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:

Anopheline mosquitoes are protected against parasite infection by tryptophan catabolism in gut microbiota

Abstract

The mosquito microbiota can influence host physiology and vector competence, but a detailed understanding of these processes is lacking. Here we found that the gut microbiota of Anopheles stephensi, a competent malaria vector, is involved in tryptophan metabolism and is responsible for the catabolism of the peritrophic matrix impairing tryptophan metabolites. Antibiotic elimination of the microbiota led to the accumulation of tryptophan and its metabolites—kynurenine, 3-hydroxykynurenine (3-HK) and xanthurenic acid. Of these metabolites, 3-HK impaired the structure of the peritrophic matrix and promoted Plasmodium berghei infection. Among the major gut microbiota members in A. stephensi, Pseudomonas alcaligenes catabolized 3-HK as revealed by whole-genome sequencing and LC–MS metabolic analysis. The genome of P. alcaligenes encodes kynureninase (KynU) that is responsible for the conversion of 3-HK to 3-hydroxyanthranilic acid. Mutation of KynU resulted in a P. alcaligenes strain that was unable to metabolize 3-HK and unable to protect the peritrophic matrix. Colonization of A. stephensi with KynU-mutated P. alcaligenes failed to protect mosquitoes against parasite infection as compared with mosquitoes colonized with wild-type P. alcaligenes. In summary, this study identifies an unexpected function of mosquito gut microbiota in controlling mosquito tryptophan metabolism, with important implications for vector competence.

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: Microbiota-regulated Trp metabolism modulates P. berghei infection.
Fig. 2: 3-HK promotes Plasmodium infection via impairing the PM.
Fig. 3: Commensal P. alcaligenes catabolizes mosquito 3-HK.
Fig. 4: P. alcaligenes KynU degrades 3-HK in mosquitoes.

Similar content being viewed by others

Data availability

All data are available in the Article and its Supplementary information. The genomic sequence data of P. alcaligenes are available at the National Center for Biotechnology Information (NCBI) Sequence Read Archive (SRA) (accession no. PRJNA686701). Transcriptomic data are available at the NCBI SRA (accession no. PRJNA686698). The 16S rRNA gene sequences are available at the NCBI SRA (accession no. PRJNA686689). Source data are provided with this paper. Further information and requests for resources should be directed to J.W. (jingwenwang@fudan.edu.cn).

References

  1. 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 

  2. Romoli, O. & Gendrin, M. The tripartite interactions between the mosquito, its microbiota and Plasmodium. Parasit. Vectors 11, 200 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Boudko, D. Y. Molecular basis of essential amino acid transport from studies of insect nutrient amino acid transporters of the SLC6 family (NAT-SLC6). J. Insect Physiol. 58, 433–449 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Han, Q., Beerntsen, B. T. & Li, J. The tryptophan oxidation pathway in mosquitoes with emphasis on xanthurenic acid biosynthesis. J. Insect Physiol. 53, 254–263 (2007).

    Article  CAS  PubMed  Google Scholar 

  5. Li, J., Beerntsen, B. T. & James, A. A. Oxidation of 3-hydroxykynurenine to produce xanthommatin for eye pigmentation: a major branch pathway of tryptophan catabolism during pupal development in the yellow fever mosquito, Aedes aegypti. Insect Biochem. Mol. Biol. 29, 329–338 (1999).

    Article  CAS  PubMed  Google Scholar 

  6. Platten, M., Nollen, E. A. A., Röhrig, U. F., Fallarino, F. & Opitz, C. A. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat. Rev. Drug Discov. 18, 379–401 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Ling, L. & Raikhel, A. S. Serotonin signaling regulates insulin-like peptides for growth, reproduction, and metabolism in the disease vector Aedes aegypti. Proc. Natl Acad. Sci USA 115, E9822–E9831 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  9. Okech, B., Arai, M. & Matsuoka, H. The effects of blood feeding and exogenous supply of tryptophan on the quantities of xanthurenic acid in the salivary glands of Anopheles stephensi (Diptera: Culicidae). Biochem. Biophys. Res. Commun. 341, 1113–1118 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Aleshnick, M., Ganusov, V. V., Nasir, G., Yenokyan, G. & Sinnis, P. Experimental determination of the force of malaria infection reveals a non-linear relationship to mosquito sporozoite loads. PLoS Pathog. 16, e1008181 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Pumpuni, C. B., Mendis, C. & Beier, J. C. Plasmodium yoelii sporozoite infectivity varies as a function of sporozoite loads in Anopheles stephensi mosquitoes. J. Parasitol. 83, 652–655 (1997).

    Article  CAS  PubMed  Google Scholar 

  12. Jiang, H. et al. Cytokine/Jak/Stat signaling mediates regeneration and homeostasis in the Drosophila midgut. Cell 137, 1343–1355 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Hegedus, D., Erlandson, M., Gillott, C. & Toprak, U. New insights into peritrophic matrix synthesis, architecture, and function. Annu Rev. Entomol. 54, 285–302 (2009).

