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:

Plasmodium UIS3 sequesters host LC3 to avoid elimination by autophagy in hepatocytes

Abstract

The causative agent of malaria, Plasmodium, replicates inside a membrane-bound parasitophorous vacuole (PV), which shields this intracellular parasite from the cytosol of the host cell1. One common threat for intracellular pathogens is the homeostatic process of autophagy, through which cells capture unwanted intracellular material for lysosomal degradation2. During the liver stage of a malaria infection, Plasmodium parasites are targeted by the autophagy machinery of the host cell, and the PV membrane (PVM) becomes decorated with several autophagy markers, including LC3 (microtubule-associated protein 1 light chain 3)3,4. Here we show that Plasmodium berghei parasites infecting hepatic cells rely on the PVM transmembrane protein UIS3 to avoid elimination by host-cell-mediated autophagy. We found that UIS3 binds host LC3 through a non-canonical interaction with a specialized surface on LC3 where host proteins with essential functions during autophagy also bind. UIS3 acts as a bona fide autophagy inhibitor by competing with host LC3-interacting proteins for LC3 binding. Our work identifies UIS3, one of the most promising candidates for a genetically attenuated vaccine against malaria5, as a unique and potent mediator of autophagy evasion in Plasmodium. We propose that the protein–protein interaction between UIS3 and host LC3 represents a target for antimalarial drug development.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Plasmodium UIS3 protects liver-stage parasites from host autophagy.
Fig. 2: Plasmodium UIS3 binds to host LC3.
Fig. 3: Non-canonical binding of Plasmodium UIS3 to the LIR pocket of LC3.
Fig. 4: UIS3 acts as a bona fide autophagy inhibitor by competing with host LC3-interacting proteins for LC3 binding.

Similar content being viewed by others

References

  1. Liehl, P., Zuzarte-Luis, V. & Mota, M. M. Unveiling the pathogen behind the vacuole. Nat. Rev. Microbiol. 13, 589–598 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Gomes, L. C. & Dikic, I. Autophagy in antimicrobial immunity. Mol. Cell 54, 224–233 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Prado, M. et al. Long-term live imaging reveals cytosolic immune responses of host hepatocytes against Plasmodium infection and parasite escape mechanisms. Autophagy 11, 1561–1579 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Thieleke-Matos, C. et al. Host cell autophagy contributes to Plasmodium liver development. Cell. Microbiol. 18, 437–450 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  6. Shen, H.-M. & Mizushima, N. At the end of the autophagic road: an emerging understanding of lysosomal functions in autophagy. Trends Biochem. Sci. 39, 61–71 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. 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  PubMed  PubMed Central  Google Scholar 

  8. Hanson, K. K. et al. Torins are potent antimalarials that block replenishment of Plasmodium liver stage parasitophorous vacuole membrane proteins. Proc. Natl Acad. Sci. USA 110, E2838–E2847 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Spielmann, T., Montagna, G. N., Hecht, L. & Matuschewski, K. Molecular make-up of the Plasmodium parasitophorous vacuolar membrane. Int. J. Med. Microbiol. 302, 179–186 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Mizushima, N. et al. Dissection of autophagosome formation using Apg5-deficient mouse embryonic stem cells. J. Cell Biol. 152, 657–667 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ganley, I. G., Wong, P.-M., Gammoh, N. & Jiang, X. Distinct autophagosomal–lysosomal fusion mechanism revealed by thapsigargin-induced autophagy arrest. Mol. Cell 42, 731–743 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sturm, A. et al. Alteration of the parasite plasma membrane and the parasitophorous vacuole membrane during exo-erythrocytic development of malaria parasites. Protist 160, 51–63 (2009).

    Article  PubMed  Google Scholar 

  13. Sharma, A., Yogavel, M., Akhouri, R. R., Gill, J. & Sharma, A. Crystal structure of soluble domain of malaria sporozoite protein UIS3 in complex with lipid. J. Biol. Chem. 283, 24077–24088 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Mikolajczak, S. A., Jacobs-Lorena, V., MacKellar, D. C., Camargo, N. & Kappe, S. H. I. L-FABP is a critical host factor for successful malaria liver stage development. Int. J. Parasitol. 37, 483–489 (2007).

    Article  CAS  PubMed  Google Scholar 

  15. Favretto, F., Assfalg, M., Molinari, H. & D’Onofrio, M. Evidence from NMR interaction studies challenges the hypothesis of direct lipid transfer from L-FABP to malaria sporozoite protein UIS3. Prot. Sci. 22, 133–138 (2013).

    Article  CAS  Google Scholar 

  16. Farré, J.-C. & Subramani, S. Mechanistic insights into selective autophagy pathways: lessons from yeast. Nat. Rev. Mol. Cell Biol. 17, 537–552 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Wacker, R. et al. LC3-association with the parasitophorous vacuole membrane of Plasmodium bergheiliver stages follows a noncanonical autophagy pathway. Cell. Microbiol. 19, e12754 (2017).

    Article  CAS  Google Scholar 

  18. Martinez, J. et al. Molecular characterization of LC3-associated phagocytosis reveals distinct roles for Rubicon, NOX2 and autophagy proteins. Nat. Cell Biol. 17, 893–906 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Noda, N. N., Ohsumi, Y. & Inagaki, F. Atg8-family interacting motif crucial for selective autophagy. FEBS Lett. 584, 1379–1385 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Birgisdottir, Å. B., Lamark, T. & Johansen, T. The LIR motif—crucial for selective autophagy. J. Cell Sci. 126, 3237–3247 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Ichimura, Y. et al. Structural basis for sorting mechanism of p62 in selective autophagy. J. Biol. Chem. 283, 22847–22857 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. Pankiv, S. et al. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J. Biol. Chem. 282, 24131–24145 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Mizushima, N., Yoshimori, T. & Levine, B. Methods in mammalian autophagy research. Cell 140, 313–326 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Klionsky, D., Abdalla, F. & Abeliovich, H. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 8, 445–544 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Barrios-Rodiles, M. et al. High-throughput mapping of a dynamic signaling network in mammalian cells. Science 307, 1621–1625 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Zaffagnini, G. & Martens, S. Mechanisms of selective autophagy. J. Mol. Biol. 428, 1714–1724 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. McEwan, D. G. et al. PLEKHM1 regulates autophagosome–lysosome fusion through HOPS complex and LC3/GABARAP proteins. Mol. Cell 57, 39–54 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Boonhok, R. et al. LAP-like process as an immune mechanism downstream of IFN-γ in control of the human malaria Plasmodium vivax liver stage. Proc. Natl Acad. Sci. USA 113, E3519–E3528 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ruivo, M. T. G. et al. Host AMPK is a modulator of Plasmodiumliver infection. Cell Rep. 16, 2539–2545 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kumar, H. et al. Protective efficacy and safety of liver stage attenuated malaria parasites. Sci. Rep. 6, 26824 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kuma, A. et al. The role of autophagy during the early neonatal starvation period. Nature 432, 1032–1036 (2004).

    Article  CAS  PubMed  Google Scholar 

  32. Sou, Y. et al. The Atg8 conjugation system is indispensable for proper development of autophagic isolation membranes in mice. Mol. Biol. Cell 19, 4762–4775 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lee, I. H. et al. Atg7 modulates p53 activity to regulate cell cycle and survival during metabolic stress. Science 336, 225–228 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhang, W. et al. PCB 126 and other dioxin-like PCBs specifically suppress hepatic PEPCK expression via the aryl hydrocarbon receptor. PLoS ONE 7, e37103 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Gonçalves, L. A., Vigário, A. M. & Penha-Gonçalves, C. Improved isolation of murine hepatocytes for in vitro malaria liver stage studies. Malar. J. 6, 169 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Itoe, M. A. A. et al. Host cell phosphatidylcholine is a key mediator of malaria parasite survival during liver stage infection. Cell Host Microbe 16, 778–786 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Janse, C. J., Ramesar, J. & Waters, A. P. High-efficiency transfection and drug selection of genetically transformed blood stages of the rodent malaria parasite Plasmodium berghei. Nat. Protoc. 1, 346–356 (2006).

    Article  CAS  PubMed  Google Scholar 

  38. Tsuji, M., Mattei, D., Nussenzweig, R. S., Eichinger, D. & Zavala, F. Demonstration of heat-shock protein 70 in the sporozoite stage of malaria parasites. Parasitol. Res. 80, 16–21 (1994).

