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:

T cell-mediated immunity to malaria

Subjects

Abstract

Immunity to malaria has been linked to the availability and function of helper CD4+ T cells, cytotoxic CD8+ T cells and γδ T cells that can respond to both the asymptomatic liver stage and the symptomatic blood stage of Plasmodium sp. infection. These T cell responses are also thought to be modulated by regulatory T cells. However, the precise mechanisms governing the development and function of Plasmodium-specific T cells and their capacity to form tissue-resident and long-lived memory populations are less well understood. The field has arrived at a point where the push for vaccines that exploit T cell-mediated immunity to malaria has made it imperative to define and reconcile the mechanisms that regulate the development and functions of Plasmodium-specific T cells. Here, we review our current understanding of the mechanisms by which T cell subsets orchestrate host resistance to Plasmodium infection on the basis of observational and mechanistic studies in humans, non-human primates and rodent models. We also examine the potential of new experimental strategies and human infection systems to inform a new generation of approaches to harness T cell responses against 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

Fig. 1: Plasmodium life cycle.
Fig. 2: Overview of tissue-specific, T cell-mediated immune resistance networks during Plasmodium infection.

Similar content being viewed by others

References

  1. World Health Organization. World Malaria Report 2018 (WHO, 2018).

  2. Regules, J. A. et al. Fractional third and fourth dose of RTS,S/AS01 malaria candidate vaccine: a phase 2a controlled human malaria parasite infection and immunogenicity study. J. Infect. Dis. 214, 762–771 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. RTS,S Clinical Trials Partnership. Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a phase 3, individually randomised, controlled trial. Lancet 386, 31–45 (2015).

    Article  CAS  Google Scholar 

  4. White, M. T. et al. Immunogenicity of the RTS,S/AS01 malaria vaccine and implications for duration of vaccine efficacy: secondary analysis of data from a phase 3 randomised controlled trial. Lancet Infect. Dis. 15, 1450–1458 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ishizuka, A. S. et al. Protection against malaria at 1 year and immune correlates following PfSPZ vaccination. Nat. Med. 22, 614–623 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Mordmuller, B. et al. Sterile protection against human malaria by chemoattenuated PfSPZ vaccine. Nature 542, 445–449 (2017).

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  8. Seder, R. A. et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science 341, 1359–1365 (2013). This study demonstrates that there is a dose-dependent immunological threshold for establishing protection against malaria that can be achieved with intravenous administration of a live-attenuated sporozoite vaccine.

    Article  CAS  PubMed  Google Scholar 

  9. Ewer, K. J. et al. Protective CD8+ T cell immunity to human malaria induced by chimpanzee adenovirus-MVA immunisation. Nat. Commun. 4, 2836 (2013).

    Article  PubMed  CAS  Google Scholar 

  10. Sissoko, M. S. et al. Safety and efficacy of PfSPZ vaccine against Plasmodium falciparum via direct venous inoculation in healthy malaria-exposed adults in Mali: a randomised, double-blind phase 1 trial. Lancet Infect. Dis. 17, 498–509 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wykes, M. N. & Lewin, S. R. Immune checkpoint blockade in infectious diseases. Nat. Rev. Immunol. 18, 91–104 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Soon, M. S. F. & Haque, A. Recent insights into CD4(+) Th cell differentiation in malaria. J. Immunol. 200, 1965–1975 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Montes de Oca, M., Good, M. F., McCarthy, J. S. & Engwerda, C. R. The impact of established immunoregulatory networks on vaccine efficacy and the development of immunity to malaria. J. Immunol. 197, 4518–4526 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Troye-Blomberg, M. et al. Production of IL 2 and IFN-gamma by T cells from malaria patients in response to Plasmodium falciparum or erythrocyte antigens in vitro. J. Immunol. 135, 3498–3504 (1985).

    CAS  PubMed  Google Scholar 

  15. Su, Z. & Stevenson, M. M. Central role of endogenous gamma interferon in protective immunity against blood-stage Plasmodium chabaudi AS infection. Infect. Immun. 68, 4399–4406 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Meding, S. J., Cheng, S. C., Simon-Haarhaus, B. & Langhorne, J. Role of gamma interferon during infection with Plasmodium chabaudi chabaudi. Infect. Immun. 58, 3671–3678 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Huang, K. Y., Schultz, W. W. & Gordon, F. B. Interferon induced by Plasmodium berghei. Science 162, 123–124 (1968).

    Article  CAS  PubMed  Google Scholar 

  18. Shear, H. L., Srinivasan, R., Nolan, T. & Ng, C. Role of IFN-gamma in lethal and nonlethal malaria in susceptible and resistant murine hosts. J. Immunol. 143, 2038–2044 (1989).

    CAS  PubMed  Google Scholar 

  19. Salles, E. M. et al. P2X7 receptor drives Th1 cell differentiation and controls the follicular helper T cell population to protect against Plasmodium chabaudi malaria. PLOS Pathog. 13, e1006595 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Gotz, A. et al. Atypical activation of dendritic cells by Plasmodium falciparum. Proc. Natl Acad. Sci. USA 114, E10568–E10577 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lazarevic, V., Glimcher, L. H. & Lord, G. M. T-bet: a bridge between innate and adaptive immunity. Nat. Rev. Immunol. 13, 777–789 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Oakley, M. S. et al. T-bet modulates the antibody response and immune protection during murine malaria. Eur. J. Immunol. 44, 2680–2691 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  24. 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).

    Article  CAS  PubMed  Google Scholar 

  25. Jaramillo, M., Gowda, D. C., Radzioch, D. & Olivier, M. Hemozoin increases IFN-gamma-inducible macrophage nitric oxide generation through extracellular signal-regulated kinase- and NF-kappa B-dependent pathways. J. Immunol. 171, 4243–4253 (2003).

    Article  CAS  PubMed  Google Scholar 

  26. Blanchette, J., Jaramillo, M. & Olivier, M. Signalling events involved in interferon-gamma-inducible macrophage nitric oxide generation. Immunology 108, 513–522 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Horowitz, A. et al. Cross-talk between T cells and NK cells generates rapid effector responses to Plasmodium falciparum-infected erythrocytes. J. Immunol. 184, 6043–6052 (2010).

    Article  CAS  PubMed  Google Scholar 

  28. Fontana, M. F. et al. Macrophage colony stimulating factor derived from CD4+ T cells contributes to control of a blood-borne infection. PLOS Pathog. 12, e1006046 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Weiss, W. R. et al. A plasmid encoding murine granulocyte-macrophage colony-stimulating factor increases protection conferred by a malaria DNA vaccine. J. Immunol. 161, 2325–2332 (1998).

    CAS  PubMed  Google Scholar 

  30. Stephens, R. & Langhorne, J. Effector memory Th1 CD4 T cells are maintained in a mouse model of chronic malaria. PLOS Pathog. 6, e1001208 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Opata, M. M. et al. Early effector cells survive the contraction phase in malaria infection and generate both central and effector memory T cells. J. Immunol. 194, 5346–5354 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Opata, M. M. et al. Protection by and maintenance of CD4 effector memory and effector T cell subsets in persistent malaria infection. PLOS Pathog. 14, e1006960 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Zander, R. A. et al. Th1-like Plasmodium-specific memory CD4(+) T cells support humoral immunity. Cell Rep. 21, 1839–1852 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Reece, W. H. et al. A CD4(+) T cell immune response to a conserved epitope in the circumsporozoite protein correlates with protection from natural Plasmodium falciparum infection and disease. Nat. Med. 10, 406–410 (2004).

