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.

  • Review Article
  • Published:

Vector biology meets disease control: using basic research to fight vector-borne diseases

Abstract

Human pathogens that are transmitted by insects are a global problem, particularly those vectored by mosquitoes; for example, malaria parasites transmitted by Anopheles species, and viruses such as dengue, Zika and chikungunya that are carried by Aedes mosquitoes. Over the past 15 years, the prevalence of malaria has been substantially reduced and virus outbreaks have been contained by controlling mosquito vectors using insecticide-based approaches. However, disease control is now threatened by alarming rates of insecticide resistance in insect populations, prompting the need to develop a new generation of specific strategies that can reduce vector-mediated transmission. Here, we review how increased knowledge in insect biology and insect–pathogen interactions is stimulating new concepts and tools for vector control. We focus on strategies that either interfere with the development of pathogens within their vectors or directly impact insect survival, including enhancement of vector-mediated immune control, manipulation of the insect microbiome, or use of powerful new genetic tools such as CRISPR–Cas systems to edit vector genomes. Finally, we offer a perspective on the implementation hurdles as well as the knowledge gaps that must be filled in the coming years to safely realize the potential of these novel strategies to eliminate the scourge of vector-borne disease.

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: The malaria and dengue transmission cycles.
Fig. 2: Immune control strategies.
Fig. 3: Manipulating the microbiome.
Fig. 4: Gene drives bias inheritance to ensure their propagation.

Similar content being viewed by others

References

  1. Kwiatkowski, D. P. How malaria has affected the human genome and what human genetics can teach us about malaria. Am. J. Hum. Genet. 77, 171–192 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Kwiatkowski, D. Malaria genomics: tracking a diverse and evolving parasite population. Int. Health 7, 82–84 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  3. World Malaria Report 196 (WHO, Geneva, 2017).

  4. The malERA Consultative Group on Vector Control. A research agenda for malaria eradication: vector control. PLoS Med. 8, e1000401 (2011).

    Article  PubMed Central  Google Scholar 

  5. Neafsey, D. E. et al. Mosquito genomics. Highly evolvable malaria vectors: the genomes of 16 Anopheles mosquitoes. Science 347, 1258522 (2015).

    Article  CAS  PubMed  Google Scholar 

  6. Zeldenryk, L. M., Gray, M., Speare, R., Gordon, S. & Melrose, W. The emerging story of disability associated with lymphatic filariasis: a critical review. PLoS Negl. Trop. Dis. 5, e1366 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Bern, C., Maguire, J. H. & Alvar, J. Complexities of assessing the disease burden attributable to leishmaniasis. PLoS Negl. Trop. Dis. 2, e313 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Garza, M. et al. Projected future distributions of vectors of Trypanosoma cruzi in North America under climate change scenarios. PLoS Negl. Trop. Dis. 8, e2818 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Gascon, J., Bern, C. & Pinazo, M. J. Chagas disease in Spain, the United States and other non-endemic countries. Acta Trop. 115, 22–27 (2010).

    Article  PubMed  Google Scholar 

  10. Ioos, S. et al. Current Zika virus epidemiology and recent epidemics. Med. Mal. Infect. 44, 302–307 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Faria, N. R. et al. Zika virus in the Americas: early epidemiological and genetic findings. Science 352, 345–349 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Reiter, P. & Sprenger, D. The used tire trade: a mechanism for the worldwide dispersal of container breeding mosquitoes. J. Am. Mosq. Control Assoc. 3, 494–501 (1987).

    CAS  PubMed  Google Scholar 

  13. Rezza, G. et al. Infection with chikungunya virus in Italy: an outbreak in a temperate region. Lancet 370, 1840–1846 (2007).

    Article  CAS  PubMed  Google Scholar 

  14. Lednicky, J. et al. Mayaro virus in child with acute febrile illness, Haiti, 2015. Emerg. Infect. Dis. 22, 2000–2002 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Hotez, P. J. & Murray, K. O. Dengue, West Nile virus, chikungunya, Zika—and now Mayaro? PLoS Negl. Trop. Dis. 11, e0005462 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  16. 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 

  17. WHO Malaria fact sheet #94, December 2011 (Innovative Vector Control Consortium); www.ivcc.com/who-malaria-fact-sheet

  18. Franco, J. R. et al. Monitoring the elimination of human African trypanosomiasis: update to 2014. PLoS Negl. Trop. Dis. 11, e0005585 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Investing to Overcome the Global Impact of Neglected Tropical Diseases—Third WHO Report on Neglected Tropical Diseases 191 (WHO, Geneva, 2015).

  20. 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 

  21. Moyes, C. L. et al. Contemporary status of insecticide resistance in the major Aedes vectors of arboviruses infecting humans. PLoS Negl. Trop. Dis. 11, e0005625 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Gomes, B. et al. Knockdown resistance mutations predict DDT resistance and pyrethroid tolerance in the visceral leishmaniasis vector Phlebotomus argentipes. PLoS Negl. Trop. Dis. 11, e0005504 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Morrison, A. C., Zielinski-Gutierrez, E., Scott, T. W. & Rosenberg, R. Defining challenges and proposing solutions for control of the virus vector Aedes aegypti. PLoS Med. 5, e68 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Killeen, G. F. Characterizing, controlling and eliminating residual malaria transmission. Malar. J. 13, 330 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Nene, V. et al. Genome sequence of Aedes aegypti, a major arbovirus vector. Science 316, 1718–1723 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. Dudchenko, O. et al. De novo assembly of the Aedes aegypti genome using Hi-C yields chromosome-length scaffolds. Science 356, 92–95 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chen, X. G. et al. Genome sequence of the Asian Tiger mosquito, Aedes albopictus, reveals insights into its biology, genetics, and evolution. Proc. Natl Acad. Sci. USA 112, E5907–5915 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Smidler, A. L., Terenzi, O., Soichot, J., Levashina, E. A. & Marois, E. Targeted mutagenesis in the malaria mosquito using TALE nucleases. PLoS ONE 8, e74511 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Dong, S. et al. Heritable CRISPR/Cas9-mediated genome editing in the yellow fever mosquito, Aedes aegypti. PLoS ONE 10, e0122353 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Itokawa, K., Komagata, O., Kasai, S., Ogawa, K. & Tomita, T. Testing the causality between CYP9M10 and pyrethroid resistance using the TALEN and CRISPR/Cas9 technologies. Sci. Rep. 6, 24652 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Gantz, V. M. et al. Highly efficient Cas9-mediated gene drive for population modification of the malaria vector mosquito Anopheles stephensi. Proc. Natl Acad. Sci. USA 112, E6736–6743 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hammond, A. et al. A CRISPR-Cas9 gene drive system targeting female reproduction in the malaria mosquito vector Anopheles gambiae. Nat. Biotechnol. 34, 78–83 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Li, M. et al. Germline Cas9 expression yields highly efficient genome engineering in a major worldwide disease vector, Aedes aegypti. Proc. Natl Acad. Sci. USA 114, E10540–E10549 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dodson, B. L. & Rasgon, J. L. Vector competence of Anopheles and Culex mosquitoes for Zika virus. PeerJ 5, e3096 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Vanlandingham, D. L. et al. Differential infectivities of o’nyong-nyong and chikungunya virus isolates in Anopheles gambiae and Aedes aegypti mosquitoes. Am. J. Trop. Med. Hyg. 72, 616–621 (2005).

    Article  PubMed  Google Scholar 

  36. Macdonald, G. Epidemiological basis of malaria control. Bull. World Health Organ. 15, 613–626 (1956).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Jordan, A. M. & Curtis, C. F. Productivity of Glossina morsitans morsitans Westwood maintained in the laboratory, with particular reference to the sterile-insect release method. Bull. World Health Organ. 46, 33–38 (1972).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Tobe, S. S. & Langley, P. A. Reproductive physiology of Glossina. Ann. Rev. Entomol. 23, 283–307 (1978).

    Article  CAS  Google Scholar 

  39. Knols, B. G., Willemse, L., Flint, S. & Mate, A. A trial to control the tsetse fly, Glossina morsitans centralis, with low densities of odour-baited targets in west Zambia. Med. Vet. Entomol. 7, 161–169 (1993).

    Article  CAS  PubMed  Google Scholar 

  40. Gurtler, R. E. et al. Domestic animal hosts strongly influence human-feeding rates of the Chagas disease vector Triatoma infestans in Argentina. PLoS Negl. Trop. Dis. 8, e2894 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Beach, R. Mosquitoes: biting behavior inhibited by ecdysone. Science 205, 829–831 (1979).

    Article  CAS  PubMed  Google Scholar 

  42. Takken, W., Klowden, M. J. & Chambers, G. M. Effect of body size on host seeking and blood meal utilization in Anopheles gambiae sensu stricto (Diptera: Culicidae): the disadvantage of being small. J. Med. Entomol. 35, 639–645 (1998).

    Article  CAS  PubMed  Google Scholar 

  43. Lefèvre, T., Vantaux, A., Dabiré, K. R., Mouline, K. & Cohuet, A. Non-genetic determinants of mosquito competence for malaria parasites. PLoS Pathog. 9, e1003365 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. 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 

  45. Takken, W. et al. Larval nutrition differentially affects adult fitness and Plasmodium development in the malaria vectors Anopheles gambiae and Anopheles stephensi. Parasites Vectors 6, 345 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Blanford, S. et al. Fungal pathogen reduces potential for malaria transmission. Science 308, 1638–1641 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Chan, M. & Johansson, M. A. The incubation periods of Dengue viruses. PLoS ONE 7, e50972 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ye, Y. H. et al. Wolbachia reduces the transmission potential of Dengue-infected Aedes aegypti. PLoS Negl. Trop. Dis. 9, e0003894 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lindsay, S. W. et al. Ability of Anopheles gambiae mosquitoes to transmit malaria during the dry and wet seasons in an area of irrigated rice cultivation in The Gambia. J. Trop. Med. Hyg. 94, 313–324 (1991).

    CAS  PubMed  Google Scholar 

  50. Dengue: Guidelines for Diagnosis, Treatment, Prevention and Control: New Edition (WHO, 2009).

  51. Achee, N. L. et al. A critical assessment of vector control for dengue prevention. PLoS Negl. Trop. Dis. 9, e0003655 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Martinez-Torres, D. et al. Molecular characterization of pyrethroid knockdown resistance (kdr) in the major malaria vector Anopheles gambiae s.s. Insect Mol. Biol. 7, 179–184 (1998).

    Article  CAS  PubMed  Google Scholar 

  53. Martinez-Torres, D. et al. Voltage-dependent Na+ channels in pyrethroid-resistant Culex pipiens L mosquitoes. Pestic. Sci. 55, 1012–1020 (1999).

    Article  CAS  Google Scholar 

  54. Ranson, H. et al. Identification of a point mutation in the voltage-gated sodium channel gene of Kenyan Anopheles gambiae associated with resistance to DDT and pyrethroids. Insect Mol. Biol. 9, 491–497 (2000).

    Article  CAS  PubMed  Google Scholar 

  55. Norris, L. C. et al. Adaptive introgression in an African malaria mosquito coincident with the increased usage of insecticide-treated bed nets. Proc. Natl Acad. Sci. USA 112, 815–820 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Toe, K. H. et al. Increased pyrethroid resistance in malaria vectors and decreased bed net effectiveness, Burkina Faso. Emerg. Infect. Dis. 20, 1691–1696 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Poupardin, R., Srisukontarat, W., Yunta, C. & Ranson, H. Identification of carboxylesterase genes implicated in temephos resistance in the dengue vector Aedes aegypti. PLoS Negl. Trop. Dis. 8, e2743 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Bariami, V., Jones, C. M., Poupardin, R., Vontas, J. & Ranson, H. Gene amplification, ABC transporters and cytochrome P450s: unraveling the molecular basis of pyrethroid resistance in the dengue vector, Aedes aegypti. PLoS Negl. Trop. Dis. 6, e1692 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Grigoraki, L. et al. Transcriptome profiling and genetic study reveal amplified carboxylesterase genes implicated in temephos resistance, in the Asian Tiger mosquito Aedes albopictus. PLoS Negl. Trop. Dis. 9, e0003771 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Balabanidou, V. et al. Cytochrome P450 associated with insecticide resistance catalyzes cuticular hydrocarbon production in Anopheles gambiae. Proc. Natl Acad. Sci. USA 113, 9268–9273 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Viana, M., Hughes, A., Matthiopoulos, J., Ranson, H. & Ferguson, H. M. Delayed mortality effects cut the malaria transmission potential of insecticide-resistant mosquitoes. Proc. Natl Acad. Sci. USA 113, 8975–8980 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Martins, A. J. et al. Effect of insecticide resistance on development, longevity and reproduction of field or laboratory selected Aedes aegypti populations. PLoS ONE 7, e31889 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Baldini, F. et al. The interaction between a sexually transferred steroid hormone and a female protein regulates oogenesis in the malaria mosquito Anopheles gambiae. PLoS Biol. 11, e1001695 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Gabrieli, P. et al. Sexual transfer of the steroid hormone 20E induces the postmating switch in Anopheles gambiae. Proc. Natl Acad. Sci. USA 111, 16353–16358 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Mitchell, S. N. et al. Mosquito biology. Evolution of sexual traits influencing vectorial capacity in anopheline mosquitoes. Science 347, 985–988 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Pitts, R. J., Liu, C., Zhou, X., Malpartida, J. C. & Zwiebel, L. J. Odorant receptor-mediated sperm activation in disease vector mosquitoes. Proc. Natl Acad. Sci. USA 111, 2566–2571 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Clifton, M. E., Correa, S., Rivera-Perez, C., Nouzova, M. & Noriega, F. G. Male Aedes aegypti mosquitoes use JH III transferred during copulation to influence previtellogenic ovary physiology and affect the reproductive output of female mosquitoes. J. Insect Physiol. 64, 40–47 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Harris, C. et al. Sterilising effects of pyriproxyfen on Anopheles arabiensis and its potential use in malaria control. Parasit. Vectors 6, 144 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Lwetoijera, D. W. et al. Comprehensive sterilization of malaria vectors using pyriproxyfen: a step closer to malaria elimination. Am. J. Trop. Med. Hyg. 90, 852–855 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Kawada, H. et al. A small-scale field trial of pyriproxyfen-impregnated bed nets against pyrethroid-resistant Anopheles gambiae s.s. in western Kenya. PLoS ONE 9, e111195 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Ohashi, K. et al. Efficacy of pyriproxyfen-treated nets in sterilizing and shortening the longevity of Anopheles gambiae (Diptera: Culicidae). J. Med. Entomol. 49, 1052–1058 (2012).

    Article  CAS  PubMed  Google Scholar 

  72. Ngufor, C. et al. Olyset Duo(R) (a pyriproxyfen and permethrin mixture net): an experimental hut trial against pyrethroid resistant Anopheles gambiae and Culex quinquefasciatus in Southern Benin. PLoS ONE 9, e93603 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Childs, L. M. et al. Disrupting mosquito reproduction and parasite development for malaria control. PLoS Pathog. 12, e1006060 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Tiono, A. B. et al. The AvecNet Trial to assess whether addition of pyriproxyfen, an insect juvenile hormone mimic, to long-lasting insecticidal mosquito nets provides additional protection against clinical malaria over current best practice in an area with pyrethroid-resistant vectors in rural Burkina Faso: study protocol for a randomised controlled trial. Trials 16, 113 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Shin, S. W. et al. REL1, a homologue of Drosophila dorsal, regulates toll antifungal immune pathway in the female mosquito Aedes aegypti. J. Biol. Chem. 280, 16499–16507 (2005).

    Article  CAS  PubMed  Google Scholar 

  76. Bian, G., Shin, S. W., Cheon, H. M., Kokoza, V. & Raikhel, A. S. Transgenic alteration of Toll immune pathway in the female mosquito Aedes aegypti. Proc. Natl Acad. Sci. USA 102, 13568–13573 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Frolet, C., Thoma, M., Blandin, S., Hoffmann, J. A. & Levashina, E. A. Boosting NF-κB-dependent basal immunity of Anopheles gambiae aborts development of Plasmodium berghei. Immunity 25, 677–685 (2006).

    Article  CAS  PubMed  Google Scholar 

  78. Garver, L. S., Dong, Y. & Dimopoulos, G. Caspar controls resistance to Plasmodium falciparum in diverse anopheline species. PLoS Pathog. 5, e1000335 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Meister, S. et al. Anopheles gambiae PGRPLC-mediated defense against bacteria modulates infections with malaria parasites. PLoS Pathog. 5, e1000542 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Mitri, C. et al. Fine pathogen discrimination within the APL1 gene family protects Anopheles gambiae against human and rodent malaria species. PLoS Pathog. 5, e1000576 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Dong, Y. et al. Engineered anopheles immunity to Plasmodium infection. PLoS Pathog. 7, e1002458 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Zou, Z. et al. Transcriptome analysis of Aedes aegypti transgenic mosquitoes with altered immunity. PLoS Pathog. 7, e1002394 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Antonova, Y., Alvarez, K. S., Kim, Y. J., Kokoza, V. & Raikhel, A. S. The role of NF-κB factor REL2 in the Aedes aegypti immune response. Insect Biochem. Mol. Biol. 39, 303–314 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Castillo, J. C., Ferreira, A. B. B., Trisnadi, N. & Barillas-Mury, C. Activation of mosquito complement antiplasmodial response requires cellular immunity. Sci. Immunol. 2, eaal1505 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Smith, R. C. & Barillas-Mury, C. Plasmodium oocysts: overlooked targets of mosquito immunity. Trends Parasitol. 32, 979–990 (2016).

    Article  PubMed  Google Scholar 

  86. Gupta, L. et al. The STAT pathway mediates late-phase immunity against Plasmodium in the mosquito Anopheles gambiae. Cell Host Microbe 5, 498–507 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Smith, R. C., Barillas-Mury, C. & Jacobs-Lorena, M. Hemocyte differentiation mediates the mosquito late-phase immune response against Plasmodium in Anopheles gambiae. Proc. Natl Acad. Sci. USA 112, E3412–3420 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Olson, K. E. & Blair, C. D. Arbovirus-mosquito interactions: RNAi pathway. Curr. Opin. Virol. 15, 119–126 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Morazzani, E. M., Wiley, M. R., Murreddu, M. G., Adelman, Z. N. & Myles, K. M. Production of virus-derived ping-pong-dependent piRNA-like small RNAs in the mosquito soma. PLoS Pathog. 8, e1002470 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Vodovar, N. et al. Arbovirus-derived piRNAs exhibit a ping-pong signature in mosquito cells. PLoS ONE 7, e30861 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Hess, A. M. et al. Small RNA profiling of Dengue virus-mosquito interactions implicates the PIWI RNA pathway in anti-viral defense. BMC Microbiol. 11, 45 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Miesen, P., Girardi, E. & van Rij, R. P. Distinct sets of PIWI proteins produce arbovirus and transposon-derived piRNAs in Aedes aegypti mosquito cells. Nucleic Acids Res. 43, 6545–6556 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Xi, Z., Ramirez, J. L. & Dimopoulos, G. The Aedes aegypti toll pathway controls dengue virus infection. PLoS Pathog. 4, e1000098 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Souza-Neto, J. A., Sim, S. & Dimopoulos, G. An evolutionary conserved function of the JAK-STAT pathway in anti-dengue defense. Proc. Natl Acad. Sci. USA 106, 17841–17846 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Ramirez, J. L. & Dimopoulos, G. The Toll immune signaling pathway control conserved anti-dengue defenses across diverse Ae. aegypti strains and against multiple dengue virus serotypes. Dev. Comp. Immunol. 34, 625–629 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Sim, S. et al. Transcriptomic profiling of diverse Aedes aegypti strains reveals increased basal-level immune activation in dengue virus-refractory populations and identifies novel virus-vector molecular interactions. PLoS Negl. Trop. Dis. 7, e2295 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Palmer, W. H., Varghese, F. S. & van Rij, R. P. Natural variation in resistance to virus infection in Dipteran insects. Viruses 10, 118 (2018).

    Article  CAS  PubMed Central  Google Scholar 

  98. Kokoza, V. et al. Engineering blood meal-activated systemic immunity in the yellow fever mosquito, Aedes aegypti. Proc. Natl Acad. Sci. USA 97, 9144–9149 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Kim, W. et al. Ectopic expression of a cecropin transgene in the human malaria vector mosquito Anopheles gambiae (Diptera: Culicidae): effects on susceptibility to Plasmodium. J. Med. Entomol. 41, 447–455 (2004).

    Article  CAS  PubMed  Google Scholar 

  100. Jupatanakul, N. et al. Engineered Aedes aegypti JAK/STAT pathway-mediated immunity to Dengue virus. PLoS Negl. Trop. Dis. 11, e0005187 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Isaacs, A. T. et al. Transgenic Anopheles stephensi coexpressing single-chain antibodies resist Plasmodium falciparum development. Proc. Natl Acad. Sci. USA 109, E1922–1930 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Clayton, A. M., Cirimotich, C. M., Dong, Y. & Dimopoulos, G. Caudal is a negative regulator of the Anopheles IMD pathway that controls resistance to Plasmodium falciparum infection. Dev. Comp. Immunol. 39, 323–332 (2013).

    Article  CAS  PubMed  Google Scholar 

  103. Garver, L. S., de Almeida Oliveira, G. & Barillas-Mury, C. The JNK pathway is a key mediator of Anopheles gambiae antiplasmodial immunity. PLoS Pathog. 9, e1003622 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Dong, Y., Cirimotich, C. M., Pike, A., Chandra, R. & Dimopoulos, G. Anopheles NF-κB-regulated splicing factors direct pathogen-specific repertoires of the hypervariable pattern recognition receptor AgDscam. Cell Host Microbe 12, 521–530 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Smith, R. C., Kizito, C., Rasgon, J. L. & Jacobs-Lorena, M. Transgenic mosquitoes expressing a phospholipase A(2) gene have a fitness advantage when fed Plasmodium falciparum-infected blood. PLoS ONE 8, e76097 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Franz, A. W. et al. Engineering RNA interference-based resistance to dengue virus type 2 in genetically modified Aedes aegypti. Proc. Natl Acad. Sci. USA 103, 4198–4203 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Pike, A. et al. Changes in the microbiota cause genetically modified Anopheles to spread in a population. Science 357, 1396–1399 (2017).

    Article  CAS  PubMed  Google Scholar 

  108. Ito, J., Ghosh, A., Moreira, L. A., Wimmer, E. A. & Jacobs-Lorena, M. Transgenic anopheline mosquitoes impaired in transmission of a malaria parasite. Nature 417, 452–455 (2002).

    Article  CAS  PubMed  Google Scholar 

  109. Ghosh, A. K., Coppens, I., Gardsvoll, H., Ploug, M. & Jacobs-Lorena, M. Plasmodium ookinetes coopt mammalian plasminogen to invade the mosquito midgut. Proc. Natl Acad. Sci. USA 108, 17153–17158 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Vega-Rodriguez, J. et al. Multiple pathways for Plasmodium ookinete invasion of the mosquito midgut. Proc. Natl Acad. Sci. USA 111, E492–500 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Ghosh, A. K. et al. Malaria parasite invasion of the mosquito salivary gland requires interaction between the Plasmodium TRAP and the Anopheles saglin proteins. PLoS Pathog. 5, e1000265 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Clayton, A. M., Dong, Y. & Dimopoulos, G. The Anopheles innate immune system in the defense against malaria infection. J. Innate Immun. 6, 169–181 (2014).

    Article  CAS  PubMed  Google Scholar 

  113. Molina-Cruz, A. et al. The human malaria parasite Pfs47 gene mediates evasion of the mosquito immune system. Science 340, 984–987 (2013).

    Article  CAS  PubMed  Google Scholar 

  114. Smith, R. C. & Jacobs-Lorena, M. Malaria parasite Pfs47 disrupts JNK signaling to escape mosquito immunity. Proc. Natl Acad. Sci. USA 112, 1250–1251 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Molina-Cruz, A. et al. Plasmodium evasion of mosquito immunity and global malaria transmission: the lock-and-key theory. Proc. Natl Acad. Sci. USA 112, 15178–15183 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Dillon, R. J. & Dillon, V. M. The gut bacteria of insects: nonpathogenic interactions. Annu. Rev. Entomol. 49, 71–92 (2004).

    Article  CAS  PubMed  Google Scholar 

  117. Dale, C. & Moran, N. A. Molecular interactions between bacterial symbionts and their hosts. Cell 126, 453–465 (2006).

    Article  CAS  PubMed  Google Scholar 

  118. Douglas, A. E. Nutritional interactions in insect-microbial symbioses: aphids and their symbiotic bacteria Buchnera. Annu. Rev. Entomol. 43, 17–37 (1998).

    Article  CAS  PubMed  Google Scholar 

  119. 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 

  120. Wang, Y., Gilbreath, T. M. 3rd, 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 

  121. Boissière, 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 

  122. da Mota, F. F. et al. Cultivation-independent methods reveal differences among bacterial gut microbiota in triatomine vectors of Chagas disease. PLoS Negl. Trop. Dis. 6, e1631 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Aksoy, E. et al. Analysis of multiple tsetse fly populations in Uganda reveals limited diversity and species-specific gut microbiota. Appl. Environ. Microbiol. 80, 4301–4312 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Segata, N. et al. The reproductive tracts of two malaria vectors are populated by a core microbiome and by gender- and swarm-enriched microbial biomarkers. Sci. Rep. 6, 24207 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Fraihi, W. et al. An integrated overview of the midgut bacterial flora composition of Phlebotomus perniciosus, a vector of zoonotic visceral leishmaniasis in the Western Mediterranean Basin. PLoS Negl. Trop. Dis. 11, e0005484 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  126. Azambuja, P., Feder, D. & Garcia, E. S. Isolation of Serratia marcescens in the midgut of Rhodnius prolixus: impact on the establishment of the parasite Trypanosoma cruzi in the vector. Exp. Parasitol. 107, 89–96 (2004).

    Article  CAS  PubMed  Google Scholar 

  127. Moraes, C. S. et al. Prodigiosin is not a determinant factor in lysis of Leishmania (Viannia) braziliensis after interaction with Serratia marcescens D-mannose sensitive fimbriae. Exp. Parasitol. 122, 84–90 (2009).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. 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 

  130. Apte-Deshpande, A., Paingankar, M., Gokhale, M. D. & Deobagkar, D. N. Serratia odorifera a midgut inhabitant of Aedes aegypti mosquito enhances its susceptibility to dengue-2 virus. PLoS ONE 7, e40401 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Carissimo, G. et al. Antiviral immunity of Anopheles gambiae is highly compartmentalized, with distinct roles for RNA interference and gut microbiota. Proc. Natl Acad. Sci. USA 112, 176–185 (2015).

    Article  CAS  Google Scholar 

  132. Scholte, E. J. et al. An entomopathogenic fungus for control of adult African malaria mosquitoes. Science 308, 1641–1642 (2005).

    Article  CAS  PubMed  Google Scholar 

  133. Fieck, A., Hurwitz, I., Kang, A. S. & Durvasula, R. Trypanosoma cruzi: synergistic cytotoxicity of multiple amphipathic anti-microbial peptides to T. cruzi and potential bacterial hosts. Exp. Parasitol. 125, 342–347 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  135. Xi, Z., Khoo, C. C. & Dobson, S. L. Wolbachia establishment and invasion in an Aedes aegypti laboratory population. Science 310, 326–328 (2005).

    Article  CAS  PubMed  Google Scholar 

  136. Bian, G. et al. Wolbachia invades Anopheles stephensi populations and induces refractoriness to Plasmodium infection. Science 340, 748–751 (2013).

    Article  CAS  PubMed  Google Scholar 

  137. Attardo, G. M. et al. Analysis of milk gland structure and function in Glossina morsitans: milk protein production, symbiont populations and fecundity. J. Insect Physiol. 54, 1236–1242 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Favia, G. et al. Bacteria of the genus Asaia stably associate with Anopheles stephensi, an Asian malarial mosquito vector. Proc. Natl Acad. Sci. USA 104, 9047–9051 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Hilgenboecker, K., Hammerstein, P., Schlattmann, P., Telschow, A. & Werren, J. H. How many species are infected with Wolbachia?A statistical analysis of current data. FEMS Microbiol. Lett. 281, 215–220 (2008).

    Article  CAS  PubMed  Google Scholar 

  140. Werren, J. H., Baldo, L. & Clark, M. E. Wolbachia: master manipulators of invertebrate biology. Nat. Rev. Micro. 6, 741–751 (2008).

    Article  CAS  Google Scholar 

  141. LePage, D. P. et al. Prophage WO genes recapitulate and enhance Wolbachia-induced cytoplasmic incompatibility. Nature 543, 243–247 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Beckmann, J. F., Ronau, J. A. & Hochstrasser, M. A Wolbachia deubiquitylating enzyme induces cytoplasmic incompatibility. Nat. Microbiol. 2, 17007 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  143. Lindsey, A. R. I. et al. Evolutionary genetics of cytoplasmic incompatibility genes cifA and cifB in prophage WO of Wolbachia. Genome Biol. Evol. 10, 434–451 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Hughes, G. L., Koga, R., Xue, P., Fukatsu, T. & Rasgon, J. L. Wolbachia infections are virulent and inhibit the human malaria parasite Plasmodium falciparum in Anopheles gambiae. PLoS Pathog. 7, e1002043 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Kambris, Z. et al. Wolbachia stimulates immune gene expression and inhibits plasmodium development in Anopheles gambiae. PLoS Pathog. 6, e1001143 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Moreira, L. A. et al. A Wolbachia symbiont in Aedes aegypti limits infection with Dengue, Chikungunya, and Plasmodium. Cell 139, 1268–1278 (2009).

    Article  PubMed  Google Scholar 

  147. Glaser, R. L. & Meola, M. A. The native Wolbachia endosymbionts of Drosophila melanogaster and Culex quinquefasciatus increase host resistance to West Nile virus infection. PLoS ONE 5, e11977 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. van den Hurk, A. F. et al. Impact of Wolbachia on infection with chikungunya and yellow fever viruses in the mosquito vector Aedes aegypti. PLoS Negl. Trop. Dis. 6, e1892 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  149. Blagrove, M. S. C., Arias-Goeta, C., Failloux, A. B. & Sinkins, S. P. Wolbachia strain wMel induces cytoplasmic incompatibility and blocks dengue transmission in Aedes albopictus. Proc. Natl Acad. Sci. USA 109, 255–260 (2012).

