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:

Innate sensing of malaria parasites

Key Points

  • Repeated cycles of Plasmodium invasion, replication and release from red blood cells result in the exponential growth of the parasite population, coinciding with a febrile paroxysms and other signs of malaria.

  • The mechanisms underlying the pathogenesis of malaria involve an overwhelming activation of innate immune cells by Plasmodium components and the systemic release of inflammatory mediators.

  • Plasmodium-derived glycosylphosphatidylinositol anchors, haemozoin and nucleic acids are sensed by distinct pattern recognition receptors (PRRs) in specific cellular compartments of macrophages and dendritic cells resulting in the release of inflammatory mediators.

  • Haemozoin acts as a vehicle to transport plasmodial DNA into phagolysosomes and, thereafter, to the host cell cytoplasm where it can be sensed by TLR9 and cytosolic innate immune receptors, respectively.

  • Interferon-γ priming promotes Toll-like receptor hyperresponsiveness and inflammasome assembly, forming the basis of systemic inflammation that is induced by Plasmodium components or microbial superinfection during malaria.

  • Further investigation of the role of PRRs in malaria may help to identify more potent and better-tolerated adjuvants for the development of a truly effective anti-malaria vaccine.

Abstract

Innate immune receptors have a key role in immune surveillance by sensing microorganisms and initiating protective immune responses. However, the innate immune system is a classic 'double-edged sword' that can overreact to pathogens, which can have deleterious effects and lead to clinical manifestations. Recent studies have unveiled the complexity of innate immune receptors that function as sensors of Plasmodium spp. in the vertebrate host. This Review highlights the cellular and molecular mechanisms by which Plasmodium infection is sensed by different families of innate immune receptors. We also discuss how these events mediate both host resistance to infection and the pathogenesis of malaria.

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: Plasmodium life cycle and the pathogenesis of malaria.
Figure 2: Innate sensors of Plasmodium PAMPs and malaria DAMPs.
Figure 3: Pattern recognition receptors and the pathophysiology of malaria.

Similar content being viewed by others

References

  1. Murray, C. J. et al. Global malaria mortality between 1980 and 2010: a systematic analysis. Lancet 379, 413–431 (2012).

    PubMed  Google Scholar 

  2. Gething, P. W. et al. A long neglected world malaria map: Plasmodium vivax endemicity in 2010. PLoS Negl. Trop. Dis. 6, e1814 (2012).

    PubMed  PubMed Central  Google Scholar 

  3. Miller, L. H., Ackerman, H. C., Su, X. Z. & Wellems, T. E. Malaria biology and disease pathogenesis: insights for new treatments. Nature Med. 19, 156–167 (2013).

    CAS  PubMed  Google Scholar 

  4. Golgi, C. On the cycle of development of malarial parasites in tertian fever: differential diagnosis between the intracellular parasites of tertian and quartant fever. Archivio per le Scienza Mediche 13, 173–196 (Italian) (1889).

    Google Scholar 

  5. O'Neill, L. A., Golenbock, D. & Bowie, A. G. The history of Toll-like receptors — redefining innate immunity. Nature Rev. Immunol. 13, 453–460 (2013).

    CAS  Google Scholar 

  6. Barbalat, R., Ewald, S. E., Mouchess, M. L. & Barton, G. M. Nucleic acid recognition by the innate immune system. Annu. Rev. Immunol. 29, 185–214 (2011).

    CAS  PubMed  Google Scholar 

  7. Schroder, K. & Tschopp, J. The inflammasomes. Cell 140, 821–832 (2010).

    CAS  PubMed  Google Scholar 

  8. Takeuchi, O. & Akira, S. Pattern recognition receptors and inflammation. Cell 140, 805–820 (2010).

    CAS  PubMed  Google Scholar 

  9. 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).

    CAS  Google Scholar 

  10. Schofield, L. & Grau, G. E. Immunological processes in malaria pathogenesis. Nature Rev. Immunol. 5, 722–735 (2005).

    CAS  Google Scholar 

  11. Clark, I. A., Budd, A. C., Alleva, L. M. & Cowden, W. B. Human malarial disease: a consequence of inflammatory cytokine release. Malar. J. 5, 85 (2006).

    PubMed  PubMed Central  Google Scholar 

  12. Del Portillo, H. A. et al. The role of the spleen in malaria. Cell. Microbiol. 14, 343–355 (2012).

    CAS  PubMed  Google Scholar 

  13. Buffet, P. A. et al. The pathogenesis of Plasmodium falciparum malaria in humans: insights from splenic physiology. Blood 117, 381–392 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Franklin, B. S. et al. MyD88-dependent activation of dendritic cells and CD4+ T lymphocytes mediates symptoms, but is not required for the immunological control of parasites during rodent malaria. Microbes Infect. 9, 881–890 (2007).

    CAS  PubMed  Google Scholar 

  15. Franklin, B. S. et al. Malaria primes the innate immune response due to interferon-γ induced enhancement of Toll-like receptor expression and function. Proc. Natl Acad. Sci. USA 106, 5789–5794 (2009). This study describes the mechanism of pro-inflammatory priming during both mouse and human malaria.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Ataide, M. A. et al. Malaria-induced NLRP12/NLRP3-dependent caspase-1 activation mediates inflammation and hypersensitivity to bacterial superinfection. PLoS Pathog. 10, e1003885 (2014). This study describes the mechanism by which inflammasomes are induced in vivo and the relevance of this process to hypersensitivity to bacterial infection during malaria.

    PubMed  PubMed Central  Google Scholar 

  17. Cunnington, A. J., Riley, E. M. & Walther, M. Stuck in a rut? Reconsidering the role of parasite sequestration in severe malaria syndromes. Trends Parasitol. 29, 585–592 (2013).