    Article  CAS  PubMed  Google Scholar 

  35. Oliveira, G. A. et al. Class II-restricted protective immunity induced by malaria sporozoites. Infect. Immun. 76, 1200–1206 (2008).

    Article  CAS  PubMed  Google Scholar 

  36. Renia, L. et al. Effector functions of circumsporozoite peptide-primed CD4+ T cell clones against Plasmodium yoelii liver stages. J. Immunol. 150, 1471–1478 (1993).

    CAS  PubMed  Google Scholar 

  37. Doolan, D. L. et al. Circumventing genetic restriction of protection against malaria with multigene DNA immunization: CD8+ cell-, interferon gamma-, and nitric oxide-dependent immunity. J. Exp. Med. 183, 1739–1746 (1996).

    Article  CAS  PubMed  Google Scholar 

  38. Sun, P. et al. Protective immunity induced with malaria vaccine, RTS,S, is linked to Plasmodium falciparum circumsporozoite protein-specific CD4+ and CD8+ T cells producing IFN-gamma. J. Immunol. 171, 6961–6967 (2003).

    Article  CAS  PubMed  Google Scholar 

  39. Burel, J. G. et al. Reduced Plasmodium parasite burden associates with CD38+ CD4+ T cells displaying cytolytic potential and impaired IFN-gamma production. PLOS Pathog. 12, e1005839 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Bijker, E. M. et al. Cytotoxic markers associate with protection against malaria in human volunteers immunized with Plasmodium falciparum sporozoites. J. Infect. Dis. 210, 1605–1615 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Tsuji, M., Romero, P., Nussenzweig, R. S. & Zavala, F. CD4+ cytolytic T cell clone confers protection against murine malaria. J. Exp. Med. 172, 1353–1357 (1990). This study identifies a protective MHC class II-restricted antigen from Plasmodium in mice.

    Article  CAS  PubMed  Google Scholar 

  42. Takita-Sonoda, Y. et al. Plasmodium yoelii: peptide immunization induces protective CD4+ T cells against a previously unrecognized cryptic epitope of the circumsporozoite protein. Exp. Parasitol. 84, 223–230 (1996).

    Article  CAS  PubMed  Google Scholar 

  43. Obeng-Adjei, N. et al. Malaria-induced interferon-gamma drives the expansion of Tbethi atypical memory B cells. PLOS Pathog. 13, e1006576 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Zander, R. A. et al. PD-1 co-inhibitory and OX40 co-stimulatory crosstalk regulates helper T cell differentiation and anti-Plasmodium humoral immunity. Cell Host Microbe 17, 628–641 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ryg-Cornejo, V. et al. Severe malaria infections impair germinal center responses by inhibiting T follicular helper cell differentiation. Cell Rep. 14, 68–81 (2016).

    Article  CAS  PubMed  Google Scholar 

  46. Guthmiller, J. J., Graham, A. C., Zander, R. A., Pope, R. L. & Butler, N. S. Cutting edge: IL-10 is essential for the generation of germinal center B cell responses and anti-Plasmodium humoral immunity. J. Immunol. 198, 617–622 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. Rivera-Correa, J. et al. Plasmodium DNA-mediated TLR9 activation of T-bet(+) B cells contributes to autoimmune anaemia during malaria. Nat. Commun. 8, 1282 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Walker, J. A. & McKenzie, A. N. J. TH2 cell development and function. Nat. Rev. Immunol. 18, 121–133 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Perez-Mazliah, D. & Langhorne, J. CD4 T cell subsets in malaria: TH1/TH2 revisited. Front. Immunol. 5, 671 (2014).

    PubMed  Google Scholar 

  50. Coomes, S. M. et al. IFNgamma and IL-12 restrict Th2 responses during Helminth/Plasmodium co-infection and promote IFNgamma from Th2 cells. PLOS Pathog. 11, e1004994 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Shimoda, K. et al. Lack of IL-4-induced Th2 response and IgE class switching in mice with disrupted Stat6 gene. Nature 380, 630–633 (1996).

    Article  CAS  PubMed  Google Scholar 

  52. von der Weid, T., Kopf, M., Kohler, G. & Langhorne, J. The immune response to Plasmodium chabaudi malaria in interleukin-4-deficient mice. Eur. J. Immunol. 24, 2285–2293 (1994).

    Article  PubMed  Google Scholar 

  53. Kumaratilake, L. M. & Ferrante, A. IL-4 inhibits macrophage-mediated killing of Plasmodium falciparum in vitro. A possible parasite-immune evasion mechanism. J. Immunol. 149, 194–199 (1992).

    CAS  PubMed  Google Scholar 

  54. Troye-Blomberg, M. et al. Production by activated human T cells of interleukin 4 but not interferon-gamma is associated with elevated levels of serum antibodies to activating malaria antigens. Proc. Natl Acad. Sci. USA 87, 5484–5488 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Carvalho, L. H. et al. IL-4-secreting CD4+ T cells are crucial to the development of CD8+ T cell responses against malaria liver stages. Nat. Med. 8, 166–170 (2002). This work demonstrates the mechanism of CD4 + T cell help in the development of optimal CD8 + T cell responses to liver-stage malaria.

    Article  CAS  PubMed  Google Scholar 

  56. Overstreet, M. G., Chen, Y. C., Cockburn, I. A., Tse, S. W. & Zavala, F. CD4+ T cells modulate expansion and survival but not functional properties of effector and memory CD8+ T cells induced by malaria sporozoites. PLOS ONE 6, e15948 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Vinuesa, C. G. & Cyster, J. G. How T cells earn the follicular rite of passage. Immunity 35, 671–680 (2011).

    Article  CAS  PubMed  Google Scholar 

  58. Victora, G. D. & Nussenzweig, M. C. Germinal centers. Annu. Rev. Immunol. 30, 429–457 (2012).

    Article  CAS  PubMed  Google Scholar 

  59. Obeng-Adjei, N. et al. Circulating Th1-cell-type Tfh cells that exhibit impaired B cell help are preferentially activated during acute malaria in children. Cell Rep. 13, 425–439 (2015). This study suggests that the circulating PD1 + CXCR5 + CXCR3 T FH cells provide B cell help that facilitates long-lived antibody-mediated protection in humans.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Butler, N. S. et al. Therapeutic blockade of PD-L1 and LAG-3 rapidly clears established blood-stage Plasmodium infection. Nat. Immunol. 13, 188–195 (2012). This study shows that blood-stage malaria may be associated with T cell dysfunction that is linked to T cell expression of inhibitory receptors.

    Article  CAS  Google Scholar 

  61. Perez-Mazliah, D. et al. Follicular helper T cells are essential for the elimination of Plasmodium infection. EBioMedicine 24, 216–230 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Figueiredo, M. M. et al. T follicular helper cells regulate the activation of B lymphocytes and antibody production during Plasmodium vivax infection. PLOS Pathog. 13, e1006484 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Perez-Mazliah, D. et al. Disruption of IL-21 signaling affects T cell-B cell interactions and abrogates protective humoral immunity to malaria. PLOS Pathog. 11, e1004715 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Sebina, I. et al. IFNAR1-signalling obstructs ICOS-mediated humoral immunity during non-lethal blood-stage Plasmodium infection. PLOS Pathog. 12, e1005999 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Wikenheiser, D. J., Ghosh, D., Kennedy, B. & Stumhofer, J. S. The costimulatory molecule ICOS regulates host Th1 and follicular Th cell differentiation in response to Plasmodium chabaudi chabaudi AS infection. J. Immunol. 196, 778–791 (2016).