    Article  CAS  PubMed  Google Scholar 

  14. Rodgers, F. H., Gendrin, M., Wyer, C. A. & Christophides, G. K. Microbiota-induced peritrophic matrix regulates midgut homeostasis and prevents systemic infection of malaria vector mosquitoes. PLoS Pathog. 13, e1006391 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Song, X., Wang, M., Dong, L., Zhu, H. & Wang, J. PGRP-LD mediates An. stephensi vector competency by regulating homeostasis of microbiota-induced peritrophic matrix synthesis. PLoS Pathog. 14, e1006899 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Phillips, R. S. Structure and mechanism of kynureninase. Arch. Biochem. Biophys. 544, 69–74 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Bortolotti, P. et al. Tryptophan catabolism in Pseudomonas aeruginosa and potential for inter-kingdom relationship. BMC Microbiol. 16, 137 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Lima, V. L. A. et al. The antioxidant role of xanthurenic acid in the Aedes aegypti midgut during digestion of a blood meal. PLoS ONE 7, e38349 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Billker, O. et al. Identification of xanthurenic acid as the putative inducer of malaria development in the mosquito. Nature 392, 289–292 (1998).

    Article  CAS  PubMed  Google Scholar 

  20. Gabrieli, P. et al. Mosquito trilogy: microbiota, immunity and pathogens, and their implications for the control of disease transmission. Front. Microbiol. 12, 630438 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Gao, H. et al. A natural symbiotic bacterium drives mosquito refractoriness to Plasmodium infection via secretion of an antimalarial lipase. Nat. Microbiol. 6, 806–817 (2021).

    Article  CAS  PubMed  Google Scholar 

  22. Bennink, S., Kiesow, M. J. & Pradel, G. The development of malaria parasites in the mosquito midgut. Cell. Microbiol. 18, 905–918 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hughes, G. L. et al. Native microbiome impedes vertical transmission of Wolbachia in Anopheles mosquitoes. Proc. Natl Acad. Sci. USA 111, 12498–12503 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ha, Y. R. et al. The effects of antibiotics on the reproductive physiology targeting ovaries in the Asian tiger mosquito, Aedes albopictus. Entomol. Res. 51, 65–73 (2021).

    Article  CAS  Google Scholar 

  25. Singh, R., Sripada, L. & Singh, R. Side effects of antibiotics during bacterial infection: mitochondria, the main target in host cell. Mitochondrion 16, 50–54 (2014).

    Article  PubMed  CAS  Google Scholar 

  26. Kalghatgi, S. et al. Bactericidal antibiotics induce mitochondrial dysfunction and oxidative damage in mammalian cells. Sci. Transl. Med. 5, 192ra185 (2013).

    Article  CAS  Google Scholar 

  27. Coon, K. L., Vogel, K. J., Brown, M. R. & Strand, M. R. Mosquitoes rely on their gut microbiota for development. Mol. Ecol. 23, 2727–2739 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Correa, M. A., Matusovsky, B., Brackney, D. E. & Steven, B. Generation of axenic Aedes aegypti demonstrate live bacteria are not required for mosquito development. Nat. Commun. 9, 4464 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Hyde, J., Correa, M. A., Hughes, G. L., Steven, B. & Brackney, D. E. Limited influence of the microbiome on the transcriptional profile of female Aedes aegypti mosquitoes. Sci. Rep. 10, 10880 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Minard, G., Mavingui, P. & Moro, C. V. Diversity and function of bacterial microbiota in the mosquito holobiont. Parasit. Vectors 6, 146 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  31. 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 

  32. Wang, M. et al. Glucose-mediated proliferation of a gut commensal bacterium promotes Plasmodium infection by increasing mosquito midgut pH. Cell Rep. 35, 108992 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ukegbu, C. V. et al. Plasmodium berghei P47 is essential for ookinete protection from the Anopheles gambiae complement-like response. Sci. Rep. 7, 6026–6026 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Tomas, A. M. et al. P25 and P28 proteins of the malaria ookinete surface have multiple and partially redundant functions. EMBO J. 20, 3975–3983 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yang, Z. et al. A malaria parasite phospholipid flippase safeguards midgut traversal of ookinetes for mosquito transmission. Sci. Adv. 7, eabf6015 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Tamura, K., Dudley, J., Nei, M. & Kumar, S. MEGA4: molecular evolutionary genetics analysis (MEGA) software version 4.0. Mol. Biol. Evol. 24, 1596–1599 (2007).

