Next Article in Journal
Non-Lethal Detection of Ranavirus in Fish
Next Article in Special Issue
The Role of NS1 Protein in the Diagnosis of Flavivirus Infections
Previous Article in Journal
HAS-Flow May Be an Adequate Method for Evaluating Human T-Cell Leukemia Virus Type 1 Infected Cells in Human T-Cell Leukemia Virus Type 1-Positive Rheumatoid Arthritis Patients Receiving Antirheumatic Therapies: A Retrospective Cross-Sectional Observation Study
Previous Article in Special Issue
Construction and Rescue of a DNA-Launched DENV2 Infectious Clone
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Aedes aegypti Metabolism with Next-Generation Insecticides

by
Michael J. Conway
1,*,
Douglas P. Haslitt
1 and
Benjamin M. Swarts
2,3
1
Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, MI 48859, USA
2
Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, MI 48859, USA
3
Biochemistry, Cell, and Molecular Biology Graduate Programs, Central Michigan University, Mount Pleasant, MI 48859, USA
*
Author to whom correspondence should be addressed.
Viruses 2023, 15(2), 469; https://doi.org/10.3390/v15020469
Submission received: 19 January 2023 / Revised: 3 February 2023 / Accepted: 6 February 2023 / Published: 8 February 2023
(This article belongs to the Special Issue Boosting Flavivirus Research: A Pandengue Net Initiative)

Abstract

:
Aedes aegypti is the primary vector of dengue virus (DENV), zika virus (ZIKV), and other emerging infectious diseases of concern. A key disease mitigation strategy is vector control, which relies heavily on the use of insecticides. The development of insecticide resistance poses a major threat to public health worldwide. Unfortunately, there is a limited number of chemical compounds available for vector control, and these chemicals can have off-target effects that harm invertebrate and vertebrate species. Fundamental basic science research is needed to identify novel molecular targets that can be exploited for vector control. Next-generation insecticides will have unique mechanisms of action that can be used in combination to limit selection of insecticide resistance. Further, molecular targets will be species-specific and limit off-target effects. Studies have shown that mosquitoes rely on key nutrients during multiple life cycle stages. Targeting metabolic pathways is a promising direction that can deprive mosquitoes of nutrition and interfere with development. Metabolic pathways are also important for the virus life cycle. Here, we review studies that reveal the importance of dietary and stored nutrients during mosquito development and infection and suggest strategies to identify next-generation insecticides with a focus on trehalase inhibitors.

1. Introduction

1.1. Vector Control Strategies

Vector control strategies can be categorized into chemical, biological, genetic, and environmental methods. Chemical methods rely on general neurotoxins, the four major categories being organochlorines, organophosphate, carbamates, and pyrethroids. Pyrethroids are the current preferred chemical strategy in mosquitoes due to their rapid onset and minimal toxicity in mammalian species [1]. Despite their widespread use, insecticide resistance is a major concern, in addition to off-target effects that can harm invertebrate and vertebrate species, the possibility of occupational exposure, and the unknown impact of sublethal concentrations on vector-borne diseases [1,2,3,4,5,6,7].
The main biological method that is being studied is infection of mosquito vectors with the bacterial symbiont Wolbachia. Wolbachia infection suppresses viral replication with a negligible risk to humans, the environment, and mosquito fitness [8,9,10,11,12,13,14,15,16,17,18,19,20,21]. Logistic and regulatory constraints currently limit the usefulness of this technique [22,23,24,25]. Additional biological methods being studied include leveraging insect viruses and other microorganisms to compete with human pathogens [26,27,28,29,30]. Biological byproducts such as toxins produced by Bacillus thuringiensis subspecies israelensis (Bti) have been used successfully to control mosquito larvae [31,32].
The oldest genetic method is the sterile insect technique (SIT). This method has a long history and relies on radiation-induced chromosome breakage to produce sterile males. The method requires the production of large numbers of sterilized males, which are released into the environment, and mate with a local population of mosquitoes. If the ratio of sterilized males to females is high enough, they will outcompete the non-sterile males and lead to a reduction in the mosquito population [33,34,35,36]. The production of sterilized males is challenging and makes it difficult to translate into largescale mosquito control programs. Oxitec expanded on this concept and used gene editing techniques to produce male mosquitos that carry a mutation that produces infertility in female progeny. This strategy is an improvement over the original SIT because male progeny continues to carry the mutation and pass it onto female progeny [37,38,39,40]. This reduces the number of genetically modified mosquitoes that need to be released into the environment to suppress a mosquito population.
In addition to SIT and Oxitec techniques, work with transcription activator-like effector nucleases (TALENs) utilized site-specific nucleases to knock out genes with essential functions such as kynurenine 3-monooxygenase (KMO), which produces eye pigmentation in Aedes embryos [41,42]. However, because they rely on sensitive DNA-binding interactions, they can be difficult to produce, and scaling up the technology is challenging. The CRISPR/Cas9 approach originally saw issues like TALEN’s with variable site-specific dependence affecting the techniques’ ability for lethality and mutagenesis; however, germline expression of Cas9 overcame the site variability. The inert site serves as a “gene drive” (GD) relying on the presence of an engineered sgRNA with a cas9 exonuclease to allow for precise and rapid dissemination of genetic payloads into mosquito populations [39,43,44,45]. Genetic methods to suppress mosquito populations have tremendous promise, although concerns regarding cost, control, species specificity, and mistrust of scientific and political institutions remain [46,47,48,49,50].
Environmental methods to control mosquito vectors are generally safe and sustainable, although they do require community participation and investment. These methods include designing communities to limit accumulation of standing water, and if needed, routine cleaning or covering of water containers. Physical barriers such as bed nets and screened windows can also reduce exposure to mosquito bites. It is also possible to trap mosquitoes and to physically remove them from the environment.

1.2. Insecticide Resistance

Aedes aegypti and Aedes albopictus are the primary and secondary vectors for several important arboviruses, including dengue virus (DENV), yellow fever virus (YFV), zika virus (ZIKV), and chikungunya virus (CHIKV) [51,52]. Insecticide application is a critical tool to reduce the population of disease vectors during an outbreak, particularly in the absence of targeted therapeutics and prophylactic vaccines [7,53,54]. Unfortunately, improper use and overuse of insecticides can select mutations that afford resistance to insecticides [7].
Insecticide resistance can arise from different mechanisms, including modification of the target site (i.e., target site resistance). Pyrethroid insecticides function by targeting voltage-gated sodium channels (VGSCs) in the insect’s nervous system. This leads to rapid paralysis and death, often described as “knockdown”. The most well-studied resistance mutations are found in the VGSC gene, and these are described as knockdown resistance (kdr) mutations [1,4]. Kdr mutations are screened in populations so that we can manage the emergence of insecticide resistance. A recent meta-analysis revealed that the rates of the major kdr mutations (i.e., F1534C, V1016G, and S989G) in Asia from 2000 to 2021 were 29%, 26%, and 22%, respectfully. Resistance to dichlorodiphenyltrichloroethane (DDT) was high in both Ae. aegypti (68%) and Ae. albopictus (64%). Resistance to permethrin (58%) and deltamethrin (27%) was also high in Ae. aegypti [55]. A very recent study showed that the L982W mutation, which confers resistance to pyrethroids, was detected at a frequency of >78% in Vietnam and Cambodia. Alleles with concomitant mutations (i.e., L982W, F1534C, V1016G, and F1534C) were confirmed in both countries at a frequency of >90% in Phnom Penh, Cambodia [56]. The spread of insecticide resistance threatens what was once an effective vector control strategy.