    Article  CAS  PubMed  Google Scholar 

  66. Hale, J. S. et al. Distinct memory CD4+ T cells with commitment to T follicular helper- and T helper 1-cell lineages are generated after acute viral infection. Immunity 38, 805–817 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Pepper, M., Pagan, A. J., Igyarto, B. Z., Taylor, J. J. & Jenkins, M. K. Opposing signals from the Bcl6 transcription factor and the interleukin-2 receptor generate T helper 1 central and effector memory cells. Immunity 35, 583–595 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Lonnberg, T. S. et al. Single-cell RNA-seq and computational analysis using temporal micture modeling resolves TH1/TFH fate bifurcation in malaria. Sci. Immunol. 2, eaal2192 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Sebina, I. et al. IL-6 promotes CD4(+) T cell and B cell activation during Plasmodium infection. Parasite Immunol. 39, e12455 (2017).

    Article  CAS  Google Scholar 

  70. James, K. R. et al. IFN regulatory factor 3 balances Th1 and T follicular helper immunity during nonlethal blood-stage Plasmodium infection. J. Immunol. 200, 1443–1456 (2018).

    Article  CAS  PubMed  Google Scholar 

  71. Wikenheiser, D. J., Brown, S. L., Lee, J. & Stumhofer, J. S. NK1.1 expression defines a population of CD4(+) effector T cells displaying Th1 and Tfh cell properties that support early antibody production during Plasmodium yoelii infection. Front. Immunol. 9, 2277 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Troye-Blomberg, M. et al. Human gamma delta T cells that inhibit the in vitro growth of the asexual blood stages of the Plasmodium falciparum parasite express cytolytic and proinflammatory molecules. Scand. J. Immunol. 50, 642–650 (1999).

    Article  CAS  PubMed  Google Scholar 

  73. Kurup, S. P. et al. Regulatory T cells impede acute and long-term immunity to blood-stage malaria through CTLA-4. Nat. Med. 23, 1220–1225 (2017). This study demonstrates a temporal role and mechanism of regulatory T cell function in blood-stage malaria.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Sage, P. T. & Sharpe, A. H. T follicular regulatory cells. Immunol. Rev. 271, 246–259 (2016).

    Article  CAS  PubMed  Google Scholar 

  75. Laidlaw, B. J. et al. Interleukin-10 from CD4(+) follicular regulatory T cells promotes the germinal center response. Sci. Immunol. 2, eaan4767 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Wing, J. B., Tekguc, M. & Sakaguchi, S. Control of germinal center responses by T-follicular regulatory cells. Front. Immunol. 9, 1910 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Xie, M. M. & Dent, A. L. Unexpected help: follicular regulatory T cells in the germinal center. Front. Immunol. 9, 1536 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Sandquist, I. & Kolls, J. Update on regulation and effector functions of Th17 cells. F1000Res 7, 205 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Sercundes, M. K. et al. Targeting neutrophils to prevent malaria-associated acute lung injury/acute respiratory distress syndrome in mice. PLOS Pathog. 12, e1006054 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Feintuch, C. M. et al. Activated neutrophils are associated with pediatric cerebral malaria vasculopathy in malawian children. MBio 7, e01300–15 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Metenou, S. et al. Filarial infection suppresses malaria-specific multifunctional Th1 and Th17 responses in malaria and filarial coinfections. J. Immunol. 186, 4725–4733 (2011).

    Article  CAS  PubMed  Google Scholar 

  82. Ishida, H. et al. Development of experimental cerebral malaria is independent of IL-23 and IL-17. Biochem. Biophys. Res. Commun. 402, 790–795 (2010).

    Article  CAS  PubMed  Google Scholar 

  83. Mastelic, B. et al. IL-22 protects against liver pathology and lethality of an experimental blood-stage malaria infection. Front. Immunol. 3, 85 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Keswani, T. & Bhattacharyya, A. Differential role of T regulatory and Th17 in Swiss mice infected with Plasmodium berghei ANKA and Plasmodium yoelii. Exp. Parasitol. 141, 82–92 (2014).

    Article  CAS  PubMed  Google Scholar 

  85. Hu, W. C. Human immune responses to Plasmodium falciparum infection: molecular evidence for a suboptimal THalphabeta and TH17 bias over ideal and effective traditional TH1 immune response. Malar J. 12, 392 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  86. Wei, L., Laurence, A., Elias, K. M. & O’Shea, J. J. IL-21 is produced by Th17 cells and drives IL-17 production in a STAT3-dependent manner. J. Biol. Chem. 282, 34605–34610 (2007).

    Article  CAS  PubMed  Google Scholar 

  87. Moretto, M. M., Hwang, S. & Khan, I. A. Downregulated IL-21 response and T follicular helper cell exhaustion correlate with compromised CD8 T cell immunity during chronic toxoplasmosis. Front. Immunol. 8, 1436 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Stumhofer, J. S., Silver, J. S. & Hunter, C. A. IL-21 is required for optimal antibody production and T cell responses during chronic Toxoplasma gondii infection. PLOS ONE 8, e62889 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Findlay, E. G. et al. Essential role for IL-27 receptor signaling in prevention of Th1-mediated immunopathology during malaria infection. J. Immunol. 185, 2482–2492 (2010).

    Article  CAS  PubMed  Google Scholar 

  90. Freitas do Rosario, A. P. et al. IL-27 promotes IL-10 production by effector Th1 CD4+ T cells: a critical mechanism for protection from severe immunopathology during malaria infection. J. Immunol. 188, 1178–1190 (2012).

    Article  CAS  PubMed  Google Scholar 

  91. Gwyer Findlay, E. et al. IL-27 receptor signaling regulates memory CD4+ T cell populations and suppresses rapid inflammatory responses during secondary malaria infection. Infect. Immun. 82, 10–20 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  92. Kimura, D. et al. Interleukin-27-producing CD4(+) T cells regulate protective immunity during malaria parasite infection. Immunity 44, 672–682 (2016).

    Article  CAS  PubMed  Google Scholar 

  93. Couper, K. N. et al. IL-10 from CD4+CD25Foxp3CD127 adaptive regulatory T cells modulates parasite clearance and pathology during malaria infection. PLOS Pathog. 4, e1000004 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Zander, R. A. et al. Type I interferons induce T regulatory 1 responses and restrict humoral immunity during experimental malaria. PLOS Pathog. 12, e1005945 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Jagannathan, P. et al. IFNgamma/IL-10 co-producing cells dominate the CD4 response to malaria in highly exposed children. PLOS Pathog. 10, e1003864 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Loevenich, K. et al. DC-derived IL-10 modulates pro-inflammatory cytokine production and promotes induction of CD4(+)IL-10(+) regulatory T cells during Plasmodium yoelii infection. Front. Immunol. 8, 152 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Draheim, M. et al. Profiling MHC II immunopeptidome of blood-stage malaria reveals that cDC1 control the functionality of parasite-specific CD4 T cells. EMBO Mol. Med. 9, 1605–1621 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Montes de Oca, M. et al. Type I interferons regulate immune responses in humans with blood-stage Plasmodium falciparum infection. Cell Rep. 17, 399–412 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Jankovic, D. et al. Conventional T-bet(+)Foxp3(-) Th1 cells are the major source of host-protective regulatory IL-10 during intracellular protozoan infection. J. Exp. Med. 204, 273–283 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Montes de Oca, M. et al. Blimp-1-dependent IL-10 production by Tr1 cells regulates TNF-mediated tissue pathology. PLOS Pathog. 12, e1005398 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Walther, M. et al. Distinct roles for FOXP3 and FOXP3 CD4 T cells in regulating cellular immunity to uncomplicated and severe Plasmodium falciparum malaria. PLOS Pathog. 5, e1000364 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Boyle, M. J. et al. The development of Plasmodium falciparum-specific IL10 CD4 T cells and protection from malaria in children in an area of high malaria transmission. Front. Immunol. 8, 1329 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  103. Miyao, T. et al. Plasticity of Foxp3(+) T cells reflects promiscuous Foxp3 expression in conventional T cells but not reprogramming of regulatory T cells. Immunity 36, 262–275 (2012).