    Article  CAS  PubMed  Google Scholar 

  37. An, Y. et al. High-fat diet induces dynamic metabolic alterations in multiple biological matrices of rats. J. Proteome Res. 12, 3755–3768 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Holmes, D. S. & Bonner, J. Preparation, molecular weight, base composition, and secondary structure of giant nuclear ribonucleic acid. Biochemistry 12, 2330–2338 (1973).

    Article  CAS  PubMed  Google Scholar 

  41. Michelsen, C. F., Glaring, P. P., Schjoerring, M. A. & Stougaard, J. K. P. Bacterial diversity in Greenlandic soils as affected by potato cropping and inorganic versus organic fertilization. Polar Biol. 37, 61–71 (2014).

    Article  Google Scholar 

  42. Edgar, R. C. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat. Methods 10, 996–998 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Oliveira, J. H. M. et al. Blood meal-derived heme decreases ROS levels in the midgut of Aedes aegypti and allows proliferation of intestinal microbiota. PLoS Pathog. 7, e1001320–e1001320 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Buchon, N., Broderick, N. A., Poidevin, M., Pradervand, S. & Lemaitre, B. Drosophila intestinal response to bacterial infection: activation of host defense and stem cell proliferation. Cell Host Microbe 5, 200–211 (2009).

    Article  CAS  PubMed  Google Scholar 

  45. Benjamini, Y., Krieger, A. M. & Yekutieli, D. Adaptive linear step-up procedures that control the false discovery rate. Biometrika 93, 491–507 (2006).

    Article  Google Scholar 

Download references

Acknowledgements

We thank G. Zhang, C. Wang and J. Gou from Fudan University for technical advice on metabolites analysis; X. Zhou from Fudan University for support on statistical analysis; S. Zhao and H. Wang from Fudan University, and G. Dimopoulos from Johns Hopkins University for comments on the manuscript; and J. Yuan from Xiamen University for providing anti-P28 polyclonal antibody. This work was supported by the National Natural Science Foundation of China (U1902211) and the National Institutes of Health Grant (R01AI129819) to J.W.

Author information

Authors and Affiliations

Authors

Contributions

Y.F., Y.P., X.S., H.W., J.W. and H.T. designed experiments, interpreted results and wrote the paper. Y.P. and Y.A. performed metabolites analysis. X.S. and H.W. conducted PM structure analysis experiments. Y.F., X.S. and W.H. conducted bacteria recolonization and Plasmodium infection experiments. Y.F. conducted and analysed results from all additional experiments. J.W. and H.T. supervised the study. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Huiru Tang or Jingwen Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Microbiology thanks Louis Lambrechts, Flaminia Catteruccia and and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1. Tryptophan metabolites analysis post normal blood meal.

Relative amount of Trp metabolites in normal (N, n = 10) and antibiotics-treated (Abx, n = 10) mosquitoes 24 h post normal blood meal. Data are presented as mean ± s.e.m. Significance was determined by two-sided Student’s t-test.

Extended Data Fig. 2. Influence of microbiota on P. berghei infection and mosquito PM formation.

a, Oocyst numbers of Normal (N) and antibiotics-treated (Abx) mosquitoes. Horizontal black bars indicate the median values. Data were pooled from two independent experiments. Significance was determined by two-sided Mann-Whitney test. b, PAS staining of PM structure in N and Abx mosquitoes 45 h post normal blood meal at 100× and 200× magnification. Red arrows indicate the PM structure. Images are representative of at least six individual mosquito midguts. Scale bars represent 100 μm.

Extended Data Fig. 3. Knocking down HKT increases 3-HK level.

a, Relative expression levels of HKT in non-injected and dsRNA injected mosquitoes. The expression level of HKT was normalized to S7. The relative expression levels of HKT in dsHKT and dsGFP mosquitoes were normalized to the gene’s expression in non-injection group, respectively. Data are presented as mean ± s.e.m (n = 8). Results from one of two independent experiments are shown. b, Relative amount of 3-HK in dsGFP and dsHKT mosquitoes right before blood meal. Data are presented as mean ± s.e.m (n = 10). Significance was determined using ANOVA with Tukey’s test in (a) and two-sided Student’s t-test in (b).

Extended Data Fig. 4. Influence of 3-HK administration on An. stephensi survival and P. berghei exflagellation.

a, Fold change of 3-HK levels in control (n = 10) and 3-HK fed (n = 10) mosquitoes at 24 h (-24 h), 0 h (0 h) prior to and 24 h (24 h) post blood meal. Data are presented as mean ± s.e.m. b, Survival curve of control (n=46) and 3-HK fed (n=43) mosquitoes post an infectious blood meal. Results from one of two independent experiments are shown. c, Exflagellation rate monitored 15 min post infection in the control (-) and 3-HK fed (+) mosquitoes. Data were pooled from two independent experiments. Each dot represents an individual mosquito. Horizontal black bars indicate the median values (c). Significance was determined by two-sided Student’s t-test in (a), Log-rank (Mantel-Cox) test in (b) and Mann-Whitney test in (c).