1.3. Novel Insecticide Targets

The risk of widespread insecticide resistance requires careful consideration of how these chemicals are used. Integrated Pest Management (IPM) programs leverage information about a pest’s life cycle and their interaction with the environment to minimize the financial costs and impact on human health and the environment. IPM programs promote judicious use of pesticides, although widespread agricultural use of the same pesticides continues to contribute to insecticide resistance [54]. An important area of research is the identification of novel insecticide targets. These studies address the rise of insecticide resistance and offer solutions that can complement current strategies and perhaps develop next-generation insecticides that have minimal off-target effects.
Identifying insecticide activity in natural products has been a strong focus of recent research, including characterization of components in essential oils, seaweed, botanicals, and medicinal plants [57,58,59,60,61]. Studies have also shown efficacy targeting specific tissues and metabolic pathways, with a focus on new mechanisms of action and avoiding off-target effects. VUO41 and VU730 are inhibitors of mosquito inward rectifier potassium (Kir) channels expressed in the Malpighian tubules of Anopheles and Aedes mosquitoes. These chemicals are specific to mosquitoes with limited activity against Kir1 channels in mammalian orthologs and honeybees [62,63].
Mosquito metabolism and developmental pathways are emerging targets for insecticide development and provide the opportunity to identify insecticides with new mechanisms of action that target molecules specific to a vector of concern (Figure 1). For example, it may be possible to generate next-generation insecticides that target insulin-like peptide (ILP) homologues that promote insulin receptor signaling and drive growth [64,65]. Further, ecdysone receptor (EcR) agonists have shown promise as novel insecticides [66]. Importantly, our study showing that the trehalase inhibitor validamycin A (ValA) inhibits mosquito development and flight supports additional research into metabolic and developmental pathways that can be targeted with next-generation insecticides [67].

1.4. Summary

Recent research has been expanding the repertoire of pharmacological targets, and basic science research has identified important roles for dietary and stored nutrients in mosquito behavior and development. Importantly, vector-borne diseases rely on a host’s metabolism to propagate, which creates an opportunity to “kill two birds with one stone”. Identification of species-specific targets involved in critical developmental stages will lead to the development of next-generation insecticides and targets for gene drive technology that have new mechanisms of action and that have minimal to non-existent off-target effects. The following is a review of promising research findings related to dietary and stored nutrients and how they are utilized by mosquitoes to drive behavior and development. Further, we discuss how these same nutrients impact infection with important vector-borne diseases and focus on the development of trehalase inhibitors as a future direction.

2. Dietary Nutrients and Impact on Mosquito Behavior and Development

Dietary nutrients drive mosquito behavior and development; however, the molecular mechanisms involved are largely unknown, which creates an opportunity to identify novel targets for insecticide development and gene drive technology. Importantly, changes in nutrition can lead to far-reaching impacts on mosquito survival, feeding behavior, oviposition, and tolerance to adverse conditions, including exposure to insecticides. Elucidating the metabolic and signaling pathways that control mosquito behavior and development is an important future direction.

2.1. Survival

Dietary nutrients are clearly important for the survival of mosquito larvae and adults. Mosquito larvae depend on organic detritus and adults depend on either plant nectar or vertebrate blood. These diets provide sugar and free fatty acids that can be converted to ATP, protein, and amino acids that are used to make more protein and deoxynucleotides, as well as lipids, which are converted into major insect hormones. Inhibiting any of these major metabolic pathways will negatively impact survival, although identifying a mosquito-specific target will likely be a challenge due to the evolutionary conservation of major biochemical pathways.
Previous research revealed that mosquitoes can survive on blood-free alternatives, but these contain different mixtures of peptides, amino acids, vitamins, carbohydrates, ATP, bovine serum albumin (BSA), and cholesterol [68,69,70,71]. Partial dietary restriction significantly influences Ae. aegypti development. Similar to studies in other biological systems, dietary restriction in both Ae. aegypti larvae and adults led to longer lifespans [72]. Specifically, females that were fed only a single or no blood meal survived 30–40% longer than those given weekly blood meals and increasing the concentration of protein in an artificial blood meal led to a decrease in survival. Larvae also lived longer when fed 50% and 25% larval diet [72]. However, this longer lifespan may be due to a developmental arrest, wherein the restriction of larval diet prolonged eclosure time and reduced the size of adult mosquitoes. Larger adult mosquitos also survive longer than small mosquitoes [73]. These data suggest that energy reserves promote development and survival of mosquitoes. In support of this, at low concentrations of larval diet, larvae spend more time foraging for food [74]. A certain threshold of nutrition and energy reserves is likely needed for larvae to progress to the next developmental stage. A recent in vitro study showed that dietary cholesterol mobilized stored triacylglycerol from lipid droplets, which supports that signals received from a mosquito’s diet can promote the utilization of stored nutrients [75].

2.2. Egg Production and Oviposition Site Selection

The size of a female mosquito positively correlates with larval diet concentration and nutrient availability and negatively correlates with larval density [73,76,77]. Larvae that have access to high concentrations of dietary nutrients have more energy reserves and can build a larger female adult mosquito. Building a larger female mosquito is important because heavier females exhibit greater blood-feeding capacity, and macronutrients in blood are critical for egg production [73]. Some studies suggest that larger mosquitoes generated from feeding larvae with higher concentrations of larval diet produce more eggs after a blood meal, although this finding is not consistent [73,77]. Although the size of a mosquito correlates with the nutrition they received as larvae, sugar feeding as adults can also modulate egg production. Importantly, high energy reserves and an empty crop correlated with higher egg production, while lower energy reserves and a full crop full of undigested sugar correlated with lower egg production [78]. These studies suggest that dietary nutrients are important for building large mosquitoes that can produce more eggs, which is likely due to the accumulation of stored nutrients.

2.3. Biting Behavior

Dietary nutrients can also modify biting behavior. It is unclear what drives this behavior, although one study revealed that supplementation of blood with 3 and 10% sugar diet significantly increased biting frequency, and continuous availability of a 5% sugar solution increased the probing response [79,80]. Feeding on specific types of sugar also influenced biting behavior [81]. These data suggest that mosquitoes can sense cues from their host, which can drive behavior, and that dietary nutrition can also impact behavior. In a seminal study, a small-molecule agonist of Ae. aegypti neuropeptide Y receptor reduced biting likely by signaling that the mosquito was fully engorged or replete with nutrients [82]. It is likely that odorant and neuropeptide receptors serve to select the best source of nutrition and ensure that sufficient nutrition is acquired prior to committing to oogenesis.

