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:

Quantitative imaging of Plasmodium transmission from mosquito to mammal

Abstract

Plasmodium, the parasite that causes malaria, is transmitted by a mosquito into the dermis and must reach the liver before infecting erythrocytes and causing disease. We present here a quantitative, real-time analysis of the fate of parasites transmitted in a rodent system. We show that only a proportion of the parasites enter blood capillaries, whereas others are drained by lymphatics. Lymph sporozoites stop at the proximal lymph node, where most are degraded inside dendritic leucocytes, but some can partially differentiate into exoerythrocytic stages. This previously unrecognized step of the parasite life cycle could influence the immune response of the host, and may have implications for vaccination strategies against the preerythrocytic stages of the parasite.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Sporozoite motility in the dermis.
Figure 2: Dermis sporozoites invade blood vessels.
Figure 3: Dermis sporozoites invade lymphatic vessels.
Figure 4: Sporozoite fate in the draining lymph node.

Similar content being viewed by others

References

  1. Beier, J.C. Malaria parasite development in mosquitoes. Annu. Rev. Entomol. 43, 519–543 (1998).

    Article  CAS  Google Scholar 

  2. Frischknecht, F. et al. Imaging movement of malaria parasites during transmission by Anopheles mosquitoes. Cell. Microbiol. 6, 687–694 (2004).

    Article  CAS  Google Scholar 

  3. Boyd, M.F. & Kitchen, S.F. The demonstration of sporozoites in human tissues. Am. J. Trop. Med. Hyg. 19, 27–31 (1939).

    Article  Google Scholar 

  4. Ponnudurai, T., Lensen, A.H., van Gemert, G.J., Bolmer, M.G. & Meuwissen, J.H. Feeding behaviour and sporozoite ejection by infected Anopheles stephensi. Trans. R. Soc. Trop. Med. Hyg. 85, 175–180 (1991).

    Article  CAS  Google Scholar 

  5. Matsuoka, H., Yoshida, S., Hirai, M. & Ishii, A. A rodent malaria, Plasmodium berghei, is experimentally transmitted to mice by merely probing of infective mosquito, Anopheles stephensi. Parasitol. Int. 51, 17–23 (2002).

    Article  Google Scholar 

  6. Sidjanski, S. & Vanderberg, J.P. Delayed migration of Plasmodium sporozoites from the mosquito bite site to the blood. Am. J. Trop. Med. Hyg. 57, 426–429 (1997).

    Article  CAS  Google Scholar 

  7. Natarajan, R. et al. Fluorescent Plasmodium berghei sporozoites and pre-erythrocytic stages: a new tool to study mosquito and mammalian host interactions with malaria parasites. Cell. Microbiol. 3, 371–379 (2001).

    Article  CAS  Google Scholar 

  8. 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 

  9. Vanderberg, J.P. Studies on the motility of Plasmodium sporozoites. J. Protozool. 21, 527–537 (1974).

    Article  CAS  Google Scholar 

  10. Vaughan, J.A., Scheller, L.F., Wirtz, R.A. & Azad, A.F. Infectivity of Plasmodium berghei sporozoites delivered by intravenous inoculation versus mosquito bite: implications for sporozoite vaccine trials. Infect. Immun. 67, 4285–4289 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Krettli, A.U. & Dantas, L.A. Which routes do Plasmodium sporozoites use for successful infections of vertebrates? Infect. Immun. 68, 3064–3065 (2000).

    Article  CAS  Google Scholar 

  12. Meis, J.F. & Verhave, J.P. Exoerythrocytic development of malaria parasites. Adv. Parasitol. 27, 1–61 (1988).

    Article  CAS  Google Scholar 

  13. Grüner, A.C. et al. Insights into the P. y. yoelii hepatic stage transcriptome reveal complex transcriptional patterns. Mol. Biochem. Parasitol. 142, 184–192 (2005).

    Article  Google Scholar 

  14. Sacci, J.B., Jr. et al. Transcriptional analysis of in vivo Plasmodium yoelii liver stage gene expression. Mol. Biochem. Parasitol. 142, 177–183 (2005).

    Article  CAS  Google Scholar 

  15. Charoenvit, Y. et al. Plasmodium yoelii: 17-kDa hepatic and erythrocytic stage protein is the target of an inhibitory monoclonal antibody. Exp. Parasitol. 80, 419–429 (1995).

    Article  CAS  Google Scholar 

  16. Luke, T.C. & Hoffman, S.L. Rationale and plans for developing a non-replicating, metabolically active, radiation-attenuated Plasmodium falciparum sporozoite vaccine. J. Exp. Biol. 206, 3803–3808 (2003).

