The Role of Insect-Microbiota Associations in Vector Competence: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Mosquitoes transmit pathogens that cause human diseases such as malaria, dengue fever, chikungunya, yellow fever, Zika fever, and filariasis. Biotechnological approaches using microorganisms have a significant potential to control mosquito populations and reduce their vector competence, making them alternatives to synthetic insecticides. Ongoing research has identified many microorganisms that can be used effectively to control mosquito populations and disease transmission. However, the successful implementation of these newly proposed approaches requires a thorough understanding of the multipronged microorganism–mosquito–pathogen–environment interactions. Although much has been achieved in discovering new entomopathogenic microorganisms, antipathogen compounds, and their mechanisms of action, only a few have been turned into viable products for mosquito control. There is a discrepancy between the number of microorganisms with the potential for the development of new insecticides and/or antipathogen products and the actual available products, highlighting the need for investments in the intersection of basic research and biotechnology.

  • biotechnology
  • microorganisms
  • bacteria
  • fungi
  • vector control
  • mosquitoes

1. Introduction

Microorganisms constitute a large group of genetically diverse biological entities found in a wide range of terrestrial and aquatic habitats, playing crucial roles in the balance of ecosystems [1][2][3]. Advances in microbiology, molecular biology, and genomics enabled the biotechnological exploration of microbes, allowing the discovery and production of antibiotics [4][5], foods [6][7], alcoholic beverages [8], bioremediators [9][10], fertilizers [11], and biopesticides [12][13]. The microorganisms associated with mosquitoes have drawn special attention to their potential applications in public health (Figure 1) [14][15][16].
Figure 1. Microorganisms and their applications for controlling vector populations and disease transmission. Microorganisms are sources of molecules with insecticide (biopesticides) antipathogen (biopharmaceuticals) activities. Interactions of environmental and symbiotic fungi and bacteria with mosquitoes and their microbiota may affect mosquito and pathogen survival, having implications for vector control and disease transmission. Research that elucidates these interactions is crucial because it underpins the development of novel biotechnological products aimed at effective vector control and reducing disease transmission. Created with BioRender.com (accessed on 5 August 2023).
Associations between mosquitoes and their microbiota have gained significant attention in scientific research due to their impact on vector competence [17][18][19][20][21][22]. In the following, we discuss ways these associations can influence vector competence. Understanding these interactions is essential for developing effective vector-borne disease control strategies and reducing their impact on public health.

2. Symbiotic Bacteria and Their Potential against Infectious Agents

The mosquito microbiota influences host development, nutrition, reproduction, and immune responses to invading organisms [23][24][25][26]. While the composition of the mosquito microbiota is largely defined by the environment in which they live [27][28][29], resident bacteria can modulate the development and replication of parasites and viruses within their vectors [30][31][32][33][34][35][36][37]. Although this modulation can enhance or reduce the survival and replication of pathogens within mosquitoes, those mosquito–microbiota interactions that negatively affect pathogens offer possibilities to control arthropod-borne diseases.
For example, the Gram-negative bacteria, Escherichia coli H243, E. coli HB101, Pseudomonas aeruginosa, and Ewingella americana inhibit the formation of Plasmodium falciparum oocysts, in Anopheles stephensi (Liston, 1901) [38]. Enterobacter sp. (Esp_Z) isolated from the intestine of Anopheles gambiae (Giles, 1902) inhibited the development of malaria parasites when reintroduced into this same vector species [39][40]. The formation of oocysts of Plasmodium berghei was affected by the presence of Serratia marcescens-HB3 in An. stephensi [41]. In An. gambiae, Escherichia coli, S. marcescens, and Pseudomonas stutzeri reduced the prevalence and intensity of P. falciparum infection [42]. The Serratia Y1 strain exerts inhibitory activity on P. berghei ookinetes by activation of the Toll immune pathway in An. stephensi [43]. Serratia ureilytica (Su_YN1) produces an antimalarial lipase (AmLip) that inhibits the formation of P. falciparum oocysts in An. stephensi and An. gambiae [44]. Asaia SF2.1 also inhibits Plasmodium development in anophelines [45].
Virus replication in their vectors is also regulated by the mosquito microbiota. Bacteria of the genera Proteus, Paenibacillus, and Chromobacterium inhibited the replication of dengue virus serotype 2 (DENV-2) when administered to mosquitoes [46][47]. Some of the mechanisms by which symbiotic bacteria can hamper pathogen development have been elucidated and can be exploited to inhibit the spread of infectious agents by mosquitoes (Figure 2).
Figure 2. Biotechnological potential of mosquito symbiotic bacteria against infectious agents. (A), secretion of toxic substances that either kill or arrest the development and replication of viruses and parasites. (B), formation of physical barriers through large population accumulation or rearrangements of molecules secreted into the midgut lumen, preventing the passage of parasites to organs essential for their successful development. (C), activation of the mosquito immune system, which not only reduces the load of symbiotic bacteria but also leads to the elimination of invading parasites through the secretion of toxic molecules, preventing their propagation in the mosquito’s body. (D), competition with infectious agents for space and nutrients can have dire consequences for these pathogens as they must compete with a vastly larger population of symbiotic bacteria in the mosquito’s midgut lumen. This results in limited resources for the pathogens, ultimately leading to their decreased survival and replication within the mosquito. (E), paratransgenesis involves populating vector insects with genetically engineered symbiotic microorganisms that effectively hinder the development of parasites through synthesizing and secreting antipathogen molecules. Created with BioRender.com (accessed on 5 August 2023).

3. Wolbachia-Based Strategy for Controlling Mosquito-Borne Viruses: Mechanisms, Efficacy, and Implications

The wMel and wAlbB strains of Wolbachia pipientis, an intracellular bacterium, inhibit dengue, CHIKUNGUNYA, and Zika virus replication within mosquito cells [48][49][50][51][52]. However, another Wolbachia strain, wPip, does not inhibit virus infection in Ae. aegypti [53] and the mechanism by which Wolbachia interferes with virus replication has not been fully elucidated. Current hypotheses include competition between Wolbachia and the virus for physical space within mosquito cells and metabolite resources [54][55] and Wolbachia-induced modulation of the host’s immune system and immune priming. Immune priming entails sensitizing or preparing the mosquito’s immune system for a faster and more efficient response to a specific pathogen, such as a virus, upon subsequent exposure [56][57].
Despite the lack of a complete understanding of the mechanism or mechanisms involved in Wolbachia-associated modulation of viral suppression, the Wolbachia-carrying mosquito-based strategy has been deployed as a public health intervention to control dengue transmission (The World Mosquito Program https://www.worldmosquitoprogram.org/) (accessed on 15 February 2023). A randomized study carried out in the city of Yogyakarta, Indonesia, compared the areas where Ae. aegypti carrying Wolbachia was released with areas without Wolbachia. The results revealed a 77% lower incidence of dengue cases, in the Wolbachia-treated area [58]. Another study conducted in the city of Niterói, Rio de Janeiro, Brazil, reported a 69% reduction in dengue, 56% in chikungunya, and a 37% reduction in Zika incidence three years after the beginning of the release of Ae. aegypti with Wolbachia [59].
Although these results bring optimism regarding the use of Wolbachia for the control of dengue transmission, these bacteria can have variable effects on mosquito-borne viruses. For example, the Wolbachia strain wMel strongly blocked Mayaro virus (MAYV) infections in Ae. aegypti, but another strain, wAlbB, did not influence MAYV infection in this same vector. Aedes aegypti infected with wAlbB and wMel showed enhanced Sindbis virus infection rates [60]. The variable effects of Wolbachia on vector competence bring into question the safety of the current release of Wolbachia-infected mosquitoes. Furthermore, the potential impact of these bacteria on biodiversity has not been thoroughly investigated [61][62], and the risk of the emergence of DENV variants that escape virus-specific inhibition in Wolbachia-infected mosquitoes [63][64], underscores the importance of further research on interactions between Wolbachia, mosquitoes, viruses, and other organisms.

4. Symbiotic Microorganisms and Paratransgenesis

Paratransgenesis involves the colonization of vector insects with genetically engineered symbiotic microorganisms that are effective in inhibiting parasite development [65][66][67][68]. Ideal symbionts for effective paratransgenesis are easily manipulated genetically, colonize mosquitoes efficiently, spread into mosquito populations (vertical and horizontal transmission), and are efficient in inhibiting pathogen development in mosquitoes [69]. Proof-of-principle experiments performed primarily with bacteria demonstrated that genetically modified microorganisms expressing antipathogen molecules are capable of interfering with or blocking the development of malaria parasites in mosquitoes [70][71][72]. Among the mosquito symbiotic bacteria, strains of Asaia, Pantoea, Serratia, Pseudomonas, and Thorsellia have been evaluated as candidates for paratransgenesis [73][74][75][76][77].
The fungus Metarhizium anisopliae has been genetically transformed to express anti-Plasmodium proteins. Mosquitoes treated with transgenic M. anisopliae had 71–98% fewer sporozoites present in their salivary glands [78]. Scorpine, one of the molecules expressed by transgenic M. anisopliae, also affects negatively dengue virus replication, expanding the application of genetically transformed fungi to control arbovirus transmission [79].
Densovirus, a small DNA virus (4–6 kb) belonging to the Parvoviridae family (Densovirinae subfamily), infects arthropods such as mosquitoes and is maintained in natural populations through both horizontal and vertical transmission from infected adults to larvae. With one of the smallest known viral genomes, Densovirus serves as a valuable molecular tool. Its compact genome can be inserted into an infectious plasmid, which can then be used to express antiparasitic genes, both in cell cultures and in live mosquitoes [80][81][82][83].