Extended Data Fig. 5. Influence of 3-HK administration on midgut barrier function.

a, DHE staining of midguts in control (-) and 3-HK-treated (+) mosquitoes at 0 h prior to (left) and 24 h (right) post normal blood meal. Scale bars represent 100 μm. Images are representative of at least eight midguts. b, TUNEL staining of midgut from controls (-) and 3-HK-treated mosquitoes (+) at 0 h prior to (left) and 24 h (right) post normal blood meal. Scale bars represent 50 μm. Images are representative of at least four midguts. c, PH3 staining of midguts from controls (-) and 3-HK-treated (+) mosquitoes at 0 h (left) prior to and 24 h (right) post infectious blood meal. Scale bars represent 100 μm. d, Quantification of PH3-positive cells in the midguts at 0 h prior to infectious blood meal. Data are presented as mean ± s.e.m (n = 9). (e) Quantification of PH3-positive cells in the midguts from controls (n=4) and 3-HK-treated (n=5) mosquitoes at 24 h post infectious blood meal. Data are presented as mean ± s.e.m. Significance was determined by two-sided Student’s t-test.

Extended Data Fig. 6. Influence of 3-HK on Per1 expression.

a, Quantification of expression levels of Per1 in An. stephensi orally administrated 3-HK prior to blood feeding. Expression levels of Per1 were analyzed at 24 h (-24 h) and 0 h (0 h) prior to a blood meal in 3-HK treated and normal mosquitoes. Data are presented as mean ± s.e.m (n = 10). Results from one of two independent experiments are shown. b, Quantification of expression levels of Per1 in An. stephensi fed with blood and 3-HK simultaneously. Expression levels of Per1 were analyzed 24 h post a blood meal. Data are presented as mean ± s.e.m (n = 10). Results from one of two independent experiments are shown. Significance was determined by two-sided Student’s t-test.

Extended Data Fig. 7. Polyoxin D treatment abolishes the PM formation.

PAS staining of PM in normal mosquitoes (Control) (a, b), polyoxin D (PD) treated (c, d) and 3-HK + polyoxin D treated mosquitoes (3-HK+PD) (e, f) 45 h post normal blood meal at 100× and 200× magnification. Red arrows indicate the PM. Images are representative of at least three individual mosquito midguts. Scale bars represent 100 μm.

Extended Data Fig. 8. Trp supplementation impairs PM structure.

a, Western blot of Per1 in the midgut of Trp-treated (+) mosquitoes and control (-) 24 h post normal blood meal. b, Effect of Trp on P. berghei infection in Abx mosquitoes. Data were pooled from two independent experiments. Each dot represents an individual mosquito. Horizontal black bars indicate the median values. Significance was determined with a two-sided Mann-Whitney test.

Extended Data Fig. 9. Influence of P. alcaligenes on PM formation.

a, Phylogenetic tree showing the relationship between P. alcaligenes and other Pseudomonas sp. based on 16S rRNA gene sequences using ClustalW and MEGA. b, Fold change of P. alcaligenes abundance in the midguts of An. stephensi 0 h prior to and 24 h post normal blood meal. The level of 16S rRNA gene was normalized to that of s7 gene. Data are presented as mean ± s.e.m (n = 10). Significance was determined by two-sided Student’s t-test. Images are representative of two independent experiments. c, Colonization efficacy of P. alcaligenes in antibiotics- treated mosquitoes. P. alcaligenes CFU were counted in the midguts of Abx (-) and P. alcaligenes (+) recolonized mosquitoes before blood feeding. Data are presented as mean ± s.e.m (n = 5). d, PAS staining of PM in Abx and P. alcaligenes recolonized mosquitoes 45 h post normal blood meal at 100× and 200× magnification. Red arrows indicate the PM. Images are representative of at least seven individual mosquito midguts. Scale bars represent 100 μm.

Extended Data Fig. 10. Mutation of KynU in P. alcaligenes.

a, Confirmation of KynU deletion by PCR. b, The growth rate of wild type P. alcaligenes (P.a.WT) and KynU mutated P. alcaligenes (P.a.ΔKynU) in vitro. Data are presented as mean ± s.e.m (n = 6).

Supplementary information

Supplementary Information

Supplementary Tables 1–3, 5 and 6, and Figs. 1 and 2.

Reporting Summary

Peer Review File

Supplementary Table 4

Differentially expressed genes between normal and 3-HK-treated mosquitoes.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

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. 8

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Feng, Y., Peng, Y., Song, X. et al. Anopheline mosquitoes are protected against parasite infection by tryptophan catabolism in gut microbiota. Nat Microbiol 7, 707–715 (2022). https://doi.org/10.1038/s41564-022-01099-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-022-01099-8

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