    Article  CAS  PubMed  Google Scholar 

  104. Ohkura, N. et al. T cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development. Immunity 37, 785–799 (2012).

    Article  CAS  PubMed  Google Scholar 

  105. Yadav, M., Stephan, S. & Bluestone, J. A. Peripherally induced tregs - role in immune homeostasis and autoimmunity. Front. Immunol. 4, 232 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Van Braeckel-Budimir, N., Kurup, S. P. & Harty, J. T. Regulatory issues in immunity to liver and blood-stage malaria. Curr. Opin. Immunol. 42, 91–97 (2016).

    Article  PubMed  CAS  Google Scholar 

  107. Finney, O. C., Riley, E. M. & Walther, M. Regulatory T cells in malaria—friend or foe? Trends Immunol. 31, 63–70 (2010).

    Article  CAS  PubMed  Google Scholar 

  108. Hansen, D. S. & Schofield, L. Natural regulatory T cells in malaria: host or parasite allies? PLOS Pathog. 6, e1000771 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Couper, K. N. et al. Incomplete depletion and rapid regeneration of Foxp3+ regulatory T cells following anti-CD25 treatment in malaria-infected mice. J. Immunol. 178, 4136–4146 (2007).

    Article  CAS  PubMed  Google Scholar 

  110. Hisaeda, H. et al. Escape of malaria parasites from host immunity requires CD4+ CD25+ regulatory T cells. Nat. Med. 10, 29–30 (2004). This work shows that T reg cells may be detrimental to the optimal control of blood-stage malaria using a mouse model.

    Article  CAS  PubMed  Google Scholar 

  111. Cambos, M., Belanger, B., Jacques, A., Roulet, A. & Scorza, T. Natural regulatory (CD4+CD25+FOXP+) T cells control the production of pro-inflammatory cytokines during Plasmodium chabaudi adami infection and do not contribute to immune evasion. Int. J. Parasitol. 38, 229–238 (2008).

    Article  CAS  PubMed  Google Scholar 

  112. Feuerer, M., Shen, Y., Littman, D. R., Benoist, C. & Mathis, D. How punctual ablation of regulatory T cells unleashes an autoimmune lesion within the pancreatic islets. Immunity 31, 654–664 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Kim, J. M., Rasmussen, J. P. & Rudensky, A. Y. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol. 8, 191–197 (2007).

    Article  CAS  PubMed  Google Scholar 

  114. Vigario, A. M. et al. Regulatory CD4+ CD25+ Foxp3+ T cells expand during experimental Plasmodium infection but do not prevent cerebral malaria. Int. J. Parasitol. 37, 963–973 (2007).

    Article  CAS  PubMed  Google Scholar 

  115. Amante, F. H. et al. A role for natural regulatory T cells in the pathogenesis of experimental cerebral malaria. Am. J. Pathol. 171, 548–559 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Steeg, C., Adler, G., Sparwasser, T., Fleischer, B. & Jacobs, T. Limited role of CD4+Foxp3+ regulatory T cells in the control of experimental cerebral malaria. J. Immunol. 183, 7014–7022 (2009).

    Article  CAS  PubMed  Google Scholar 

  117. Jangpatarapongsa, K. et al. Plasmodium vivax parasites alter the balance of myeloid and plasmacytoid dendritic cells and the induction of regulatory T cells. Eur. J. Immunol. 38, 2697–2705 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Torcia, M. G. et al. Functional deficit of T regulatory cells in Fulani, an ethnic group with low susceptibility to Plasmodium falciparum malaria. Proc. Natl Acad. Sci. USA 105, 646–651 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Walther, M. et al. Upregulation of TGF-beta, FOXP3, and CD4+CD25+ regulatory T cells correlates with more rapid parasite growth in human malaria infection. Immunity 23, 287–296 (2005). This study demonstrates that T reg cell expansions during blood-stage malaria in humans correlate with poor control of the infection.

    Article  CAS  PubMed  Google Scholar 

  120. Todryk, S. M. et al. Correlation of memory T cell responses against TRAP with protection from clinical malaria, and CD4 CD25 high T cells with susceptibility in Kenyans. PLOS ONE 3, e2027 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Apostolou, I. & von Boehmer, H. In vivo instruction of suppressor commitment in naive T cells. J. Exp. Med. 199, 1401–1408 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Benoist, C. & Mathis, D. Treg cells, life history, and diversity. Cold Spring Harb. Perspect. Biol. 4, a007021 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Finney, C. A., Taylor, M. D., Wilson, M. S. & Maizels, R. M. Expansion and activation of CD4(+)CD25(+) regulatory T cells in Heligmosomoides polygyrus infection. Eur. J. Immunol. 37, 1874–1886 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Haribhai, D. et al. A central role for induced regulatory T cells in tolerance induction in experimental colitis. J. Immunol. 182, 3461–3468 (2009).

    Article  CAS  PubMed  Google Scholar 

  125. Haribhai, D. et al. A requisite role for induced regulatory T cells in tolerance based on expanding antigen receptor diversity. Immunity 35, 109–122 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Lathrop, S. K. et al. Peripheral education of the immune system by colonic commensal microbiota. Nature 478, 250–254 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Elkord, E. Helios should not be cited as a marker of human thymus-derived Tregs. Commentary: helios(+) and helios(-) cells coexist within the natural FOXP3(+) T regulatory cell subset in humans. Front. Immunol. 7, 276 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Scholzen, A., Mittag, D., Rogerson, S. J., Cooke, B. M. & Plebanski, M. Plasmodium falciparum-mediated induction of human CD25Foxp3 CD4 T. cells is independent of direct TCR stimulation and requires IL-2, IL-10 and TGFbeta. PLOS Pathog. 5, e1000543 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  129. Liu, Y. et al. A critical function for TGF-beta signaling in the development of natural CD4+CD25+Foxp3+ regulatory T cells. Nat. Immunol. 9, 632–640 (2008).

    Article  CAS  PubMed  Google Scholar 

  130. Zheng, Y. et al. Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T cell fate. Nature 463, 808–812 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Abel, S. et al. Plasmodium yoelii infection of BALB/c mice results in expansion rather than induction of CD4(+) Foxp3(+) regulatory T cells. Immunology 148, 197–205 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Abel, S. et al. Strong impact of CD4+ Foxp3+ regulatory T cells and limited effect of T cell-derived IL-10 on pathogen clearance during Plasmodium yoelii infection. J. Immunol. 188, 5467–5477 (2012).

    Article  CAS  PubMed  Google Scholar 

  133. Sedegah, M. et al. Naturally acquired CD8+ cytotoxic T lymphocytes against the Plasmodium falciparum circumsporozoite protein. J. Immunol. 149, 966–971 (1992).

    CAS  PubMed  Google Scholar 

  134. Doolan, D. L. et al. Degenerate cytotoxic T cell epitopes from P. falciparum restricted by multiple HLA-A and HLA-B supertype alleles. Immunity 7, 97–112 (1997).

    Article  CAS  PubMed  Google Scholar 

  135. Romero, P. et al. Cloned cytotoxic T cells recognize an epitope in the circumsporozoite protein and protect against malaria. Nature 341, 323–326 (1989).