2.4. Tolerance to Adverse Conditions

Mosquitoes encounter many stressors in the laboratory and natural environment, and receiving nutrition improves survival. In one study, Ae. aegypti were more likely to survive dehydrating conditions if they recently engorged on blood [83]. Ae. aegypti were also more likely to escape during an exito-repellency avoidance assay after exposure to deltamethrin and cypermethrin insecticides if they had fed on blood or sugar [84]. A separate study revealed that hydration alone improved survival in a CDC bottle bioassay, suggesting that mosquitoes adequately hydrated with water, sugar water, or blood are more likely to resist insecticide treatment [84,85].

3. Dietary Nutrients and Impact on Virus Infection

Dengue virus (DENV), Zika virus (ZIKV), yellow fever virus (YFV), and chikungunya virus (CHIKV) are transmitted by Ae. aegypti. All viruses are obligate intracellular parasites and depend on the host cell’s metabolism to produce raw materials and chemical energy to produce progeny virions. Studies in animal models have previously linked dietary nutrition and disease outcome. Recent research revealed the importance of host-derived dietary nutrients during arbovirus acquisition in mosquito vectors.

3.1. Protein

One study compared ZIKV infection in Ae. aegypti that were fed either infectious whole blood or Dulbecco’s phosphate buffered saline solution containing 250 mg/mL BSA. ZIKV acquisition in midgut tissue was worse 7- and 14-days post-infection (dpi) when mosquitoes were fed the infectious protein solution, although dissemination to peripheral tissues was not affected [86]. These data suggest that blood protein is not a critical factor that promotes ZIKV acquisition, and that other blood nutrients may be important for virus infection.

3.2. Sugar

Mosquitoes acquire sugar in vertebrate blood and plant nectar—mostly sucrose, glucose, and fructose—which are primarily used for the energy demands found in mosquitoes to promote flight, survival, and reproduction [87]. What is less recognized, but also important, is how sugar impacts the virus acquisition in the mosquito. In one study, a sugar meal just prior to administration of an infected blood meal protected mosquitoes from infection with arboviruses from different families [87]. In contrast, a blood meal supplemented with glucose promoted an increase in mosquito infection compared to blood meal alone [88]. These results require further investigation since they are in conflict. It is important to note that administration of a sugar meal would place nutrients into the crop, whereas glucose supplemented into a blood meal would place nutrients into the midgut. These are dramatically different environments, and dietary sugar likely integrates with mosquito metabolism and immunity in unique ways depending on where it resides.

3.3. Lipid

The lipid fraction of vertebrate blood is arguably the most complex and contains free fatty acids (FFAs), triacylglycerol, cholesterol, and cell-associated phospholipids. The contribution of blood meal-derived vertebrate lipids to virus infection is largely unknown [8,11,12,13,14]. Previous research has shown that intracellular lipids are important for DENV replication in both vertebrate and invertebrate cells, and that DENV manipulates its host’s lipidome to facilitate replication [15,16,17,18,19,20,21,22]. The importance of lipids in the DENV life cycle is clear, although it is not known how blood-derived lipids impact infection. In mosquitoes, alterations in cholesterol and lipid trafficking through Wolbachia infection or chemical/genetic manipulation interfered with DENV infection [17,18,20,23]. In contrast, human low-density lipoprotein (LDL) inhibited flavivirus infection in vitro and in vivo [8]. Further research revealed that extracellular vesicles (EVs) in serum restricted DENV fusion in the Ae. aegypti-derived (Aag2) cell line but not in mammalian cells [13]. Vertebrate lipids appear to inhibit DENV at an early stage in its life cycle and promote DENV at a later stage in its life cycle. In support of this, DENV reduced protein expression of low-density lipoprotein receptor-related protein 1 (LRP-1) in Aag2 cells, leading to increased intracellular cholesterol levels and enhanced virus replication [24]. These studies reveal a complicated relationship with blood-derived lipids, where some species may inhibit acquisition and others facilitate replication and dissemination.

4. Stored Nutrients and Impact on Mosquito Development

Mosquitoes store nutrients in the form of glycogen, triacylglycerols, and trehalose. Glycogen and triacylglycerols are stored in adipocytes in the fat body [89]. Trehalose is synthesized from two glucose molecules and is the major blood sugar in mosquitoes [90]. Mosquito larvae and adults acquire nutrients from either organic detritus, vertebrate blood, or plant nectar, and store nutrients as needed to fuel key life cycle transitions. Recent research has shown the importance of stored nutrients on mosquito development. The molecular mechanisms that contribute to metabolism of stored nutrients are well-established and largely conserved across species, which creates an opportunity to quickly identify targets for insecticide development and gene drive technology. Optimizing compounds that can specifically target Ae. aegypti metabolic pathways is an important future direction.

4.1. Lipid

Fatty acids stored in lipid droplets within cells in the fat body take the form of triacylglycerols and are mobilized through lipolysis to maintain metabolic activity of cells and tissues, and provide energy needs for long-term flight, oogenesis, and resisting starvation [91,92,93,94]. Insect adipokinetic hormone (AKH), juvenile hormone (JH), and 20-hydroxyexdysone (20E) act as metabolic switches to promote the mobilization of stored lipids [95,96]. AKHs activate fat body lipases, which convert triacylglycerols to free fatty acids and glycerol, and allow insects to generate ATP though beta oxidation [95]. JH and 20E regulate expression of genes involved in triacyclglycerol catabolism and β-oxidation [96]. Preliminary studies have shown that dibenzoylhydrazine compounds can function as ecdysone receptor agonists and have insecticidal activity through the promotion of premature development [97,98]. Much of the research that has described insect lipid metabolism has been performed in model organisms, although the limited research available in Ae. aegypti supports that many of these pathways and individual genes are highly conserved and would serve as broad targets for the development of next-generation insecticides or gene drive technology.
Research focused on Aedes spp. reveals that triacylglycerol is an important regulator of the gonadotropic cycle—it increases the strength of eggshells and facilities overwintering in diapausing eggs [94]. It is unclear how stored lipids drive the development of mosquito larvae and pupae, although crowding of larvae increases triacylglycerol storage and limits the size of adult mosquitoes and downstream reproductive fitness [99]. Limiting triacylglycerol also reduces the size of male mosquito accessory organs [100]. These data suggest that targeting lipid metabolism would impact multiple life cycle stages, undermining mosquito fitness at the level of larvae, adults, and eggs.

