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.

  • Letter
  • Published:

Host-mediated regulation of superinfection in malaria

Abstract

In regions of high rates of malaria transmission, mosquitoes repeatedly transmit liver-tropic Plasmodium sporozoites to individuals who already have blood-stage parasitemia1. This manifests itself in semi-immune children (who have been exposed since birth to Plasmodium infection and as such show low levels of peripheral parasitemia but can still be infected) older than 5 years of age by concurrent carriage of different parasite genotypes at low asymptomatic parasitemias2. Superinfection presents an increased risk of hyperparasitemia and death in less immune individuals but counterintuitively is not frequently observed in the young3,4. Here we show in a mouse model that ongoing blood-stage infections, above a minimum threshold, impair the growth of subsequently inoculated sporozoites such that they become growth arrested in liver hepatocytes and fail to develop into blood-stage parasites. Inhibition of the liver-stage infection is mediated by the host iron regulatory hormone hepcidin5, whose synthesis we found to be stimulated by blood-stage parasites in a density-dependent manner. We mathematically modeled this phenomenon and show how density-dependent protection against liver-stage malaria can shape the epidemiological patterns of age-related risk and the complexity of malaria infections seen in young children. The interaction between these two Plasmodium stages and host iron metabolism has relevance for the global efforts to reduce malaria transmission and for evaluation of iron supplementation programs in malaria-endemic regions.

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: Malaria blood-stage infection protects against secondary Plasmodium liver infection.
Figure 2: Effect of blood-stage infection on superinfecting sporozoite EEF number or development and on sporozoite infection of hepatocytes.
Figure 3: Hepcidin induction during blood-stage infection leads to iron redistribution and restricts Plasmodium liver infection.
Figure 4: Model predictions for age-related incidence and multiplicity of infections under different inhibitory effects and transmission intensities.

Similar content being viewed by others

References

  1. Robert, V. et al. Malaria transmission in urban sub-Saharan Africa. Am. J. Trop. Med. Hyg. 68, 169–176 (2003).

    Article  Google Scholar 

  2. al-Yaman, F. et al. Reduced risk of clinical malaria in children infected with multiple clones of Plasmodium falciparum in a highly endemic area: a prospective community study. Trans. R. Soc. Trop. Med. Hyg. 91, 602–605 (1997).

    Article  CAS  Google Scholar 

  3. Smith, T.A., Leuenberger, R. & Lengeler, C. Child mortality and malaria transmission intensity in Africa. Trends Parasitol. 17, 145–149 (2001).

    Article  CAS  Google Scholar 

  4. Molineaux, L. & Gramiccia, G. The Garki Project: Research on the Epidemiology and Control of Malaria in the Sudan Savanna of West Africa (World Health Organization, Geneva, 1980).

  5. Ganz, T. Hepcidin—a peptide hormone at the interface of innate immunity and iron metabolism. Curr. Top. Microbiol. Immunol. 306, 183–198 (2006).

    CAS  PubMed  Google Scholar 

  6. Prudêncio, M., Rodriguez, A. & Mota, M.M. The silent path to thousands of merozoites: the Plasmodium liver stage. Nat. Rev. Microbiol. 4, 849–856 (2006).

    Article  Google Scholar 

  7. Haldar, K., Murphy, S.C., Milner, D.A. & Taylor, T.E. Malaria: mechanisms of erythrocytic infection and pathological correlates of severe disease. Annu. Rev. Pathol. 2, 217–249 (2007).

    Article  CAS  Google Scholar 

  8. Ploemen, I.H. et al. Visualisation and quantitative analysis of the rodent malaria liver stage by real time imaging. PLoS ONE 4, e7881 (2009).

    Article  Google Scholar 

  9. Henke, J.M. & Bassler, B.L. Bacterial social engagements. Trends Cell Biol. 14, 648–656 (2004).

    Article  CAS  Google Scholar 

  10. Guha, M., Kumar, S., Choubey, V., Maity, P. & Bandyopadhyay, U. Apoptosis in liver during malaria: role of oxidative stress and implication of mitochondrial pathway. FASEB J. 20, 1224–1226 (2006).

    Article  CAS  Google Scholar 

  11. Heppner, D.G., Hallaway, P.E., Kontoghiorghes, G.J. & Eaton, J.W. Antimalarial properties of orally active iron chelators. Blood 72, 358–361 (1988).

