1
|
Bahrami R, Quaranta S, Perdomo HD, Bonizzoni M, Khorramnejad A. Carry-over effects of Bacillus thuringiensis on tolerant Aedes albopictus mosquitoes. Parasit Vectors 2024; 17:456. [PMID: 39511654 PMCID: PMC11545555 DOI: 10.1186/s13071-024-06556-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND The biological larvicide Bacillus thuringiensis subsp. israelensis (Bti) represents a safe and effective alternative to chemical insecticides for mosquito control. Efficient control of mosquitoes implicates continuous and extensive application of Bti. This massive use of Bti imposes strong selective pressure, but the complex mode of action of the numerous synergistic Bti endotoxins lower the risk of the emergence of resistance. Although resistance to Bti has not been identified at the population level in nature, some larvae can survive Bti exposure, suggesting tolerance mechanisms. Here we investigated whether Bti-tolerant Aedes albopictus larvae experience any fitness costs. We also studied how this tolerance affects different aspects of the phenotype of the emerging adults that could be relevant for arboviral transmission. METHODS We exposed Ae. albopictus larvae to lethal concentration of Bti and studied the fitness and gut microbiota of tolerant larvae and their adult counterparts. We further compared the transcript abundance of nine key immunity genes in the gut of Bti-tolerant larvae and their emerging adults versus those not exposed to Bti. RESULTS Our results showed that Bti exposure has multifaceted impacts on Ae. albopictus mosquitoes during both larval and adult stages. The carry-over effect of Bti exposure on tolerant larvae manifested in reduced adult emergence rate, shorter lifespan, and decreased fecundity. Bti also alters the gut microbiota of both larvae and adults. We observed higher microbial diversity in Bti-tolerant larvae and changes in the richness of core microbiota. Bti infection and the altered microbiota triggered immune responses in the larval and adult guts. CONCLUSIONS The observed reduction in mosquito fitness and changes in the composition of the microbiota of adults emerging from tolerant larvae could negatively influence mosquito vectorial capacity. Understanding these impacts is crucial for evaluating the broader implications of Bti-based insecticides in mosquito control programs.
Collapse
|
2
|
Wu-Chuang A, Rojas A, Bernal C, Cardozo F, Valenzuela A, Romero C, Mateos-Hernández L, Cabezas-Cruz A. Influence of microbiota-driven natural antibodies on dengue transmission. Front Immunol 2024; 15:1368599. [PMID: 38558802 PMCID: PMC10978734 DOI: 10.3389/fimmu.2024.1368599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Dengue has had a significant global health impact, with a dramatic increase in incidence over the past 50 years, affecting more than 100 countries. The absence of a specific treatment or widely applicable vaccine emphasizes the urgent need for innovative strategies. This perspective reevaluates current evidence supporting the concept of dual protection against the dengue virus (DENV) through natural antibodies (NAbs), particularly anti-α-Gal antibodies induced by the host's gut microbiome (GM). These anti-α-Gal antibodies serve a dual purpose. Firstly, they can directly identify DENV, as mosquito-derived viral particles have been observed to carry α-Gal, thereby providing a safeguard against human infections. Secondly, they possess the potential to impede virus development in the vector by interacting with the vector's microbiome and triggering infection-refractory states. The intricate interplay between human GM and NAbs on one side and DENV and vector microbiome on the other suggests a novel approach, using NAbs to directly target DENV and simultaneously disrupt vector microbiome to decrease pathogen transmission and vector competence, thereby blocking DENV transmission cycles.
Collapse
Affiliation(s)
- Alejandra Wu-Chuang
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR Virologie, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Alejandra Rojas
- Universidad Nacional de Asunción, Instituto de Investigaciones en Ciencias de la Salud, San Lorenzo, Paraguay
| | - Cynthia Bernal
- Universidad Nacional de Asunción, Instituto de Investigaciones en Ciencias de la Salud, San Lorenzo, Paraguay
| | - Fátima Cardozo
- Universidad Nacional de Asunción, Instituto de Investigaciones en Ciencias de la Salud, San Lorenzo, Paraguay
| | - Adriana Valenzuela
- Universidad Nacional de Asunción, Instituto de Investigaciones en Ciencias de la Salud, San Lorenzo, Paraguay
| | - Cristina Romero
- Universidad Nacional de Asunción, Facultad de Ciencias Químicas, San Lorenzo, Paraguay
| | - Lourdes Mateos-Hernández
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Alejandro Cabezas-Cruz
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, France
| |
Collapse
|
3
|
Ricardo Dos Santos Correia P, Duarte de Freitas J, André Zeoly L, Silva Porto R, José da Paz Lima D. Discovery and structure-activity relationship of Morita-Baylis-Hillman adducts as larvicides against dengue mosquito vector, Aedes aegypti (Diptera: Culicidae). Bioorg Med Chem 2023; 90:117315. [PMID: 37253304 DOI: 10.1016/j.bmc.2023.117315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/24/2023] [Accepted: 05/02/2023] [Indexed: 06/01/2023]
Abstract
Neglected tropical diseases (NTDs) have become a significant public health problem worldwide, notably the life-threatening dengue hemorrhagic fever borne by the Aedes aegypti mosquito. Thus, mosquito vector control measures remain essential in public health vector surveillance and control to combat Aedes-borne infections. Therefore, a series of MBH adducts were synthesized and assessed towards the fourth instar mosquito larvae, Aedes aegypti, along with the preliminary structure-activity relationship (SAR). Noteworthy, this compound class might be synthetized by an efficient eco-friendly synthesismethod and a rapid route for the synthesis of commercial larvicide through a single synthetic step. The bioassays showed that this compound class is a promising larvicide to control Aedes aegypti mosquito larvae, mainly 3g, with an LC50 of 41.35 µg/mL, which was higher than evaluated positive controls. Nevertheless, it is a viable larvicidalhit candidate for further hit-to-leadproperties optimization of its biphenyl backbone scaffold with enhanced insecticidalbioactivity. Moreover, scanning electron microscopy analysis suggested a disruption of the osmoregulatory/ionoregulatory functions by the complete deterioration of the terminal exoskeleton hindgut and anal papillae. Therefore, this new study shows the larvicidal efficacy of the tested compounds against the Aedes aegypti larvae.