4.2. Sugar

Transcriptomic studies have shown that metabolism of stored sugars plays an important role in multiple life cycle stages in insects, including eclosure and oogenesis [101]. Insects, including mosquitoes, store sugar in the form of glycogen and trehalose, which have interconnected metabolic pathways (Figure 2A) [89,102,103,104]. Similar to animals, insect glycogen is a branched polymer of glucose that can be degraded as needed to release glucose to support glycolysis and other activities. Glycogen is synthesized in adipocytes in the fat body from UDP-glucose, which itself is generated mainly from dietary carbohydrate or amino acids [89,104]. When required, glycogen breakdown liberates glucose in the form of glucose-1-phosphate that is isomerized to glucose-6-phosphate, with the latter then being utilized in glycolysis or other pathways [89,104]. Glycogen is also stored in eggs and promotes overwintering. Reduction of glycogen levels using RNAi to knock down glycogen synthase kinase-3 (GSK-3) led to embryonic lethality [105,106,107,108]. Glycogen is clearly an important stored nutrient that supports several stages in the mosquito life cycle.
The other major storage sugar in insects is trehalose, which is the main sugar found in hemolymph. Trehalose, which is also found in fungi, bacteria, and plants, is a non-mammalian disaccharide consisting of two glucose units that are linked together by a 1,1-α,α-glycosidic bond (Figure 2A) [109]. Trehalose is synthesized in fat body adipocytes through trehalose phosphate synthase (TPS)-catalyzed conversion of UDP-glucose and glucose-6-phosphate to trehalose-6-phosphate, which is then dephosphorylated by trehalose-6-phosphate phosphatase (TPP) to give trehalose [104,110]. Trehalose is then secreted via trehalose-specific transporters (e.g., TRET1) into the hemolymph, where it serves as a circulating form of stored glucose [90]. Trehalose is a multifunctional molecule in mosquitoes, as it provides energy and promotes growth, metamorphosis, stress recovery, chitin synthesis, and flight [90,111,112,113,114,115,116,117,118,119,120]. When needed for these functions, trehalose is hydrolyzed by an extracellular trehalose-specific glycoside hydrolase, or trehalase (TREH), yielding two molecules of glucose that can be imported into cells via glucose transporters (GLUTs) [90]. There are two forms of trehalase in insects, including: (i) trehalase 1 (Tre-1), which is soluble and has been identified in insect hemolymph, midgut goblet cells, and eggs; and (ii) trehalase 2 (Tre-2), which is membrane-bound and has been identified in flight muscle, follicle cells, ovary cells, spermatophore, midgut, and brain tissue [90].
Because humans and other vertebrates do not synthesize trehalose nor require it as a nutrient, trehalose metabolism represents an attractive target for the development of safe and specific next-generation insecticides. Trehalases have received significant attention as targets because they are required for the mobilization of trehalose into glucose, which is critical to insect physiology [90,121]. For example, multiple RNAi studies have demonstrated that trehalase deficiency causes abnormal growth and development in various insect species, including brown plant hopper and beet armyworm [122,123]. In addition, various small-molecule trehalase inhibitors, such as validamycin A (Val A, Figure 2B), have been shown to impair development at several stages of the insect life cycle [111,112,116,117,119]. With respect to mosquitoes specifically, the prospects of targeting trehalase are also encouraging. Recently, we demonstrated that Val A delays larval and pupal development of Ae. aegypti, and it inhibits the flight of adults, likely due to hypoglycemia [67].
The promising results of Val A in impairing Ae. aegypti development, coupled with the availability of an array of other trehalase inhibitors, motivates continued development and testing of this class of compounds [90]. To complement other existing trehalose mimetics and pseudosaccharides, we recently introduced a variety of synthetic trehalose analogues, a number of which were shown to inhibit bacterial trehalose utilization, including 5-deoxy-5-thio-D-trehalose (5-ThioTre, Figure 2B) [124]. In contrast to Val A, however, 5-ThioTre had no impact on Ae. aegypti development or flight [67]. In light of the differential activities of Val A and 5-ThioTre, future research should broadly investigate structure-activity relationships of trehalase inhibitors toward the identification of optimal insecticides for targeting Ae. aegypti with minimal off-target effects. In addition, other components of Ae. aegypti trehalose metabolism, such as trehalose synthesis via TPS/TPP or transport via TRET1, can also be explored as potential molecular targets. Given the noted absence of trehalose from humans and other vertebrates, such strategies are expected to exhibit higher specificity and limited off-target effects compared to existing insecticides.

5. Stored Nutrients and Impact on Virus Infection

Very few studies utilize Ae. aegypti to directly test the role of stored sugars (i.e., glycogen or trehalose) on virus infection, although other mosquito models reveal that stored nutrients are important modulators of infection. One study shows that limiting the concentration of sucrose available to Culex mosquitoes reduced total glycogen and lipid per mosquito, and this correlated with an increased ability to orally transmit virus [125]. Another study showed that overexpression of the cellular energy sensor AMP-activated protein kinase (AMPK) reduced glycogen and trehalose concentrations in Anopheles stephensi. This led to reduced permissiveness to infection by Plasmodium falciparum and a reduction in egg production [126]. Another study using Anopheles gambiae showed that knockdown of trehalose transporter AgTreT1 reduced hemolymph trehalose concentration, which correlated with decreased resistance to low humidity, heat, and reduced the number of Plasmodium falciparum oocytes [127].
One study that used Ae. aegypti as a model organism showed that infection with the nematode Brugia malayi reduced glycogen and lipid concentration in mosquitoes and reduced maximum flight speed [128]. A set of in vitro and in vivo studies also confirmed the importance of intracellular lipid trafficking for mosquitoes and DENV and identified thiosemicarbazones as a potential class of insecticides that can inhibit Ae aegypti sterol carrier protein 2 (SCP-2) [129,130,131,132]. Although not classically considered a stored nutrient, polyamines are formed in a cycle that requires amino acids: L-methionine and L-ornithine. Inhibiting this pathway is detrimental to arbovirus infection [133]. These data are limiting, although the emerging consensus is that the energy and physical material provided by stored nutrients is important for the replication of pathogens and for the cell to mount an immune response to infection, and that competition for this limiting resource can negatively impact mosquito life history traits.

6. Conclusions

The literature focused on insect metabolism is rich and has described the significant impact nutrient deprivation has on mosquito behavior and development. Conserved genes and pathways exist that can be investigated as targets for next-generation insecticides or gene drive technology. A more interesting direction is the identification of unique targets and strategies to control disease vectors while limiting off-target effects. The most promising research areas related to development of new vector control technologies, which include next-generation insecticides, are focused on ecdysone signaling and trehalose metabolism. Ecdysone receptor agonists and trehalose analogues have been used in several insect models and can clearly influence mosquito behavior and development. Interestingly, some trehalose analogues appear to work in bacteria but not in insects [67,134]. This observation opens the door to optimizing next-generation insecticides with enhanced activities and minimal off-target effects.