    CAS  PubMed  Google Scholar 

  12. Goma, J., Renia, L., Miltgen, F. & Mazier, D. Effects of iron deficiency on the hepatic development of Plasmodium yoelii. Parasite 2, 351–356 (1995).

    CAS  PubMed  Google Scholar 

  13. Goma, J., Renia, L., Miltgen, F. & Mazier, D. Iron overload increases hepatic development of Plasmodium yoelii in mice. Parasitology 112, 165–168 (1996).

    Article  CAS  Google Scholar 

  14. Stahel, E. et al. Iron chelators: in vitro inhibitory effect on the liver stage of rodent and human malaria. Am. J. Trop. Med. Hyg. 39, 236–240 (1988).

    Article  CAS  Google Scholar 

  15. Armitage, A.E., Pinches, R., Eddowes, L.A., Newbold, C.I. & Drakesmith, H. Plasmodium falciparum infected erythrocytes induce hepcidin (HAMP) mRNA synthesis by peripheral blood mononuclear cells. Br. J. Haematol. 147, 769–771 (2009).

    Article  CAS  Google Scholar 

  16. de Mast, Q. et al. Assessment of urinary concentrations of hepcidin provides novel insight into disturbances in iron homeostasis during malarial infection. J. Infect. Dis. 199, 253–262 (2009).

    Article  Google Scholar 

  17. Nemeth, E. et al. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 306, 2090–2093 (2004).

    Article  CAS  Google Scholar 

  18. Babitt, J.L. et al. Modulation of bone morphogenetic protein signaling in vivo regulates systemic iron balance. J. Clin. Invest. 117, 1933–1939 (2007).

    Article  CAS  Google Scholar 

  19. Nemeth, E. et al. Hepcidin, a putative mediator of anemia of inflammation, is a type II acute-phase protein. Blood 101, 2461–2463 (2003).

    Article  CAS  Google Scholar 

  20. Katagiri, T. et al. Identification of a BMP-responsive element in Id1, the gene for inhibition of myogenesis. Genes Cells 7, 949–960 (2002).

    Article  CAS  Google Scholar 

  21. Yu, P.B. et al. Dorsomorphin inhibits BMP signals required for embryogenesis and iron metabolism. Nat. Chem. Biol. 4, 33–41 (2008).

    Article  CAS  Google Scholar 

  22. Albuquerque, S.S. et al. Host cell transcriptional profiling during malaria liver stage infection reveals a coordinated and sequential set of biological events. BMC Genomics 10, 270 (2009).

    Article  Google Scholar 

  23. Drakesmith, H. et al. Resistance to hepcidin is conferred by hemochromatosis-associated mutations of ferroportin. Blood 106, 1092–1097 (2005).

    Article  CAS  Google Scholar 

  24. Rivera, S. et al. Synthetic hepcidin causes rapid dose-dependent hypoferremia and is concentrated in ferroportin-containing organs. Blood 106, 2196–2199 (2005).

    Article  CAS  Google Scholar 

  25. Roy, C.N. et al. Hepcidin antimicrobial peptide transgenic mice exhibit features of the anemia of inflammation. Blood 109, 4038–4044 (2007).

    Article  CAS  Google Scholar 

  26. Sama, W., Owusu-Agyei, S., Felger, I., Dietz, K. & Smith, T. Age and seasonal variation in the transition rates and detectability of Plasmodium falciparum malaria. Parasitology 132, 13–21 (2006).

    Article  CAS  Google Scholar 

  27. Mayor, A. et al. Plasmodium falciparum multiple infections in Mozambique, its relation to other malariological indices and to prospective risk of malaria morbidity. Trop. Med. Int. Health 8, 3–11 (2003).

    Article  Google Scholar 

  28. Owusu-Agyei, S., Smith, T., Beck, H.P., Amenga-Etego, L. & Felger, I. Molecular epidemiology of Plasmodium falciparum infections among asymptomatic inhabitants of a holoendemic malarious area in northern Ghana. Trop. Med. Int. Health 7, 421–428 (2002).

    Article  CAS  Google Scholar 

  29. Mmbando, B.P. et al. Epidemiology of malaria in an area prepared for clinical trials in Korogwe, north-eastern Tanzania. Malar. J. 8, 165 (2009).