Collapse
Affiliation(s)
- Paulo Ricardo Dos Santos Correia
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Avenida Lourival Melo Mota, Maceió, Alagoas 57072-970, Brazil
| | | | - Lucas André Zeoly
- Department of Organic Chemistry, Institute of Chemistry, University of Campinas, PO Box 6154, 13083-970 Campinas, São Paulo, Brazil
| | - Ricardo Silva Porto
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Avenida Lourival Melo Mota, Maceió, Alagoas 57072-970, Brazil
| | - Dimas José da Paz Lima
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Avenida Lourival Melo Mota, Maceió, Alagoas 57072-970, Brazil.
| |
Collapse
|
4
|
Wang L, Pang Z, Chen Q, Song Z, Lu Y, Yang M, Huang J, Yu XQ, Wang X. Sublethal exposure to spinetoram impacts life history traits and dengue virus replication in Aedes aegypti. INSECT SCIENCE 2023; 30:486-500. [PMID: 36069276 DOI: 10.1111/1744-7917.13116] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/04/2022] [Accepted: 08/14/2022] [Indexed: 06/15/2023]
Abstract
Insecticides are anthropogenic environmental stressors and also a common stressor for mosquito vectors. However, the use of insecticides is often guided by short-term efficacy, and the sublethal effect on their target or nontarget species has long been ignored. Here, we analyzed how sublethal exposure of the promising vector-control bioinsecticide spinetoram to Aedes aegypti larvae alter adult performance and susceptibility to dengue virus (DENV) infection. We found that the surviving adult mosquitoes were significantly smaller and exhibited weaker blood-feeding capacity than control females, apart from the extended immature development period. In terms of reproductive potential, although the F0 generation produced a similar number of eggs and offspring during the first gonotrophic cycle, the survival rates of the F1 generations were significantly lower as compared to the control group, suggesting transgenerational sublethal effects on the F1 generation. Notably, surviving adult females had higher DENV-2 viral loads than the control group after spinetoram sublethal exposure. Mechanistically, transcriptomic analysis showed that inhibition of oxidative phosphorylation may function in stimulating DENV production in adult Ae. aegypti. In Aag2 cells, significant accumulation of apoptosis, mitochondrial reactive oxygen species production, and DENV-2 replication by spinetoram exposure consistently support our conclusion. Our study highlights the threat of sublethal spinetoram exposure on outbreaks of mosquito-borne viruses.
Collapse
Affiliation(s)
- Luoluo Wang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Zhichang Pang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Qi Chen
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Zhentao Song
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yi Lu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Meng Yang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jia Huang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiao-Qiang Yu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiaoyun Wang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Factors Affecting Arbovirus Midgut Escape in Mosquitoes. Pathogens 2023; 12:pathogens12020220. [PMID: 36839492 PMCID: PMC9963182 DOI: 10.3390/pathogens12020220] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Arboviral diseases spread by mosquitoes cause significant morbidity and mortality throughout much of the world. The treatment and prevention of these diseases through medication and vaccination is often limited, which makes controlling arboviruses at the level of the vector ideal. One way to prevent the spread of an arbovirus would be to stop its vector from developing a disseminated infection, which is required for the virus to make its way to the saliva of the mosquito to be potentially transmitted to a new host. The midgut of the mosquito provides one such opportunity to stop an arbovirus in its tracks. It has been known for many years that in certain arbovirus-vector combinations, or under certain circumstances, an arbovirus can infect and replicate in the midgut but is unable to escape from the tissue to cause disseminated infection. This situation is known as a midgut escape barrier. If we better understand why this barrier occurs, it might aid in the development of more informed control strategies. In this review, we discuss how the midgut escape barrier contributes to virus-vector specificity and possible mechanisms that may allow this barrier to be overcome in successful virus-vector combinations. We also discuss several of the known factors that either increase or decrease the likelihood of midgut escape.
Collapse
|
6
|
Meier CJ, Rouhier MF, Hillyer JF. Chemical Control of Mosquitoes and the Pesticide Treadmill: A Case for Photosensitive Insecticides as Larvicides. INSECTS 2022; 13:1093. [PMID: 36555003 PMCID: PMC9783766 DOI: 10.3390/insects13121093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 06/17/2023]
Abstract
Insecticides reduce the spread of mosquito-borne disease. Over the past century, mosquito control has mostly relied on neurotoxic chemicals-such as pyrethroids, neonicotinoids, chlorinated hydrocarbons, carbamates and organophosphates-that target adults. However, their persistent use has selected for insecticide resistance. This has led to the application of progressively higher amounts of insecticides-known as the pesticide treadmill-and negative consequences for ecosystems. Comparatively less attention has been paid to larvae, even though larval death eliminates a mosquito's potential to transmit disease and reproduce. Larvae have been targeted by source reduction, biological control, growth regulators and neurotoxins, but hurdles remain. Here, we review methods of mosquito control and argue that photoactive molecules that target larvae-called photosensitive insecticides or PSIs-are an environmentally friendly addition to our mosquitocidal arsenal. PSIs are ingested by larvae and produce reactive oxygen species (ROS) when activated by light. ROS then damage macromolecules resulting in larval death. PSIs are degraded by light, eliminating environmental accumulation. Moreover, PSIs only harm small translucent organisms, and their broad mechanism of action that relies on oxidative damage means that resistance is less likely to evolve. Therefore, PSIs are a promising alternative for controlling mosquitoes in an environmentally sustainable manner.
