Skip to main content

REVIEW article

Front. Genet., 10 March 2020
Sec. Evolutionary and Population Genetics
This article is part of the Research Topic The Arms Race Between Vectors and Human Pathogens View all 9 articles

Use of Microbiota to Fight Mosquito-Borne Disease

  • 1Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
  • 2CAS Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China

Mosquito-borne diseases cause more than 700 million people infected and one million people die (Caraballo and King, 2014). With the limitations of progress toward elimination imposed by insecticide- and drug-resistance, combined with the lack of vaccines, innovative strategies to fight mosquito-borne disease are urgently needed. In recent years, the use of mosquito microbiota has shown great potential for cutting down transmission of mosquito-borne pathogens. Here we review what is known about the mosquito microbiota and how this knowledge is being used to develop new ways to control mosquito-borne disease. We also discuss the challenges for the eventual release of genetically modified (GM) symbionts in the field.

Introduction

Mosquito vectors mainly include three genera, Anopheles, Aedes, and Culex. Spread of disease is via the bite of infected female mosquitoes. The pathogens include malaria, dengue, Chikungunya, Zika, Yellow fever, and West Nile and they lead to more than one million deaths every year (WHO, 2016; Rosenberg et al., 2018). Presently, strategies to control mosquito-borne diseases are limited to mosquito population reduction and in case of malaria, to drugs. No drugs are available to treat viral diseases. With the current unavailability of a vaccine (with the exception of yellow fever) that protects from any of the mosquito-borne pathogens (Cheeseman et al., 2012; Ferguson, 2018) and with the widespread of insecticide resistance of mosquitoes (Ranson and Lissenden, 2016), new weapons to fight these diseases are urgently needed.

Insect microbiota are involved in many important biological processes such as nutrition, digestion, sexual reproduction, development, and refractoriness to pathogens (Douglas, 2014). Bacteria such as Wolbachia, can shorten the life span of some mosquito species (McMeniman et al., 2009) and block virus mosquito infection and dissemination (Moreira et al., 2009; van den Hurk et al., 2012). In recent years, increasing interest has been shown in employing symbiotic bacteria to control mosquito-borne diseases.

Gut Microbiota Diversity and Distribution in Mosquitoes

The mosquito gut microbiota includes prokaryotes, viruses, and eukaryotic microbes. In this review, we focus on prokaryotes and eukaryotic microbes. The mosquito gut microbiota is mainly acquired from the environment (Wang et al., 2017; Strand, 2018), and its composition is highly dynamic, varying greatly with species, nutrition, stage of mosquito development, and geography (Tchioffo et al., 2015; Minard et al., 2017; Novakova et al., 2017; Bascunan et al., 2018; Krajacich et al., 2018; Muturi et al., 2018; Telang et al., 2018; Duguma et al., 2019). Sequencing of the 16S rRNA or18S rRNA hypervariable regions is used as a culture-independent approach to study mosquito microbiota composition (Pidiyar et al., 2004; Belda et al., 2017).

The mosquito gut microbiota is dominated by Gram-negative bacteria. A previous study identified 98 bacteria genera in anophelines, Pseudomonas, Aeromonas, Asaia, Comamonas, Elizabethkingia, Enterobacter, Klebsiella, Pantoea, and Serratia being the most common ones (Gendrin and Christophides, 2013). Similarly, Gram-negative bacteria are also dominant in Aedes spp. (Scolari et al., 2019).

However, unlike for the prokaryotic bacteria, the abundant 18S rRNA of the mosquito host strongly interferes with the definition of the eukaryotic microbiota composition via 18S rRNA gene sequencing. Thus, the mosquito eukaryotic microbiota remains poorly studied. Belda designed V4-region peptide-nucleic acid (PNA) oligonucleotide blockers to reduce by more than 80% mosquito 18S rRNA background for the detection of eukaryotic microbes (Belda et al., 2017). Most eukaryotic microbiota identified from mosquitoes belong to single cell eukaryotic phyla, such as Candida, Pichia with some Penicillium also being identified (Jupatanakul et al., 2014; Romoli and Gendrin, 2018; Thongsripong et al., 2018).

Bacteria colonize different mosquito organs, mainly midgut and rarely salivary glands, ovaries and male accessory glands (Tchioffo et al., 2015; Muturi et al., 2018). Most studies have focused on midgut microbiota. Mosquito salivary gland, ovaries and hemolymph are also important for virus or parasite replication and transmission. The adult mosquito midgut and ovary share some dominant bacteria classes, while other bacteria are only found in specific tissues or development stages (Tchioffo et al., 2015). Ovary bacteria can be vertically transmitted. Wolbachia is an intracellular bacterium that infects not only somatic tissue cells, but importantly also stably infects the germ cells of the ovary leading to vertical transmission (Hughes et al., 2014; Fraser et al., 2017; Jiggins, 2017). Asaia, an extracellular bacterium, can colonize the ovary of Anopheles mosquitoes and be vertically transmitted (Favia et al., 2007; Damiani et al., 2010). Serratia AS1, also an extracellular bacterium, was originally isolated from Anopheles ovaries, stably colonizes ovaries, and is transmitted vertically from female to progeny (Wang et al., 2017). Interestingly, Serratia AS1 also colonizes the accessory glands of male Anopheles mosquitoes, leading to sexually transmission (Wang et al., 2017).