    Article  Google Scholar 

  17. Mueller, A.K., Labaied, M., Kappe, S.H. & Matuschewski, K. Genetically modified Plasmodium parasites as a protective experimental malaria vaccine. Nature 433, 164–167 (2005).

    Article  CAS  Google Scholar 

  18. Mueller, A.K. et al. Plasmodium liver stage developmental arrest by depletion of a protein at the parasite-host interface. Proc. Natl. Acad. Sci. USA 102, 3022–3027 (2005).

    Article  CAS  Google Scholar 

  19. Good, M.F. Genetically modified Plasmodium highlights the potential of whole parasite vaccine strategies. Trends Immunol. 26, 295–297 (2005).

    Article  CAS  Google Scholar 

  20. Tongren, J.E., Zavala, F., Roos, D.S. & Riley, E.M. Malaria vaccines: if at first you don't succeed.... Trends Parasitol. 20, 604–610 (2004).

    Article  CAS  Google Scholar 

  21. Bousso, P. & Robey, E. Dynamics of CD8+ T cell priming by dendritic cells in intact lymph nodes. Nat. Immunol. 4, 579–585 (2003).

    Article  CAS  Google Scholar 

  22. Hugues, S. et al. Distinct T cell dynamics in lymph nodes during the induction of tolerance and immunity. Nat. Immunol. 5, 1235–1242 (2004).

    Article  CAS  Google Scholar 

  23. Sumen, C., Mempel, T.R., Mazo, I.B. & von Andrian, U.H. Intravital microscopy: visualizing immunity in context. Immunity 21, 315–329 (2004).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Genovesio, C. Zimmer and J.-C. Olivo-Marin for help with tracking analysis, P. Roux for help with confocal microscopy, the members of the Center for Production and Infection of Anopheles of the Pasteur Institute for mosquitoes rearing, B. Boisson for help in RT-PCR and C. Janse for providing PbGFPCON parasites. We are grateful to G. Milon, C. Bourgouin, P. Sinnis, S. Mecheri and F. Zavala for comments on the manuscript. The work was supported by funds from the Pasteur Institute (Strategic project 'Grand Programme Horizontal Anopheles'), the Howard Hughes Medical Institute and the European Commission (FP6 BioMalPar Network of Excellence). R.A. was supported by the Pasteur Institute Grand Programme Horizontal fellowship and F.F. by a Human Frontier Science Program long-term fellowship. R.M. is a Howard Hughes Medical Institute International Scholar.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Rogerio Amino, Friedrich Frischknecht or Robert Ménard.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Fig. 1

The number of sporozoites at the site of mosquito bite decreases with time. (PDF 53 kb)

Supplementary Table 1

Lymph sporozoites end their journey in the first draining lymph node. (PDF 19 kb)

Supplementary Movie 1

Sporozoite gliding in the skin. Two time-lapse series showing 200 seconds of sporozoite movement in the dermis of a hairless mouse at 3 minutes and 19 minutes after a single mosquito bite. The maximum projections of the fluorescent signal at the end of the respective time-lapse series show that the sporozoite gliding velocity decreases with time. Image series acquired with an epifluorescent wide-field microscope. (MOV 3296 kb)

Supplementary Movie 2

A sporozoite glides for 114 seconds with high velocity in the dermis, before slowing down upon encountering a blood vessel and invading the blood vessel wall; note the constriction (arrowhead) of the parasite at 282 seconds. After invading the blood vessel, the sporozoite rests several seconds inside the vessel before being taken away with the blood stream between 300 and 306 seconds. The red color represents projected fluorescent signals after injection of fluorescently labeled BSA, which was used to detect blood vessels with the spinning disk confocal microscope (BSA is taken up by endothelial and other dermal cells). The green signal of the sporozoite corresponds to a single confocal plane. Image series acquired with a spinning disk confocal microscope. (MOV 1608 kb)

Supplementary Movie 3

A sporozoite glides in the skin for 106 seconds before slowing down its speed and displaying a moving constriction (arrowhead). From 130 seconds onwards, the sporozoite drifts sideways for several hundred seconds. A second sporozoite (entering the field at 83 seconds) is also seen drifting sideways. The fluorescent signal of the sporozoite corresponds to a single confocal plane. Image series acquired with a spinning disk confocal microscope. (MOV 4761 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Amino, R., Thiberge, S., Martin, B. et al. Quantitative imaging of Plasmodium transmission from mosquito to mammal. Nat Med 12, 220–224 (2006). https://doi.org/10.1038/nm1350

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm1350

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