Collapse
Affiliation(s)
- Cole J. Meier
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
| | | | - Julián F. Hillyer
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
7
|
Alomar AA, Eastmond BH, Rapti Z, Walker ED, Alto BW. Ingestion of spinosad-containing toxic sugar bait alters Aedes albopictus vector competence and vectorial capacity for dengue virus. Front Microbiol 2022; 13:933482. [PMID: 36090120 PMCID: PMC9459233 DOI: 10.3389/fmicb.2022.933482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 08/05/2022] [Indexed: 12/01/2022] Open
Abstract
Dengue virus (DENV) is a highly prevalent vector-borne virus that causes life-threatening illnesses to humans worldwide. The development of a tool to control vector populations has the potential to reduce the burden of DENV. Toxic sugar bait (TSB) provides a form of vector control that takes advantage of the sugar-feeding behavior of adult mosquitoes. However, studies on the effect of ingestion of toxins in TSB on vector competence and vectorial capacity for viruses are lacking. This study evaluated vector competence for DENV serotype-1 of Aedes albopictus at 7 and 14 days post-ingestion of TSB formulated with spinosad (of bacteria origin) as an oral toxin. Our results and others were modeled to estimate effects on Ae. albopictus vectorial capacity for DENV. Ingestion of TSB caused a reduction in survival of females, but increased mosquito susceptibility to DENV infection, disseminated infection, and transmission. However, this increase in vector competence was obviated by the reduction in survival, leading to a lower predicted vectorial capacity. The findings of this study highlight the importance of evaluating the net impact of TSB ingestion on epidemiological parameters of vectorial capacity in the context of vector control efforts to reduce the risk of transmission of vector-borne viruses.
Collapse
Affiliation(s)
- Abdullah A. Alomar
- Florida Medical Entomology Laboratory, University of Florida, Vero Beach, FL, United States
- *Correspondence: Abdullah A. Alomar,
| | - Bradley H. Eastmond
- Florida Medical Entomology Laboratory, University of Florida, Vero Beach, FL, United States
| | - Zoi Rapti
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Department of Mathematics, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Edward D. Walker
- Department of Entomology, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Barry W. Alto
- Florida Medical Entomology Laboratory, University of Florida, Vero Beach, FL, United States
| |
Collapse
|
8
|
Juache-Villagrana AE, Pando-Robles V, Garcia-Luna SM, Ponce-Garcia G, Fernandez-Salas I, Lopez-Monroy B, Rodriguez-Sanchez IP, Flores AE. Assessing the Impact of Insecticide Resistance on Vector Competence: A Review. INSECTS 2022; 13:377. [PMID: 35447819 PMCID: PMC9024519 DOI: 10.3390/insects13040377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 01/09/2023]
Abstract
The primary strategy to avoid adverse impacts from insect-mediated pathogen transmission is the chemical control of vector populations through insecticides; its continued use has led to insecticide resistance and unknown consequences on vector competence. This review aims to systematically analyze and synthesize the research on the influence of insecticide resistance (IR) on vector competence (VC). Thirty studies met the inclusion criteria. Twenty studies, conducted either in laboratory or field settings, described the influence of phenotypic insecticide resistance and mechanisms on VC in vectors of human pathogens. Seven studies showed the effect of exposure to insecticides on VC in vectors of human pathogens. Three studies reported the influence of phenotypic resistance and mechanisms on VC in crop pests. The evidence shows that IR could enhance, impair, or have no direct effect on VC in either field or laboratory-designed studies. Similar positive and negative trends are found in pest vectors in crops and studies of insecticide exposure and VC. Even though there is evidence that exposure to insecticides and IR can enhance VC, thus increasing the risk of pathogen transmission, more investigations are needed to confirm the observed patterns and what implications these factors could have in vector control programs.
Collapse
Affiliation(s)
- Alan E. Juache-Villagrana
- Facultad de Ciencias Biologicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n Cd. Universitaria, San Nicolas de los Garza 66455, Nuevo Leon, Mexico; (A.E.J.-V.); (S.M.G.-L.); (G.P.-G.); (I.F.-S.); (B.L.-M.); (I.P.R.-S.)
| | - Victoria Pando-Robles
- Centro de Investigacion Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Publica, Cuernavaca 62100, Morelos, Mexico;
| | - Selene M. Garcia-Luna
- Facultad de Ciencias Biologicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n Cd. Universitaria, San Nicolas de los Garza 66455, Nuevo Leon, Mexico; (A.E.J.-V.); (S.M.G.-L.); (G.P.-G.); (I.F.-S.); (B.L.-M.); (I.P.R.-S.)
| | - Gustavo Ponce-Garcia
- Facultad de Ciencias Biologicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n Cd. Universitaria, San Nicolas de los Garza 66455, Nuevo Leon, Mexico; (A.E.J.-V.); (S.M.G.-L.); (G.P.-G.); (I.F.-S.); (B.L.-M.); (I.P.R.-S.)
| | - Ildefonso Fernandez-Salas
- Facultad de Ciencias Biologicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n Cd. Universitaria, San Nicolas de los Garza 66455, Nuevo Leon, Mexico; (A.E.J.-V.); (S.M.G.-L.); (G.P.-G.); (I.F.-S.); (B.L.-M.); (I.P.R.-S.)
| | - Beatriz Lopez-Monroy
- Facultad de Ciencias Biologicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n Cd. Universitaria, San Nicolas de los Garza 66455, Nuevo Leon, Mexico; (A.E.J.-V.); (S.M.G.-L.); (G.P.-G.); (I.F.-S.); (B.L.-M.); (I.P.R.-S.)
| | - Iram P. Rodriguez-Sanchez
- Facultad de Ciencias Biologicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n Cd. Universitaria, San Nicolas de los Garza 66455, Nuevo Leon, Mexico; (A.E.J.-V.); (S.M.G.-L.); (G.P.-G.); (I.F.-S.); (B.L.-M.); (I.P.R.-S.)
| | - Adriana E. Flores
- Facultad de Ciencias Biologicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n Cd. Universitaria, San Nicolas de los Garza 66455, Nuevo Leon, Mexico; (A.E.J.-V.); (S.M.G.-L.); (G.P.-G.); (I.F.-S.); (B.L.-M.); (I.P.R.-S.)