Impact of Microbiota on Mosquito Physiology and Pathogen Transmission

Mosquito microbiota play critical roles in many mosquito biology processes, including nutrition, digestion, mating and sexual reproduction, development, immune response functions, and refractoriness to pathogens (Douas, 2011).

Impact of Microbiota on Mosquito Nutrition, Reproduction and Development

Dong et al. (2009) compared transcriptome between septic and aseptic adult female mosquitoes that had been fed different diets and found that some genes involved in digestion and metabolic processes such as glycolysis, gluconeogenesis and sugar transport, are stimulated by the presence of the microbiota. In Aedes aegypti, midgut microbiota, especially Enterobacter sp. and Serratia sp. isolates possess hemolytic activity that can lead to red blood cell (RBC) lysis and hemoglobin release (Gaio Ade et al., 2011). In A. aegypti, antibiotics treatment of female mosquitoes decreased the lysis of RBCs and egg production (Gaio Ade et al., 2011). However, egg production is not supported by every bacterium. Individual bacteria genera were used to populate adult mosquitoes emerged from gnotobiotic larvae. Five bacteria (Aquitalea, Sphingobacterium, Chryseobacterium, Paenibacillus, and Comamonas) were tested which supported egg production in A. aegypti, while in A. atropalpus only Comamonas supported egg production (Coon et al., 2016).

Mosquito microbiota can affect mosquito development. In Anopheles, a higher load of bacteria in the food diet sped larva growth and development (Linenberg et al., 2016). In A. gambiae, larvae carrying Asaia developed faster as it took 2 days less to reach the pupal stage than no-Asaia larvae (Mitraka et al., 2013). In A. aegypti, larval gut bacteria are crucial for growth and molting (Coon et al., 2017). Axenic larvae which are produced by surface sterilizing eggs, don’t molt and die as first instars; some species of bacteria which include Escherichia coli can colonize the midgut of axenic larvae and rescue larvae growth, while dead bacteria do not (Coon et al., 2014; Thongsripong et al., 2018). Larva gut microbiota consume oxygen and mediate hypoxia in the midgut. The hypoxia signal activates hypoxia-inducible transcription factors (HIFs) which activate several processes essential for larval growth, such as the insulin/insulin growth factor and mitogen activated kinases pathway (MAPK) (Vogel et al., 2017; Valzania et al., 2018). However, another study found that live bacteria are not required for A. aegypti larvae and adult development (Correa et al., 2018). In this study, a mixture of liver powder, yeast extract and heat-killed bacteria rescued axenic A. aegypti growth from larvae to adults. This result implies that a diet with the appropriate concentration of nutrients but not containing live bacteria appears to be sufficient to rescue larval development. In Drosophila, larval microbiota is essential for scavenging amino acids (Yamada et al., 2015). So, these studies suggest that larval gut microbiota may provide some essential nutrition (such as amino acids and proteins) which rescue axenic larvae growth and molting.

Impact of Microbiota on Mosquito Refractoriness to Pathogens

Gut bacteria can influence the outcome of pathogen infections. Mosquito midgut microbiota induces peritrophic matrix formation and stimulate basal immune activity that protects the mosquito from pathogen infection (Barletta et al., 2017; Rodgers et al., 2017; Song et al., 2018; Yordanova et al., 2018). However, the effect of mosquito gut bacteria on parasite infection is complicated. A previous study showed that different strains of the genus Serratia can induce different outcomes on Plasmodium infections (Bando et al., 2013). Interestingly, a recent study reported that a Serratia marcescens strain isolated from a lab-adapted A. aegypti mosquito strain facilitates arboviral infection (Wu et al., 2019). Gloria-Soria studied more than 2,000 A. aegypti from 63 populations in 27 countries and did not find any natural infection by Wolbachia in A. aegypti (Gloria-Soria et al., 2018). Wolbachia has been applied to control arboviruses spread in A. aegypti mosquitoes. Moreira reported for the first time that Wolbachia infection reduces the ability of dengue and Chikungunya virus (CHIKV) to infect A. aegypti (Moreira et al., 2009). More recently Wolbachia was shown to also be a strong inhibitor of A. aegypti Zika virus infection (Dutra et al., 2016). Infection by the wMel strain of Wolbachia also can significantly reduce CHIKV and Yellow Fever virus (YFV) infection and dissemination rate (van den Hurk et al., 2012). However, a Wolbachia strain was reported to enhance vertical densovirus transmission by Culex pipiens (Altinli et al., 2018; King et al., 2018).