    PubMed  Google Scholar 

  18. Chang, K. H. & Stevenson, M. M. Malarial anaemia: mechanisms and implications of insufficient erythropoiesis during blood-stage malaria. Int. J. Parasitol. 34, 1501–1516 (2004).

    CAS  PubMed  Google Scholar 

  19. Awandare, G. A. et al. Mechanisms of erythropoiesis inhibition by malarial pigment and malaria-induced proinflammatory mediators in an in vitro model. Am. J. Hematol. 86, 155–162 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Dinarello, C. A. Immunological and inflammatory functions of the interleukin-1 family. Annu. Rev. Immunol. 27, 519–550 (2009).

    CAS  PubMed  Google Scholar 

  21. Tracey, K. J. & Cerami, A. Tumor necrosis factor: a pleiotropic cytokine and therapeutic target. Annu. Rev. Med. 45, 491–503 (1994).

    CAS  PubMed  Google Scholar 

  22. Wagner-Jauregg, J. & Bruetsch, W. L. The history of the malaria treatment of general paralysis. Am. J. Psychiatry 102, 577–582 (1946).

    CAS  PubMed  Google Scholar 

  23. Taylor, W. R., Hanson, J., Turner, G. D., White, N. J. & Dondorp, A. M. Respiratory manifestations of malaria. Chest 142, 492–505 (2012).

    PubMed  Google Scholar 

  24. Van den Steen, P. E. et al. Pathogenesis of malaria-associated acute respiratory distress syndrome. Trends Parasitol. 29, 346–358 (2013).

    CAS  PubMed  Google Scholar 

  25. White, N. J., Turner, G. D., Day, N. P. & Dondorp, A. M. Lethal malaria: Marchiafava and Bignami were right. J. Infect. Dis. 208, 192–198 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Scherf, A., Lopez-Rubio, J. J. & Riviere, L. Antigenic variation in Plasmodium falciparum. Annu. Rev. Microbiol. 62, 445–470 (2008).

    CAS  PubMed  Google Scholar 

  27. Kraemer, S. M. & Smith, J. D. A family affair: var genes, PfEMP1 binding, and malaria disease. Curr. Opin. Microbiol. 9, 374–380 (2006).

    CAS  PubMed  Google Scholar 

  28. Shikani, H. J. et al. Cerebral malaria: we have come a long way. Am. J. Pathol. 181, 1484–1492 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Idro, R., Jenkins, N. E. & Newton, C. R. Pathogenesis, clinical features, and neurological outcome of cerebral malaria. Lancet Neurol. 4, 827–840 (2005).

    PubMed  Google Scholar 

  30. Rogerson, S. J., Hviid, L., Duffy, P. E., Leke, R. F. & Taylor, D. W. Malaria in pregnancy: pathogenesis and immunity. Lancet Infect. Dis. 7, 105–117 (2007).

    CAS  PubMed  Google Scholar 

  31. Dondorp, A. M. et al. Direct in vivo assessment of microcirculatory dysfunction in severe falciparum malaria. J. Infect. Dis. 197, 79–84 (2008).

    CAS  PubMed  Google Scholar 

  32. Francischetti, I. M. et al. Plasmodium falciparum-infected erythrocytes induce tissue factor expression in endothelial cells and support the assembly of multimolecular coagulation complexes. J. Thromb. Haemost. 5, 155–165 (2007).

    CAS  PubMed  Google Scholar 

  33. Anstey, N. M. et al. Nitric oxide in Tanzanian children with malaria: inverse relationship between malaria severity and nitric oxide production/nitric oxide synthase type 2 expression. J. Exp. Med. 184, 557–567 (1996).

    CAS  PubMed  Google Scholar 

  34. Ong, P. K. et al. Nitric oxide synthase dysfunction contributes to impaired cerebroarteriolar reactivity in experimental cerebral malaria. PLoS Pathog. 9, e1003444 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Janeway, C. A. Jr. Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb. Symp. Quant. Biol. 54, 1–13 (1989).

    CAS  PubMed  Google Scholar 

  36. Almeida, I. C. & Gazzinelli, R. T. Proinflammatory activity of glycosylphosphatidylinositol anchors derived from Trypanosoma cruzi: structural and functional analyses. J. Leukoc. Biol. 70, 467–477 (2001).

    CAS  PubMed  Google Scholar 

  37. Krishnegowda, G. et al. Induction of proinflammatory responses in macrophages by the glycosylphosphatidylinositols of Plasmodium falciparum: cell signaling receptors, glycosylphosphatidylinositol (GPI) structural requirement, and regulation of GPI activity. J. Biol. Chem. 280, 8606–8616 (2005). This study demonstrates that malaria GPI anchors induce pro-inflammatory cytokines via TLR1–TLR2 heterodimers.

    CAS  PubMed  Google Scholar 

  38. Durai, P., Govindaraj, R. G. & Choi, S. Structure and dynamic behavior of Toll-like receptor 2 subfamily triggered by malarial glycosylphosphatidylinositols of Plasmodium falciparum. FEBS J. 280, 6196–6212 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Schofield, L. & Hackett, F. Signal transduction in host cells by a glycosylphosphatidylinositol toxin of malaria parasites. J. Exp. Med. 177, 145–153 (1993).

    CAS  PubMed  Google Scholar 

  40. Tachado, S. D. et al. Glycosylphosphatidylinositol toxin of Plasmodium induces nitric oxide synthase expression in macrophages and vascular endothelial cells by a protein tyrosine kinase-dependent and protein kinase C-dependent signaling pathway. J. Immunol. 156, 1897–1907 (1996).