| |
Collapse
|
9
|
Alomar AA, Alto BW. Evaluation of Pyriproxyfen Effects on Aedes aegypti and Predatory Mosquito Toxorhynchites rutilus (Diptera: Culicidae). JOURNAL OF MEDICAL ENTOMOLOGY 2022; 59:585-590. [PMID: 34865101 DOI: 10.1093/jme/tjab193] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Indexed: 06/13/2023]
Abstract
Control of mosquito vectors of pathogens remains heavily dependent on the application of conventional insecticides. Pyriproxyfen (PPF) is a novel insecticide that has been proposed for use in autodissemination techniques to control mosquito vectors. The use of PPF can inhibit adult emergence but does not inhibit larval development. This feature is favorable for controlling Aedes aegypti because PPF has the potential to work in combination with natural sources of mortality (competition, predation) during the immature stages, and other control methods, including biocontrol agents that further suppress recruitment of adult mosquitoes. However, the PPF effects on life-history traits of Ae. aegypti in comparison to predatory mosquito Toxorhynchites rutilus, a source of mortality, are not fully understood. Here, we show that larval exposure to PPF concentrations that inhibit 50-90% of adult emergence in Ae. aegypti had a negligible effect on adult emergence and lifespan of Tx. rutilus. Weights of adult Ae. aegypti and Tx. rutilus were not influenced by PPF. These findings suggest that the use of PPF to control mosquito vectors may have low effects on mosquito biocontrol agents. Our results extend and confirm earlier data showing that PPF has potential to implement with Tx. rutilus to suppress Ae. aegypti and provide an additional advantage of PPF use in autodissemination control strategies.
Collapse
Affiliation(s)
- Abdullah A Alomar
- Florida Medical Entomology Laboratory, University of Florida, Vero Beach, FL, 32962, USA
| | - Barry W Alto
- Florida Medical Entomology Laboratory, University of Florida, Vero Beach, FL, 32962, USA
| |
Collapse
|
10
|
Giraud É, Varet H, Legendre R, Sismeiro O, Aubry F, Dabo S, Dickson LB, Moro CV, Lambrechts L. Mosquito-bacteria interactions during larval development trigger metabolic changes with carry-over effects on adult fitness. Mol Ecol 2021; 31:1444-1460. [PMID: 34905257 DOI: 10.1111/mec.16327] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 11/30/2022]
Abstract
In animals with distinct life stages such as holometabolous insects, adult phenotypic variation is often shaped by the environment of immature stages, including their interactions with microbes colonizing larval habitats. Such carry-over effects were previously observed for several adult traits of the mosquito Aedes aegypti after larval exposure to different bacteria, but the mechanistic underpinnings are unknown. Here, we investigated the molecular changes triggered by gnotobiotic larval exposure to different bacteria in Ae. aegypti. We initially screened a panel of 16 bacterial isolates from natural mosquito breeding sites to determine their ability to influence adult life-history traits. We subsequently focused on four bacterial isolates (belonging to Flavobacterium, Lysobacter, Paenibacillus, and Enterobacteriaceae) with significant carry-over effects on adult survival and found that they were associated with distinct transcriptomic profiles throughout mosquito development. Moreover, we detected carry-over effects at the level of gene expression for the Flavobacterium and Paenibacillus isolates. The most prominent transcriptomic changes in gnotobiotic larvae reflected a profound remodeling of lipid metabolism, which translated into phenotypic differences in lipid storage and starvation resistance at the adult stage. Together, our findings indicate that larval exposure to environmental bacteria trigger substantial physiological changes that impact adult fitness, uncovering a possible mechanism underlying carry-over effects of mosquito-bacteria interactions during larval development.
Collapse
Affiliation(s)
- Émilie Giraud
- Insect-Virus Interactions Unit, Institut Pasteur, UMR2000, CNRS, 75015, Paris, France
| | - Hugo Varet
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, 75015, Paris, France.,Plate-forme Technologique Biomics - Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75015, Paris, France
| | - Rachel Legendre
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, 75015, Paris, France.,Plate-forme Technologique Biomics - Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75015, Paris, France
| | - Odile Sismeiro
- Plate-forme Technologique Biomics - Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75015, Paris, France.,Unité Biologie des Bactéries Pathogènes à Gram-positif, CNRS, UMR2001 Microbiologie Intégrative et Moléculaire, Institut Pasteur, 75015, Paris, France
| | - Fabien Aubry
- Insect-Virus Interactions Unit, Institut Pasteur, UMR2000, CNRS, 75015, Paris, France
| | - Stéphanie Dabo
- Insect-Virus Interactions Unit, Institut Pasteur, UMR2000, CNRS, 75015, Paris, France
| | - Laura B Dickson
- Insect-Virus Interactions Unit, Institut Pasteur, UMR2000, CNRS, 75015, Paris, France.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Claire Valiente Moro
- Univ Lyon, CNRS, INRAE, UMR Écologie Microbienne, Université Claude Bernard Lyon 1, VetAgro Sup, F-69622, Villeurbanne, France
| | - Louis Lambrechts
- Insect-Virus Interactions Unit, Institut Pasteur, UMR2000, CNRS, 75015, Paris, France
| |
Collapse
|
11
|
Juvenile hormone analog enhances Zika virus infection in Aedes aegypti. Sci Rep 2021; 11:21062. [PMID: 34702871 PMCID: PMC8548497 DOI: 10.1038/s41598-021-00432-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 10/12/2021] [Indexed: 12/17/2022] Open
Abstract
In recent years, there has been a rise in the emergence of arboviruses of public health importance, including Zika, chikungunya, dengue, and yellow fever viruses. Insecticide-based mosquito control has been the primary method for mitigating transmission of arboviruses. The consequences for the application of insecticides include both lethal and sublethal effects, and associated development of insecticide resistance. However, little is known about the influence on arboviral transmission. Mosquitoes with phenotypes that exhibit insecticide resistance or experience sublethal effects may be associated with altered susceptibility to arbovirus infection and transmission. Juvenile hormone analogs (JHAs) are insecticides that prevent pupa to adult molting of mosquitoes by mimicking the action of their natural juvenile hormone. Here, we examined whether the JHA pyriproxyfen interacts with ambient temperature (20 °C and 30 °C) during juvenile stages to influence life-history traits, population growth (λ'), and Zika virus (ZIKV) infection in Aedes aegypti. Development time of females was lengthened at 20 °C and in the presence of JHA. Prevention of pupa to adult molting by JHA was differentially higher at elevated temperature than low temperature. Size of females was larger at 20 °C and smaller at 30 °C. Infection, disseminated infection, and transmission of ZIKV in females were enhanced by JHA at both 20 °C and 30 °C relative to the controls. These results demonstrate that mosquito life-history and vector competence parameters are strongly influenced by interactive effects of JHA and temperature. The JHA-induced enhancement of ZIKV infection in females should be a consideration when implementing JHA in vector control strategies.
Collapse
|
12
|
Andreazza F, Oliveira EE, Martins GF. Implications of Sublethal Insecticide Exposure and the Development of Resistance on Mosquito Physiology, Behavior, and Pathogen Transmission. INSECTS 2021; 12:insects12100917. [PMID: 34680686 PMCID: PMC8539869 DOI: 10.3390/insects12100917] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/03/2021] [Accepted: 10/04/2021] [Indexed: 12/31/2022]
Abstract
Simple Summary Mosquitoes are one of the greatest threats to human lives; they transmit a wide range of pathogens, including viruses that cause lethal diseases. Mosquitoes are found in both aquatic (as larvae or pupae) and terrestrial (as adults) environments during their complex life cycle. For decades, insecticides have been systematically used on mosquitoes with the aim to reduce their population. Little is known about how the stress resulting from the exposure of mosquitoes to insecticides impacts the tri-partite relationship between the mosquitoes, their vertebrate hosts, and the pathogens they transmit. In this work, we review existing experimental evidence to obtain a broad picture on the potential effects of the (sub)lethal exposure of hematophagous mosquitoes to different insecticides. We have focused on studies that have advanced our understanding of their physiological and behavioral responses (including the mechanisms behind insecticide resistance) and the spread of pathogens by these vectors—understudied but critically important issues for epidemiology. Studying these exposure-related effects is of paramount importance for predicting how they respond to insecticide exposure and whether this exposure makes them more or less likely to transmit pathogens. Abstract For many decades, insecticides have been used to control mosquito populations in their larval and adult stages. Although changes in the population genetics, physiology, and behavior of mosquitoes exposed to lethal and sublethal doses of insecticides are expected, the relationships between these changes and their abilities to transmit pathogens remain unclear. Thus, we conducted a comprehensive review on the sublethal effects of insecticides and their contributions to insecticide resistance in mosquitoes, with the main focus on pyrethroids. We discuss the direct and acute effects of sublethal concentrations on individuals and populations, the changes in population genetics caused by the selection for resistance after insecticide exposure, and the major mechanisms underlying such resistance. Sublethal exposures negatively impact the individual’s performance by affecting their physiology and behavior and leaving them at a disadvantage when compared to unexposed organisms. How these sublethal effects could change mosquito population sizes and diversity so that pathogen transmission risks can be affected is less clear. Furthermore, despite the beneficial and acute aspects of lethality, exposure to higher insecticide concentrations clearly impacts the population genetics by selecting resistant individuals, which may bring further and complex interactions for mosquitoes, vertebrate hosts, and pathogens. Finally, we raise several hypotheses concerning how the here revised impacts of insecticides on mosquitoes could interplay with vector-mediated pathogens’ transmission.
Collapse
Affiliation(s)
- Felipe Andreazza
- Departamento de Entomologia, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (F.A.); (E.E.O.)
| | - Eugênio E. Oliveira
- Departamento de Entomologia, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil; (F.A.); (E.E.O.)
| | - Gustavo Ferreira Martins
- Departamento de Biologia Geral, Universidade Federal de Viçosa, Viçosa 36570-900, MG, Brazil
- Correspondence:
| |
Collapse
|
13
|
Carvalho KDS, Guedes DRD, Crespo MM, de Melo-Santos MAV, Silva-Filha MHNL. Aedes aegypti continuously exposed to Bacillus thuringiensis svar. israelensis does not exhibit changes in life traits but displays increased susceptibility for Zika virus. Parasit Vectors 2021; 14:379. [PMID: 34321098 PMCID: PMC8317411 DOI: 10.1186/s13071-021-04880-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/16/2021] [Indexed: 11/20/2022] Open
Abstract
Background Aedes aegypti can transmit arboviruses worldwide, and Bacillus thuringiensis svar. israelensis (Bti)-based larvicides represent an effective tool for controlling this species. The safety of Bti and lack of resistance have been widely reported; however, little is known regarding the impact of the extensive use of these larvicides on the life traits of mosquitoes. Therefore, this study investigated biological parameters, including susceptibility to arbovirus, of an Ae. aegypti strain (RecBti) subjected to 29 generations of exposure to Bti compared with the RecL reference strain. Methods The biological parameters of individuals reared under controlled conditions were compared. Also, the viral susceptibility of females not exposed to Bti during their larval stage was analysed by oral infection and followed until 14 or 21 days post-infection (dpi). Results RecBti individuals did not display alterations in the traits that were assessed (fecundity, fertility, pupal weight, developmental time, emergence rate, sex ratio and haematophagic capacity) compared to RecL individuals. Females from both strains were susceptible to dengue serotype 2 (DENV-2) and Zika virus (ZIKV). However, RecBti females showed significantly higher rates of ZIKV infection compared with RecL females at 7 (90% versus 68%, Chi-square: χ2 = 7.27, df = 1, P = 0.006) and 14 dpi (100% versus 87%, Chi-square: χ2 = 7.69, df = 1, P = 0.005) and for dissemination at 7 dpi (83.3% versus 36%, Fisher’s exact test: P < 0.0001, OR = 0.11, 95% CI 0.03–0.32). Quantification of DENV-2 and ZIKV viral particles produced statistically similar results for females from both strains. Conclusions Prolonged exposure of Ae. aegypti larvae to Bti did not alter most of the evaluated biological parameters, except that RecBti females exhibited a higher vector susceptibility for ZIKV. This finding is related to a background of Bti exposure for several generations but not to a previous exposure of the tested females during the larval stage. This study highlights mosquito responses that could be associated with the chronic exposure to Bti in addition to the primary larvicidal effect elicited by this control agent. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-021-04880-6.
Collapse
Affiliation(s)
| | | | - Mônica Maria Crespo
- Department of Entomology, Instituto Aggeu Magalhães-Fiocruz, Recife, Pernambuco, Brazil
| | | | | |
Collapse
|
14
|
Bacterial Toxins Active against Mosquitoes: Mode of Action and Resistance. Toxins (Basel) 2021; 13:toxins13080523. [PMID: 34437394 PMCID: PMC8402332 DOI: 10.3390/toxins13080523] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 12/25/2022] Open
Abstract
Larvicides based on the bacteria Bacillus thuringiensis svar. israelensis (Bti) and Lysinibacillus sphaericus are effective and environmentally safe compounds for the control of dipteran insects of medical importance. They produce crystals that display specific and potent insecticidal activity against larvae. Bti crystals are composed of multiple protoxins: three from the three-domain Cry type family, which bind to different cell receptors in the midgut, and one cytolytic (Cyt1Aa) protoxin that can insert itself into the cell membrane and act as surrogate receptor of the Cry toxins. Together, those toxins display a complex mode of action that shows a low risk of resistance selection. L. sphaericus crystals contain one major binary toxin that display an outstanding persistence in field conditions, which is superior to Bti. However, the action of the Bin toxin based on its interaction with a single receptor is vulnerable for resistance selection in insects. In this review we present the most recent data on the mode of action and synergism of these toxins, resistance issues, and examples of their use worldwide. Data reported in recent years improved our understanding of the mechanism of action of these toxins, showed that their combined use can enhance their activity and counteract resistance, and reinforced their relevance for mosquito control programs in the future years.
Collapse
|
15
|
Caragata EP, Dong S, Dong Y, Simões ML, Tikhe CV, Dimopoulos G. Prospects and Pitfalls: Next-Generation Tools to Control Mosquito-Transmitted Disease. Annu Rev Microbiol 2021; 74:455-475. [PMID: 32905752 DOI: 10.1146/annurev-micro-011320-025557] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mosquito-transmitted diseases, including malaria and dengue, are a major threat to human health around the globe, affecting millions each year. A diverse array of next-generation tools has been designed to eliminate mosquito populations or to replace them with mosquitoes that are less capable of transmitting key pathogens. Many of these new approaches have been built on recent advances in CRISPR/Cas9-based genome editing. These initiatives have driven the development of pathogen-resistant lines, new genetics-based sexing methods, and new methods of driving desirable genetic traits into mosquito populations. Many other emerging tools involve microorganisms, including two strategies involving Wolbachia that are achieving great success in the field. At the same time, other mosquito-associated bacteria, fungi, and even viruses represent untapped sources of new mosquitocidal or antipathogen compounds. Although there are still hurdles to be overcome, the prospect that such approaches will reduce the impact of these diseases is highly encouraging.
Collapse
Affiliation(s)
- E P Caragata
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - S Dong
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - Y Dong
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - M L Simões
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - C V Tikhe
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - G Dimopoulos
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| |
Collapse
|
16
|
Fogaça AC, Sousa G, Pavanelo DB, Esteves E, Martins LA, Urbanová V, Kopáček P, Daffre S. Tick Immune System: What Is Known, the Interconnections, the Gaps, and the Challenges. Front Immunol 2021; 12:628054. [PMID: 33737931 PMCID: PMC7962413 DOI: 10.3389/fimmu.2021.628054] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Ticks are ectoparasitic arthropods that necessarily feed on the blood of their vertebrate hosts. The success of blood acquisition depends on the pharmacological properties of tick saliva, which is injected into the host during tick feeding. Saliva is also used as a vehicle by several types of pathogens to be transmitted to the host, making ticks versatile vectors of several diseases for humans and other animals. When a tick feeds on an infected host, the pathogen reaches the gut of the tick and must migrate to its salivary glands via hemolymph to be successfully transmitted to a subsequent host during the next stage of feeding. In addition, some pathogens can colonize the ovaries of the tick and be transovarially transmitted to progeny. The tick immune system, as well as the immune system of other invertebrates, is more rudimentary than the immune system of vertebrates, presenting only innate immune responses. Although simpler, the large number of tick species evidences the efficiency of their immune system. The factors of their immune system act in each tick organ that interacts with pathogens; therefore, these factors are potential targets for the development of new strategies for the control of ticks and tick-borne diseases. The objective of this review is to present the prevailing knowledge on the tick immune system and to discuss the challenges of studying tick immunity, especially regarding the gaps and interconnections. To this end, we use a comparative approach of the tick immune system with the immune system of other invertebrates, focusing on various components of humoral and cellular immunity, such as signaling pathways, antimicrobial peptides, redox metabolism, complement-like molecules and regulated cell death. In addition, the role of tick microbiota in vector competence is also discussed.
Collapse
Affiliation(s)
- Andréa C. Fogaça
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Géssica Sousa
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniel B. Pavanelo
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Eliane Esteves
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Larissa A. Martins
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
- Laboratory of Bacteriology, Tick-Pathogen Transmission Unit, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Veronika Urbanová
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Petr Kopáček
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Sirlei Daffre
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
Hery L, Guidez A, Durand AA, Delannay C, Normandeau-Guimond J, Reynaud Y, Issaly J, Goindin D, Legrave G, Gustave J, Raffestin S, Breurec S, Constant P, Dusfour I, Guertin C, Vega-Rúa A. Natural Variation in Physicochemical Profiles and Bacterial Communities Associated with Aedes aegypti Breeding Sites and Larvae on Guadeloupe and French Guiana. MICROBIAL ECOLOGY 2021; 81:93-109. [PMID: 32621210 PMCID: PMC7794107 DOI: 10.1007/s00248-020-01544-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/15/2020] [Indexed: 05/10/2023]
Abstract
Aedes aegypti develop in aquatic habitats in which mosquito larvae are exposed to physicochemical elements and microorganisms that may influence their life cycle and their ability to transmit arboviruses. Little is known about the natural bacterial communities associated with A. aegypti or their relation to the biotic and abiotic characteristics of their aquatic habitats. We characterized the physicochemical properties and bacterial microbiota of A. aegypti breeding sites and larvae on Guadeloupe and in French Guiana. In addition, we explored whether geographic location, the type of breeding site and physicochemical parameters influenced the microbiota associated with this mosquito species. We used large-scale 16S rRNA gene sequencing of 160 breeding sites and 147 pools of A. aegypti larvae and recorded 12 physicochemical parameters at the sampled breeding sites. Ordination plots and multiple linear regression were used to assess the influence of environmental factors on the bacterial microbiota of water and larvae. We found territory-specific differences in physicochemical properties (dissolved oxygen, conductivity) and the composition of bacterial communities in A. aegypti breeding sites that influenced the relative abundance of several bacteria genera (e.g., Methylobacterium, Roseoccocus) on the corresponding larvae. A significant fraction of the bacterial communities identified on larvae, dominated by Herbiconiux and Microvirga genera, were consistently enriched in mosquitoes regardless the location. In conclusion, territory-specific differences observed in the biotic and abiotic properties of A. aegypti breeding sites raise concern about the impact of these changes on pathogen transmission by different A. aegypti populations.
Collapse
Affiliation(s)
- Lyza Hery
- Laboratory of Vector Control Research, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur of Guadeloupe, Morne Jolivière, Guadeloupe France
| | - Amandine Guidez
- Vector Control and Adaptation Unit, Cayenne, Institut Pasteur of French Guiana, Vectopôle Amazonien Emile Abonnenc, Cayenne, French Guiana France
| | | | - Christelle Delannay
- Laboratory of Vector Control Research, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur of Guadeloupe, Morne Jolivière, Guadeloupe France
| | | | - Yann Reynaud
- Laboratory of Vector Control Research, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur of Guadeloupe, Morne Jolivière, Guadeloupe France
| | - Jean Issaly
- Vector Control and Adaptation Unit, Cayenne, Institut Pasteur of French Guiana, Vectopôle Amazonien Emile Abonnenc, Cayenne, French Guiana France
| | - Daniella Goindin
- Laboratory of Vector Control Research, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur of Guadeloupe, Morne Jolivière, Guadeloupe France
| | - Grégory Legrave
- Laboratory of Environment and Food Hygiene, Institut Pasteur of Guadeloupe, Morne Jolivière, Guadeloupe France
| | - Joel Gustave
- Regional Health Agency of Guadeloupe, Gourbeyre, Guadeloupe France
| | - Stéphanie Raffestin
- Laboratory of Environment and Hygiene, Institut Pasteur of French Guiana, Cayenne, French Guiana France
| | - Sebastien Breurec
- Transmission, Reservoir and Diversity of Pathogens Unit, Institut Pasteur of Guadeloupe, Pointe-à-Pitre, France
- Hyacinthe Bastaraud Faculty of Medicine, University of Antilles, Pointe-à-Pitre, France
- INSERM Centre for Clinical Investigation 1424, Pointe-à-Pitre, Les Abymes France
| | - Philippe Constant
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec Canada
| | - Isabelle Dusfour
- Vector Control and Adaptation Unit, Cayenne, Institut Pasteur of French Guiana, Vectopôle Amazonien Emile Abonnenc, Cayenne, French Guiana France
| | - Claude Guertin
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec Canada
| | - Anubis Vega-Rúa
- Laboratory of Vector Control Research, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur of Guadeloupe, Morne Jolivière, Guadeloupe France
| |
Collapse
|
18
|
Alomar AA, Eastmond BH, Alto BW. The effects of exposure to pyriproxyfen and predation on Zika virus infection and transmission in Aedes aegypti. PLoS Negl Trop Dis 2020; 14:e0008846. [PMID: 33201875 PMCID: PMC7707533 DOI: 10.1371/journal.pntd.0008846] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/01/2020] [Accepted: 09/24/2020] [Indexed: 12/21/2022] Open
Abstract
Zika virus (ZIKV) is an emerging mosquito-borne pathogen that can cause global public health threats. In the absence of effective antiviral medications, prevention measures rely largely on reducing the number of adult mosquito vectors by targeting juvenile stages. Despite the importance of juvenile mosquito control measures in reducing adult population size, a full understanding of the effects of these measures in determining mosquito phenotypic traits and in mosquito-arbovirus interactions is poorly understood. Pyriproxyfen is a juvenile hormone analog that primarily blocks adult emergence, but does not cause mortality in larvae. This mechanism has the potential to work in combination with other juvenile sources of mortality in nature such as predation to affect mosquito populations. Here, we experimentally evaluated the effects of juvenile exposure to pyriproxyfen and predatory mosquito Toxorhynchites rutilus on Aedes aegypti phenotypes including susceptibility to ZIKV infection and transmission. We discovered that combined effects of pyriproxyfen and Tx. rutilus led to higher inhibition of adult emergence in Ae. aegypti than observed in pyriproxyfen or Tx. rutilus treatments alone. Adult body size was larger in treatments containing Tx. rutilus and in treatments mimicking the daily mortality of predation compared to control or pyriproxyfen treatments. Susceptibility to infection with ZIKV in Ae. aegypti was reduced in predator treatment relative to those exposed to pyriproxyfen. Disseminated infection, transmission, and titers of ZIKV in Ae. aegypti were similar in all treatments relative to controls. Our data suggest that the combination of pyriproxyfen and Tx. rutilus can inhibit adult Ae. aegypti emergence but may confer a fitness advantage in survivors and does not inhibit their vector competence for ZIKV relative to controls. Understanding the ultimate consequences of juvenile mosquito control measures on subsequent adults’ ability to transmit pathogens is critical to fully understand their overall impacts. Mosquito control approaches primarily depend on lowering the number of potential adult mosquito vectors by inhibiting juvenile stages to reduce the risk of pathogen transmission. Pyriproxyfen is a juvenile hormone analog that inhibits the emergence of adult mosquitoes by interrupting metamorphosis, but does not target larvae. This mechanism allows natural sources of mortality like predation to act in combination with pyriproxyfen to affect mosquito population size. Here, we determined the effects of juvenile exposure to pyriproxyfen and predatory mosquito Toxorhynchites rutilus on adult Aedes aegypti traits, including infection with Zika virus. Combined effects of pyriproxyfen and Tx. rutilus led to strong inhibition of adult emergence in Ae. aegypti. Treatments containing predators or those mimicking the daily mortality of predation produced larger sized adults. Susceptibility to ZIKV infection was lowest in the predator treatment and highest in the pyriproxyfen treatment. Disseminated infection, transmission, and viral titers of ZIKV were similar between treatments. Our data suggest that the combination of pyriproxyfen and predators can enhance inhibition of adult Ae. aegypti emergence, but survivors may have fitness benefits such being larger mosquitoes. Understanding the consequences of control approaches in mosquito-pathogen interactions will assist to evaluate their suitability in mosquito control programs.
Collapse
Affiliation(s)
- Abdullah A. Alomar
- University of Florida, Department of Entomology and Nematology, Florida Medical Entomology Laboratory, Vero Beach, Florida, United States of America
| | - Bradley H. Eastmond
- University of Florida, Department of Entomology and Nematology, Florida Medical Entomology Laboratory, Vero Beach, Florida, United States of America
| | - Barry W. Alto
- University of Florida, Department of Entomology and Nematology, Florida Medical Entomology Laboratory, Vero Beach, Florida, United States of America
- * E-mail:
| |
Collapse
|
19
|
Dacey DP, Chain FJJ. The Challenges of Microbial Control of Mosquito-Borne Diseases Due to the Gut Microbiome. Front Genet 2020; 11:504354. [PMID: 33133140 PMCID: PMC7575760 DOI: 10.3389/fgene.2020.504354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 09/18/2020] [Indexed: 01/09/2023] Open
Abstract
Mosquitoes are one of the deadliest animals on earth because of their ability to transmit a wide range of human pathogens. Traditional mosquito control methods use chemical insecticides, but with dwindling long-term effectiveness and negative effects on the environment, microbial forms of control have become common alternatives. The insecticide Bacillus thuringiensis subspecies israelensis (Bti) is the most popular of these alternatives, although it can also have direct effects on lowering environmental biodiversity and indirect effects on food-web relationships in the ecosystems where it is deployed. In addition, microbial control agents that impede pathogen development or transmission from mosquito to human are under investigation, including Wolbachia and Asaia, but unexpected interactions with mosquito gut bacteria can hinder their effectiveness. Improved characterization of mosquito gut bacterial communities is needed to determine the taxa that interfere with microbial controls and their effectiveness in wild populations. This mini-review briefly discusses relationships between mosquito gut bacteria and microbial forms of control, and the challenges in ensuring their success.
Collapse
Affiliation(s)
- Daniel P Dacey
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA, United States
| | - Frédéric J J Chain
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA, United States
| |
Collapse
|
20
|
Caputo B, Manica M. Mosquito surveillance and disease outbreak risk models to inform mosquito-control operations in Europe. CURRENT OPINION IN INSECT SCIENCE 2020; 39:101-108. [PMID: 32403040 DOI: 10.1016/j.cois.2020.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/09/2020] [Accepted: 03/25/2020] [Indexed: 06/11/2023]
Abstract
Surveillance programs are needed to guide mosquito-control operations to reduce both nuisance and the spread of mosquito-borne diseases. Understanding the thresholds for action to reduce both nuisance and the risk of arbovirus transmission is becoming critical. To date, mosquito surveillance is mainly implemented to inform about pathogen transmission risks rather than to reduce mosquito nuisance even though lots of control efforts are aimed at the latter. Passive surveillance, such as digital monitoring (validated by entomological trapping), is a powerful tool to record biting rates in real time. High-quality data are essential to model the risk of arbovirus diseases. For invasive pathogens, efforts are needed to predict the arrival of infected hosts linked to the small-scale vector to host contact ratio, while for endemic pathogens efforts are needed to set up region-wide highly structured surveillance measures to understand seasonal re-activation and pathogen transmission in order to carry out effective control operations.
Collapse
Affiliation(s)
- Beniamino Caputo
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, Piazzale A. Moro 5, 38010, 00185 Rome, Italy.
| | - Mattia Manica
- Department of Biodiversity and Molecular Ecology, Research and Innovation Centre, Fondazione Edmund Mach, Via E. Mach 1, 38010 San Michele all' Adige, Italy
| |
Collapse
|
21
|
Caragata EP, Tikhe CV, Dimopoulos G. Curious entanglements: interactions between mosquitoes, their microbiota, and arboviruses. Curr Opin Virol 2019; 37:26-36. [PMID: 31176069 PMCID: PMC6768729 DOI: 10.1016/j.coviro.2019.05.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 11/22/2022]
Abstract
Mosquitoes naturally harbor a diverse community of microorganisms that play a crucial role in their biology. Mosquito-microbiota interactions are abundant and complex. They can dramatically alter the mosquito immune response, and impede or enhance a mosquito's ability to transmit medically important arboviral pathogens. Yet critically, given the massive public health impact of arboviral disease, few such interactions have been well characterized. In this review, we describe the current state of knowledge of the role of microorganisms in mosquito biology, how microbial-induced changes to mosquito immunity moderate infection with arboviruses, cases of mosquito-microbial-virus interactions with a defined mechanism, and the molecular interactions that underlie the endosymbiotic bacterium Wolbachia's ability to block virus infection in mosquitoes.
Collapse
Affiliation(s)
- Eric P Caragata
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Chinmay V Tikhe
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|