Using Microbiota for Mosquito Population Reduction

Chemical insecticides have long been used for mosquito population control. However, a major problem is the development of insecticide resistance. Also, insecticides may have adverse effects, such as non-target killing and environmental disturbance. In contrast, use of the mosquito microbiota for population control minimizes the problem of resistance and show minimal negative effects to the environment. The best studied bacteria belong to the Wolbachia genus. Intracellular bacteria Wolbachia can infect approximately 2/3 of insect species. Wolbachia can vertical spread through the female germline to regulate insect reproduction. Cytoplasmic incompatibility (CI) is the main feature caused by Wolbachia in insects. when the uninfected females mate with Wolbachia-infected males, and lay eggs which cannot develop to larvae; however, if both of female and male parents are infected, embryos develop normally (Jiggins, 2017). Mosquito population reduction is achieved by releasing Wolbachia-infected male mosquitoes in the field. The understanding of the molecular bases for CI has long been an enigma. Recent studies showed that the Wolbachia deubiquitylating (DUB) enzymes cidA and cidB contribute to CI of mosquito zygotes (Beckmann et al., 2017). Wolbachia pipientis Type IV Effector WD0830 also plays an important role in CI (Sheehan et al., 2016). The Wolbachia genome encodes more than 20 ankyrin-repeat proteins, which may contribute to mosquito male offspring killing. Moreover, infection with some Wolbachia strains can shorten A. aegypti life-span (McMeniman et al., 2009). Harumoto and Lemaitre (2018) identified a toxin produced by the endosymbiont Spiroplasma poulsonii that selectively kills male Drosophila offspring. Recently, Zheng released Wolbachia infected Aedes albopictus to reduce mosquito population by offspring CI, and successfully reduce mosquito 88.7–96.6% biting in two isolated riverine islands in Guangzhou, China (Zheng et al., 2019).

Exploitation of Microbiota to Combat Mosquito-Borne Diseases

Mosquito microbiota shows much potential to combat mosquito-borne diseases by rendering mosquito refractory to arthropod-borne human pathogens. For this purpose, the ideal microbe should have the following characteristics: easy genetic manipulation, efficient colonization of mosquitoes, be able to spread into mosquito populations (vertical and horizontal transmission), and effectively inhibit pathogen development in mosquitoes (Wang and Jacobs-Lorena, 2013; Wang et al., 2017).

The use of symbiotic bacteria to reduce the mosquito vectorial competence has gained increasing interest as an alternative approach toward disease control. This is based on two facts: (1) in initial stages of infection, the commensal microbiota and mosquito-borne pathogens share the same midgut compartment; (2) midgut microbiota proliferate dramatically after a mosquito blood meal, resulting in a corresponding increase of effector molecules secreted by the bacteria (Wang and Jacobs-Lorena, 2013).

Several reports have shown that the midgut microbiota can affect the infection of malaria parasite in its host mosquitoes (Pumpuni et al., 1993; Gendrin and Christophides, 2013; Wang et al., 2017). Some mosquito gut bacteria including S. marcescens, Acinetobacter sp. inhibit malaria parasite infection in mosquitoes (Cirimotich et al., 2011; Wang et al., 2017). However, mechanisms by which specific gut bacteria negatively impact malaria parasite development in the mosquito is largely unknown.

To exploit gut symbionts in the control of vector-borne disease transmission, genetic engineering has been used to modify certain symbiotic bacteria to produce anti-pathogen effector molecules (paratransgenesis) without affecting the fitness of the host vectors. In 1997, Rhodnius prolixus engineered with a gene encoding cecropin A, a peptide lethal to the parasite Trypanosoma cruzi, was introduced into the R. prolixus vector to control transmission of T. cruzi (Durvasula et al., 1997). The mosquito symbiotic bacterium Pantoea agglomerans was engineered to express anti-malaria effectors to interfere with malaria parasite development in mosquitoes (Wang et al., 2012). Recently, a new bacterium strain (AS1) of the genus Serratia isolated from the Anopheles ovary, was shown to stably colonize the mosquito midgut and reproductive organs. Serratia AS1 is transmitted vertically from the female to offspring and horizontally from male to female during mating, and spreads rapidly into mosquito populations. Moreover, Serratia AS1 can be engineered to express anti-malaria genes and mosquitoes that carry these bacteria are substantially refractory to the human malaria parasite Plasmodium falciparum. Thus, Serratia AS1 provides a powerful tool for driving mosquito refractoriness to Plasmodium infection (Wang et al., 2017). Another symbiotic bacterium Asaia can also colonize the mosquito midgut and reproductive organs (Favia et al., 2007). Recently, Asaia was also modified to express anti-malaria effectors and the engineered strains inhibit the development of malaria parasite (Shane et al., 2018). Reveillaud reported that Wolbachia from four wild Culex pipiens mosquitoes carry a plasmid (pWCP), opening the possibility of future paratransgenesis utilizing Wolbachia (Reveillaud et al., 2019).