    CAS  PubMed  Google Scholar 

  41. Schofield, L. et al. Glycosylphosphatidylinositol toxin of Plasmodium up-regulates intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selectin expression in vascular endothelial cells and increases leukocyte and parasite cytoadherence via tyrosine kinase-dependent signal transduction. J. Immunol. 156, 1886–1896 (1996).

    CAS  PubMed  Google Scholar 

  42. Jani, D. et al. HDP — a novel heme detoxification protein from the malaria parasite. PLoS Pathog. 4, e1000053 (2008).

    PubMed  PubMed Central  Google Scholar 

  43. Nguyen, P. H., Day, N., Pram, T. D., Ferguson, D. J. & White, N. J. Intraleucocytic malaria pigment and prognosis in severe malaria. Trans. R. Soc. Trop. Med. Hyg. 89, 200–204 (1995).

    CAS  PubMed  Google Scholar 

  44. Jaramillo, M. et al. Synthetic Plasmodium-like hemozoin activates the immune response: a morphology - function study. PLoS ONE 4, e6957 (2009).

    PubMed  PubMed Central  Google Scholar 

  45. Parroche, P. et al. Malaria hemozoin is immunologically inert but radically enhances innate responses by presenting malaria DNA to Toll-like receptor 9. Proc. Natl Acad. Sci. USA 104, 1919–1924 (2007). This study demonstrates the importance of Plasmodium DNA in the activation of TLR9 by native haemozoin.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Shio, M. T. et al. Malarial hemozoin activates the NLRP3 inflammasome through Lyn and Syk kinases. PLoS Pathog. 5, e1000559 (2009).

    PubMed  Google Scholar 

  47. Dostert, C. et al. Malarial hemozoin is a Nalp3 inflammasome activating danger signal. PLoS ONE 4, e6510 (2009). References 46 and 47 demonstrate that haemozoin crystals activate NLRP3 inflammasomes.

    PubMed  PubMed Central  Google Scholar 

  48. Coban, C. et al. Toll-like receptor 9 mediates innate immune activation by the malaria pigment hemozoin. J. Exp. Med. 201, 19–25 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Giribaldi, G. et al. Involvement of inflammatory chemokines in survival of human monocytes fed with malarial pigment. Infect. Immun. 78, 4912–4921 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Skorokhod, O. A., Schwarzer, E., Ceretto, M. & Arese, P. Malarial pigment haemozoin, IFN-γ, TNF-α, IL-1β and LPS do not stimulate expression of inducible nitric oxide synthase and production of nitric oxide in immuno-purified human monocytes. Malar. J. 6, 73 (2007).

    PubMed  PubMed Central  Google Scholar 

  51. Goldie, P., Roth, E. F. Jr., Oppenheim, J. & Vanderberg, J. P. Biochemical characterization of Plasmodium falciparum hemozoin. Am. J. Trop. Med. Hyg. 43, 584–596 (1990).

    CAS  PubMed  Google Scholar 

  52. Barrera, V. et al. Host fibrinogen stably bound to hemozoin rapidly activates monocytes via TLR-4 and CD11b/CD18-integrin: a new paradigm of hemozoin action. Blood 117, 5674–5682 (2011).

    CAS  PubMed  Google Scholar 

  53. Kalantari, P. et al. Dual engagement of the NLRP3 and AIM2 inflammasomes by Plasmodium-derived hemozoin and DNA during malaria. Cell Rep. 6, 196–210 (2014). This study describes the trafficking of haemozoin in phagocytic cells and the relevance of this process for activating endosomal and cytosolic innate immune receptors.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Coban, C. et al. Immunogenicity of whole-parasite vaccines against Plasmodium falciparum involves malarial hemozoin and host TLR9. Cell Host Microbe 7, 50–61 (2010). This study proposes the use of haemozoin as a vaccine adjuvant.

    CAS  PubMed  Google Scholar 

  55. Torgbor, C. et al. A multifactorial role for P. falciparum malaria in endemic Burkitt's lymphoma pathogenesis. PLoS Pathog. 10, e1004170 (2014).

    PubMed  PubMed Central  Google Scholar 

  56. Reimer, T. et al. Experimental cerebral malaria progresses independently of the Nlrp3 inflammasome. Eur. J. Immunol. 40, 764–769 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Rock, K. L., Latz, E., Ontiveros, F. & Kono, H. The sterile inflammatory response. Annu. Rev. Immunol. 28, 321–342 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Griffith, J. W., Sun, T., McIntosh, M. T. & Bucala, R. Pure hemozoin is inflammatory in vivo and activates the NALP3 inflammasome via release of uric acid. J. Immunol. 183, 5208–5220 (2009).

    CAS  PubMed  Google Scholar 

  59. Caetano, B. C. et al. Requirement of UNC93B1 reveals a critical role for TLR7 in host resistance to primary infection with Trypanosoma cruzi. J. Immunol. 187, 1903–1911 (2011).

    CAS  PubMed  Google Scholar 

  60. Schamber-Reis, B. L. et al. UNC93B1 and nucleic acid-sensing Toll-like receptors mediate host resistance to infection with Leishmania major. J. Biol. Chem. 288, 7127–7136 (2013).

    PubMed  PubMed Central  Google Scholar 

  61. Andrade, W. A. et al. Combined action of nucleic acid-sensing Toll-like receptors and TLR11/TLR12 heterodimers imparts resistance to Toxoplasma gondii in mice. Cell Host Microbe 13, 42–53 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Pichyangkul, S. et al. Malaria blood stage parasites activate human plasmacytoid dendritic cells and murine dendritic cells through a Toll-like receptor 9-dependent pathway. J. Immunol. 172, 4926–4933 (2004).