    Article  CAS  PubMed  Google Scholar 

  136. Doolan, D. L. et al. HLA-DR-promiscuous T cell epitopes from Plasmodium falciparum pre-erythrocytic-stage antigens restricted by multiple HLA class II alleles. J. Immunol. 165, 1123–1137 (2000).

    Article  CAS  PubMed  Google Scholar 

  137. Doolan, D. L. et al. Identification of Plasmodium falciparum antigens by antigenic analysis of genomic and proteomic data. Proc. Natl Acad. Sci. USA 100, 9952–9957 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Epstein, J. E. et al. Live attenuated malaria vaccine designed to protect through hepatic CD8(+) T cell immunity. Science 334, 475–480 (2011). This study demonstrates the ability of RAS vaccines to stimulate sterilizing immunity to malaria in humans.

    Article  CAS  PubMed  Google Scholar 

  139. Hafalla, J. C. et al. Identification of targets of CD8(+) T cell responses to malaria liver stages by genome-wide epitope profiling. PLOS Pathog. 9, e1003303 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Van Braeckel-Budimir, N. & Harty, J. T. CD8 T cell-mediated protection against liver-stage malaria: lessons from a mouse model. Front. Microbiol. 5, 272 (2014).

    PubMed  PubMed Central  Google Scholar 

  141. Villarino, N. & Schmidt, N. W. CD8(+) T cell responses to Plasmodium and intracellular parasites. Curr. Immunol. Rev. 9, 169–178 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Medica, D. L. & Sinnis, P. Quantitative dynamics of Plasmodium yoelii sporozoite transmission by infected anopheline mosquitoes. Infect. Immun. 73, 4363–4369 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Churcher, T. S. et al. Probability of transmission of malaria from mosquito to human is regulated by mosquito parasite density in naive and vaccinated hosts. PLOS Pathog. 13, e1006108 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  144. Cowman, A. F., Healer, J., Marapana, D. & Marsh, K. Malaria: biology and disease. Cell 167, 610–624 (2016).

    Article  CAS  PubMed  Google Scholar 

  145. Crompton, P. D. et al. Malaria immunity in man and mosquito: insights into unsolved mysteries of a deadly infectious disease. Annu. Rev. Immunol. 32, 157–187 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Miller, J. D. et al. Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity 28, 710–722 (2008).

    Article  CAS  PubMed  Google Scholar 

  147. Sturm, A. et al. Manipulation of host hepatocytes by the malaria parasite for delivery into liver sinusoids. Science 313, 1287–1290 (2006).

    Article  CAS  PubMed  Google Scholar 

  148. Tran, T. M. et al. An intensive longitudinal cohort study of Malian children and adults reveals no evidence of acquired immunity to Plasmodium falciparum infection. Clin. Infect. Dis. 57, 40–47 (2013). This study shows that, while repeated exposure to P. falciparum does induce clinical immunity, it does not induce immune responses capable of preventing infection.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Offeddu, V., Thathy, V., Marsh, K. & Matuschewski, K. Naturally acquired immune responses against Plasmodium falciparum sporozoites and liver infection. Int. J. Parasitol. 42, 535–548 (2012).

    Article  CAS  PubMed  Google Scholar 

  150. Doolan, D. L., Dobano, C. & Baird, J. K. Acquired immunity to malaria. Clin. Microbiol. Rev. 22, 13–36 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Weiss, W. R. & Jiang, C. G. Protective CD8+ T lymphocytes in primates immunized with malaria sporozoites. PLOS ONE 7, e31247 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Weiss, W. R., Sedegah, M., Beaudoin, R. L., Miller, L. H. & Good, M. F. CD8+ T cells (cytotoxic/suppressors) are required for protection in mice immunized with malaria sporozoites. Proc. Natl Acad. Sci. USA 85, 573–576 (1988). This work indicates that CD8 + T cells are critical for mediating immunity to liver-stage malaria.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Schmidt, N. W., Butler, N. S., Badovinac, V. P. & Harty, J. T. Extreme CD8 T cell requirements for anti-malarial liver-stage immunity following immunization with radiation attenuated sporozoites. PLOS Pathog. 6, e1000998 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Corradin, G. & Levitskaya, J. Priming of CD8(+) T cell responses to liver stage malaria parasite antigens. Front. Immunol. 5, 527 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. Zhang, N. & Bevan, M. J. CD8(+) T cells: foot soldiers of the immune system. Immunity 35, 161–168 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Jung, S. et al. In vivo depletion of CD11c+ dendritic cells abrogates priming of CD8+ T cells by exogenous cell-associated antigens. Immunity 17, 211–220 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Chakravarty, S. et al. CD8+ T lymphocytes protective against malaria liver stages are primed in skin-draining lymph nodes. Nat. Med. 13, 1035–1041 (2007). This study demonstrates the mechanism by which sporozoite-specific CD8 + T cells are primed in the local skin-draining lymph nodes.

    Article  CAS  PubMed  Google Scholar 

  158. Radtke, A. J. et al. Lymph-node resident CD8alpha+ dendritic cells capture antigens from migratory malaria sporozoites and induce CD8+ T cell responses. PLOS Pathog. 11, e1004637 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  159. Butler, N. S. et al. Superior antimalarial immunity after vaccination with late liver stage-arresting genetically attenuated parasites. Cell Host Microbe 9, 451–462 (2011). This study shows that live-attenuated Plasmodium parasites that progress further in their liver-stage development and exhibit greater antigenic breadth induce enhanced protection.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Kurup, S. P. et al. Monocyte-derived CD11c+ cells acquire Plasmodium from hepatocytes to prime CD8 T cell immunity to liver-stage malaria. Cell Host Microbe https://doi.org/10.1016/j.chom.2019.02.014 (2019). This study demonstrates a fundamental mechanism of priming CD8 + T cell responses against liver-stage malaria.

    Article  CAS  Google Scholar 

  161. Douradinha, B. et al. Genetically attenuated P36p-deficient Plasmodium berghei sporozoites confer long-lasting and partial cross-species protection. Int. J. Parasitol. 37, 1511–1519 (2007).

    Article  CAS  PubMed  Google Scholar 

  162. Kramer, L. D. & Vanderberg, J. P. Intramuscular immunization of mice with irradiated Plasmodium berghei sporozoites. Enhancement of protection with albumin. Am. J. Trop. Med. Hyg. 24, 913–916 (1975).

    Article  CAS  PubMed  Google Scholar 

  163. Holz, L. E., Fernandez-Ruiz, D. & Heath, W. R. Protective immunity to liver-stage malaria. Clin. Transl Immunol. 5, e105 (2016).

    Article  CAS  Google Scholar 

  164. Ryg-Cornejo, V. et al. NK cells and conventional dendritic cells engage in reciprocal activation for the induction of inflammatory responses during Plasmodium berghei ANKA infection. Immunobiology 218, 263–271 (2013).

    Article  CAS  PubMed  Google Scholar 

  165. Morrot, A., Hafalla, J. C., Cockburn, I. A., Carvalho, L. H. & Zavala, F. IL-4 receptor expression on CD8+ T cells is required for the development of protective memory responses against liver stages of malaria parasites. J. Exp. Med. 202, 551–560 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. da Silva, H. B. et al. Early skin immunological disturbance after Plasmodium-infected mosquito bites. Cell. Immunol. 277, 22–32 (2012).