The discovery of mosquito symbiotic bacteria [71][84][85][86], viruses [80][81][82][83], and fungi [87] is an active area of research. Advances toward deploying paratransgenesis as a tool for blocking pathogen transmission by mosquitoes also include the identification of antipathogen effector peptides and mechanisms of cellular secretion [66][71]. In bacteria, one efficient way of secreting effector molecules from the bacterial cytoplasm into the lumen of the mosquito intestine was engineered using Escherichia coli’s hemolysin-A secretion system. This process involves the export of proteins, such as hemolysin HlyA, through a specific mechanism that includes an export signal at the C-terminal end of the protein, along with the membrane proteins HlyB, HlyD, and the outer membrane protein TolC [88]. The use of bacteria for paratransgenesis is the most advanced alternative compared with viruses and fungi applications. However, concerns about the safety of releasing engineered bacteria into the environment and the potential unforeseen consequences still require attention when contemplating field tests for paratransgenesis.
Self-limiting paratransgenesis [69] has been suggested as an alternative for initial field trials. This approach proposes the utilization of transient expression of antipathogen compounds from a plasmid that is gradually lost, reverting bacteria to their original wild type. Risk assessment still needs to be carried out and laws and regulations need to be created and enacted before paratransgenesis can be tested in field conditions. However, the processes by which genetically modified microorganisms (GMs) can be spread in nature and how they should act to inhibit the development of target parasites in mosquitoes have already been envisaged. This is illustrated in Figure 3, which presents the paratransgenesis process as a multifaceted approach to combating mosquito-borne diseases.
Figure 3. Strategies for dissemination of GM microorganisms in the wild and their continual circulation among mosquitoes. (A) Male and female mosquitoes are fed in the laboratory with a sucrose solution containing the GM microorganisms and then released into the wild to mate with other wild mosquitoes. The spread of GM microorganisms can also occur through the provision of sucrose baits in the field enriched with GM microorganisms. This enables the GM microorganisms to be transmitted forward, allowing them to spread throughout the wild mosquito population and aiding in reducing vector-borne disease transmission. (B) Release of GM microorganisms into natural larval breeding sites. The GM microorganisms are ingested by the larvae and remain associated with them until adulthood. If mosquitoes become infected with a parasite that is a target of the effector molecules produced by the GM microorganisms, these molecules will interfere with the development of the target pathogen, thereby preventing its transmission. (C) Persistence of GM microorganisms in mosquitoes for generations through vertical and horizontal transmission. Vertical transmission occurs from parents to offspring, while horizontal transmission takes place between mosquitoes during mating or sharing of breeding sites. The presence of GM microorganisms can continue to impact mosquito populations for an extended period. Created with BioRender.com (accessed on 5 August 2023).

This entry is adapted from the peer-reviewed paper 10.3390/insects14090718

References

  1. Onen, O.; Aboh, A.A.; Mfam, A.N.; Akor, M.O.; Nweke, C.N.; Osuagwu, A.N. Microbial Diversity: Values and Roles in Ecosystems. Asian J. Biol. 2020, 9, 10–22.
  2. Rousk, J.; Bengtson, P. Microbial Regulation of Global Biogeochemical Cycles. Front. Microbiol. 2014, 5, 103.
  3. Rodríguez-Frías, F.; Quer, J.; Tabernero, D.; Cortese, M.F.; Garcia-Garcia, S.; Rando-Segura, A.; Pumarola, T. Microorganisms as Shapers of Human Civilization, from Pandemics to Even Our Genomes: Villains or Friends? A Historical Approach. Microorganisms 2021, 9, 2518.
  4. Raaijmakers, J.M.; Vlami, M.; De Souza, J.T. Antibiotic Production by Bacterial Biocontrol Agents. Antonie Leeuwenhoek 2002, 81, 537–547.
  5. Uchida, R.; Imasato, R.; Tomoda, H.; Ōmura, S. Yaequinolones, New Insecticidal Antibiotics Produced by Penicillium sp. FKI-2140. J. Antibiot. 2006, 59, 652–658.
  6. Bintsis, T. Lactic Acid Bacteria: Their Applications in Foods. J. Bacteriol. Mycol. 2018, 6, 89–94.
  7. Barzee, T.J.; Cao, L.; Pan, Z.; Zhang, R. Fungi for Future Foods. J. Future Foods 2021, 1, 25–37.
  8. Torres-Guardado, R.; Esteve-Zarzoso, B.; Reguant, C.; Bordons, A. Microbial Interactions in Alcoholic Beverages. Int. Microbiol. 2022, 25, 1–15.
  9. Sevak, P.I.; Pushkar, B.K.; Kapadne, P.N. Lead Pollution and Bacterial Bioremediation: A Review. Environ. Chem. Lett. 2021, 19, 4463–4488.
  10. Cowan, A.R.; Costanzo, C.M.; Benham, R.; Loveridge, E.J.; Moody, S.C. Fungal Bioremediation of Polyethylene: Challenges and Perspectives. J. Appl. Microbiol. 2022, 132, 78–89.
  11. Jiménez-Gómez, A.; García-Estévez, I.; Escribano-Bailón, M.T.; García-Fraile, P.; Rivas, R. Bacterial Fertilizers Based on Rhizobium laguerreae and Bacillus halotolerans Enhance Cichorium endivia L. Phenolic Compound and Mineral Contents and Plant Development. Foods 2021, 10, 424.
  12. Dunham, B. Microbial Pesticides: A Key Role in the Multinational Portfolio. New Ag Int. 2015, 32–36.
  13. Ruiu, L. Microbial Biopesticides in Agroecosystems. Agronomy 2018, 8, 235.
  14. Thongsripong, P.; Chandler, J.A.; Green, A.B.; Kittayapong, P.; Wilcox, B.A.; Kapan, D.D.; Bennett, S.N. Mosquito Vector-associated Microbiota: Metabarcoding Bacteria and Eukaryotic Symbionts across Habitat Types in Thailand Endemic for Dengue and Other Arthropod-borne Diseases. Ecol. Evol. 2018, 8, 1352–1368.
  15. da Silva Gonçalves, D.; Iturbe-Ormaetxe, I.; Martins-da-Silva, A.; Telleria, E.L.; Rocha, M.N.; Traub-Csekö, Y.M.; O’Neill, S.L.; Sant’Anna, M.R.V.; Moreira, L.A. Wolbachia Introduction into Lutzomyia longipalpis (Diptera: Psychodidae) Cell Lines and Its Effects on Immune-Related Gene Expression and Interaction with Leishmania infantum. Parasites Vectors 2019, 12, 33.
  16. Caragata, E.P.; Short, S.M. Vector Microbiota and Immunity: Modulating Arthropod Susceptibility to Vertebrate Pathogens. Curr. Opin. Insect Sci. 2022, 50, 100875.
  17. Dennison, N.J.; Jupatanakul, N.; Dimopoulos, G. The Mosquito Microbiota Influences Vector Competence for Human Pathogens. Curr. Opin. Insect Sci. 2014, 3, 6–13.
  18. Carlson, J.S.; Short, S.M.; Angleró-Rodríguez, Y.I.; Dimopoulos, G. Larval Exposure to Bacteria Modulates Arbovirus Infection and Immune Gene Expression in Adult Aedes aegypti. Dev. Comp. Immunol. 2020, 104, 103540.
  19. Gao, H.; Cui, C.; Wang, L.; Jacobs-Lorena, M.; Wang, S. Mosquito Microbiota and Implications for Disease Control. Trends Parasitol. 2020, 36, 98–111.
  20. Gabrieli, P.; Caccia, S.; Varotto-Boccazzi, I.; Arnoldi, I.; Barbieri, G.; Comandatore, F.; Epis, S. Mosquito Trilogy: Microbiota, Immunity and Pathogens, and Their Implications for the Control of Disease Transmission. Front. Microbiol. 2021, 12, 630438.
  21. Cansado-Utrilla, C.; Zhao, S.Y.; McCall, P.J.; Coon, K.L.; Hughes, G.L. The Microbiome and Mosquito Vectorial Capacity: Rich Potential for Discovery and Translation. Microbiome 2021, 9, 111.
  22. Wang, J.; Gao, L.; Aksoy, S. Microbiota in Disease-Transmitting Vectors. Nat. Rev. Microbiol. 2023, 21, 604–618.
  23. Douglas, A.E. Lessons from Studying Insect Symbioses. Cell Host Microbe 2011, 10, 359–367.
  24. Minard, G.; Mavingui, P.; Moro, C.V. Diversity and Function of Bacterial Microbiota in the Mosquito Holobiont. Parasites Vectors 2013, 6, 146.
  25. Kumar, A.; Srivastava, P.; Sirisena, P.; Dubey, S.K.; Kumar, R.; Shrinet, J.; Sunil, S. Mosquito Innate Immunity. Insects 2018, 9, 95.
  26. Ferreira, Q.R.; Lemos, F.F.B.; Moura, M.N.; de Nascimento, J.O.S.; Novaes, A.F.; Barcelos, I.S.; Fernandes, L.A.; de Amaral, L.S.B.; Barreto, F.K.; de Melo, F.F. Role of the Microbiome in Aedes spp. Vector Competence: What Do We Know? Viruses 2023, 15, 779.
  27. Saab, S.A.; Dohna, H.Z.; Nilsson, L.K.; Onorati, P.; Nakhleh, J.; Terenius, O.; Osta, M.A. The Environment and Species Affect Gut Bacteria Composition in Laboratory Co-Cultured Anopheles gambiae and Aedes albopictus Mosquitoes. Sci. Rep. 2020, 10, 3352.
  28. Mosquera, K.D.; Nilsson, L.K.J.; de Oliveira, M.R.; Rocha, E.M.; Marinotti, O.; Håkansson, S.; Tadei, W.P.; de Souza, A.Q.L.; Terenius, O. Comparative Assessment of the Bacterial Communities Associated with Anopheles darlingi Immature Stages and Their Breeding Sites in the Brazilian Amazon. Parasites Vectors 2023, 16, 156.
  29. Dos Santos, N.A.C.; de Carvalho, V.R.; Souza Neto, J.; Alonso, D.P.; Ribolla, P.E.M.; Medeiros, J.F.; da Araujo, M.S. Bacterial Microbiota from Lab-Reared and Field-Captured Anopheles darlingi Midgut and Salivary Gland. Microorganisms 2023, 11, 1145.
  30. Dong, Y.; Manfredini, F.; Dimopoulos, G. Implication of the Mosquito Midgut Microbiota in the Defense against Malaria Parasites. PLoS Pathog. 2009, 5, e1000423.
  31. Cirimotich, C.M.; Dong, Y.; Garver, L.S.; Sim, S.; Dimopoulos, G. Mosquito Immune Defenses against Plasmodium Infection. Dev. Comp. Immunol. 2010, 34, 387–395.
  32. Wang, Y.; Gilbreath III, T.M.; Kukutla, P.; Yan, G.; Xu, J. Dynamic Gut Microbiome across Life History of the Malaria Mosquito Anopheles gambiae in Kenya. PLoS ONE 2011, 6, e24767.
  33. Gendrin, M.; Christophides, G.K. The Anopheles Mosquito Microbiota and Their Impact on Pathogen Transmission. In Anopheles Mosquitoes-New Insights into Malaria Vectors; IntechOpen: Rijeka, Croatia, 2013; ISBN 953-51-1188-4.
  34. Ricci, I.; Valzano, M.; Ulissi, U.; Epis, S.; Cappelli, A.; Favia, G. Symbiotic Control of Mosquito Borne Disease. Pathog. Glob. Health 2012, 106, 380–385.
  35. Eappen, A.G.; Smith, R.C.; Jacobs-Lorena, M. Enterobacter-Activated Mosquito Immune Responses to Plasmodium Involve Activation of SRPN6 in Anopheles stephensi. PLoS ONE 2013, 8, e62937.
  36. Romoli, O.; Gendrin, M. The Tripartite Interactions between the Mosquito, Its Microbiota and Plasmodium. Parasites Vectors 2018, 11, 200.
  37. Shi, C.; Beller, L.; Wang, L.; Rosales Rosas, A.; De Coninck, L.; Héry, L.; Mousson, L.; Pagès, N.; Raes, J.; Delang, L. Bidirectional Interactions between Arboviruses and the Bacterial and Viral Microbiota in Aedes aegypti and Culex quinquefasciatus. MBio 2022, 13, e01021-22.
  38. Pumpuni, C.B.; Beier, M.S.; Nataro, J.P.; Guers, L.D.; Davis, J.R. Plasmodium falciparum: Inhibition of Sporogonic Development in Anopheles stephensi by Gram-Negative Bacteria. Exp. Parasitol. 1993, 77, 195–199.
  39. Cirimotich, C.M.; Dong, Y.; Clayton, A.M.; Sandiford, S.L.; Souza-Neto, J.A.; Mulenga, M.; Dimopoulos, G. Natural Microbe-Mediated Refractoriness to Plasmodium Infection in Anopheles gambiae. Science 2011, 332, 855–858.
  40. Dennison, N.J.; Saraiva, R.G.; Cirimotich, C.M.; Mlambo, G.; Mongodin, E.F.; Dimopoulos, G. Functional Genomic Analyses of Enterobacter, Anopheles and Plasmodium Reciprocal Interactions That Impact Vector Competence. Malar. J. 2016, 15, 425.
  41. Bando, H.; Okado, K.; Guelbeogo, W.M.; Badolo, A.; Aonuma, H.; Nelson, B.; Fukumoto, S.; Xuan, X.; Sagnon, N.; Kanuka, H. Intra-Specific Diversity of Serratia marcescens in Anopheles Mosquito Midgut Defines Plasmodium Transmission Capacity. Sci. Rep. 2013, 3, 1641.
  42. Tchioffo, M.T.; Boissiere, A.; Churcher, T.S.; Abate, L.; Gimonneau, G.; Nsango, S.E.; Awono-Ambene, P.H.; Christen, R.; Berry, A.; Morlais, I. Modulation of Malaria Infection in Anopheles gambiae Mosquitoes Exposed to Natural Midgut Bacteria. PLoS ONE 2013, 8, e81663.
  43. Bai, L.; Wang, L.; Vega-Rodríguez, J.; Wang, G.; Wang, S. A Gut Symbiotic Bacterium Serratia marcescens Renders Mosquito Resistance to Plasmodium Infection through Activation of Mosquito Immune Responses. Front. Microbiol. 2019, 10, 1580.
  44. Gao, H.; Bai, L.; Jiang, Y.; Huang, W.; Wang, L.; Li, S.; Zhu, G.; Wang, D.; Huang, Z.; Li, X. A Natural Symbiotic Bacterium Drives Mosquito Refractoriness to Plasmodium Infection via Secretion of an Antimalarial Lipase. Nat. Microbiol. 2021, 6, 806–817.
  45. Cappelli, A.; Damiani, C.; Mancini, M.V.; Valzano, M.; Rossi, P.; Serrao, A.; Ricci, I.; Favia, G. Asaia Activates Immune Genes in Mosquito Eliciting an Anti-Plasmodium Response: Implications in Malaria Control. Front. Genet. 2019, 10, 836.
  46. Ramirez, J.L.; Short, S.M.; Bahia, A.C.; Saraiva, R.G.; Dong, Y.; Kang, S.; Tripathi, A.; Mlambo, G.; Dimopoulos, G. Chromobacterium Csp_P Reduces Malaria and Dengue Infection in Vector Mosquitoes and Has Entomopathogenic and in Vitro Anti-Pathogen Activities. PLoS Pathog. 2014, 10, e1004398.
  47. Ramirez, J.L.; Souza-Neto, J.; Torres Cosme, R.; Rovira, J.; Ortiz, A.; Pascale, J.M.; Dimopoulos, G. Reciprocal Tripartite Interactions between the Aedes aegypti Midgut Microbiota, Innate Immune System and Dengue Virus Influences Vector Competence. PLoS Neglected Trop. Dis. 2012, 6, e1561.
  48. Moreira, L.A.; Iturbe-Ormaetxe, I.; Jeffery, J.A.; Lu, G.; Pyke, A.T.; Hedges, L.M.; Rocha, B.C.; Hall-Mendelin, S.; Day, A.; Riegler, M. A Wolbachia Symbiont in Aedes aegypti Limits Infection with Dengue, Chikungunya, and Plasmodium. Cell 2009, 139, 1268–1278.
  49. Walker, T.; Johnson, P.H.; Moreira, L.A.; Iturbe-Ormaetxe, I.; Frentiu, F.D.; McMeniman, C.J.; Leong, Y.S.; Dong, Y.; Axford, J.; Kriesner, P. The w Mel Wolbachia Strain Blocks Dengue and Invades Caged Aedes aegypti Populations. Nature 2011, 476, 450–453.
  50. 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. 2016, 6, 28792.
  51. Ryan, P.A.; Turley, A.P.; Wilson, G.; Hurst, T.P.; Retzki, K.; Brown-Kenyon, J.; Hodgson, L.; Kenny, N.; Cook, H.; Montgomery, B.L. Establishment of WMel Wolbachia in Aedes aegypti Mosquitoes and Reduction of Local Dengue Transmission in Cairns and Surrounding Locations in Northern Queensland, Australia. Gates Open Res. 2019, 3, 1547.
  52. Nazni, W.A.; Hoffmann, A.A.; NoorAfizah, A.; Cheong, Y.L.; Mancini, M.V.; Golding, N.; Kamarul, G.M.; Arif, M.A.; Thohir, H.; NurSyamimi, H. Establishment of Wolbachia Strain WAlbB in Malaysian Populations of Aedes aegypti for Dengue Control. Curr. Biol. 2019, 29, 4241–4248.e5.
  53. Fraser, J.E.; O’Donnell, T.B.; Duyvestyn, J.M.; O’Neill, S.L.; Simmons, C.P.; Flores, H.A. Novel Phenotype of Wolbachia Strain w Pip in Aedes aegypti Challenges Assumptions on Mechanisms of Wolbachia-Mediated Dengue Virus Inhibition. PLoS Pathog. 2020, 16, e1008410.
  54. Caragata, E.P.; Rancès, E.; Hedges, L.M.; Gofton, A.W.; Johnson, K.N.; O’Neill, S.L.; McGraw, E.A. Dietary Cholesterol Modulates Pathogen Blocking by Wolbachia. PLoS Pathog. 2013, 9, e1003459.
  55. Geoghegan, V.; Stainton, K.; Rainey, S.M.; Ant, T.H.; Dowle, A.A.; Larson, T.; Hester, S.; Charles, P.D.; Thomas, B.; Sinkins, S.P. Perturbed Cholesterol and Vesicular Trafficking Associated with Dengue Blocking in Wolbachia-Infected Aedes aegypti Cells. Nat. Commun. 2017, 8, 526.
  56. Kambris, Z.; Cook, P.E.; Phuc, H.K.; Sinkins, S.P. Immune Activation by Life-Shortening Wolbachia and Reduced Filarial Competence in Mosquitoes. Science 2009, 326, 134–136.
  57. Rancès, E.; Ye, Y.H.; Woolfit, M.; McGraw, E.A.; O’Neill, S.L. The Relative Importance of Innate Immune Priming in Wolbachia-Mediated Dengue Interference. PLoS Pathog. 2012, 8, e1002548.
  58. Utarini, A.; Indriani, C.; Ahmad, R.A.; Tantowijoyo, W.; Arguni, E.; Ansari, M.R.; Supriyati, E.; Wardana, D.S.; Meitika, Y.; Ernesia, I. Efficacy of Wolbachia-Infected Mosquito Deployments for the Control of Dengue. N. Engl. J. Med. 2021, 384, 2177–2186.
  59. Pinto, S.B.; Riback, T.I.; Sylvestre, G.; Costa, G.; Peixoto, J.; Dias, F.B.; Tanamas, S.K.; Simmons, C.P.; Dufault, S.M.; Ryan, P.A. Effectiveness of Wolbachia-Infected Mosquito Deployments in Reducing the Incidence of Dengue and Other Aedes-Borne Diseases in Niterói, Brazil: A Quasi-Experimental Study. PLoS Neglected Trop. Dis. 2021, 15, e0009556.
  60. Dodson, B.L.; Pujhari, S.; Brustolin, M.L.; Metz, H.C.; Rasgon, J.L. Variable Effects of Wolbachia on Alphavirus Infection in Aedes aegypti. bioRxiv 2023.
  61. Loreto, E.L.S.; Wallau, G.L. Risks of Wolbachia Mosquito Control. Science 2016, 351, 1273.
  62. Sanaei, E.; Charlat, S.; Engelstädter, J. Wolbachia Host Shifts: Routes, Mechanisms, Constraints and Evolutionary Consequences. Biol. Rev. 2021, 96, 433–453.
  63. Edenborough, K.M.; Flores, H.A.; Simmons, C.P.; Fraser, J.E. Using Wolbachia to Eliminate Dengue: Will the Virus Fight Back? J. Virol. 2021, 95, e02203-20.
  64. Thi Hue Kien, D.; Edenborough, K.M.; da Silva Goncalves, D.; Thuy Vi, T.; Casagrande, E.; Thi Le Duyen, H.; Thi Long, V.; Thi Dui, L.; Thi Tuyet Nhu, V.; Thi Giang, N. Genome Evolution of Dengue Virus Serotype 1 under Selection by Wolbachia pipientis in Aedes aegypti Mosquitoes. Virus Evol. 2023, 3, vead016.
  65. Wilke, A.B.B.; Marrelli, M.T. Paratransgenesis: A Promising New Strategy for Mosquito Vector Control. Parasites Vectors 2015, 8, 342.
  66. Wang, S.; Jacobs-Lorena, M. Paratransgenesis Applications: Fighting Malaria with Engineered Mosquito Symbiotic Bacteria. In Arthropod Vector: Controller of Disease Transmission, Volume 1; Elsevier: Amsterdam, The Netherlands, 2017; pp. 219–234.
  67. Ratcliffe, N.A.; Furtado Pacheco, J.P.; Dyson, P.; Castro, H.C.; Gonzalez, M.S.; Azambuja, P.; Mello, C.B. Overview of Paratransgenesis as a Strategy to Control Pathogen Transmission by Insect Vectors. Parasites Vectors 2022, 15, 112.
  68. Wang, S.; Jacobs-Lorena, M. Transgenesis and Paratransgenesis for the Control of Malaria. In Mosquito Gene Drives and the Malaria Eradication Agenda; Jenny Stanford Publishing, United Square: Singapore, 2023; pp. 21–37.
  69. Huang, W.; Wang, S.; Jacobs-Lorena, M. Use of Microbiota to Fight Mosquito-Borne Disease. Front. Genet. 2020, 11, 196.
  70. Yoshida, S.; Ioka, D.; Matsuoka, H.; Endo, H.; Ishii, A. Bacteria Expressing Single-Chain Immunotoxin Inhibit Malaria Parasite Development in Mosquitoes. Mol. Biochem. Parasitol. 2001, 113, 89–96.
  71. Wang, S.; Ghosh, A.K.; Bongio, N.; Stebbings, K.A.; Lampe, D.J.; Jacobs-Lorena, M. Fighting Malaria with Engineered Symbiotic Bacteria from Vector Mosquitoes. Proc. Natl. Acad. Sci. USA 2012, 109, 12734–12739.
  72. Wang, S.; Dos-Santos, A.L.; Huang, W.; Liu, K.C.; Oshaghi, M.A.; Wei, G.; Agre, P.; Jacobs-Lorena, M. Driving Mosquito Refractoriness to Plasmodium falciparum with Engineered Symbiotic Bacteria. Science 2017, 357, 1399–1402.
  73. Villegas, L.M.; Pimenta, P.F.P. Metagenomics, Paratransgenesis and the Anopheles Microbiome: A Portrait of the Geographical Distribution of the Anopheline Microbiota Based on a Meta-Analysis of Reported Taxa. Memórias Inst. Oswaldo Cruz 2014, 109, 672–684.
  74. Bongio, N.J.; Lampe, D.J. Inhibition of Plasmodium berghei Development in Mosquitoes by Effector Proteins Secreted from Asaia sp. Bacteria Using a Novel Native Secretion Signal. PLoS ONE 2015, 10, e0143541.
  75. Mancini, M.V.; Spaccapelo, R.; Damiani, C.; Accoti, A.; Tallarita, M.; Petraglia, E.; Rossi, P.; Cappelli, A.; Capone, A.; Peruzzi, G. Paratransgenesis to Control Malaria Vectors: A Semi-Field Pilot Study. Parasites Vectors 2016, 9, 140.
  76. Raharimalala, F.N.; Boukraa, S.; Bawin, T.; Boyer, S.; Francis, F. Molecular Detection of Six (Endo-) Symbiotic Bacteria in Belgian Mosquitoes: First Step towards the Selection of Appropriate Paratransgenesis Candidates. Parasitol. Res. 2016, 115, 1391–1399.
  77. Rocha, E.M.; Marinotti, O.; Serrão, D.M.; Correa, L.V.; de Katak, R.M.; de Oliveira, J.C.; Muniz, V.A.; de Oliveira, M.R.; do Nascimento Neto, J.F.; Pessoa, M.C.F. Culturable Bacteria Associated with Anopheles darlingi and Their Paratransgenesis Potential. Malar. J. 2021, 20, 40.
  78. Fang, W.; Vega-Rodríguez, J.; Ghosh, A.K.; Jacobs-Lorena, M.; Kang, A.; St. Leger, R.J. Development of Transgenic Fungi That Kill Human Malaria Parasites in Mosquitoes. Science 2011, 331, 1074–1077.
  79. Carballar-Lejarazú, R.; Rodriguez, M.H.; de la Cruz Hernández-Hernández, F.; Ramos-Castaneda, J.; Possani, L.D.; Zurita-Ortega, M.; Reynaud-Garza, E.; Hernández-Rivas, R.; Loukeris, T.; Lycett, G. Recombinant Scorpine: A Multifunctional Antimicrobial Peptide with Activity against Different Pathogens. Cell. Mol. Life Sci. 2008, 65, 3081–3092.
  80. Ward, T.W.; Jenkins, M.S.; Afanasiev, B.N.; Edwards, M.; Duda, B.A.; Suchman, E.; Jacobs-Lorena, M.; Beaty, B.J.; Carlson, J.O. Aedes aegypti Transducing Densovirus Pathogenesis and Expression in Aedes aegypti and Anopheles gambiae Larvae. Insect Mol. Biol. 2001, 10, 397–405.
  81. Carlson, J.; Suchman, E.; Buchatsky, L. Densoviruses for Control and Genetic Manipulation of Mosquitoes. Adv. Virus Res. 2006, 68, 361–392.
  82. Ren, X.; Hoiczyk, E.; Rasgon, J.L. Viral Paratransgenesis in the Malaria Vector Anopheles gambiae. PLoS Pathog. 2008, 4, e1000135.
  83. Johnson, R.M.; Rasgon, J.L. Densonucleosis Viruses (‘Densoviruses’) for Mosquito and Pathogen Control. Curr. Opin. Insect Sci. 2018, 28, 90–97.
  84. Riehle, M.A.; Moreira, C.K.; Lampe, D.; Lauzon, C.; Jacobs-Lorena, M. Using Bacteria to Express and Display Anti-Plasmodium Molecules in the Mosquito Midgut. Int. J. Parasitol. 2007, 37, 595–603.
  85. Favia, G.; Ricci, I.; Marzorati, M.; Negri, I.; Alma, A.; Sacchi, L.; Bandi, C.; Daffonchio, D. Bacteria of the Genus Asaia: A Potential Paratransgenic Weapon against Malaria. Transgenes. Manag. Vector Borne Dis. 2008, 627, 49–59.
  86. Dehghan, H.; Mosa-Kazemi, S.H.; Yakhchali, B.; Maleki-Ravasan, N.; Vatandoost, H.; Oshaghi, M.A. Evaluation of Anti-Malaria Potency of Wild and Genetically Modified Enterobacter Cloacae Expressing Effector Proteins in Anopheles stephensi. Parasites Vectors 2022, 15, 63.
  87. Rasgon, J.L. Using Infections to Fight Infections: Paratransgenic Fungi Can Block Malaria Transmission in Mosquitoes. Future Microbiol. 2011, 6, 851–853.
  88. Tzschaschel, B.D.; Guzmán, C.A.; Timmis, K.N.; Lorenzo, V.d. An Escherichia coli Hemolysin Transport System-Based Vector for the Export of Polypeptides: Export of Shiga-like Toxin IIeB Subunit by Salmonella typhimurium AroA. Nat. Biotechnol. 1996, 14, 765–769.
More
This entry is offline, you can click here to edit this entry!
ScholarVision Creations