    CAS  PubMed  Google Scholar 

  63. Wu, X., Gowda, N. M., Kumar, S. & Gowda, D. C. Protein–DNA complex is the exclusive malaria parasite component that activates dendritic cells and triggers innate immune responses. J. Immunol. 184, 4338–4348 (2010).

    CAS  PubMed  Google Scholar 

  64. Sharma, S. et al. Innate immune recognition of an AT-rich stem-loop DNA motif in the Plasmodium falciparum genome. Immunity 35, 194–207 (2011). This study defines the importance of AT-rich DNA motifs from the P. falciparum genome in inducing type I IFN production via the STING pathway.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Anstey, N. M., Douglas, N. M., Poespoprodjo, J. R. & Price, R. N. Plasmodium vivax: clinical spectrum, risk factors and pathogenesis. Adv. Parasitol. 80, 151–201 (2012).

    PubMed  Google Scholar 

  66. Wu, J. et al. Strain-specific innate immune signaling pathways determine malaria parasitemia dynamics and host mortality. Proc. Natl Acad. Sci. USA 111, E511–E520 (2014). This study demonstrates the importance of parasite strain on the activation of various innate immune receptors during Plasmodium infection.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Liehl, P. et al. Host-cell sensors for Plasmodium activate innate immunity against liver-stage infection. Nature Med. 20, 47–53 (2014). This study demonstrates the ability of sporozoite RNA to induce type I IFN in an MDA5-dependent manner.

    CAS  PubMed  Google Scholar 

  68. Baccarella, A., Fontana, M. F., Chen, E. C. & Kim, C. C. Toll-like receptor 7 mediates early innate immune responses to malaria. Infect. Immun. 81, 4431–4442 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Miller, J. L., Sack, B. K., Baldwin, M., Vaughan, A. M. & Kappe, S. H. Interferon-mediated innate immune responses against malaria parasite liver stages. Cell Rep. 7, 436–447 (2014). This study describes the mechanism by which type I IFNs mediate control of Plasmodium replication in the liver.

    CAS  PubMed  Google Scholar 

  70. Matzinger, P. Tolerance, danger, and the extended family. Annu. Rev. Immunol. 12, 991–1045 (1994).

    CAS  PubMed  Google Scholar 

  71. Mantel, P. Y. & Marti, M. The role of extracellular vesicles in Plasmodium and other protozoan parasites. Cell. Microbiol. 16, 344–354 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Gallego-Delgado, J., Ty, M., Orengo, J. M., van de Hoef, D. & Rodriguez, A. A surprising role for uric acid: the inflammatory malaria response. Curr. Rheumatol Rep. 16, 401 (2014).

    PubMed  PubMed Central  Google Scholar 

  73. Figueiredo, R. T. et al. Characterization of heme as activator of Toll-like receptor 4. J. Biol. Chem. 282, 20221–20229 (2007).

    CAS  PubMed  Google Scholar 

  74. Butt, A. N. & Swaminathan, R. Overview of circulating nucleic acids in plasma/serum. Ann. NY Acad. Sci. 1137, 236–242 (2008).

    CAS  PubMed  Google Scholar 

  75. Barrat, F. J. & Coffman, R. L. Development of TLR inhibitors for the treatment of autoimmune diseases. Immunol. Rev. 223, 271–283 (2008).

    CAS  PubMed  Google Scholar 

  76. Franklin, B. S. et al. Plasma circulating nucleic acids levels increase according to the morbidity of Plasmodium vivax malaria. PLoS ONE 6, e19842 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Orengo, J. M. et al. Plasmodium-induced inflammation by uric acid. PLoS Pathog. 4, e1000013 (2008). This study indicates the potential role of uric acid as a pro-inflammatory agent during malaria.

    PubMed  PubMed Central  Google Scholar 

  78. Lopera-Mesa, T. M. et al. Plasma uric acid levels correlate with inflammation and disease severity in Malian children with Plasmodium falciparum malaria. PLoS ONE 7, e46424 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Couper, K. N. et al. Parasite-derived plasma microparticles contribute significantly to malaria infection-induced inflammation through potent macrophage stimulation. PLoS Pathog. 6, e1000744 (2010).

    PubMed  PubMed Central  Google Scholar 

  80. Combes, V. et al. Circulating endothelial microparticles in malawian children with severe falciparum malaria complicated with coma. JAMA 291, 2542–2544 (2004).

    CAS  PubMed  Google Scholar 

  81. Campos, F. M. et al. Augmented plasma microparticles during acute Plasmodium vivax infection. Malar. J. 9, 327 (2010).

    PubMed  PubMed Central  Google Scholar 

  82. Wassmer, S. C., Combes, V., Candal, F. J., Juhan-Vague, I. & Grau, G. E. Platelets potentiate brain endothelial alterations induced by Plasmodium falciparum. Infect. Immun. 74, 645–653 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Wagener, F. A. et al. Different faces of the heme-heme oxygenase system in inflammation. Pharmacol. Rev. 55, 551–571 (2003).

    CAS  PubMed  Google Scholar 

  84. Pamplona, A. et al. Heme oxygenase-1 and carbon monoxide suppress the pathogenesis of experimental cerebral malaria. Nature Med. 13, 703–710 (2007).

    CAS  PubMed  Google Scholar 

  85. Ferreira, A. et al. Sickle hemoglobin confers tolerance to Plasmodium infection. Cell 145, 398–409 (2011). References 84 and 85 demonstrate the importance of haem oxygenase 1 in protecting against the toxic effects of haem during malaria.

    CAS  PubMed  Google Scholar 

  86. Yeo, T. W. et al. Increased carboxyhemoglobin in adult falciparum malaria is associated with disease severity and mortality. J. Infect. Dis. 208, 813–817 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Gozzelino, R. et al. Metabolic adaptation to tissue iron overload confers tolerance to malaria. Cell Host Microbe 12, 693–704 (2012).