    Article  PubMed  CAS  Google Scholar 

  167. Kimura, K. et al. CD8+ T cells specific for a malaria cytoplasmic antigen form clusters around infected hepatocytes and are protective at the liver stage of infection. Infect. Immun. 81, 3825–3834 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Cockburn, I. A. et al. In vivo imaging of CD8+ T cell-mediated elimination of malaria liver stages. Proc. Natl Acad. Sci. USA 110, 9090–9095 (2013). This work provides insight into the possible mechanisms of CD8 + T cell-mediated elimination of infected hepatocytes in the liver.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Harty, J. T., Tvinnereim, A. R. & White, D. W. CD8+ T cell effector mechanisms in resistance to infection. Annu. Rev. Immunol. 18, 275–308 (2000).

    Article  CAS  PubMed  Google Scholar 

  170. Halle, S., Halle, O. & Forster, R. Mechanisms and dynamics of T cell-mediated cytotoxicity in vivo. Trends Immunol. 38, 432–443 (2017).

    Article  CAS  PubMed  Google Scholar 

  171. Butler, N. S., Schmidt, N. W. & Harty, J. T. Differential effector pathways regulate memory CD8 T cell immunity against Plasmodium berghei versus P. yoelii sporozoites. J. Immunol. 184, 2528–2538 (2010).

    Article  CAS  PubMed  Google Scholar 

  172. Morrot, A. & Zavala, F. Regulation of the CD8+ T cell responses against Plasmodium liver stages in mice. Int. J. Parasitol. 34, 1529–1534 (2004).

    Article  CAS  PubMed  Google Scholar 

  173. Renggli, J. et al. Elimination of P. berghei liver stages is independent of Fas (CD95/Apo-I) or perforin-mediated cytotoxicity. Parasite Immunol. 19, 145–148 (1997).

    Article  CAS  PubMed  Google Scholar 

  174. Nganou-Makamdop, K., van Gemert, G. J., Arens, T., Hermsen, C. C. & Sauerwein, R. W. Long term protection after immunization with P. berghei sporozoites correlates with sustained IFNgamma responses of hepatic CD8+ memory T cells. PLOS ONE 7, e36508 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Clark, I. A., Hunt, N. H., Butcher, G. A. & Cowden, W. B. Inhibition of murine malaria (Plasmodium chabaudi) in vivo by recombinant interferon-gamma or tumor necrosis factor, and its enhancement by butylated hydroxyanisole. J. Immunol. 139, 3493–3496 (1987).

    CAS  PubMed  Google Scholar 

  176. Jobe, O. et al. Genetically attenuated Plasmodium berghei liver stages induce sterile protracted protection that is mediated by major histocompatibility complex class I-dependent interferon-gamma-producing CD8+ T cells. J. Infect. Dis. 196, 599–607 (2007).

    Article  CAS  PubMed  Google Scholar 

  177. Mellouk, S. et al. Inhibitory activity of interferons and interleukin 1 on the development of Plasmodium falciparum in human hepatocyte cultures. J. Immunol. 139, 4192–4195 (1987).

    CAS  PubMed  Google Scholar 

  178. Seguin, M. C. et al. Induction of nitric oxide synthase protects against malaria in mice exposed to irradiated Plasmodium berghei infected mosquitoes: involvement of interferon gamma and CD8+ T cells. J. Exp. Med. 180, 353–358 (1994).

    Article  CAS  PubMed  Google Scholar 

  179. Chakravarty, S., Baldeviano, G. C., Overstreet, M. G. & Zavala, F. Effector CD8+ T lymphocytes against liver stages of Plasmodium yoelii do not require gamma interferon for antiparasite activity. Infect. Immun. 76, 3628–3631 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Doolan, D. L. & Hoffman, S. L. The complexity of protective immunity against liver-stage malaria. J. Immunol. 165, 1453–1462 (2000).

    Article  CAS  PubMed  Google Scholar 

  181. Schmidt, N. W., Butler, N. S. & Harty, J. T. Plasmodium-host interactions directly influence the threshold of memory CD8 T cells required for protective immunity. J. Immunol. 186, 5873–5884 (2011).

    Article  CAS  PubMed  Google Scholar 

  182. Schmidt, N. W. et al. Memory CD8 T cell responses exceeding a large but definable threshold provide long-term immunity to malaria. Proc. Natl Acad. Sci. USA 105, 14017–14022 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Fernandez-Ruiz, D. et al. Liver-resident memory CD8(+) T cells form a front-line defense against malaria liver-stage infection. Immunity 45, 889–902 (2016). This seminal study demonstrates the role of Plasmodium antigen-specific, non-recirculating (resident) CD8 + T cells in the liver in protection from liver-stage malaria.

    Article  CAS  PubMed  Google Scholar 

  184. Gola, A. et al. Prime and target immunization protects against liver-stage malaria in mice. Sci. Transl Med. 10, eaap9128 (2018).

    Article  PubMed  CAS  Google Scholar 

  185. Olsen, T. M., Stone, B. C., Chuenchob, V. & Murphy, S. C. Prime-and-trap malaria vaccination to generate protective CD8(+) liver-resident memory T cells. J. Immunol. 201, 1984–1993 (2018).

    Article  CAS  PubMed  Google Scholar 

  186. Holz, L. E. et al. CD8(+) T cell activation leads to constitutive formation of liver tissue-resident memory T cells that seed a large and flexible niche in the liver. Cell Rep. 25, 68–79 (2018).

    Article  CAS  PubMed  Google Scholar 

  187. McNamara, H. A. et al. Up-regulation of LFA-1 allows liver-resident memory T cells to patrol and remain in the hepatic sinusoids. Sci. Immunol. 2, eaaj1996 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  188. Schenkel, J. M., Fraser, K. A., Vezys, V. & Masopust, D. Sensing and alarm function of resident memory CD8(+) T cells. Nat. Immunol. 14, 509–513 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Kumar, S. & Miller, L. H. Cellular mechanisms in immunity to blood stage infection. Immunol. Lett. 25, 109–114 (1990).

    Article  CAS  PubMed  Google Scholar 

  190. Vinetz, J. M. et al. Adoptive transfer of CD8+ T cells from immune animals does not transfer immunity to blood stage Plasmodium yoelii malaria. J. Immunol. 144, 1069–1074 (1990).

    CAS  PubMed  Google Scholar 

  191. Miyakoda, M. et al. Development of memory CD8+ T cells and their recall responses during blood-stage infection with Plasmodium berghei ANKA. J. Immunol. 189, 4396–4404 (2012).

    Article  CAS  PubMed  Google Scholar 

  192. Chandele, A., Mukerjee, P., Das, G., Ahmed, R. & Chauhan, V. S. Phenotypic and functional profiling of malaria-induced CD8 and CD4 T cells during blood-stage infection with Plasmodium yoelii. Immunology 132, 273–286 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Lundie, R. J. et al. Blood-stage Plasmodium infection induces CD8+ T lymphocytes to parasite-expressed antigens, largely regulated by CD8alpha+ dendritic cells. Proc. Natl Acad. Sci. USA 105, 14509–14514 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Junqueira, C. et al. Cytotoxic CD8(+) T cells recognize and kill Plasmodium vivax-infected reticulocytes. Nat. Med. 24, 1330–1336 (2018). This study investigates the mechanism by which CD8 + T cells contribute to the control of blood-stage malaria.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Swanson, P. A. 2nd et al. CD8+ T cells induce fatal brainstem pathology during cerebral malaria via luminal antigen-specific engagement of brain vasculature. PLOS Pathog. 12, e1006022 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  196. Belnoue, E. et al. On the pathogenic role of brain-sequestered alphabeta CD8+ T cells in experimental cerebral malaria. J. Immunol. 169, 6369–6375 (2002).