Concerns Relating to Potential Release of Genetically Modified Symbionts

While the feasibility of using paratransgenesis to contain the spread of malaria was demonstrated with laboratory experiments, translation of these findings to field application will need to overcome major regulatory barriers, as it involves the release of genetically modified (GM) organisms in nature. A basic requirement for the release of GM organisms is that benefits considerably outweigh the risks (Durvasula et al., 1997). Among issues that need to be considered is horizontal gene transfer (HGT). For mosquitoes, no study has been performed to evaluate potential transgene dispersion via HGT. For R. prolixus, a theoretical model was designed to predict HGT from a GM bacteria Rhodococcus rhodnii to a closely related bacterium, Gordona rubropertinctus, and predicted HGT frequency is less than 1.14 × 10–16 per 100,000 bacterial generations (Matthews et al., 2011).

Concluding Remarks

The mosquito microbiota is acquired from the environment, and its composition is highly dynamic, varying depending on species, nutrition, development stage, and geography. Microbiota mostly colonize the midgut and rarely salivary glands and reproductive organs. The mosquito microbiota plays important roles in host nutrition, digestion, mating, sexual reproduction, development, immune functions and refractoriness to pathogens. Microbiota, GM or not, have been proposed for mosquito population control and combating mosquito-borne diseases. The introduction of GM symbionts engineered to produce anti-pathogen molecules into mosquitoes in the field shows much promise, but this can happen only after regulatory and public concerns are overcome.

A number of scientific questions remain to be addressed. First, many commensal bacteria may not always stop pathogen development in the mosquito. For example, Serratia inhibits malaria parasite infection of mosquitoes (Gonzalez-Ceron et al., 2003; Bando et al., 2013; Wang et al., 2017), while it promotes dengue virus infection of a culicine mosquito (Wu et al., 2019); A Wolbachia species reduces arbovirus infection of A. aegypti mosquitoes (Moreira et al., 2009; van den Hurk et al., 2012; Dutra et al., 2016) whereas another species enhances vertical densovirus transmission by Culex pipiens (Altinli et al., 2018; King et al., 2018). These apparently contradictory observations will only be clarified when the mechanisms underlying the observed effects are understood. Second, except for Wolbachia, no naturally occurring symbiont that can both inhibit pathogen infection and spread through mosquito populations has been identified. Wolbachia are effective in blocking viral transmission by A. aegypti but not to control transmission of the malaria parasite by anopheline mosquitoes. Identification of a naturally occurring bacterium that can inhibit Plasmodium transmission and spread through mosquito populations is an important future goal. Thirdly, the identification of effector proteins that specifically inhibit transmission of viruses such as dengue, zika, yellow fever and Chikungunya, and are harmless to the host vector, would allow implementation of disease control via paratransgenesis and mosquito transgenesis. Lastly, laboratory experimentation has demonstrated the high promise of paratransgenesis to fight mosquito-borne disease and a high priority should be given to address regulatory, ethical, and public acceptance issues.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Funding

This work was supported by NIH grant R01AI031478, grants from the National Natural Science Foundation of China (grants 31830086, 31772534, 31830086, and 31472044), the National Key R&D Program of China (2017YFD0200400 and 2018YFA0900502), and the Strategic Priority Research Program of Chinese Academy of Sciences (grant XDB11010500) and the Bloomberg Philanthropies.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

Altinli, M., Soms, J., Ravallec, M., Justy, F., Bonneau, M., Weill, M., et al. (2018). Sharing cells with Wolbachia: the transovarian vertical transmission of Culex pipiens densovirus. Environ. Microbiol. 21, 3284–3298. doi: 10.1111/1462-2920.14511

PubMed Abstract | CrossRef Full Text | Google Scholar

Bando, H., Okado, K., Guelbeogo, W. M., Badolo, A., Aonuma, H., Nelson, B., et al. (2013). Intra-specific diversity of Serratia marcescens in Anopheles mosquito midgut defines Plasmodium transmission capacity. Sci. Rep. 3:1641. doi: 10.1038/srep01641

PubMed Abstract | CrossRef Full Text | Google Scholar

Barletta, A. B., Nascimento-Silva, M. C., Talyuli, O. A., Oliveira, J. H., Pereira, L. O., Oliveira, P. L., et al. (2017). Microbiota activates IMD pathway and limits Sindbis infection in Aedes aegypti. Parasit. Vectors 10:103. doi: 10.1186/s13071-017-2040-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Bascunan, P., Nino-Garcia, J. P., Galeano-Castaneda, Y., Serre, D., and Correa, M. M. (2018). Factors shaping the gut bacterial community assembly in two main Colombian malaria vectors. Microbiome 6:148. doi: 10.1186/s40168-018-0528-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Beckmann, J. F., Ronau, J. A., and Hochstrasser, M. (2017). A Wolbachia deubiquitylating enzyme induces cytoplasmic incompatibility. Nat. Microbiol. 2:17007. doi: 10.1038/nmicrobiol.2017.7

PubMed Abstract | CrossRef Full Text | Google Scholar

Belda, E., Coulibaly, B., Fofana, A., Beavogui, A. H., Traore, S. F., Gohl, D. M., et al. (2017). Preferential suppression of Anopheles gambiae host sequences allows detection of the mosquito eukaryotic microbiome. Sci. Rep. 7:3241. doi: 10.1038/s41598-017-03487-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Caraballo, H., and King, K. (2014). Emergency department management of mosquito-borne illness: malaria, dengue, and West Nile virus. Emerg. Med. Pract. 16, 1–23.