    CAS  PubMed  Google Scholar 

  88. Riley, E. M. & Stewart, V. A. Immune mechanisms in malaria: new insights in vaccine development. Nature Med. 19, 168–178 (2013).

    CAS  PubMed  Google Scholar 

  89. Stevenson, M. M. & Riley, E. M. Innate immunity to malaria. Nature Rev. Immunol. 4, 169–180 (2004).

    CAS  Google Scholar 

  90. Sexton, A. C. et al. Transcriptional profiling reveals suppressed erythropoiesis, up-regulated glycolysis, and interferon-associated responses in murine malaria. J. Infect. Dis. 189, 1245–1256 (2004).

    CAS  PubMed  Google Scholar 

  91. Ockenhouse, C. F. et al. Common and divergent immune response signaling pathways discovered in peripheral blood mononuclear cell gene expression patterns in presymptomatic and clinically apparent malaria. Infect. Immun. 74, 5561–5573 (2006). This study characterizes the expression of pro-inflammatory genes during human malaria.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Bastos, K. R. et al. Impaired macrophage responses may contribute to exacerbation of blood-stage Plasmodium chabaudi chabaudi malaria in interleukin-12-deficient mice. J. Interferon Cytokine Res. 22, 1191–1199 (2002).

    CAS  PubMed  Google Scholar 

  93. Artavanis-Tsakonas, K. & Riley, E. M. Innate immune response to malaria: rapid induction of IFN-γ from human NK cells by live Plasmodium falciparum-infected erythrocytes. J. Immunol. 169, 2956–2963 (2002).

    CAS  PubMed  Google Scholar 

  94. Walther, M. et al. Innate immune responses to human malaria: heterogeneous cytokine responses to blood-stage Plasmodium falciparum correlate with parasitological and clinical outcomes. J. Immunol. 177, 5736–5745 (2006).

    CAS  PubMed  Google Scholar 

  95. Sponaas, A. M. et al. Migrating monocytes recruited to the spleen play an important role in control of blood stage malaria. Blood 114, 5522–5531 (2009).

    CAS  PubMed  Google Scholar 

  96. Sedegah, M., Finkelman, F. & Hoffman, S. L. Interleukin 12 induction of interferon γ-dependent protection against malaria. Proc. Natl Acad. Sci. USA 91, 10700–10702 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Stevenson, M. M., Tam, M. F., Wolf, S. F. & Sher, A. IL-12-induced protection against blood-stage Plasmodium chabaudi AS requires IFN-γ and TNF-α and occurs via a nitric oxide-dependent mechanism. J. Immunol. 155, 2545–2556 (1995).

    CAS  PubMed  Google Scholar 

  98. Su, Z. & Stevenson, M. M. IL-12 is required for antibody-mediated protective immunity against blood-stage Plasmodium chabaudi AS malaria infection in mice. J. Immunol. 168, 1348–1355 (2002).

    CAS  PubMed  Google Scholar 

  99. Wykes, M. N. & Good, M. F. What really happens to dendritic cells during malaria? Nature Rev. Microbiol. 6, 864–870 (2008).

    CAS  Google Scholar 

  100. Ing, R., Segura, M., Thawani, N., Tam, M. & Stevenson, M. M. Interaction of mouse dendritic cells and malaria-infected erythrocytes: uptake, maturation, and antigen presentation. J. Immunol. 176, 441–450 (2006). This study demonstrates the activation of DCs by infected RBCs.

    CAS  PubMed  Google Scholar 

  101. Torgler, R. et al. Sporozoite-mediated hepatocyte wounding limits Plasmodium parasite development via MyD88-mediated NF-κ B activation and inducible NO synthase expression. J. Immunol. 180, 3990–3999 (2008).

    CAS  PubMed  Google Scholar 

  102. Gowda, N. M., Wu, X. & Gowda, D. C. TLR9 and MyD88 are crucial for the development of protective immunity to malaria. J. Immunol. 188, 5073–5085 (2012).

    CAS  PubMed  Google Scholar 

  103. Griffith, J. W. et al. Toll-like receptor modulation of murine cerebral malaria is dependent on the genetic background of the host. J. Infect. Dis. 196, 1553–1564 (2007).

    CAS  PubMed  Google Scholar 

  104. Wykes, M. N. et al. Plasmodium strain determines dendritic cell function essential for survival from malaria. PLoS Pathog. 3, e96 (2007). This study demonstrates that lethal strains of Plasmodium avoid activation of DCs to evade protective immune responses in mice.

    PubMed  PubMed Central  Google Scholar 

  105. da Silva, H. B. et al. IFN-γ-induced priming maintains long-term strain-transcending immunity against blood-stage Plasmodium chabaudi malaria. J. Immunol. 191, 5160–5169 (2013). This study defines the importance of IFN γ priming in maintaining long-term immunity to mouse malaria.

    CAS  PubMed  Google Scholar 

  106. Cramer, J. P. et al. MyD88/IL-18-dependent pathways rather than TLRs control early parasitaemia in non-lethal Plasmodium yoelii infection. Microbes Infect. 10, 1259–1265 (2008).

    CAS  PubMed  Google Scholar 

  107. Guermonprez, P. et al. Inflammatory Flt3l is essential to mobilize dendritic cells and for T cell responses during Plasmodium infection. Nature Med. 19, 730–738 (2013).

    CAS  PubMed  Google Scholar 

  108. Haque, A. et al. Type I interferons suppress CD4+ T-cell-dependent parasite control during blood-stage Plasmodium infection. Eur. J. Immunol. 41, 2688–2698 (2011).