Author Contributions

Conceptualization, M.J.C. and B.M.S.; writing—original draft preparation, M.J.C., D.P.H. and B.M.S.; writing—review and editing, M.J.C. and B.M.S.; funding acquisition, B.M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This review was funded by National Institutes of Health (R15AI117670). The APC was waived.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fang, Y.; Shi, W.Q.; Wu, J.T.; Li, Y.Y.; Xue, J.B.; Zhang, Y. Resistance to pyrethroid and organophosphate insecticides, and the geographical distribution and polymorphisms of target-site mutations in voltage-gated sodium channel and acetylcholinesterase 1 genes in Anopheles sinensis populations in Shanghai, China. Parasit. Vectors 2019, 12, 396. [Google Scholar] [CrossRef]
  2. Amelia-Yap, Z.H.; Chen, C.D.; Sofian-Azirun, M.; Low, V.L. Pyrethroid resistance in the dengue vector Aedes aegypti in Southeast Asia: Present situation and prospects for management. Parasit. Vectors 2018, 11, 332. [Google Scholar] [CrossRef]
  3. Andreazza, F.; Oliveira, E.E.; Martins, G.F. Implications of Sublethal Insecticide Exposure and the Development of Resistance on Mosquito Physiology, Behavior and Pathogen Transmission. Insects 2021, 12, 917. [Google Scholar] [CrossRef]
  4. Du, Y.; Nomura, Y.; Zhorov, B.S.; Dong, K. Sodium Channel Mutations and Pyrethroid Resistance in Aedes aegypti. Insects 2016, 7, 60. [Google Scholar] [CrossRef]
  5. Liu, N. Insecticide resistance in mosquitoes: Impact, mechanisms, and research directions. Annu. Rev. Entomol. 2015, 60, 537–559. [Google Scholar] [CrossRef]
  6. Moyes, C.L.; Vontas, J.; Martins, A.J.; Ng, L.C.; Koou, S.Y.; Dusfour, I.; Raghavendra, K.; Pinto, J.; Corbel, V.; David, J.P.; et al. Contemporary status of insecticide resistance in the major Aedes vectors of arboviruses infecting humans. PLoS Negl. Trop. Dis. 2017, 11, e0005625. [Google Scholar] [CrossRef]
  7. Roberts, D.R.; Andre, R.G. Insecticide resistance issues in vector-borne disease control. Am. J. Trop. Med. Hyg. 1994, 50, 21–34. [Google Scholar] [CrossRef]
  8. Bian, G.; Xu, Y.; Lu, P.; Xie, Y.; Xi, Z. The endosymbiotic bacterium Wolbachia induces resistance to dengue virus in Aedes aegypti. PLoS Pathog. 2010, 6, e1000833. [Google Scholar] [CrossRef] [PubMed]
  9. Blagrove, M.S.; Arias-Goeta, C.; Di Genua, C.; Failloux, A.B.; Sinkins, S.P. A Wolbachia wMel transinfection in Aedes albopictus is not detrimental to host fitness and inhibits Chikungunya virus. PLoS Negl. Trop. Dis. 2013, 7, e2152. [Google Scholar] [CrossRef]
  10. Dobson, S.L.; Fox, C.W.; Jiggins, F.M. The effect of Wolbachia-induced cytoplasmic incompatibility on host population size in natural and manipulated systems. Proc. Biol. Sci. 2002, 269, 437–445. [Google Scholar] [CrossRef] [Green Version]
  11. 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. [Google Scholar] [CrossRef] [PubMed]
  12. Glaser, R.L.; Meola, M.A. The native Wolbachia endosymbionts of Drosophila melanogaster and Culex quinquefasciatus increase host resistance to West Nile virus infection. PLoS ONE 2010, 5, e11977. [Google Scholar] [CrossRef]
  13. Hoffmann, A.A.; Montgomery, B.L.; Popovici, J.; Iturbe-Ormaetxe, I.; Johnson, P.H.; Muzzi, F.; Greenfield, M.; Durkan, M.; Leong, Y.S.; Dong, Y.; et al. Successful establishment of Wolbachia in Aedes populations to suppress dengue transmission. Nature 2011, 476, 454–457. [Google Scholar] [CrossRef]
  14. McMeniman, C.J.; Lane, R.V.; Cass, B.N.; Fong, A.W.; Sidhu, M.; Wang, Y.F.; O’Neill, S.L. Stable introduction of a life-shortening Wolbachia infection into the mosquito Aedes aegypti. Science 2009, 323, 141–144. [Google Scholar] [CrossRef] [PubMed]
  15. Molloy, J.C.; Sommer, U.; Viant, M.R.; Sinkins, S.P. Wolbachia Modulates Lipid Metabolism in Aedes albopictus Mosquito Cells. Appl. Environ. Microbiol. 2016, 82, 3109–3120. [Google Scholar] [CrossRef] [PubMed]
  16. 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.; et al. A Wolbachia symbiont in Aedes aegypti limits infection with dengue, Chikungunya, and Plasmodium. Cell 2009, 139, 1268–1278. [Google Scholar] [CrossRef]
  17. Mousson, L.; Zouache, K.; Arias-Goeta, C.; Raquin, V.; Mavingui, P.; Failloux, A.B. The native Wolbachia symbionts limit transmission of dengue virus in Aedes albopictus. PLoS Negl. Trop. Dis. 2012, 6, e1989. [Google Scholar] [CrossRef]
  18. Ross, P.A.; Endersby, N.M.; Hoffmann, A.A. Costs of Three Wolbachia Infections on the Survival of Aedes aegypti Larvae under Starvation Conditions. PLoS Negl. Trop. Dis. 2016, 10, e0004320. [Google Scholar] [CrossRef]
  19. Shaw, W.R.; Catteruccia, F. Vector biology meets disease control: Using basic research to fight vector-borne diseases. Nat. Microbiol. 2019, 4, 20–34. [Google Scholar] [CrossRef]
  20. 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.; et al. The wMel Wolbachia strain blocks dengue and invades caged Aedes aegypti populations. Nature 2011, 476, 450–453. [Google Scholar] [CrossRef]
  21. Xi, Z.; Khoo, C.C.; Dobson, S.L. Wolbachia establishment and invasion in an Aedes aegypti laboratory population. Science 2005, 310, 326–328. [Google Scholar] [CrossRef]
  22. Soh, L.T.; Ong, Z.; Vasquez, K.; Chen, I.; Li, X.; Niah, W.; Panchapakesan, C.; Sheldenkar, A.; Sim, S.; Ng, L.C.; et al. A Household-Based Survey to Understand Factors Influencing Awareness, Attitudes and Knowledge towards Wolbachia-Aedes Technology. Int. J. Environ. Res. Public. Health 2021, 18, 11997. [Google Scholar] [CrossRef]
  23. Liew, C.; Soh, L.T.; Chen, I.; Ng, L.C. Public sentiments towards the use of Wolbachia-Aedes technology in Singapore. BMC Public Health 2021, 21, 1417. [Google Scholar] [CrossRef]