PubMed Abstract | Google Scholar

Cheeseman, I. H., Miller, B. A., Nair, S., Nkhoma, S., Tan, A., Tan, J. C., et al. (2012). A major genome region underlying artemisinin resistance in malaria. Science 336, 79–82. doi: 10.1126/science.1215966

PubMed Abstract | CrossRef Full Text | Google Scholar

Cirimotich, C. M., Dong, Y., Clayton, A. M., Sandiford, S. L., Souza-Neto, J. A., Mulenga, M., et al. (2011). Natural microbe-mediated refractoriness to Plasmodium infection in Anopheles gambiae. Science 332, 855–858. doi: 10.1126/science.1201618

PubMed Abstract | CrossRef Full Text | Google Scholar

Coon, K. L., Brown, M. R., and Strand, M. R. (2016). Gut bacteria differentially affect egg production in the anautogenous mosquito Aedes aegypti and facultatively autogenous mosquito Aedes atropalpus (Diptera: Culicidae). Parasit. Vectors 9, 1–12. doi: 10.1186/s13071-016-1660-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Coon, K. L., Valzania, L., McKinney, D. A., Vogel, K. J., Brown, M. R., and Strand, M. R. (2017). Bacteria-mediated hypoxia functions as a signal for mosquito development. Proc. Natl. Acad. Sci. U.S.A. 114, E5362–E5369. doi: 10.1073/pnas.1702983114

PubMed Abstract | CrossRef Full Text | Google Scholar

Coon, K. L., Vogel, K. J., Brown, M. R., and Strand, M. R. (2014). Mosquitoes rely on their gut microbiota for development. Mol. Ecol. 23, 2727–2739. doi: 10.1111/mec.12771

PubMed Abstract | CrossRef Full Text | Google Scholar

Correa, M. A., Matusovsky, B., Brackney, D. E., and Steven, B. (2018). Generation of axenic Aedes aegypti demonstrate live bacteria are not required for mosquito development. Nat. Commun. 9:4464. doi: 10.1038/s41467-018-07014-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Damiani, C., Ricci, I., Crotti, E., Rossi, P., Rizzi, A., Scuppa, P., et al. (2010). Mosquito-bacteria symbiosis: the case of Anopheles gambiae and Asaia. Microb. Ecol. 60, 644–654. doi: 10.1007/s00248-010-9704-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Dong, Y., Manfredini, F., and Dimopoulos, G. (2009). Implication of the mosquito midgut microbiota in the defense against malaria parasites. PLoS Pathog. 5:e1000423. doi: 10.1371/journal.ppat.1000423

PubMed Abstract | CrossRef Full Text | Google Scholar

Douas, A. E. (2011). Lessons from studying insect symbioses. Cell Host Microbe 10, 359–367. doi: 10.1016/j.chom.2011.09.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Douglas, A. E. (2014). The molecular basis of bacterial-insect symbiosis. J. Mol. Biol. 426, 3830–3837. doi: 10.1016/j.jmb.2014.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Duguma, D., Hall, M. W., Smartt, C. T., Debboun, M., and Neufeld, J. D. (2019). Microbiota variations in Culex nigripalpus disease vector mosquito of West Nile virus and Saint Louis encephalitis from different geographic origins. PeerJ 6:e6168. doi: 10.7717/peerj.6168

PubMed Abstract | CrossRef Full Text | Google Scholar

Durvasula, R. V., Gumbs, A., Panackal, A., Kruglov, O., Aksoy, S., Merrifield, R. B., et al. (1997). Prevention of insect-borne disease: an approach using transgenic symbiotic bacteria. Proc. Natl. Acad. Sci. U.S.A. 94, 3274–3278. doi: 10.1073/pnas.94.7.3274

PubMed Abstract | CrossRef Full Text | Google Scholar

Dutra, H. L., Rocha, M. N., Dias, F. B., Mansur, S. B., Caragata, E. P., and Moreira, L. A. (2016). Wolbachia blocks currently circulating Zika virus Isolates in Brazilian Aedes aegypti mosquitoes. Cell Host Microbe 19, 771–774. doi: 10.1016/j.chom.2016.04.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Favia, G., Ricci, I., Damiani, C., Raddadi, N., Crotti, E., Marzorati, M., et al. (2007). Bacteria of the genus Asaia stably associate with Anopheles stephensi, an Asian malarial mosquito vector. Proc. Natl. Acad. Sci. U.S.A. 104, 9047–9051. doi: 10.1073/pnas.0610451104

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferguson, N. M. (2018). Challenges and opportunities in controlling mosquito-borne infections. Nature 559, 490–497. doi: 10.1038/s41586-018-0318-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Fraser, J. E., De Bruyne, J. T., Iturbe-Ormaetxe, I., Stepnell, J., Burns, R. L., Flores, H. A., et al. (2017). Novel Wolbachia-transinfected Aedes aegypti mosquitoes possess diverse fitness and vector competence phenotypes. PLoS Pathog. 13:e1006751. doi: 10.1371/journal.ppat.1006751