    Article  Google Scholar 

  30. Smith, T. et al. Relationship between the entomologic inoculation rate and the force of infection for Plasmodium falciparum malaria. Am. J. Trop. Med. Hyg. 75, 11–18 (2006).

    Article  Google Scholar 

  31. Fuqua, C., Winans, S.C. & Greenberg, E.P. Census and consensus in bacterial ecosystems: the LuxR-LuxI family of quorum-sensing transcriptional regulators. Annu. Rev. Microbiol. 50, 727–751 (1996).

    Article  CAS  Google Scholar 

  32. López, D., Vlamakis, H., Losick, R. & Kolter, R. Paracrine signaling in a bacterium. Genes Dev. 23, 1631–1638 (2009).

    Article  Google Scholar 

  33. Hughes, D.T. & Sperandio, V. Inter-kingdom signalling: communication between bacteria and their hosts. Nat. Rev. Microbiol. 6, 111–120 (2008).

    Article  CAS  Google Scholar 

  34. Sazawal, S. et al. Effects of routine prophylactic supplementation with iron and folic acid on admission to hospital and mortality in preschool children in a high malaria transmission setting: community-based, randomised, placebo-controlled trial. Lancet 367, 133–143 (2006).

    Article  CAS  Google Scholar 

  35. Franke-Fayard, B. et al. A Plasmodium berghei reference line that constitutively expresses GFP at a high level throughout the complete life cycle. Mol. Biochem. Parasitol. 137, 23–33 (2004).

    Article  CAS  Google Scholar 

  36. Tarun, A.S. et al. Quantitative isolation and in vivo imaging of malaria parasite liver stages. Int. J. Parasitol. 36, 1283–1293 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank L. Renia (Agency for Science, Technology and Research, Singapore) for helpful comments and S-methylthiourea and N-acetylcysteine, as well as R. Appelberg (Instituto de Biologia Molecular e Celular (IBMC), Portugal), J. Di Santo (Institut Pasteur) and J. Rivera (US National Institutes of Arthritis and Musculoskeletal and Skin Diseases) for IFN-γ–, RAG2γc- and Mcpt4-deficient mice, respectively, S. Rivella (Weill Cornell Medical College) for GFP- and HAMP.GFP-expressing adenoviruses, and P. Arosio (Faculty of Medicine, University of Brescia) for antibodies against mouse heavy and light ferritin chains. P. berghei ANKA, P. berghei NK65, P. yoelii 17X NL and P. chabaudi chabaudi AS were obtained from C. Jansen (Leiden University), A. Rodriguez (New York University), D. Walliker (Edinburgh University) and W. Jarra (UK National Institute for Medical Research), respectively. This work was supported by the European Community's Seventh Framework Programme (FP7/2007-2013) under grant agreement 242095 (M.M.M.), Fundação para a Ciência e Tecnologia, European Science Foundation (European Young Investigator Award to M.M.M.), Howard Hughes Medical Institute (to M.M.M.) and the Medical Research Council UK (grant ID 92044). H.D. is a Beit Memorial Fellow for Medical Research and a Medical Research Council New Investigator. S.P. and C.C. were supported by Fundação para a Ciência e a Tecnologia fellowships (SFRH/BD/31523/2006 and SFRH/BPD/40965/2007, respectively). M.R. is supported by a Royal Society University Research Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

S.P. performed the majority of the experimental work. C.C. performed the microarray analysis, and M.R. and C.I.N. initiated and tested the mathematical model. L.A.G. produced mouse primary hepatocytes. S.E., D.S. and C.R. provided intellectual input, and C.R. provided HAMP-transgenic mice. M.M.M. conceived of and supervised the study. S.P., A.E.A., C.I.N., H.D. and M.M.M. designed the experimental procedures and wrote the manuscript. All authors read and approved of the manuscript.

Corresponding author

Correspondence to Maria M Mota.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Methods (PDF 1323 kb)

Supplementary Table

Supplementary Table 1 (XLS 759 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Portugal, S., Carret, C., Recker, M. et al. Host-mediated regulation of superinfection in malaria. Nat Med 17, 732–737 (2011). https://doi.org/10.1038/nm.2368

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2368

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