  24. Lwin, M.O.; Ong, Z.; Panchapakesan, C.; Sheldenkar, A.; Soh, L.T.; Chen, I.; Li, X.; Niah, W.; Vasquez, K.; Sim, S.; et al. Influence of public hesitancy and receptivity on reactive behaviours towards releases of male Wolbachia-Aedes mosquitoes for dengue control. PLoS Negl. Trop. Dis. 2022, 16, e0010910. [Google Scholar] [CrossRef]
  25. Arham, A.F.; Amin, L.; Mustapa, M.A.C.; Mahadi, Z.; Yaacob, M.; Ibrahim, M. Determinants of stakeholders’ attitudes and intentions toward supporting the use of Wolbachia-infected Aedes mosquitoes for dengue control. BMC Public Health 2021, 21, 2314. [Google Scholar] [CrossRef]
  26. Hegazy, M.I.; Hegazy, A.M.; Saad, A.M.; Salem, H.M.; El-Tahan, A.M.; El-Saadony, M.T.; Soliman, S.M.; Taha, A.E.; Alshehri, M.A.; Ezzat Ahmed, A.; et al. ٍSome biologically active microorganisms have the potential to suppress mosquito larvae (Culex pipiens, Diptera: Culicidae). Saudi J. Biol. Sci. 2022, 29, 1998–2006. [Google Scholar] [CrossRef]
  27. Ye, G.; Wang, Y.; Liu, X.; Dong, Q.; Cai, Q.; Yuan, Z.; Xia, H. Transmission competence of a new mesonivirus, Yichang virus, in mosquitoes and its interference with representative flaviviruses. PLoS Negl. Trop. Dis. 2020, 14, e0008920. [Google Scholar] [CrossRef]
  28. White, A.V.; Fan, M.; Mazzara, J.M.; Roper, R.L.; Richards, S.L. Mosquito-infecting virus Espirito Santo virus inhibits replication and spread of dengue virus. J. Med. Virol. 2021, 93, 3362–3373. [Google Scholar] [CrossRef]
  29. Schultz, M.J.; Frydman, H.M.; Connor, J.H. Dual Insect specific virus infection limits Arbovirus replication in Aedes mosquito cells. Virology 2018, 518, 406–413. [Google Scholar] [CrossRef]
  30. Goenaga, S.; Kenney, J.L.; Duggal, N.K.; Delorey, M.; Ebel, G.D.; Zhang, B.; Levis, S.C.; Enria, D.A.; Brault, A.C. Potential for Co-Infection of a Mosquito-Specific Flavivirus, Nhumirim Virus, to Block West Nile Virus Transmission in Mosquitoes. Viruses 2015, 7, 5801–5812. [Google Scholar] [CrossRef] [Green Version]
  31. Novak, R.J.; Gubler, D.J.; Underwood, D. Evaluation of slow-release formulations of temephos (Abate) and Bacillus thuringiensis var. israelensis for the control of Aedes aegypti in Puerto Rico. J. Am. Mosq. Control Assoc. 1985, 1, 449–453. [Google Scholar] [PubMed]
  32. Hare, S.G.; Nasci, R.S. Effects of sublethal exposure to Bacillus thuringiensis var. israelensis on larval development and adult size in Aedes aegypti. J. Am. Mosq. Control Assoc. 1986, 2, 325–328. [Google Scholar] [PubMed]
  33. Chen, C.; Aldridge, R.L.; Gibson, S.; Kline, J.; Aryaprema, V.; Qualls, W.; Xue, R.D.; Boardman, L.; Linthicum, K.J.; Hahn, D.A. Developing the radiation-based sterile insect technique (SIT) for controlling Aedes aegypti: Identification of a sterilizing dose. Pest Manag. Sci. 2022, 79, 1175–1183. [Google Scholar] [CrossRef] [PubMed]
  34. Martín-Park, A.; Che-Mendoza, A.; Contreras-Perera, Y.; Pérez-Carrillo, S.; Puerta-Guardo, H.; Villegas-Chim, J.; Guillermo-May, G.; Medina-Barreiro, A.; Delfín-González, H.; Méndez-Vales, R.; et al. Pilot trial using mass field-releases of sterile males produced with the incompatible and sterile insect techniques as part of integrated Aedes aegypti control in Mexico. PLoS Negl. Trop. Dis. 2022, 16, e0010324. [Google Scholar] [CrossRef]
  35. Silva, E.B.; Mendonça, C.M.; Mendonça, J.A.; Dias, E.S.F.; Florêncio, S.G.L.; Guedes, D.R.D.; Paiva, M.H.S.; Amaral, A.; Netto, A.M.; Melo-Santos, M.A.V. Effects of gamma radiation on the reproductive viability of Aedes aegypti and its descendants (Diptera: Culicidae). Acta Trop. 2022, 228, 106284. [Google Scholar] [CrossRef]
  36. Hallinan, E.; Rai, K.S. Radiation sterilization of Aedes aegypti in nitrogen and implications for sterile male technique. Nature 1973, 244, 368–369. [Google Scholar] [CrossRef]
  37. Fu, G.; Lees, R.S.; Nimmo, D.; Aw, D.; Jin, L.; Gray, P.; Berendonk, T.U.; White-Cooper, H.; Scaife, S.; Kim Phuc, H.; et al. Female-specific flightless phenotype for mosquito control. Proc. Natl. Acad. Sci. USA 2010, 107, 4550–4554. [Google Scholar] [CrossRef]
  38. Bargielowski, I.; Nimmo, D.; Alphey, L.; Koella, J.C. Comparison of life history characteristics of the genetically modified OX513A line and a wild type strain of Aedes aegypti. PLoS ONE 2011, 6, e20699. [Google Scholar] [CrossRef]
  39. Chen, J.; Luo, J.; Wang, Y.; Gurav, A.S.; Li, M.; Akbari, O.S.; Montell, C. Suppression of female fertility in Aedes aegypti with a CRISPR-targeted male-sterile mutation. Proc. Natl. Acad. Sci. USA 2021, 118, e2105075118. [Google Scholar] [CrossRef]
  40. Phuc, H.K.; Andreasen, M.H.; Burton, R.S.; Vass, C.; Epton, M.J.; Pape, G.; Fu, G.; Condon, K.C.; Scaife, S.; Donnelly, C.A.; et al. Late-acting dominant lethal genetic systems and mosquito control. BMC Biol. 2007, 5, 11. [Google Scholar] [CrossRef] [Green Version]
  41. Aryan, A.; Anderson, M.A.; Myles, K.M.; Adelman, Z.N. TALEN-based gene disruption in the dengue vector Aedes aegypti. PLoS ONE 2013, 8, e60082. [Google Scholar] [CrossRef]
  42. Han, Q.; Calvo, E.; Marinotti, O.; Fang, J.; Rizzi, M.; James, A.A.; Li, J. Analysis of the wild-type and mutant genes encoding the enzyme kynurenine monooxygenase of the yellow fever mosquito, Aedes aegypti. Insect Mol. Biol. 2003, 12, 483–490. [Google Scholar] [CrossRef] [PubMed]
  43. Kistler, K.E.; Vosshall, L.B.; Matthews, B.J. Genome engineering with CRISPR-Cas9 in the mosquito Aedes aegypti. Cell Rep. 2015, 11, 51–60. [Google Scholar] [CrossRef] [PubMed]
  44. Li, M.; Bui, M.; Yang, T.; Bowman, C.S.; White, B.J.; Akbari, O.S. Germline Cas9 expression yields highly efficient genome engineering in a major worldwide disease vector, Aedes aegypti. Proc. Natl. Acad. Sci. USA. 2017, 114, E10540–E10549. [Google Scholar] [CrossRef]