PubMed Abstract | CrossRef Full Text | Google Scholar

Gaio Ade, O., Gusmao, D. S., Santos, A. V., Berbert-Molina, M. A., Pimenta, P. F., and Lemos, F. J. (2011). Contribution of midgut bacteria to blood digestion and egg production in Aedes aegypti (diptera: culicidae) (L.). Parasit. Vectors 4:105. doi: 10.1186/1756-3305-4-105

PubMed Abstract | CrossRef Full Text | Google Scholar

Gendrin, M., and Christophides, G. K. (2013). “The Anopheles mosquito microbiota and their impact on pathogen transmission,” in Anopheles Mosquitoes-New Insights into Malaria Vectors, ed. S. Manguin (London: IntechOpen).

Google Scholar

Gloria-Soria, A., Chiodo, T. G., and Powell, J. R. (2018). Lack of evidence for natural Wolbachia infections in Aedes aegypti (Diptera: Culicidae). J. Med. Entomol. 55, 1354–1356. doi: 10.1093/jme/tjy084

PubMed Abstract | CrossRef Full Text | Google Scholar

Gonzalez-Ceron, L., Santillan, F., Rodriguez, M. H., Mendez, D., and Hernandez-Avila, J. E. (2003). Bacteria in midguts of field-collected Anopheles albimanus block Plasmodium vivax sporogonic development. J. Med. Entomol. 40, 371–374. doi: 10.1603/0022-2585-40.3.371

PubMed Abstract | CrossRef Full Text | Google Scholar

Harumoto, T., and Lemaitre, B. (2018). Male-killing toxin in a bacterial symbiont of Drosophila. Nature 557, 252–255. doi: 10.1038/s41586-018-0086-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Hughes, G. L., Dodson, B. L., Johnson, R. M., Murdock, C. C., Tsujimoto, H., Suzuki, Y., et al. (2014). Native microbiome impedes vertical transmission of Wolbachia in Anopheles mosquitoes. Proc. Natl. Acad. Sci. U.S.A. 111, 12498–12503. doi: 10.1073/pnas.1408888111

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiggins, F. M. (2017). The spread of Wolbachia through mosquito populations. PLoS Biol. 15:e2002780. doi: 10.1371/journal.pbio.2002780

PubMed Abstract | CrossRef Full Text | Google Scholar

Jupatanakul, N., Sim, S., and Dimopoulos, G. (2014). The insect microbiome modulates vector competence for arboviruses. Viruses 6, 4294–4313. doi: 10.3390/v6114294

PubMed Abstract | CrossRef Full Text | Google Scholar

King, J. G., Souto-Maior, C., Sartori, L. M., Maciel-de-Freitas, R., and Gomes, M. G. M. (2018). Variation in Wolbachia effects on Aedes mosquitoes as a determinant of invasiveness and vectorial capacity. Nat. Commun. 9:1483. doi: 10.1038/s41467-018-03981-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Krajacich, B. J., Huestis, D. L., Dao, A., Yaro, A. S., Diallo, M., Krishna, A., et al. (2018). Investigation of the seasonal microbiome of Anopheles coluzzii mosquitoes in Mali. PLoS One 13:e0194899. doi: 10.1371/journal.pone.0194899

PubMed Abstract | CrossRef Full Text | Google Scholar

Linenberg, I., Christophides, G. K., and Gendrin, M. (2016). Larval diet affects mosquito development and permissiveness to Plasmodium infection. Sci. Rep. 6:38230. doi: 10.1038/srep38230

PubMed Abstract | CrossRef Full Text | Google Scholar

Matthews, S., Rao, V. S., and Durvasula, R. V. (2011). Modeling horizontal gene transfer (HGT) in the gut of the Chagas disease vector Rhodnius prolixus. Parasit. Vectors 4:77. doi: 10.1186/1756-3305-4-77

PubMed Abstract | CrossRef Full Text | Google Scholar

McMeniman, C. J., Lane, R. V., Cass, B. N., Fong, A. W., Sidhu, M., Wang, Y. F., et al. (2009). Stable introduction of a life-shortening Wolbachia infection into the mosquito Aedes aegypti. Science 323, 141–144. doi: 10.1126/science.1165326

PubMed Abstract | CrossRef Full Text | Google Scholar

Minard, G., Tran Van, V., Tran, F. H., Melaun, C., Klimpel, S., Koch, L. K., et al. (2017). Identification of sympatric cryptic species of Aedes albopictus subgroup in Vietnam: new perspectives in phylosymbiosis of insect vector. Parasit. Vectors 10:276. doi: 10.1186/s13071-017-2202-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Mitraka, E., Stathopoulos, S., Siden-Kiamos, I., Christophides, G. K., and Louis, C. (2013). Asaia accelerates larval development of Anopheles gambiae. Pathog. Glob. Health 107, 305–311. doi: 10.1179/2047773213Y.0000000106