    Article  CAS  PubMed  Google Scholar 

  197. Nitcheu, J. et al. Perforin-dependent brain-infiltrating cytotoxic CD8+ T lymphocytes mediate experimental cerebral malaria pathogenesis. J. Immunol. 170, 2221–2228 (2003).

    Article  CAS  PubMed  Google Scholar 

  198. Randall, L. M. et al. Common strategies to prevent and modulate experimental cerebral malaria in mouse strains with different susceptibilities. Infect. Immun. 76, 3312–3320 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Claser, C. et al. CD8+ T cells and IFN-gamma mediate the time-dependent accumulation of infected red blood cells in deep organs during experimental cerebral malaria. PLOS ONE 6, e18720 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Haque, A. et al. Granzyme B expression by CD8+ T cells is required for the development of experimental cerebral malaria. J. Immunol. 186, 6148–6156 (2011).

    Article  CAS  PubMed  Google Scholar 

  201. Farooq, U., Dubey, M. L., Shrivastava, S. K. & Mahajan, R. C. Genetic polymorphism in Plasmodium falciparum: differentiation of parasite isolates of high & low virulence by RAPD. Indian J. Med. Res. 136, 292–295 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Kyriacou, H. M. et al. Differential var gene transcription in Plasmodium falciparum isolates from patients with cerebral malaria compared to hyperparasitaemia. Mol. Biochem. Parasitol. 150, 211–218 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Bertin, G. I. et al. Proteomic analysis of Plasmodium falciparum parasites from patients with cerebral and uncomplicated malaria. Sci. Rep. 6, 26773 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Mazier, D., Nitcheu, J. & Idrissa-Boubou, M. Cerebral malaria and immunogenetics. Parasite Immunol. 22, 613–623 (2000).

    Article  CAS  PubMed  Google Scholar 

  205. Mackey, L. J., Hochmann, A., June, C. H., Contreras, C. E. & Lambert, P. H. Immunopathological aspects of Plasmodium berghei infection in five strains of mice. II. Immunopathology of cerebral and other tissue lesions during the infection. Clin. Exp. Immunol. 42, 412–420 (1980).

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Van Braeckel-Budimir, N. et al. A T cell receptor locus harbors a malaria-specific immune response gene. Immunity 47, 835–847 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  207. White, N. J., Turner, G. D., Medana, I. M., Dondorp, A. M. & Day, N. P. The murine cerebral malaria phenomenon. Trends Parasitol. 26, 11–15 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  208. Hunt, N. H. et al. Murine cerebral malaria: the whole story. Trends Parasitol. 26, 272–274 (2010).

    Article  PubMed  Google Scholar 

  209. Chien, Y. H., Meyer, C. & Bonneville, M. γδ T cells: first line of defense and beyond. Annu. Rev. Immunol. 32, 121–155 (2014).

    Article  CAS  PubMed  Google Scholar 

  210. Holtmeier, W. & Kabelitz, D. γδ T cells link innate and adaptive immune responses. Chem. Immunol. Allergy 86, 151–183 (2005).

    Article  CAS  PubMed  Google Scholar 

  211. Hayday, A. C. γδ cells: a right time and a right place for a conserved third way of protection. Annu. Rev. Immunol. 18, 975–1026 (2000).

    Article  CAS  PubMed  Google Scholar 

  212. Carding, S. R. & Egan, P. J. Gammadelta T cells: functional plasticity and heterogeneity. Nat. Rev. Immunol. 2, 336–345 (2002).

    Article  CAS  PubMed  Google Scholar 

  213. Zhao, Y., Niu, C. & Cui, J. Gamma-delta (γδ) T cells: friend or foe in cancer development? J. Transl Med. 16, 3 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Kurup, S. P. & Harty, J. T. γδ T cells and immunity to human malaria in endemic regions. Ann. Transl Med. 3, S22 (2015).

    PubMed  PubMed Central  Google Scholar 

  215. Hviid, L. et al. Perturbation and proinflammatory type activation of V delta 1(+) gamma delta T cells in African children with Plasmodium falciparum malaria. Infect. Immun. 69, 3190–3196 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Langhorne, J., Morris-Jones, S., Casabo, L. G. & Goodier, M. The response of gamma delta T cells in malaria infections: a hypothesis. Res. Immunol. 145, 429–436 (1994).

    Article  CAS  PubMed  Google Scholar 

  217. Roussilhon, C., Agrapart, M., Ballet, J. J. & Bensussan, A. T lymphocytes bearing the gamma delta T cell receptor in patients with acute Plasmodium falciparum malaria. J. Infect. Dis. 162, 283–285 (1990).

    Article  CAS  PubMed  Google Scholar 

  218. Teirlinck, A. C. et al. Longevity and composition of cellular immune responses following experimental Plasmodium falciparum malaria infection in humans. PLOS Pathog. 7, e1002389 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. D’Ombrain, M. C. et al. Association of early interferon-gamma production with immunity to clinical malaria: a longitudinal study among Papua New Guinean children. Clin. Infect. Dis. 47, 1380–1387 (2008).

    Article  PubMed  CAS  Google Scholar 

  220. Goodier, M. et al. Gamma delta T cells in the peripheral blood of individuals from an area of holoendemic Plasmodium falciparum transmission. Trans. R. Soc. Trop. Med. Hyg. 87, 692–696 (1993).

    Article  CAS  PubMed  Google Scholar 

  221. Jagannathan, P. et al. Loss and dysfunction of Vdelta2(+) γδ T cells are associated with clinical tolerance to malaria. Sci. Transl Med. 6, 251ra117 (2014). This work investigates the role of γδ T cells in blood-stage malaria in humans in a malaria-endemic region and possible disease outcomes.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  222. De Rosa, S. C. et al. Ontogeny of gamma delta T cells in humans. J. Immunol. 172, 1637–1645 (2004).

    Article  PubMed  Google Scholar 

  223. Dondorp, A. M. et al. The relationship between age and the manifestations of and mortality associated with severe malaria. Clin. Infect. Dis. 47, 151–157 (2008).

    Article  PubMed  Google Scholar 

  224. Schwartz, E., Sadetzki, S., Murad, H. & Raveh, D. Age as a risk factor for severe Plasmodium falciparum malaria in nonimmune patients. Clin. Infect. Dis. 33, 1774–1777 (2001).

    Article  CAS  PubMed  Google Scholar 

  225. Inoue, S. I., Niikura, M., Asahi, H., Kawakami, Y. & Kobayashi, F. γδ T cells modulate humoral immunity against Plasmodium berghei infection. Immunology 155, 519–532 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Mamedov, M. R. et al. A macrophage colony-stimulating-factor-producing γδ T cell subset prevents malarial parasitemic recurrence. Immunity 48, 350–363 (2018). This study identifies a protective, M-CSF-producing γδ T cell subset in the later stage of malaria infection.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Zaidi, I. et al. γδ T cells are required for the induction of sterile immunity during irradiated sporozoite vaccinations. J. Immunol. 199, 3781–3788 (2017).

    Article  CAS  PubMed  Google Scholar 

  228. Zeng, X. et al. γδ T cells recognize a microbial encoded B cell antigen to initiate a rapid antigen-specific interleukin-17 response. Immunity 37, 524–534 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Cai, Y. et al. Differential developmental requirement and peripheral regulation for dermal Vgamma4 and Vgamma6T17 cells in health and inflammation. Nat. Commun. 5, 3986 (2014).

    Article  CAS  PubMed  Google Scholar 

  230. Paul, S. & Lal, G. Regulatory and effector functions of gamma-delta (γδ) T cells and their therapeutic potential in adoptive cellular therapy for cancer. Int. J. Cancer 139, 976–985 (2016).

