation is 1%. Named in honour of Thomas Hunt Morgan.

Arginase

An enzyme that mediates the consumption of arginine in the urea cycle and thus has an antagonistic role to nitric oxide synthase, as they both rely on the same substrate.

Alternatively activated macrophages

Macrophages that have a T helper 2 type phenotype, dampening inflammation and promoting repair; these cells also typically express arginase. This is one of two polar phenotypes that macrophages can adopt following stimulation by cytokines.

TIR domain-containing adaptor inducing interferon-β

(TRIF). An adaptor protein for signalling from Toll-like receptors, which also contain TIR domains.

TNF receptor-associated factor 6

(TRAF6). A protein adaptor that is involved in signalling through tumour necrosis factor (TNF), interleukin-1 and Toll-like receptors.

IκB kinase

(IKK). A kinase comprising two catalytic subunits, IKKα and IKKβ, and a regulatory subunit, IKKγ (also known as NEMO). IKK targets inhibitor of NF-κB (IκB) to induce its degradation, allowing nuclear factor-κB (NF-κB) to translocate to the nucleus.

Inhibitor of NF-κB

(IκB). A protein that binds nuclear factor-κB (NF-κB), preventing its translocation to the nucleus. Under certain conditions, IκB becomes phosphorylated by inhibitor of IκB (IKK), and this triggers IκB ubiquitylation and proteasome-mediated degradation allowing active NF-κB to translocate to the nucleus.

Classically activated macrophage

A macrophages that has a T helper 1 type phenotype, promoting inflammation and the expression of nitric oxide synthase. This is one of two polar phenotypes that macrophages can adopt following stimulation by cytokines.

Unfolded-protein response

A cellular stress response triggered by unfolded proteins accumulating in the ER. This response leads to stalled translation and to upregulation of chaperones for protein folding.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hunter, C., Sibley, L. Modulation of innate immunity by Toxoplasma gondii virulence effectors. Nat Rev Microbiol 10, 766–778 (2012). https://doi.org/10.1038/nrmicro2858

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrmicro2858

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology