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.

  • Article
  • Published:

A natural symbiotic bacterium drives mosquito refractoriness to Plasmodium infection via secretion of an antimalarial lipase

Abstract

The stalling global progress in the fight against malaria prompts the urgent need to develop new intervention strategies. Whilst engineered symbiotic bacteria have been shown to confer mosquito resistance to parasite infection, a major challenge for field implementation is to address regulatory concerns. Here, we report the identification of a Plasmodium-blocking symbiotic bacterium, Serratia ureilytica Su_YN1, isolated from the midgut of wild Anopheles sinensis in China that inhibits malaria parasites via secretion of an antimalarial lipase. Analysis of Plasmodium vivax epidemic data indicates that local malaria cases in Tengchong (Yunnan province, China) are significantly lower than imported cases and importantly, that the local vector A. sinensis is more resistant to infection by P. vivax than A. sinensis from other regions. Analysis of the gut symbiotic bacteria of mosquitoes from Yunnan province led to the identification of S. ureilytica Su_YN1. This bacterium renders mosquitoes resistant to infection by the human parasite Plasmodium falciparum or the rodent parasite Plasmodium berghei via secretion of a lipase that selectively kills parasites at various stages. Importantly, Su_YN1 rapidly disseminates through mosquito populations by vertical and horizontal transmission, providing a potential tool for blocking malaria transmission in the field.

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: Annual P. vivax cases and A. sinensis susceptibility to P. vivax infection.
Fig. 2: Effect of gut symbiotic Serratia bacteria of wild mosquitoes on Plasmodium development.
Fig. 3: Identification of the secreted antimalarial lipase AmLip in S. ureilytica Su_YN1.
Fig. 4: AmLip disrupts and kills parasites.
Fig. 5: S. ureilytica Su_YN1 bacteria spread efficiently through mosquito populations.

Similar content being viewed by others

Data availability

The entire 16S rRNA gene sequence dataset reported in this paper has been deposited in the National Center for Biotechnology Information Sequence Read Archive (accession no. PRJNA642229). This whole-genome shotgun project has been deposited at DDBJ/ENA/GenBank under the accession numbers JACAAS000000000, JACAAT000000000, JACAAU000000000 and JACAAV000000000. Source data are provided with this paper.

References

  1. World Malaria Report (World Health Organization, 2019).

  2. Global Vector Control Response 2017–2030 (World Health Organization, 2017).

  3. Dondorp, A. M. et al. Artemisinin resistance: current status and scenarios for containment. Nat. Rev. Microbiol. 8, 272–280 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Ranson, H. & Lissenden, N. Insecticide resistance in African Anopheles mosquitoes: a worsening situation that needs urgent action to maintain malaria control. Trends Parasitol. 32, 187–196 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Hamilton, W. L. et al. Evolution and expansion of multidrug-resistant malaria in southeast Asia: a genomic epidemiology study. Lancet Infect. Dis. 19, 943–951 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  6. van der Pluijm, R. W. et al. Determinants of dihydroartemisinin-piperaquine treatment failure in Plasmodium falciparum malaria in Cambodia, Thailand, and Vietnam: a prospective clinical, pharmacological, and genetic study. Lancet Infect. Dis. 19, 952–961 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Imwong, M., Hien, T. T., Thuy-Nhien, N. T., Dondorp, A. M. & White, N. J. Spread of a single multidrug resistant malaria parasite lineage (PfPailin) to Vietnam. Lancet Infect. Dis. 17, 1022–1023 (2017).

    Article  PubMed  Google Scholar 

  8. Haldar, K., Bhattacharjee, S. & Safeukui, I. Drug resistance in Plasmodium. Nat. Rev. Microbiol. 16, 156–170 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bhatt, S. et al. The effect of malaria control on Plasmodium falciparum in Africa between 2000 and 2015. Nature 526, 207–211 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Alonso, P. L. & Tanner, M. Public health challenges and prospects for malaria control and elimination. Nat. Med. 19, 150–155 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Talapko, J., Skrlec, I., Alebic, T., Jukic, M. & Vcev, A. Malaria: the past and the present. Microorganisms 7, 179 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  12. Vythilingam, I. & Hii, J. in Anopheles Mosquitoes—New Insights into Malaria Vectors (ed. Manguin, S.) 1st edn (InTech, 2013).

  13. Ghosh, A., Edwards, M. J. & Jacobs-Lorena, M. The journey of the malaria parasite in the mosquito: hopes for the new century. Parasitol. Today 16, 196–201 (2000).

    Article  CAS  PubMed  Google Scholar 

  14. Simon, N. et al. Sexual stage adhesion proteins form multi-protein complexes in the malaria parasite Plasmodium falciparum. J. Biol. Chem. 284, 14537–14546 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Pradel, G. Proteins of the malaria parasite sexual stages: expression, function and potential for transmission blocking strategies. Parasitology 134, 1911–1929 (2007).

    Article  CAS  PubMed  Google Scholar 

  16. Sinden, R. E., Alavi, Y. & Raine, J. D. Mosquito–malaria interactions: a reappraisal of the concepts of susceptibility and refractoriness. Insect Biochem. Mol. Biol. 34, 625–629 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Boissiere, A. et al. Midgut microbiota of the malaria mosquito vector Anopheles gambiae and interactions with Plasmodium falciparum infection. PLoS Pathog. 8, e1002742 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Heu, K. & Gendrin, M. Mosquito microbiota and its influence on disease vectorial transmission (in French). Biol. Aujourdhui 212, 119–136 (2018).

    Article  PubMed  Google Scholar 

  19. Wilke, A. B. B. & Marrelli, M. T. Paratransgenesis: a promising new strategy for mosquito vector control. Parasit. Vectors 8, 342 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Coutinho-Abreu, I. V., Zhu, K. Y. & Ramalho-Ortigao, M. Transgenesis and paratransgenesis to control insect-borne diseases: current status and future challenges. Parasitol. Int. 59, 1–8 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Wang, S. et al. Fighting malaria with engineered symbiotic bacteria from vector mosquitoes. Proc. Natl Acad. Sci. USA 109, 12734–12739 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wang, S. & Jacobs-Lorena, M. Genetic approaches to interfere with malaria transmission by vector mosquitoes. Trends Biotechnol. 31, 185–193 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bahia, A. C. et al. Exploring Anopheles gut bacteria for Plasmodium blocking activity. Environ. Microbiol. 16, 2980–2994 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wang, S. et al. Driving mosquito refractoriness to Plasmodium falciparum with engineered symbiotic bacteria. Science 357, 1399–1402 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Cirimotich, C. M. et al. Natural microbe mediated refractoriness to Plasmodium infection in Anopheles gambiae. Science 332, 855–858 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Gao, H., Cui, C., Wang, L., Jacobs-Lorena, M. & Wang, S. Mosquito microbiota and implications for disease control. Trends Parasitol. 36, 98–111 (2020).

    Article  PubMed  Google Scholar 

  27. Dong, Y. M., Manfredini, F. & Dimopoulos, G. Implication of the mosquito midgut microbiota in the defense against malaria parasites. PLoS Pathog. 5, e1000423 (2009).

  28. Angkawidjaja, C. & Kanaya, S. Family I.3 lipase: bacterial lipases secreted by the type I secretion system. Cell Mol. Life Sci. 63, 2804–2817 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. Arpigny, J. L. & Jaeger, K.-E. Bacterial lipolytic enzymes: classification and properties. Biochem. J. 343, 177–183 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Osei-Poku, J., Mbogo, C. M., Palmer, W. J. & Jiggins, F. M. Deep sequencing reveals extensive variation in the gut microbiota of wild mosquitoes from Kenya. Mol. Ecol. 21, 5138–5150 (2012).

    Article  CAS  PubMed  Google Scholar 

  31. Wang, Y., Gilbreath, T. M. III, Kukutla, P., Yan, G. & Xu, J. Dynamic gut microbiome across life history of the malaria mosquito Anopheles gambiae in Kenya. PLoS ONE 6, e24767 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bando, H. et al. Intra-specific diversity of Serratia marcescens in Anopheles mosquito midgut defines Plasmodium transmission capacity. Sci. Rep. 3, 1641 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Pumpuni, C. B., Demaio, J., Kent, M., Davis, J. R. & Beier, J. C. Bacterial population dynamics in three anopheline species: the impact on Plasmodium sporogonic development. Am. J. Trop. Med Hyg. 54, 214–218 (1996).

    Article  CAS  PubMed  Google Scholar 

  34. Rani, A., Sharma, A., Rajagopal, R., Adak, T. & Bhatnagar, R. K. Bacterial diversity analysis of larvae and adult midgut microflora using culture-dependent and culture-independent methods in lab-reared and field-collected Anopheles stephensi—an Asian malarial vector. BMC Microbiol. 9, 1471–2180 (2009).

    Article  Google Scholar 

  35. Gonzalez-Ceron, L., Santillan, F., Rodriguez, M. H., Mendez, D. & Hernandez-Avila, J. E. Bacteria in midguts of field-collected Anopheles albimanus block Plasmodium vivax sporogonic development. J. Med. Entomol. 40, 371–374 (2003).

    Article  PubMed  Google Scholar 

  36. Villegas, L. M. & Pimenta, P. F. P. Metagenomics, paratransgenesis and the Anopheles microbiome: a portrait of the geographical distribution of the anopheline microbiota based on a meta-analysis of reported taxa. Mem. Inst. Oswaldo Cruz 109, 672–684 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Bai, L., Wang, L., Vega-Rodriguez, J., Wang, G. & Wang, S. A gut gymbiotic bacterium Serratia marcescens renders mosquito resistance to Plasmodium infection through activation of mosquito immune responses. Front. Microbiol. 10, 1580 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Garcia, C. J. et al. Serralysin family metalloproteases protects Serratia marcescens from predation by the predatory bacteria Micavibrio aeruginosavorus. Sci. Rep. 8, 14025 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Mitamura, T. & Palacpac, N. M. Q. Lipid metabolism in Plasmodium falciparum-infected erythrocytes: possible new targets for malaria chemotherapy. Microbes Infect. 5, 545–552 (2003).

    Article  CAS  PubMed  Google Scholar 

  40. Zhu, G. et al. Susceptibility of Anopheles sinensis to Plasmodium vivax in malarial outbreak areas of central China. Parasit. Vectors 6, 176 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Reyon, D. et al. FLASH assembly of TALENs for high-throughput genome editing. Nat. Biotechnol. 30, 460–465 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Caporaso, J. G. et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 7, 335–336 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Rognes, T., Flouri, T., Nichols, B., Quince, C. & Mahe, F. VSEARCH: a versatile open source tool for metagenomics. PeerJ 4, e2584 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Wang, Q., Garrity, G. M., Tiedje, J. M. & Cole, J. R. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl. Environ. Microbiol. 73, 5261–5267 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kumar, S., Stecher, G. & Tamura, K. MEGA7: Molecular Evolutionary Genetics Analysis version 7.0 for bigger datasets. Mol. Biol. Evol. 33, 1870–1874 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Berlin, K. et al. Assembling large genomes with single-molecule sequencing and locality-sensitive hashing. Nat. Biotechnol. 33, 623–630 (2014).

    Article  Google Scholar 

  48. Chen, F., Mackey, A. J., Vermunt, J. K. & Roos, D. S. Assessing performance of orthology detection strategies applied to eukaryotic genomes. PLoS ONE 2, e383 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Edgar, R. C. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 32, 1792–1797 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Capella-Gutierrez, S., Silla-Martinez, J. M. & Gabaldon, T. trimAl: a tool for automated alignment trimming in large-scale phylogenetic analyses. Bioinformatics 25, 1972–1973 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Guindon, S. et al. New algorithms and methods to estimate maximum-likelihood phylogenies: assessing the performance of PhyML 3.0. Syst. Biol. 59, 307–321 (2010).

    Article  CAS  PubMed  Google Scholar 

  52. Abascal, F., Zardoya, R. & Posada, D. ProtTest: selection of best-fit models of protein evolution. Bioinformatics 21, 2104–2105 (2005).

    Article  CAS  PubMed  Google Scholar 

  53. Wei, G. et al. Insect pathogenic fungus interacts with the gut microbiota to accelerate mosquito mortality. Proc. Natl Acad. Sci. USA 114, 5994–5999 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sinden, R. E. et al. Ookinete antigens of Plasmodium berghei: a light and electron-microscope immunogold study of expression of the 21 kDa determinant recognized by a transmission-blocking antibody. Proc. R. Soc. Lond. B Biol. Sci. 230, 443–458 (1987).

    Article  CAS  PubMed  Google Scholar 

  55. De Niz, M., Stanway, R. R., Wacker, R., Keller, D. & Heussler, V. T. An ultrasensitive NanoLuc-based luminescence system for monitoring Plasmodium berghei throughout its life cycle. Malar. J. 15, 232–232 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Gao, H. et al. ISP1-anchored polarization of GCβ/CDC50A complex initiates malaria ookinete gliding motility. Curr. Biol. 28, 2763–2776 (2018).

    Article  CAS  PubMed  Google Scholar 

  57. Kumar, S., Molina-Cruz, A., Gupta, L., Rodrigues, J. & Barillas-Mury, C. A peroxidase/dual oxidase system modulates midgut epithelial immunity in Anopheles. Science 327, 1644–1648 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. McNamara, C. W. et al. Targeting Plasmodium PI(4)K to eliminate malaria. Nature 504, 248–253 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Huang, H., Song, X. & Yang, S. Development of a RecE/T-Assisted CRISPR–Cas9 toolbox for Lactobacillus. Biotechnol. J. 14, e1800690 (2019).

    Article  PubMed  Google Scholar 

  60. Liu, P. C., Lee, K. K. & Chen, S. N. Pathogenicity of different isolates of Vibrio harveyi in tiger prawn, Penaeus monodon. Lett. Appl. Microbiol. 22, 413–416 (1996).

    Article  Google Scholar 

  61. Kourker, G. & Jaeger, K. E. Specific and sensitive plate assay for bacterial lipases. Appl. Environ. Microbiol. 53, 211–213 (1987).

    Article  Google Scholar 

  62. Studier, F. W. Protein production by auto-induction in high density shaking cultures. Protein Expr. Purif. 41, 207–234 (2005).

    Article  CAS  PubMed  Google Scholar 

  63. Fantappie, L. et al. Antibody-mediated immunity induced by engineered Escherichia coli OMVs carrying heterologous antigens in their lumen. J. Extracell. Vesicles https://doi.org/10.3402/jev.v3.24015 (2014).

  64. Peach, M., Marsh, N., Miskiewicz, E. I. & MacPhee, D. J. in Western Blotting: Methods and Protocols (eds Kurien, B. T. & Scofield, R. H.) 49–60 (Springer New York, 2015).

  65. Javed, S. et al. Bacterial lipases: a review on purification and characterization. Prog. Biophys. Mol. Biol. 132, 23–34 (2018).

    Article  CAS  PubMed  Google Scholar 

  66. Saitou, N. & Nei, M. The neighbor-joining method: a new method for reconstructing phylogenetic trees. Mol. Biol. Evol. 4, 406–425 (1987).

    CAS  PubMed  Google Scholar 

  67. Felsenstein, J. Confidence limits on phylogenies: an approach using the bootstrap. Evolution 39, 783–791 (1985).

    Article  PubMed  Google Scholar 

  68. Nei, M., & Kumar, S. Molecular Evolution and Phylogenetics (Oxford University Press, 2000).

Download references

Acknowledgements

This work was supported by grants from the National Natural Science Foundation of China (grants nos. 31830086, 32021001 and 31472044) to S.W.; the National Key R&D Program of China (grant no. 2019YFC1200800) to S.W.; the Strategic Priority Research Program of the Chinese Academy of Sciences (grant no. XDB11010500) to S.W.; the Key Research Program of the Chinese Academy of Sciences (grant no. KFZD-SW-219) to S.W.; the National Institutes of Health (grant no. R01AI031478) to M.J.-L.; the Johns Hopkins Malaria Research Institute Insectary, Parasite Core Facilities; the Bloomberg Philanthropies and the Jiangsu Provincial Department of Science and Technology (grant no. BM2018020) to J.C. We thank F. Li for rearing mosquitoes.

Author information

Authors and Affiliations

Authors

Contributions

S.W. conceived the project. S.W., H.G. and L.B. designed the study. L.B., X.L., S.L., G.Z. and J.C. collected wild mosquitoes. L.B. conducted the gut symbiotic bacteria isolation, gut colonization, RNA interference and effects of isolated bacteria on P. berghei infection and mosquito biology assays. L.B. and X.L. performed the A. sinensis susceptibility to P. vivax infection assays. H.G. conducted the in vitro anti-Plasmodium activity, mass spectrometry, gene disruption, western blot, AmLip-mediated anti-Plasmodium activity and immunofluorescence assays. Y.J. conducted the bacterial transmission and cage experiments. W.H. conducted assays to determine the effect of Serratia bacteria on P. falciparum infection. Z.H. and L.B. investigated the effect of Serratia culture supernatant on P. falciparum gametocyte development. D.W. provided P. vivax epidemic data. S.Z. performed phylogenomic analysis. L.J. and M.J.-L. provided materials. H.G., L.B. and S.W. analysed the data. H.G., L.W. and S.W. wrote the manuscript. M.J.-L. and S.W. edited the manuscript.

Corresponding author

Correspondence to Sibao Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Microbiology thanks Hitotaka Kanuka and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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

Extended data

Extended Data Fig. 1 Persistence of non-symbiotic bacteria in mosquito midguts.

GFP-tagged E. coli and S. aureus were administered to newly emerged female mosquitoes via sugar feeding. Bacteria midgut colonization was determined by plating serially diluted midgut homogenates on LB agar plates containing 100 μg/ml kanamycin. a,b, E. coli and S. aureus bacteria numbers in mosquitoes maintained with sugar. c,d, E. coli and S. aureus bacteria numbers in mosquitoes post a blood meal. Data points are mean ± s.d. The dots represent biologically independent replicates (n = 2). Each replicate contains 10 mosquito midguts.

Source data

Extended Data Fig. 2 Serratia strains stably colonize the mosquito midgut and do not impact mosquito longevity.

a, b, Effect of Serratia strains on An. sinensis (a) and An. stephensi (b) survival post blood meal. Bacteria were administered to two-day-old female mosquitoes via sugar meal and then fed blood. Survival was monitored daily. Data points are mean ± s.d. The dots represent biologically independent replicates (n = 2). Each replicate contains 10 mosquito midguts. c,d, Serratia bacteria numbers in the midgut of female An. sinensis (c) and An. stephensi (d) post blood meal. Data points are mean ± s.d. The dots represent biologically independent replicates (n = 2). Each replicate contains 10 mosquito midguts. e, Visualization of GFP-tagged Su_YN1 and Sm_YN3 bacteria in the midgut of An. stephensi at 24 h after a blood meal. Bright-field images (left) are paired with the corresponding fluorescent images (right). The experiments were repeated twice with similar results.

Source data

Extended Data Fig. 3 Effect of bacteria on P. berghei oocyst formation in An. stephensi.

a, P. berghei oocyst load in An. stephensi mosquitoes carrying Asaia, Acinetobacter or Pantoea bacteria from YN wild caught mosquitoes. b, P. berghei oocyst load in An. stephensi mosquitoes fed with different Serratia strains from different wild-caught mosquito populations. Circles represent the number of oocysts in individual midguts, and horizontal lines indicate the median number of oocysts per midgut. The sample size (n Number) of each group is listed in the table of the lower panel. The statistical significance of the oocyst intensity between the bacteria-fed mosquitoes and PBS-fed mosquitoes (Ctrl) was analysed using the two-tailed Mann-Whitney test. ****P < 0.0001, P > 0.05, not significant (ns). The exact P values in (a) were as follows: Asaia, 0.4508; Acinetobacter, 0.8728; Pantoea, 0.4265. The exact P values in (b) were as follows: Su_YN1, < 0.0001; Sm_YN3, < 0.0001; Sf_JS1, 0.0271; Sf_JS2, 0.0135; Su_JS3, 0.0117; Sm_LN1, 0.0220.

Source data

Extended Data Fig. 4 The effect of Serratia Su_YN1 and Sm_YN3 on An. stephensi blood feeding, fecundity and oviposition rate.

Serratia Su_YN1 and Sm_YN3 do not impact An. stephensi mosquito blood feeding behaviour (n = 100 mosquitoes each group) (a), egg production (Ctrl, n = 46, Su_YN1, n = 39, Sm_YN3, n = 44) (b) or oviposition rate (n = 100 mosquitoes each group) c, Two-day-old An. stephensi mosquitoes were fed on a sugar meal containing bacteria or 5% sugar alone (Ctrl). Three days later, female mosquitoes were fed on a mouse and three days later eggs were collected from individual females. The experiments were repeated three times with similar results. No significant differences were detected among the groups (one-way ANOVA or two-tailed Mann-Whitney test).

Source data

Extended Data Fig. 5 Effect of Rel1 and Rel2 silencing on Su_YN1- or Sm_YN3-mediated anti-Plasmodium activity.

Rel1 and Rel2 were silenced in An. stephensi by systemic injection of double-stranded RNA dsRel1, dsRel2 or dsGFP. The injected mosquitoes were fed on a sugar meal containing Su_YN1 or Sm_YN3. Three days later, the mosquitoes were allowed to feed on the same P. berghei infected mouse. The injected double-stranded RNA (ds) and presence of bacteria are indicated below each column. Each dot represents the oocyst number of an individual midgut, and the horizontal lines indicate the median number of oocysts per midgut. Data are from n = 25 to 28 mosquitoes per group. The sample size (n Number) of each group is listed in the table of the lower panel. The statistical significance of oocyst intensity differences between the dsRel1- or dsRel2-injected and dsGFP-injected mosquitoes carrying the same bacteria (Su_YN1 or Sm_YN3) was analysed using the two-tailed Mann-Whitney test. ****P < 0.0001, P > 0.05, not significant (ns). The exact values were as follows: dsRel1+Su_YN1, 0.8519; dsRel2+Su_YN1, 0.5605; dsRel1+Sm_YN3, < 0.0001; sRel2+Sm_YN3, 0.4851.

Source data

Extended Data Fig. 6 Su_YN1 culture supernatant inhibited P. berghei ookinete formation.

a, Inhibition by Serratia culture supernatants of P. yoelii ookinete formation in vitro. Ookinete formation was quantified by luminescence measurements using the Py.17XNL reporter strain. RLU, relative light units. The experiments were repeated three times with similar results. Data points are mean ± s.d. The dots represent biologically independent replicates (n = 2). b, Effect of Su_YN1 culture supernatant on P. berghei ookinete formation in vitro assay using Giemsa staining. Transformation rate was quantified by comparison with the PBS control. The experiments were repeated three times with similar results. Data points are mean ± s.d. The dots represent biologically independent replicates (n = 3). Statistical significance of the ookinete transformation rate was compared with the PBS control using two-tailed Student’s t-test, ****P < 0.0001. The exact P value was, < 0.0001.

Source data

Extended Data Fig. 7 Haemolytic activity assay of Su_YN1 and Sm_YN3.

a, Bacteria culture supernatant was added (10% V/V) and the mixture incubated with erythrocytes for 12 h. b, The supernatants were collected and the absorbance at 540 nm was measured to evaluate haemoglobin release. Saponin was used as a haemolytic positive control. Data points are mean ± s.d. The dots represent biologically independent replicates (n = 3). Statistical significance of haemolytic activity was compared with the PBS control using two-tailed Student’s t-test. P > 0.05, not significant (ns). The exact P values were as follows: Su_YN1, 0.1672; Sm_YN3, 0.3754.

Source data

Extended Data Fig. 8 Antimalarial activity of different Su_YN1 culture supernatant fractions.

a, Antimalarial activity of organic solvent extracts. Su_YN1 culture supernatant was extracted with solvents of various polarities. The extracted fractions were dried and dissolved in DMSO and the remaining aqueous phase was vacuum treated to remove residual organic reagents. All fractions were tested for antimalarial activity. b, Antimalarial activity assay of Su_YN1 culture supernatant separated using a 3 kDa cut-off centrifugal filter. The retentate and the filtrate were tested. c, Trypsin digestion of Su_YN1 culture supernatant abolishes antimalarial activity. Coomassie Brilliant Blue staining in the lower panel shows the protein patterns before and after treatment. Data points in (b) and (c) are mean ± s.d. The dots represent biologically independent replicates (n = 3). Statistical significance of the ookinete inhibition rate was compared with the PBS control using two-tailed Student’s t-test, ****P < 0.0001, P > 0.05, not significant (ns). The exact P values in (b) were: Trypsin, < 0.0001; Trypsin-inactivated, 0.0552. The exact P values in (c) were: Retentate (compared with PBS), < 0.0001; Filtrate (compared with Retentate), < 0.0001.

Source data

Extended Data Fig. 9 AmLip gene expression in different Serratia strains.

Detection of AmLip transcript abundance in different Serratia strains by qRT-PCR using Serratia 16 s rRNA as an internal reference. Data points are mean ± s.d. The dots represent biologically independent replicates (n = 3). The experiments were repeated twice with similar results.

Source data

Extended Data Fig. 10 Synthesis and purification of Serratia AmLip protein.

a, Western blot detection using HA antibody, of 3HA-tagged AmLip protein in bacterial extracts of Su_YN1 wild type, knock-out mutant AmLip-KO and a mutant AmLip-KO complemented with the AmLip:HA gene. The experiments were repeated twice with similar results. b, Knockout of AmLip was verified by Western blot assay using an AmLip mouse antiserum. The experiments were repeated twice with similar results. c, Expression and purification of AmLip protein expressed E. coli BL21 (DE3). Coomassie blue staining showing the AmLip protein before and after purification on a nickel column. The experiments were repeated twice with similar results. d, Lipase activity test of the purified AmLip protein using the egg yolk lipoprotein plate degradation assay. e, Lipase activity assay of the purified AmLip protein using the trioleoylglycerol- rhodamine B plate degradation assay.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–7 and Tables 1–4.

Reporting Summary

Peer Review File

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 8

Unmodified gels.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed western blots and unmodified gels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gao, H., Bai, L., Jiang, Y. et al. A natural symbiotic bacterium drives mosquito refractoriness to Plasmodium infection via secretion of an antimalarial lipase. Nat Microbiol 6, 806–817 (2021). https://doi.org/10.1038/s41564-021-00899-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-021-00899-8

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