    Article  CAS  PubMed  Google Scholar 

  150. Dutra, H. L. et al. Wolbachia blocks currently circulating Zika virus isolates in Brazilian Aedes aegypti mosquitoes. Cell Host Microbe 19, 771–774 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Aliota, M. T., Peinado, S. A., Velez, I. D. & Osorio, J. E. The wMel strain of Wolbachia reduces transmission of Zika virus by Aedes aegypti. Sci. Rep. 6, 28792 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Caragata, E. P. et al. Dietary cholesterol modulates pathogen blocking by Wolbachia. PLoS Pathog. 9, e1003459 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Geoghegan, V. et al. Perturbed cholesterol and vesicular trafficking associated with dengue blocking in Wolbachia-infected Aedes aegypti cells. Nat. Commun. 8, 526 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Brelsfoard, C. L., Sechan, Y. & Dobson, S. L. Interspecific hybridization yields strategy for South Pacific filariasis vector elimination. PLoS Negl. Trop. Dis. 2, e129 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Atyame, C. M. et al. Cytoplasmic incompatibility as a means of controlling Culex pipiens quinquefasciatus mosquito in the islands of the south-western Indian Ocean. PLoS Negl. Trop. Dis. 5, e1440 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  156. World Mosquito Program (2017); www.worldmosquitoprogram.org

  157. Frentiu, F. D. et al. Limited dengue virus replication in field-collected Aedes aegypti mosquitoes infected with Wolbachia. PLoS Negl. Trop. Dis. 8, e2688 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  158. Schmidt, T. L. et al. Local introduction and heterogeneous spatial spread of dengue-suppressing Wolbachia through an urban population of Aedes aegypti. PLoS Biol. 15, e2001894 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Baldini, F. et al. Evidence of natural Wolbachia infections in field populations of Anopheles gambiae. Nat. Commun. 5, 3985 (2014).

    Article  CAS  PubMed  Google Scholar 

  160. Shaw, W. R. et al. Wolbachia infections in natural Anopheles populations affect egg laying and negatively correlate with Plasmodium development. Nat. Commun. 7, 11772 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Gomes, F. M. et al. Effect of naturally occurring Wolbachia in Anopheles gambiae s.l. mosquitoes from Mali on Plasmodium falciparum malaria transmission. Proc. Natl Acad. Sci. USA 114, 12566–12571 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Dodson, B. L. et al. Wolbachia enhances West Nile virus (WNV) infection in the mosquito Culex tarsalis. PLoS Negl. Trop. Dis. 8, e2965 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Hughes, G. L., Rivero, A. & Rasgon, J. L. Wolbachia can enhance Plasmodium infection in mosquitoes: implications for malaria control? PLoS Pathog. 10, e1004182 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Burt, A. Site-specific selfish genes as tools for the control and genetic engineering of natural populations. Proc. Biol. Sci. 270, 921–928 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Lindquist, D. A., Abusowa, M. & Hall, M. J. The New World screwworm fly in Libya: a review of its introduction and eradication. Med. Vet. Entomol. 6, 2–8 (1992).

    Article  CAS  PubMed  Google Scholar 

  166. Oliva, C. F. et al. The sterile insect technique for controlling populations of Aedes albopictus (Diptera: Culicidae) on Reunion Island: mating vigour of sterilized males. PLoS ONE 7, e49414 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Bellini, R., Medici, A., Puggioli, A., Balestrino, F. & Carrieri, M. Pilot field trials with Aedes albopictus irradiated sterile males in Italian urban areas. J. Med. Entomol. 50, 317–325 (2013).

    Article  CAS  PubMed  Google Scholar 

  168. Vreysen, M. J. et al. Glossina austeni (Diptera: Glossinidae) eradicated on the island of Unguja, Zanzibar, using the sterile insect technique. J. Econ. Entomol. 93, 123–135 (2000).

    Article  CAS  PubMed  Google Scholar 

  169. Dame, D. A., Curtis, C. F., Benedict, M. Q., Robinson, A. S. & Knols, B. G. Historical applications of induced sterilisation in field populations of mosquitoes. Malar. J. 2, S2 (2009).

    Article  Google Scholar 

  170. Harris, A. F. et al. Field performance of engineered male mosquitoes. Nat. Biotechnol. 29, 1034–1037 (2011).

    Article  CAS  PubMed  Google Scholar 

  171. Harris, A. F. et al. Successful suppression of a field mosquito population by sustained release of engineered male mosquitoes. Nat. Biotechnol. 30, 828–830 (2012).

    Article  CAS  PubMed  Google Scholar 

  172. Carvalho, D. O. et al. Suppression of a field population of Aedes aegypti in Brazil by sustained release of transgenic male mosquitoes. PLoS Negl. Trop. Dis. 9, e0003864 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Chen, C. H. et al. A synthetic maternal-effect selfish genetic element drives population replacement in Drosophila. Science 316, 597–600 (2007).

    Article  CAS  PubMed  Google Scholar 

  174. Windbichler, N. et al. A synthetic homing endonuclease-based gene drive system in the human malaria mosquito. Nature 473, 212–215 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Simoni, A. et al. Development of synthetic selfish elements based on modular nucleases in Drosophila melanogaster. Nucleic Acids Res. 42, 7461–7472 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Esvelt, K. M., Smidler, A. L., Catteruccia, F. & Church, G. M. Concerning RNA-guided gene drives for the alteration of wild populations. eLife 3, e03401 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Hsu, P. D., Lander, E. S. & Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 1262–1278 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Hammond, A. M. et al. The creation and selection of mutations resistant to a gene drive over multiple generations in the malaria mosquito. PLoS Genet. 13, e1007039 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Champer, J. et al. Novel CRISPR/Cas9 gene drive constructs reveal insights into mechanisms of resistance allele formation and drive efficiency in genetically diverse populations. PLoS Genet. 13, e1006796 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. The Anopheles gambiae 1000 Genomes Consortium. Genetic diversity of the African malaria vector Anopheles gambiae. Nature 552, 96–100 (2017).