    CAS  PubMed  Google Scholar 

  109. Haque, A. et al. Type I IFN signaling in CD8 DCs impairs Th1-dependent malaria immunity. J. Clin. Invest. 124, 2483–2496 (2014). References 108 and 109 demonstrate that signalling via the type I IFN receptor impairs DC function and the development of protective T cell responses in mouse malaria.

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Perry, J. A., Olver, C. S., Burnett, R. C. & Avery, A. C. Cutting edge: the acquisition of TLR tolerance during malaria infection impacts T cell activation. J. Immunol. 174, 5921–5925 (2005).

    CAS  PubMed  Google Scholar 

  111. Urban, B. C. et al. Plasmodium falciparum-infected erythrocytes modulate the maturation of dendritic cells. Nature 400, 73–77 (1999).

    CAS  PubMed  Google Scholar 

  112. Schwarzer, E., Alessio, M., Ulliers, D. & Arese, P. Phagocytosis of the malarial pigment, hemozoin, impairs expression of major histocompatibility complex class II antigen, CD54, and CD11c in human monocytes. Infect. Immun. 66, 1601–1606 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Pinzon-Charry, A. et al. Apoptosis and dysfunction of blood dendritic cells in patients with falciparum and vivax malaria. J. Exp. Med. 210, 1635–1646 (2013). This study demonstrates that the frequency of circulating DCs is decreased during acute episodes of human malaria.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Roestenberg, M. et al. Protection against a malaria challenge by sporozoite inoculation. N. Engl. J. Med. 361, 468–477 (2009).

    CAS  PubMed  Google Scholar 

  115. Seder, R. A. et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science 341, 1359–1365 (2013).

    CAS  PubMed  Google Scholar 

  116. Agnandji, S. T. et al. First results of phase 3 trial of RTS,S/AS01 malaria vaccine in African children. N. Engl. J. Med. 365, 1863–1875 (2011).

    PubMed  Google Scholar 

  117. Olotu, A. et al. Four-year efficacy of RTS,S/AS01E and its interaction with malaria exposure. N. Engl. J. Med. 368, 1111–1120 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Schofield, L., Hewitt, M. C., Evans, K., Siomos, M. A. & Seeberger, P. H. Synthetic GPI as a candidate anti-toxic vaccine in a model of malaria. Nature 418, 785–789 (2002).

    CAS  PubMed  Google Scholar 

  119. Genton, B. et al. A recombinant blood-stage malaria vaccine reduces Plasmodium falciparum density and exerts selective pressure on parasite populations in a phase 1-2b trial in Papua New Guinea. J. Infect. Dis. 185, 820–827 (2002).

    PubMed  Google Scholar 

  120. Thera, M. A. et al. A field trial to assess a blood-stage malaria vaccine. N. Engl. J. Med. 365, 1004–1013 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. King, C. L. et al. Naturally acquired Duffy-binding protein-specific binding inhibitory antibodies confer protection from blood-stage Plasmodium vivax infection. Proc. Natl Acad. Sci. USA 105, 8363–8368 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Kester, K. E. et al. Randomized, double-blind, phase 2a trial of falciparum malaria vaccines RTS,S/AS01B and RTS,S/AS02A in malaria-naive adults: safety, efficacy, and immunologic associates of protection. J. Infect. Dis. 200, 337–346 (2009).

    CAS  PubMed  Google Scholar 

  123. Onishi, M. et al. Hemozoin is a potent adjuvant for hemagglutinin split vaccine without pyrogenicity in ferrets. Vaccine 32, 3004–3009 (2014).

    CAS  PubMed  Google Scholar 

  124. McCall, M. B. et al. Plasmodium falciparum infection causes proinflammatory priming of human TLR responses. J. Immunol. 179, 162–171 (2007).

    CAS  PubMed  Google Scholar 

  125. Hartgers, F. C. et al. Enhanced Toll-like receptor responsiveness associated with mitogen-activated protein kinase activation in Plasmodium falciparum-infected children. Infect. Immun. 76, 5149–5157 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Leoratti, F. M. et al. Neutrophil paralysis in Plasmodium vivax malaria. PLoS Negl. Trop. Dis. 6, e1710 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Franklin, B. S. et al. Therapeutical targeting of nucleic acid-sensing Toll-like receptors prevents experimental cerebral malaria. Proc. Natl Acad. Sci. USA 108, 3689–3694 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Mabey, D. C., Brown, A. & Greenwood, B. M. Plasmodium falciparum malaria and Salmonella infections in Gambian children. J. Infect. Dis. 155, 1319–1321 (1987).

    CAS  PubMed  Google Scholar 

  129. Berkley, J., Mwarumba, S., Bramham, K., Lowe, B. & Marsh, K. Bacteraemia complicating severe malaria in children. Trans. R. Soc. Trop. Med. Hyg. 93, 283–286 (1999).

    CAS  PubMed  Google Scholar 

  130. Bronzan, R. N. et al. Bacteremia in Malawian children with severe malaria: prevalence, etiology, HIV coinfection, and outcome. J. Infect. Dis. 195, 895–904 (2007).

    PubMed  Google Scholar 

  131. Were, T. et al. Bacteremia in Kenyan children presenting with malaria. J. Clin. Microbiol. 49, 671–676 (2011). This study reports an enhanced risk of developing severe malaria in patients with secondary bacterial infection.

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Morpeth, S. C., Ramadhani, H. O. & Crump, J. A. Invasive non-Typhi Salmonella disease in Africa. Clin. Infect. Dis. 49, 606–611 (2009).

    PubMed  Google Scholar 

  133. Scott, J. A. et al. Relation between falciparum malaria and bacteraemia in Kenyan children: a population-based, case-control study and a longitudinal study. Lancet 378, 1316–1323 (2011). This study reports an enhanced frequency of bacterial infections in children undergoing acute malaria episodes.