    Article  CAS  PubMed  Google Scholar 

  39. Fentress, S. J. et al. Phosphorylation of immunity-related GTPases by a Toxoplasma gondii-secreted kinase promotes macrophage survival and virulence. Cell Host Microbe 8, 484–495 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Xu, D. & Zhang, Y. Improving the physical realism and structural accuracy of protein models by a two-step atomic-level energy minimization. Biophys. J. 101, 2525–2534 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Torchala, M., Moal, I. H., Chaleil, R. A. G., Fernandez-Recio, J. & Bates, P. A. SwarmDock: a server for flexible protein–protein docking. Bioinformatics 29, 807–809 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Pronk, S. et al. GROMACS 4.5: a high-throughput and highly parallel open source molecular simulation toolkit. Bioinformatics 29, 845–854 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Vangone, A., Oliva, R. & Cavallo, L. CONS-COCOMAPS: a novel tool to measure and visualize the conservation of inter-residue contacts in multiple docking solutions. BMC Bioinformatics 13(Suppl. 4), S19 (2012).

    Article  Google Scholar 

  44. Laskowski, R. A. & Swindells, M. B. LigPlot+: multiple ligand–protein interaction diagrams for drug discovery. J. Chem. Inf. Model. 51, 2778–2786 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Kelly, S. M., Jess, T. J. & Price, N. C. How to study proteins by circular dichroism. Biochim. Biophys. Acta 1751, 119–139 (2005).

    Article  CAS  PubMed  Google Scholar 

  46. Van Wesenbeeck, L. et al. Involvement of PLEKHM1 in osteoclastic vesicular transport and osteopetrosis in incisors absent rats and humans. J. Clin. Invest. 117, 919–930 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank M. Komatsu (Tokyo Metropolitan Institute of Medical Science) for providing Atg3 MEFs, T. Finkel (NIH National Heart, Lung, and Blood Institute) for the Atg7 MEFs, F. Randow (MRC Laboratory of Molecular Biology) for the gift of p62-luciferase and GST-LC3 expression plasmids, Jacobus Pharmaceuticals for the WR99210 compound, A. Parreira for producing P. berghei-infected Anopheles mosquitoes, and S. Marques and K. Slavic for their help with parasite cloning. This work was supported by grants from the European Research Council (ERC‐2012‐StG_311502 to M.M.M.), Fundação para a Ciência e Tecnologia (EXCL/IMI-MIC/0056/2012 to M.M.M. and PTDC/IMI-MICC/1568/2012 to G.G.C.) and Institut Mérieux (MRG_20052016 to M.M.M). E.R. was the recipient of EMBO (ALTF 949-2008) and FCT (SFRH/BPD/68709/2010) fellowships. L.R. is the recipient of FCT fellowship SFRH/BPD/111323/2015. G.G.C. was sponsored by Marie Curie (PIEF-GA-2009-235864) and FCT (SFRH/BPD/74151/2010) fellowships. L.M.S. was supported by the European Community's Seventh Framework Programme (FP7/2007-2013) under grant agreement no. 242095 (EVIMalaR). V.Z.L. was sponsored by EMBO (ALTF 357-2009) and FCT (BPD-81953-2011) fellowships. I.M.V. and J.M.-V. were supported by NIH (1F32A11042-5 021) and FCT (SFRH/BD/52226/2013) fellowships, respectively.

Author information

Authors and Affiliations

Authors

Contributions

E.R. and M.M.M. conceived and led the study and wrote the manuscript. E.R, L.R., G.G.C. and J.M.-V. performed the experiments, acquired the data, performed data analysis and interpreted results. F.J.E. performed molecular docking, protein purification and SPR analysis. Animal experimentation was conducted by L.M.-S., L.R., I.M.V. and V.Z.-L. W.B. performed electron microscopy analysis. Circular dichroism was performed by T.N.F. G.R.M. constructed plasmids for parasite transfections. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Maria M. Mota.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Supplementary Information

Supplementary Figures 1–11, Supplementary Tables 1–5, Supplementary References.

Life Sciences Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Real, E., Rodrigues, L., Cabal, G.G. et al. Plasmodium UIS3 sequesters host LC3 to avoid elimination by autophagy in hepatocytes. Nat Microbiol 3, 17–25 (2018). https://doi.org/10.1038/s41564-017-0054-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-017-0054-x

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