  45. Li, M.; Yang, T.; Bui, M.; Gamez, S.; Wise, T.; Kandul, N.P.; Liu, J.; Alcantara, L.; Lee, H.; Edula, J.R.; et al. Suppressing mosquito populations with precision guided sterile males. Nat. Commun. 2021, 12, 5374. [Google Scholar] [CrossRef]
  46. Schairer, C.E.; Triplett, C.; Akbari, O.S.; Bloss, C.S. California Residents’ Perceptions of Gene Drive Systems to Control Mosquito-Borne Disease. Front. Bioeng. Biotechnol. 2022, 10, 848707. [Google Scholar] [CrossRef] [PubMed]
  47. Connolly, J.B.; Mumford, J.D.; Glandorf, D.C.M.; Hartley, S.; Lewis, O.T.; Evans, S.W.; Turner, G.; Beech, C.; Sykes, N.; Coulibaly, M.B.; et al. Recommendations for environmental risk assessment of gene drive applications for malaria vector control. Malar. J. 2022, 21, 152. [Google Scholar] [CrossRef]
  48. Wise, I.J.; Borry, P. An Ethical Overview of the CRISPR-Based Elimination of Anopheles gambiae to Combat Malaria. J. Bioethical Inq. 2022, 19, 371–380. [Google Scholar] [CrossRef]
  49. James, S.L.; Marshall, J.M.; Christophides, G.K.; Okumu, F.O.; Nolan, T. Toward the Definition of Efficacy and Safety Criteria for Advancing Gene Drive-Modified Mosquitoes to Field Testing. Vector Borne Zoonotic Dis. 2020, 20, 237–251. [Google Scholar] [CrossRef]
  50. Famakinde, D.O. Public health concerns over gene-drive mosquitoes: Will future use of gene-drive snails for schistosomiasis control gain increased level of community acceptance? Pathog. Glob. Health 2020, 114, 55–63. [Google Scholar] [CrossRef]
  51. Fauci, A.S.; Morens, D.M. Zika Virus in the Americas—Yet Another Arbovirus Threat. N. Engl. J. Med. 2016, 374, 601–604. [Google Scholar] [CrossRef]
  52. Bhatt, S.; Gething, P.W.; Brady, O.J.; Messina, J.P.; Farlow, A.W.; Moyes, C.L.; Drake, J.M.; Brownstein, J.S.; Hoen, A.G.; Sankoh, O.; et al. The global distribution and burden of dengue. Nature 2013, 496, 504–507. [Google Scholar] [CrossRef]
  53. Roiz, D.; Wilson, A.L.; Scott, T.W.; Fonseca, D.M.; Jourdain, F.; Muller, P.; Velayudhan, R.; Corbel, V. Integrated Aedes management for the control of Aedes-borne diseases. PLoS Negl. Trop. Dis. 2018, 12, e0006845. [Google Scholar] [CrossRef]
  54. Rose, R.I. Pesticides and public health: Integrated methods of mosquito management. Emerg. Infect. Dis. 2001, 7, 17–23. [Google Scholar] [CrossRef]
  55. Zulfa, R.; Lo, W.C.; Cheng, P.C.; Martini, M.; Chuang, T.W. Updating the Insecticide Resistance Status of Aedes aegypti and Aedes albopictus in Asia: A Systematic Review and Meta-Analysis. Trop. Med. Infect. Dis. 2022, 7, 306. [Google Scholar] [CrossRef]
  56. Kasai, S.; Itokawa, K.; Uemura, N.; Takaoka, A.; Furutani, S.; Maekawa, Y.; Kobayashi, D.; Imanishi-Kobayashi, N.; Amoa-Bosompem, M.; Murota, K.; et al. Discovery of super-insecticide-resistant dengue mosquitoes in Asia: Threats of concomitant knockdown resistance mutations. Sci. Adv. 2022, 8, eabq7345. [Google Scholar] [CrossRef]
  57. Rants’o, T.A.; Koekemoer, L.L.; Panayides, J.L.; van Zyl, R.L. Potential of Essential Oil-Based Anticholinesterase Insecticides against Anopheles Vectors: A Review. Molecules 2022, 27, 7026. [Google Scholar] [CrossRef]
  58. Yu, K.X.; Jantan, I.; Ahmad, R.; Wong, C.L. The major bioactive components of seaweeds and their mosquitocidal potential. Parasitol. Res. 2014, 113, 3121–3141. [Google Scholar] [CrossRef]
  59. Zeni, V.; Baliota, G.V.; Benelli, G.; Canale, A.; Athanassiou, C.G. Diatomaceous Earth for Arthropod Pest Control: Back to the Future. Molecules 2021, 26, 7487. [Google Scholar] [CrossRef]
  60. Acheuk, F.; Basiouni, S.; Shehata, A.A.; Dick, K.; Hajri, H.; Lasram, S.; Yilmaz, M.; Emekci, M.; Tsiamis, G.; Spona-Friedl, M.; et al. Status and Prospects of Botanical Biopesticides in Europe and Mediterranean Countries. Biomolecules 2022, 12, 311. [Google Scholar] [CrossRef]
  61. Marston, A.; Maillard, M.; Hostettmann, K. Search for antifungal, molluscicidal and larvicidal compounds from African medicinal plants. J. Ethnopharmacol. 1993, 38, 215–223. [Google Scholar] [CrossRef] [PubMed]
  62. Piermarini, P.M.; Esquivel, C.J.; Denton, J.S. Malpighian Tubules as Novel Targets for Mosquito Control. Int. J. Environ. Res. Public Health 2017, 14, 111. [Google Scholar] [CrossRef] [PubMed]
  63. Swale, D.R.; Engers, D.W.; Bollinger, S.R.; Gross, A.; Inocente, E.A.; Days, E.; Kanga, F.; Johnson, R.M.; Yang, L.; Bloomquist, J.R.; et al. An insecticide resistance-breaking mosquitocide targeting inward rectifier potassium channels in vectors of Zika virus and malaria. Sci. Rep. 2016, 6, 36954. [Google Scholar] [CrossRef] [PubMed]
  64. Xu, J.; Hopkins, K.; Sabin, L.; Yasunaga, A.; Subramanian, H.; Lamborn, I.; Gordesky-Gold, B.; Cherry, S. ERK signaling couples nutrient status to antiviral defense in the insect gut. Proc. Natl. Acad. Sci. USA 2013, 110, 15025–15030. [Google Scholar] [CrossRef]
  65. Zhang, X.; Zhu, X.; Bi, X.; Huang, J.; Zhou, L. The Insulin Receptor: An Important Target for the Development of Novel Medicines and Pesticides. Int. J. Mol. Sci. 2022, 23, 7793. [Google Scholar] [CrossRef] [PubMed]
  66. Ekoka, E.; Maharaj, S.; Nardini, L.; Dahan-Moss, Y.; Koekemoer, L.L. 20-Hydroxyecdysone (20E) signaling as a promising target for the chemical control of malaria vectors. Parasit. Vectors 2021, 14, 86. [Google Scholar] [CrossRef] [PubMed]
  67. Marten, A.D.; Stothard, A.I.; Kalera, K.; Swarts, B.M.; Conway, M.J. Validamycin A Delays Development and Prevents Flight in Aedes aegypti (Diptera: Culicidae). J. Med. Entomol. 2020, 57, 1096–1103. [Google Scholar] [CrossRef]
  68. Marques, J.; Cardoso, J.C.R.; Felix, R.C.; Santana, R.A.G.; Guerra, M.; Power, D.; Silveira, H. Fresh-blood-free diet for rearing malaria mosquito vectors. Sci. Rep. 2018, 8, 17807. [Google Scholar] [CrossRef]