    PubMed  PubMed Central  Google Scholar 

  134. Cunnington, A. J., de Souza, J. B., Walther, M. & Riley, E. M. Malaria impairs resistance to Salmonella through heme- and heme oxygenase-dependent dysfunctional granulocyte mobilization. Nature Med. 18, 120–127 (2012).

    CAS  Google Scholar 

  135. Lokken, K. L. et al. Malaria parasite infection compromises control of concurrent systemic non-typhoidal Salmonella infection via IL-10-mediated alteration of myeloid cell function. PLoS Pathog. 10, e1004049 (2014). References 134 and 135 describe the mechanism of enhanced susceptibility to bacterial infection in mouse malaria.

    PubMed  PubMed Central  Google Scholar 

  136. Cunnington, A. J. et al. Prolonged neutrophil dysfunction after Plasmodium falciparum malaria is related to hemolysis and heme oxygenase-1 induction. J. Immunol. 189, 5336–5346 (2012).

    CAS  PubMed  Google Scholar 

  137. Adachi, K. et al. Plasmodium berghei infection in mice induces liver injury by an IL-12- and toll-like receptor/myeloid differentiation factor 88-dependent mechanism. J. Immunol. 167, 5928–5934 (2001).

    CAS  PubMed  Google Scholar 

  138. Barboza, R. et al. MyD88 signaling is directly involved in the development of murine placental malaria. Infect. Immun. 82, 830–838 (2014).

    PubMed  PubMed Central  Google Scholar 

  139. Coban, C. et al. Pathological role of Toll-like receptor signaling in cerebral malaria. Int. Immunol. 19, 67–79 (2007). References 137, 138 and 139 demonstrate the importance of MYD88 and TLRs in different aspects of the pathogenesis of mouse malaria.

    CAS  PubMed  Google Scholar 

  140. Kordes, M., Matuschewski, K. & Hafalla, J. C. Caspase-1 activation of interleukin-1β (IL-1β) and IL-18 is dispensable for induction of experimental cerebral malaria. Infect. Immun. 79, 3633–3641 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Grau, G. E. et al. Monoclonal antibody against interferon-γ can prevent experimental cerebral malaria and its associated overproduction of tumor necrosis factor. Proc. Natl Acad. Sci. USA 86, 5572–5574 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Campanella, G. S. et al. Chemokine receptor CXCR3 and its ligands CXCL9 and CXCL10 are required for the development of murine cerebral malaria. Proc. Natl Acad. Sci. USA 105, 4814–4819 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Grau, G. E. et al. Tumor necrosis factor (cachectin) as an essential mediator in murine cerebral malaria. Science 237, 1210–1212 (1987).

    CAS  PubMed  Google Scholar 

  144. Kwiatkowski, D. et al. Anti-TNF therapy inhibits fever in cerebral malaria. Q. J. Med. 86, 91–98 (1993).

    CAS  PubMed  Google Scholar 

  145. Looareesuwan, S. et al. Pentoxifylline as an ancillary treatment for severe falciparum malaria in Thailand. Am. J. Trop. Med. Hyg. 58, 348–353 (1998).

    CAS  PubMed  Google Scholar 

  146. Ball, E. A. et al. IFNAR1 controls progression to cerebral malaria in children and CD8+ T cell brain pathology in Plasmodium berghei-infected mice. J. Immunol. 190, 5118–5127 (2013).

    CAS  PubMed  Google Scholar 

  147. Palomo, J. et al. Type I interferons contribute to experimental cerebral malaria development in response to sporozoite or blood-stage Plasmodium berghei ANKA. Eur. J. Immunol. 43, 2683–2695 (2013).

    CAS  PubMed  Google Scholar 

  148. Vigario, A. M. et al. Recombinant human IFN-α inhibits cerebral malaria and reduces parasite burden in mice. J. Immunol. 178, 6416–6425 (2007).

    CAS  PubMed  Google Scholar 

  149. McGuire, W., Hill, A. V., Allsopp, C. E., Greenwood, B. M. & Kwiatkowski, D. Variation in the TNF-α promoter region associated with susceptibility to cerebral malaria. Nature 371, 508–510 (1994).

    CAS  PubMed  Google Scholar 

  150. Aucan, C. et al. Interferon-α receptor-1 (IFNAR1) variants are associated with protection against cerebral malaria in the Gambia. Genes Immun. 4, 275–282 (2003).

    CAS  PubMed  Google Scholar 

  151. Phawong, C. et al. Haplotypes of IL12B promoter polymorphisms condition susceptibility to severe malaria and functional changes in cytokine levels in Thai adults. Immunogenetics 62, 345–356 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Koch, O. et al. IFNGR1 gene promoter polymorphisms and susceptibility to cerebral malaria. J. Infect. Dis. 185, 1684–1687 (2002).

    CAS  PubMed  Google Scholar 

  153. Cabantous, S. et al. Evidence that interferon-γ plays a protective role during cerebral malaria. J. Infect. Dis. 192, 854–860 (2005).

    CAS  PubMed  Google Scholar 

  154. Sortica, V. A. et al. IL1B, IL4R, IL12RB1 and TNF gene polymorphisms are associated with Plasmodium vivax malaria in Brazil. Malar. J. 11, 409 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Sabeti, P. et al. CD40L association with protection from severe malaria. Genes Immun. 3, 286–291 (2002).

    CAS  PubMed  Google Scholar 

  156. Burgner, D. et al. Inducible nitric oxide synthase polymorphism and fatal cerebral malaria. Lancet 352, 1193–1194 (1998).