    Article  CAS  PubMed  Google Scholar 

  231. Constant, P. et al. Stimulation of human gamma delta T cells by nonpeptidic mycobacterial ligands. Science 264, 267–270 (1994).

    Article  CAS  PubMed  Google Scholar 

  232. Sheridan, B. S. et al. γδ T cells exhibit multifunctional and protective memory in intestinal tissues. Immunity 39, 184–195 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Elloso, M. M., van der Heyde, H. C., vande Waa, J. A., Manning, D. D. & Weidanz, W. P. Inhibition of Plasmodium falciparum in vitro by human gamma delta T cells. J. Immunol. 153, 1187–1194 (1994).

    CAS  PubMed  Google Scholar 

  234. Hensmann, M. & Kwiatkowski, D. Cellular basis of early cytokine response to Plasmodium falciparum. Infect. Immun. 69, 2364–2371 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Behr, C. et al. Plasmodium falciparum stimuli for human γδ T cells are related to phosphorylated antigens of mycobacteria. Infect. Immun. 64, 2892–2896 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Elloso, M. M., van der Heyde, H. C., Troutt, A., Manning, D. D. & Weidanz, W. P. Human gamma delta T cell subset-proliferative response to malarial antigen in vitro depends on CD4+ T cells or cytokines that signal through components of the IL-2R. J. Immunol. 157, 2096–2102 (1996).

    CAS  PubMed  Google Scholar 

  237. Jones, S. M., Goodier, M. R. & Langhorne, J. The response of gamma delta T cells to Plasmodium falciparum is dependent on activated CD4+ T cells and the recognition of MHC class I molecules. Immunology 89, 405–412 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Pichyangkul, S., Saengkrai, P., Yongvanitchit, K., Stewart, A. & Heppner, D. G. Activation of γδ T cells in malaria: interaction of cytokines and a schizont-associated Plasmodium falciparum antigen. J. Infect. Dis. 176, 233–241 (1997).

    Article  CAS  PubMed  Google Scholar 

  239. Cockburn, I. A. & Seder, R. A. Malaria prevention: from immunological concepts to effective vaccines and protective antibodies. Nat. Immunol. 19, 1199–1211 (2018).

    Article  CAS  PubMed  Google Scholar 

  240. Draper, S. J. et al. Malaria vaccines: recent advances and new horizons. Cell Host Microbe 24, 43–56 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Butler, N. S., Vaughan, A. M., Harty, J. T. & Kappe, S. H. Whole parasite vaccination approaches for prevention of malaria infection. Trends Immunol. 33, 247–254 (2012).

    Article  CAS  PubMed  Google Scholar 

  242. Hoffman, S. L., Vekemans, J., Richie, T. L. & Duffy, P. E. The march toward malaria vaccines. Am. J. Prev. Med. 49, S319–S333 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  243. Hill, A. V. Vaccines against malaria. Phil. Trans. R. Soc. B 366, 2806–2814 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  244. Ewer, K. J. et al. Progress with viral vectored malaria vaccines: A multi-stage approach involving “unnatural immunity”. Vaccine 33, 7444–7451 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. Li, Y. et al. Enhancing immunogenicity and transmission-blocking activity of malaria vaccines by fusing Pfs25 to IMX313 multimerization technology. Sci. Rep. 6, 18848 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  246. Doll, K. L., Pewe, L. L., Kurup, S. P. & Harty, J. T. Discriminating protective from nonprotective Plasmodium-specific CD8+ T cell responses. J. Immunol. 196, 4253–4262 (2016).

    Article  CAS  PubMed  Google Scholar 

  247. Lau, L. S. et al. CD8+ T cells from a novel T cell receptor transgenic mouse induce liver-stage immunity that can be boosted by blood-stage infection in rodent malaria. PLOS Pathog. 10, e1004135 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  248. Fernandez-Ruiz, D. et al. Development of a novel CD4(+) TCR transgenic line that reveals a dominant role for CD8(+) dendritic cells and CD40 signaling in the generation of helper and CTL responses to blood-stage malaria. J. Immunol. 199, 4165–4179 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Shekalaghe, S. et al. Controlled human malaria infection of Tanzanians by intradermal injection of aseptic, purified, cryopreserved Plasmodium falciparum sporozoites. Am. J. Trop. Med. Hyg. 91, 471–480 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  250. Hodgson, S. H. et al. Evaluating controlled human malaria infection in Kenyan adults with varying degrees of prior exposure to Plasmodium falciparum using sporozoites administered by intramuscular injection. Front. Microbiol. 5, 686 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  251. Obiero, J. M. et al. Impact of malaria preexposure on antiparasite cellular and humoral immune responses after controlled human malaria infection. Infect. Immun. 83, 2185–2196 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors apologize to the countless researchers whose contributions are not discussed in this manuscript owing to space limitations. They also thank the Butler and Harty laboratory members for helpful discussions. Work in the laboratory of N.S.B. is supported by grants from the US National Institutes of Health (NIH) (AI125446 and AI127481). Work in the laboratory of J.T.H. is supported by grants from the NIH (AI42767, AI95178, AI100527 and AI114543).

Reviewer information

Nature Reviews Immunology thanks A. Haque, M. Good and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the discussion of content and to the writing and the editing of the manuscript.

Corresponding author

Correspondence to John T. Harty.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Sporozoite

A Plasmodium parasite life form transmitted by mosquito bite and capable of initiating the asexual cycle of growth and differentiation in the vertebrate host.

Merozoites

Plasmodium parasite life forms that first develop in infected hepatocytes and are capable of initiating either sexual or asexual cycles of development in host red blood cells.

Merosome

Host cell-derived, membrane-bound structure containing multiple merozoites that buds from infected hepatocytes during Plasmodium egress from the liver. Merosomes release merozoites into circulation after rupture.

RTS,S

The candidate anti-malarial vaccine furthest along in global development. RTS,S comprises two subdomains of the Plasmodium falciparum circumsporozoite protein (CSP) that are associated with units of the hepatitis B surface antigen and formulated with the adjuvant AS01 (3-O-desacyl-4′-monophosphoryl lipid A and the saponin QS-21). Infection is prevented by inducing antibodies that either immobilize sporozoites in the skin or prevent sporozoites from infecting the liver.

Circumsporozoite protein

(CSP). Immunodominant, high-density surface antigen expressed by Plasmodium sporozoites that is the target of humoral and cellular immune responses that either block sporozoite infection of the liver or eliminate infected hepatocytes, respectively.

Chemoprophylaxis and sporozoite (CPS) immunization

A vaccination strategy whereby virulent sporozoites are delivered by either mosquito bites or needle injection with prophylactic delivery of a drug targeting Plasmodium blood stages. Parasites initiate and complete liver-stage development, release merozoites and initiate the first wave of blood-stage infection before being eliminated by the drug. Vaccinated individuals are thereby exposed to antigens that derive from multiple parasite life cycle stages while remaining protected against clinical disease by the anti-blood-stage drug.

Apicoplasts

Non-photosynthetic organelles that characterize protists within the phylum Apicomplexa and are likely derived from an algal endosymbiont. All apicoplast functions are not fully known, but defined activities primarily relate to essential metabolic pathways necessary for the viability of Plasmodium and other apicomplexans.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kurup, S.P., Butler, N.S. & Harty, J.T. T cell-mediated immunity to malaria. Nat Rev Immunol 19, 457–471 (2019). https://doi.org/10.1038/s41577-019-0158-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41577-019-0158-z

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