PubMed Abstract | CrossRef Full Text | Google Scholar

Moreira, L. A., Iturbe-Ormaetxe, I., Jeffery, J. A., Lu, G., Pyke, A. T., Hedges, L. M., et al. (2009). A Wolbachia symbiont in Aedes aegypti limits infection with dengue, Chikungunya, and Plasmodium. Cell 139, 1268–1278. doi: 10.1016/j.cell.2009.11.042

PubMed Abstract | CrossRef Full Text | Google Scholar

Muturi, E. J., Lagos-Kutz, D., Dunlap, C., Ramirez, J. L., Rooney, A. P., Hartman, G. L., et al. (2018). Mosquito microbiota cluster by host sampling location. Parasit. Vectors 11:468. doi: 10.1186/s13071-018-3036-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Novakova, E., Woodhams, D. C., Rodriguez-Ruano, S. M., Brucker, R. M., Leff, J. W., Maharaj, A., et al. (2017). Mosquito microbiome dynamics, a background for prevalence and seasonality of West Nile virus. Front. Microbiol. 8:526. doi: 10.3389/fmicb.2017.00526

PubMed Abstract | CrossRef Full Text | Google Scholar

Pidiyar, V. J., Jangid, K., Patole, M. S., and Shouche, Y. S. (2004). Studies on cultured and uncultured microbiota of wild Culex quinquefasciatus mosquito midgut based on 16s ribosomal RNA gene analysis. Am. J. Trop. Med. Hyg. 70, 597–603. doi: 10.4269/ajtmh.2004.70.597

PubMed Abstract | CrossRef Full Text | Google Scholar

Pumpuni, C. B., Beier, M. S., Nataro, J. P., Guers, L. D., and Davis, J. R. (1993). Plasmodium falciparum: inhibition of sporogonic development in Anopheles stephensi by gram-negative bacteria. Exp. Parasitol. 77, 195–199. doi: 10.1006/expr.1993.1076

PubMed Abstract | CrossRef Full Text | Google Scholar

Ranson, H., and Lissenden, N. (2016). Insecticide resistance in African Anopheles mosquitoes: a worsening situation that needs urgent action to maintain malaria control. Trends Parasitol. 32, 187–196. doi: 10.1016/j.pt.2015.11.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Reveillaud, J., Bordenstein, S. R., Cruaud, C., Shaiber, A., Esen, O. C., Weill, M., et al. (2019). Author correction: the Wolbachia mobilome in Culex pipiens includes a putative plasmid. Nat. Commun. 10:3153. doi: 10.1038/s41467-019-11234-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodgers, F. H., Gendrin, M., Wyer, C. A. S., and Christophides, G. K. (2017). Microbiota-induced peritrophic matrix regulates midgut homeostasis and prevents systemic infection of malaria vector mosquitoes. PLoS Pathog. 13:e1006391. doi: 10.1371/journal.ppat.1006391

PubMed Abstract | CrossRef Full Text | Google Scholar

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

Google Scholar

Rosenberg, R., Lindsey, N. P., Fischer, M., Gregory, C. J., Hinckley, A. F., Mead, P. S., et al. (2018). Vital signs: trends in reported vectorborne disease cases – United States and Territories, 2004-2016. MMWR Morb. Mortal. Wkly. Rep. 67, 496–501. doi: 10.15585/mmwr.mm6717e1

PubMed Abstract | CrossRef Full Text | Google Scholar

Scolari, F., Casiraghi, M., and Bonizzoni, M. (2019). Aedes spp. and their microbiota: a review. Front. Microbiol. 10:2036. doi: 10.3389/fmicb.2019.02036

PubMed Abstract | CrossRef Full Text | Google Scholar

Shane, J. L., Grogan, C. L., Cwalina, C., and Lampe, D. J. (2018). Blood meal-induced inhibition of vector-borne disease by transgenic microbiota. Nat. Commun. 9:4127. doi: 10.1038/s41467-018-06580-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Sheehan, K. B., Martin, M., Lesser, C. F., Isberg, R. R., and Newton, I. L. (2016). Identification and characterization of a candidate Wolbachia pipientis Type IV effector that interacts with the actin cytoskeleton. MBio 7:e00622-16. doi: 10.1128/mBio.00622-16

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, X., Wang, M., Dong, L., Zhu, H., and Wang, J. (2018). PGRP-LD mediates A. stephensi vector competency by regulating homeostasis of microbiota-induced peritrophic matrix synthesis. PLoS Pathog. 14:e1006899. doi: 10.1371/journal.ppat.1006899

PubMed Abstract | CrossRef Full Text | Google Scholar

Strand, M. R. (2018). Composition and functional roles of the gut microbiota in mosquitoes. Curr. Opin. Insect. Sci. 28, 59–65. doi: 10.1016/j.cois.2018.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Tchioffo, M. T., Boissiere, A., Abate, L., Nsango, S. E., Bayibeki, A. N., Awono-Ambene, P. H., et al. (2015). Dynamics of bacterial community composition in the malaria mosquito’s epithelia. Front. Microbiol. 6:1500. doi: 10.3389/fmicb.2015.01500