    CAS  PubMed  Google Scholar 

  157. Mockenhaupt, F. P. et al. Toll-like receptor (TLR) polymorphisms in African children: common TLR-4 variants predispose to severe malaria. Proc. Natl Acad. Sci. USA 103, 177–182 (2006).

    CAS  PubMed  Google Scholar 

  158. Mockenhaupt, F. P. et al. Common polymorphisms of toll-like receptors 4 and 9 are associated with the clinical manifestation of malaria during pregnancy. J. Infect. Dis. 194, 184–188 (2006).

    CAS  PubMed  Google Scholar 

  159. Sam-Agudu, N. A. et al. TLR9 polymorphisms are associated with altered IFN-γ levels in children with cerebral malaria. Am. J. Trop. Med. Hyg. 82, 548–555 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Leoratti, F. M. et al. Variants in the toll-like receptor signaling pathway and clinical outcomes of malaria. J. Infect. Dis. 198, 772–780 (2008).

    CAS  PubMed  Google Scholar 

  161. Khor, C. C. et al. A Mal functional variant is associated with protection against invasive pneumococcal disease, bacteremia, malaria and tuberculosis. Nature Genet. 39, 523–528 (2007).

    CAS  PubMed  Google Scholar 

  162. Shanks, G. D. & White, N. J. The activation of vivax malaria hypnozoites by infectious diseases. Lancet Infect. Dis. 13, 900–906 (2013).

    PubMed  Google Scholar 

  163. Lopes, S. C. et al. Paucity of Plasmodium vivax mature schizonts in peripheral blood is associated with their increased cytoadhesive potential. J. Infect. Dis. 209, 1403–1407 (2014).

    PubMed  Google Scholar 

  164. Mantel, P. Y. et al. Malaria-infected erythrocyte-derived microvesicles mediate cellular communication within the parasite population and with the host immune system. Cell Host Microbe 13, 521–534 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Antonelli, L. R. et al. The CD14+CD16+ inflammatory monocyte subset displays increased mitochondrial activity and effector function during acute Plasmodium vivax malaria. PLoS Pathog. 10, e1004393 (2014).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank M. Trombly for critically reviewing this manuscript. The authors are also grateful to all of the members and collaborators of the R.T.G., K.A.F. and D.T.G. laboratories for their invaluable contributions to the work that is reviewed here. R.T.G. is a recipient of a Scholar Fellowship from Coordenação de Aperfeiçoamento de Pessoal de Ensino Superior (CAPES), Brazil, and the David Rockefeller Center for Latin American Studies at Harvard School of Public Health, USA. The R.T.G. laboratory in Brazil is funded by the National Institute of Science and Technology for Vaccines, Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) and Fundação de Amparo a Pesquisa de Minas Gerai (Fapemig). D.T.G. is a recipient of a Visiting Scientists Fellowship from CNPq. K.A.F., D.T.G. and R.T.G. are funded by the US National Institutes of Health.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ricardo T. Gazzinelli.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Glossary

Apicomplexa

A large group of protozoa that includes the Plasmodium genus. They possess an apical complex structure that is involved in penetrating host cells.

Sporozoites

The Plasmodium stage that develops in the salivary glands of the mosquito and infects humans during the blood meal. When they are in the bloodstream, the sporozoites are transported to the liver where they invade hepatocytes for the first round of replication in the vertebrate host.

Merozoites

The pathogenic Plasmodium stage that infects red blood cells (RBCs) and rapidly reproduces asexually, forming schizonts that contain multiple parasites. RBCs are destroyed by the end of this process, which releases many merozoites that will infect other host cells.

Schizogeny

The replicative process of malaria parasites, either in hepatocytes or red blood cells, that involves nuclear division without cytoplasmic segmentation, followed by a cell budding to form the progeny called merozoites.

Pathogen-associated molecular patterns

(PAMPs). Molecular structures that are normally abundant in certain pathogens and are detected by innate immune receptors known as pattern recognition receptors. PAMPs are often used as vaccine adjuvants.

Malaria-associated syndromes

A collection of signs and symptoms that are characteristic of a specific manifestation of malaria, such as anaemia, respiratory distress or cerebral malaria.

Paroxysms

In malaria, paroxysms refer to sudden attacks of disease that are characterized by high fever, chills and various other symptoms. This sudden worsening of malaria symptoms is cyclic and coincides with the synchronous rupture of Plasmodium-infected red blood cells.

Glycosylphosphatidylinositol anchors

(GPI anchors). Glycolipid structures that link surface proteins to the surface membrane of eukaryotic cells. In protozoa, most surface proteins are linked via GPIs, and free GPIs are also expressed at the surface.

Haemozoin

The crystalline product resulting from the digestion of haemoglobin during different intraerythrocytic Plasmodium stages.

Toll-like receptors

(TLRs). A family of pattern recognition receptors that sense pathogen-associated molecular patterns and initiate pro-inflammatory responses during microbial infection.

Inflammasomes

Molecular platforms formed by members of the NOD-like receptors family, which activate caspase 1 to cleave pro-IL-1β and pro-IL-18 and release the active form of these cytokines.

Cytosolic sensors

Different families of pattern recognition receptors located in the cytoplasm that are often activated by DNA and RNA, and that induce the production of type I interferons.

Microvesicles

Vesicles derived from platelets, red blood cells, endothelial cells and leukocytes that serve as a communication system between host cells, and display important pro-inflammatory activity.

Adjuvant

An agent that is mixed with an antigen to increase the immune response to that antigen following immunization.

Reticulocyte

Immature red blood cells (RBCs) that correspond to 1% of total circulating RBCs in humans.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gazzinelli, R., Kalantari, P., Fitzgerald, K. et al. Innate sensing of malaria parasites. Nat Rev Immunol 14, 744–757 (2014). https://doi.org/10.1038/nri3742

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri3742

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