  181. Marshall, J. M., Buchman, A., Sanchez, C. H. & Akbari, O. S. Overcoming evolved resistance to population-suppressing homing-based gene drives. Sci. Rep. 7, 3776 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Sawadogo, S. P. et al. Effects of age and size on Anopheles gambiae s.s. male mosquito mating success. J. Med. Entomol. 50, 285–293 (2013).

    Article  PubMed  Google Scholar 

  183. Sawadogo, S. P. et al. Targeting male mosquito swarms to control malaria vector density. PLoS ONE 12, e0173273 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Diabate, A. & Tripet, F. Targeting male mosquito mating behaviour for malaria control. Parasit. Vectors 8, 347 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  185. Cator, L. J., Arthur, B. J., Harrington, L. C. & Hoy, R. R. Harmonic convergence in the love songs of the dengue vector mosquito. Science 323, 1077–1079 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Cator, L. J. & Harrington, L. C. The harmonic convergence of fathers predicts the mating success of sons in Aedes aegypti. Anim. Behav. 82, 627–633 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  187. Oye, K. A. et al. Biotechnology. Regulating gene drives. Science 345, 626–628 (2014).

    Article  CAS  PubMed  Google Scholar 

  188. Ferguson, H. M. et al. Ecology: a prerequisite for malaria elimination and eradication. PLoS Med. 7, e1000303 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  189. Norris, E. J. & Coats, J. R. Current and future repellent technologies: the potential of spatial repellents and their place in mosquito-borne disease control. Int. J. Environ. Res. Public Health 14, 124 (2017).

    Article  CAS  PubMed Central  Google Scholar 

  190. Ernst, K. C. et al. Awareness and support of release of genetically modified “sterile” mosquitoes, Key West, Florida, USA. Emerg. Infect. Dis. 21, 320–324 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Adalja, A., Sell, T. K., McGinty, M. & Boddie, C. Genetically modified (gm) mosquito use to reduce mosquito-transmitted disease in the US: a community opinion survey. PLoS Curr. https://doi.org/10.1371/currents.outbreaks.1c39ec05a743d41ee39391ed0f2ed8d3 (2016).

  192. Mathers, C. D., Ezzati, M. & Lopez, A. D. Measuring the burden of neglected tropical diseases: the global burden of disease framework. PLoS Negl. Trop. Dis. 1, e114 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  193. Global Program to Eliminate Lymphatic Filariasis Progress Report 594–608 (WHO, Geneva, 2017).

  194. Lymphatic filariasis (WHO, Geneva, 2017); www.who.int/mediacentre/factsheets/fs102/en/

  195. GBD 2015 Disease and Injury Incidence Prevalence Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet 388, 1545–1602 (2016).

    Article  Google Scholar 

  196. GBD 2015 Mortality and Cause of Death Collaborators. Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet 388, 1459–1544 (2016).

    Article  Google Scholar 

  197. What is Chagas disease? (WHO, Geneva, 2017); www.who.int/chagas/disease/en/

  198. Control of the leishmaniases — Report of a WHO Expert Committee 158 (World Health Organization, Geneva, 1990).

  199. Parasites—Onchocerciasis (also known as River Blindness) (Centers for Disease Control and Prevention, 2017); www.cdc.gov/parasites/onchocerciasis/epi.html

  200. Brady, O. J. et al. Refining the global spatial limits of dengue virus transmission by evidence-based consensus. PLoS Negl. Trop. Dis. 6, e1760 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  201. Shearer, F. M. et al. Global yellow fever vaccination coverage from 1970 to 2016: an adjusted retrospective analysis. Lancet Infect. Dis. 17, 1209–1217 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  202. CDC: Zika Travel Information (2017); wwwnc.cdc.gov/travel/page/zika-information

  203. Messina, J. P. et al. Mapping global environmental suitability for Zika virus. eLife 5, e15272 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Nsoesie, E. O. et al. Global distribution and environmental suitability for chikungunya virus, 1952 to 2015. Euro. Surveill. https://doi.org/10.2807/1560-7917.ES.2016.21.20.30234 (2016).

  205. Chikungunya (WHO, Geneva, 2017); www.who.int/mediacentre/factsheets/fs327/en/

  206. Gibson, G. & Torr, S. J. Visual and olfactory responses of haematophagous Diptera to host stimuli. Med. Vet. Entomol. 13, 2–23 (1999).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank M. Bernardi for graphic assistance and D. Abernathy, P. Marcenac and D. Paton for careful reading of this manuscript. F.C. is funded on research related to the topic discussed here by a Faculty Research Scholar Award by the Howard Hughes Medical Institute (HHMI) and the Bill & Melinda Gates Foundation (BMGF) (grant ID OPP1158190), and by the National Institutes of Health (NIH) (R01 AI124165). The findings and conclusions within this publication are those of the authors and do not necessarily reflect positions or policies of the HHMI, the BMGF or the NIH.

Author information

Authors and Affiliations

Authors

Contributions

W.R.S. and F.C. wrote the paper.

Corresponding authors

Correspondence to W. Robert Shaw or Flaminia Catteruccia.

Ethics declarations

Competing interests

Harvard University has filed a patent application on behalf of the investigators related to this research.

Additional information

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shaw, W.R., Catteruccia, F. Vector biology meets disease control: using basic research to fight vector-borne diseases. Nat Microbiol 4, 20–34 (2019). https://doi.org/10.1038/s41564-018-0214-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-018-0214-7

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