PubMed Abstract | CrossRef Full Text | Google Scholar

Telang, A., Skinner, J., Nemitz, R. Z., and McClure, A. M. (2018). Metagenome and culture-based methods reveal candidate bacterial mutualists in the southern house mosquito (Diptera: Culicidae). J. Med. Entomol. 55, 1170–1181. doi: 10.1093/jme/tjy056

PubMed Abstract | CrossRef Full Text | Google Scholar

Thongsripong, P., Chandler, J. A., Green, A. B., Kittayapong, P., Wilcox, B. A., Kapan, D. D., et al. (2018). Mosquito vector-associated microbiota: metabarcoding bacteria and eukaryotic symbionts across habitat types in Thailand endemic for dengue and other arthropod-borne diseases. Ecol. Evol. 8, 1352–1368. doi: 10.1002/ece3.3676

PubMed Abstract | CrossRef Full Text | Google Scholar

Valzania, L., Coon, K. L., Vogel, K. J., Brown, M. R., and Strand, M. R. (2018). Hypoxia-induced transcription factor signaling is essential for larval growth of the mosquito Aedes aegypti. Proc. Natl. Acad. Sci. U.S.A. 115, 457–465. doi: 10.1073/pnas.1719063115

PubMed Abstract | CrossRef Full Text | Google Scholar

van den Hurk, A. F., Hall-Mendelin, S., Pyke, A. T., Frentiu, F. D., McElroy, K., Day, A., et al. (2012). Impact of Wolbachia on infection with chikungunya and yellow fever viruses in the mosquito vector Aedes aegypti. PLoS Negl. Trop. Dis. 6:e1892. doi: 10.1371/journal.pntd.0001892

PubMed Abstract | CrossRef Full Text | Google Scholar

Vogel, K. J., Valzania, L., Coon, K. L., Brown, M. R., and Strand, M. R. (2017). Transcriptome sequencing reveals large-scale changes in axenic Aedes aegypti larvae. PLoS Negl. Trop. Dis. 11:e0005273. doi: 10.1371/journal.pntd.0005273

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S., Dos-Santos, A. L. A., Huang, W., Liu, K. C., Oshaghi, M. A., Wei, G., et al. (2017). Driving mosquito refractoriness to Plasmodium falciparum with engineered symbiotic bacteria. Science 357, 1399–1402. doi: 10.1126/science.aan5478

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S., Ghosh, A. K., Bongio, N., Stebbings, K. A., Lampe, D. J., and Jacobs-Lorena, M. (2012). Fighting malaria with engineered symbiotic bacteria from vector mosquitoes. Proc. Natl. Acad. Sci. U.S.A. 109, 12734–12739. doi: 10.1073/pnas.1204158109

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S., and Jacobs-Lorena, M. (2013). Genetic approaches to interfere with malaria transmission by vector mosquitoes. Trends Biotechnol. 31, 185–193. doi: 10.1016/j.tibtech.2013.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

WHO (2016). Global Health Impacts of Vector-Borne Diseases: Workshop Summary. Washington, DC: National Academies Press.

Google Scholar

Wu, P., Sun, P., Nie, K., Zhu, Y., Shi, M., Xiao, C., et al. (2019). A gut commensal bacterium promotes mosquito permissiveness to arboviruses. Cell Host Microbe 25, 101.e5–112.e5. doi: 10.1016/j.chom.2018.11.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamada, R., Deshpande, S. A., Bruce, K. D., Mak, E. M., and Ja, W. W. (2015). Microbes promote amino acid harvest to rescue undernutrition in Drosophila. Cell Rep. 10, 865–872. doi: 10.1016/j.celrep.2015.01.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Yordanova, I. A., Zakovic, S., Rausch, S., Costa, G., Levashina, E., and Hartmann, S. (2018). Micromanaging immunity in the murine host vs. the mosquito vector: microbiota-dependent immune responses to intestinal parasites. Front. Cell. Infect. Microbiol. 8:308. doi: 10.3389/fcimb.2018.00308

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, X., Zhang, D., Li, Y., Yang, C., Wu, Y., Liang, X., et al. (2019). Incompatible and sterile insect techniques combined eliminate mosquitoes. Nature 572, 56–61. doi: 10.1038/s41586-019-1407-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: insect microbiota, arboviruses, malaria, paratransgenesis, mosquito-pathogen interactions

Citation: Huang W, Wang S and Jacobs-Lorena M (2020) Use of Microbiota to Fight Mosquito-Borne Disease. Front. Genet. 11:196. doi: 10.3389/fgene.2020.00196

Received: 26 June 2019; Accepted: 19 February 2020;
Published: 10 March 2020.

Edited by:

Jayme A. Souza-Neto, São Paulo State University, Brazil

Reviewed by:

Claire Valiente Moro, Université Claude Bernard Lyon 1, France
Guido Favia, University of Camerino, Italy
Pedro L. Oliveira, Federal University of Rio de Janeiro, Brazil

Copyright © 2020 Huang, Wang and Jacobs-Lorena. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Marcelo Jacobs-Lorena, ljacob13@jhu.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.