  69. da Silva Costa, G.; Rodrigues, M.M.S.; Silva, A.A.E. Toward a blood-free diet for Anopheles darlingi (Diptera: Culicidae). J. Med. Entomol. 2020, 57, 947–951. [Google Scholar] [CrossRef]
  70. Gonzales, K.K.; Hansen, I.A. Artificial Diets for Mosquitoes. Int. J. Environ. Res. Public Health 2016, 13, 1267. [Google Scholar] [CrossRef] [Green Version]
  71. Gonzales, K.K.; Rodriguez, S.D.; Chung, H.N.; Kowalski, M.; Vulcan, J.; Moore, E.L.; Li, Y.; Willette, S.M.; Kandel, Y.; Van Voorhies, W.A.; et al. The Effect of SkitoSnack, an Artificial Blood Meal Replacement, on Aedes aegypti Life History Traits and Gut Microbiota. Sci. Rep. 2018, 8, 11023. [Google Scholar] [CrossRef]
  72. Joy, T.K.; Arik, A.J.; Corby-Harris, V.; Johnson, A.A.; Riehle, M.A. The impact of larval and adult dietary restriction on lifespan, reproduction and growth in the mosquito Aedes aegypti. Exp. Gerontol. 2010, 45, 685–690. [Google Scholar] [CrossRef]
  73. Rocha-Santos, C.; Dutra, A.; Fróes Santos, R.; Cupolillo, C.; de Melo Rodovalho, C.; Bellinato, D.F.; Dos Santos Dias, L.; Jablonka, W.; Lima, J.B.P.; Silva Neto, M.A.C.; et al. Effect of Larval Food Availability on Adult Aedes Aegypti (Diptera: Culicidae) Fitness and Susceptibility to Zika Infection. J. Med. Entomol. 2021, 58, 535–547. [Google Scholar] [CrossRef]
  74. Reiskind, M.H.; Janairo, M.S. Late-instar Behavior of Aedes aegypti (Diptera: Culicidae) Larvae in Different Thermal and Nutritive Environments. J. Med. Entomol. 2015, 52, 789–796. [Google Scholar] [CrossRef]
  75. Marten, A.D.; Tift, C.T.; Tree, M.O.; Bakke, J.; Conway, M.J. Chronic depletion of vertebrate lipids in Aedes aegypti. cells dysregulates lipid metabolism and inhibits innate immunity without altering dengue infectivity. PLoS Negl. Trop. Dis. 2022, 16, e0010890. [Google Scholar] [CrossRef]
  76. Talyuli, O.A.; Bottino-Rojas, V.; Taracena, M.L.; Soares, A.L.; Oliveira, J.H.; Oliveira, P.L. The use of a chemically defined artificial diet as a tool to study Aedes aegypti physiology. J. Insect Physiol. 2015, 83, 1–7. [Google Scholar] [CrossRef]
  77. Naksathit, A.T.; Scott, T.W. Effect of female size on fecundity and survivorship of Aedes aegypti fed only human blood versus human blood plus sugar. J. Am. Mosq. Control Assoc. 1998, 14, 148–152. [Google Scholar]
  78. Mostowy, W.M.; Foster, W.A. Antagonistic effects of energy status on meal size and egg-batch size of Aedes aegypti (Diptera: Culicidae). J. Vector Ecol. 2004, 29, 84–93. [Google Scholar]
  79. Canyon, D.V.; Hii, J.L.; Muller, R. Effect of diet on biting, oviposition, and survival of Aedes aegypti (Diptera: Culicidae). J. Med. Entomol. 1999, 36, 301–308. [Google Scholar] [CrossRef]
  80. Khan, A.A.; Maibach, H.I. A study of the probing response of Aedes aegypti. 1. Effect of nutrition on probing. J. Econ. Entomol. 1970, 63, 974–976. [Google Scholar] [CrossRef]
  81. Kessler, S.; Vlimant, M.; Guerin, P.M. Sugar-sensitive neurone responses and sugar feeding preferences influence lifespan and biting behaviours of the Afrotropical malaria mosquito, Anopheles gambiae. J. Comp. Physiol. Neuroethol. Sens. Neural Behav. Physiol. 2015, 201, 317–329. [Google Scholar] [CrossRef]
  82. Duvall, L.B.; Ramos-Espiritu, L.; Barsoum, K.E.; Glickman, J.F.; Vosshall, L.B. Small-Molecule Agonists of Ae. aegypti Neuropeptide Y Receptor Block Mosquito Biting. Cell 2019, 176, 687–701.e685. [Google Scholar] [CrossRef] [Green Version]
  83. Holmes, C.J.; Brown, E.S.; Sharma, D.; Nguyen, Q.; Spangler, A.A.; Pathak, A.; Payton, B.; Warden, M.; Shah, A.J.; Shaw, S.; et al. Bloodmeal regulation in mosquitoes curtails dehydration-induced mortality, altering vectorial capacity. J. Insect Physiol. 2022, 137, 104363. [Google Scholar] [CrossRef]
  84. Chareonviriyaphap, T.; Kongmee, M.; Bangs, M.J.; Sathantriphop, S.; Meunworn, V.; Parbaripai, A.; Suwonkerd, W.; Akratanakul, P. Influence of nutritional and physiological status on behavioral responses of Aedes aegypti (Diptera: Culicidae) to deltamethrin and cypermethrin. J. Vector Ecol. 2006, 31, 89–101. [Google Scholar] [CrossRef]
  85. Norris, E.J.; Bloomquist, J.R. Nutritional status significantly affects toxicological endpoints in the CDC bottle bioassay. Pest Manag. Sci. 2022, 78, 743–748. [Google Scholar] [CrossRef]
  86. Huang, Y.S.; Lyons, A.C.; Hsu, W.W.; Park, S.L.; Higgs, S.; Vanlandingham, D.L. Differential outcomes of Zika virus infection in Aedes aegypti orally challenged with infectious blood meals and infectious protein meals. PLoS ONE 2017, 12, e0182386. [Google Scholar] [CrossRef]
  87. Almire, F.; Terhzaz, S.; Terry, S.; McFarlane, M.; Gestuveo, R.J.; Szemiel, A.M.; Varjak, M.; McDonald, A.; Kohl, A.; Pondeville, E. Sugar feeding protects against arboviral infection by enhancing gut immunity in the mosquito vector Aedes aegypti. PLoS Pathog. 2021, 17, e1009870. [Google Scholar] [CrossRef]
  88. Weng, S.C.; Tsao, P.N.; Shiao, S.H. Blood glucose promotes dengue virus infection in the mosquito Aedes aegypti. Parasit. Vectors 2021, 14, 376. [Google Scholar] [CrossRef]
  89. Arrese, E.L.; Soulages, J.L. Insect fat body: Energy, metabolism and regulation. Annu. Rev. Entomol. 2010, 55, 207–225. [Google Scholar] [CrossRef]
  90. Shukla, E.; Thorat, L.J.; Nath, B.B.; Gaikwad, S.M. Insect trehalase: Physiological significance and potential applications. Glycobiology 2015, 25, 357–367. [Google Scholar] [CrossRef]
  91. Alabaster, A.; Isoe, J.; Zhou, G.; Lee, A.; Murphy, A.; Day, W.A.; Miesfeld, R.L. Deficiencies in acetyl-CoA carboxylase and fatty acid synthase 1 differentially affect eggshell formation and blood meal digestion in Aedes aegypti. Insect Biochem. Mol. Biol. 2011, 41, 946–955. [Google Scholar] [CrossRef] [PubMed]
  92. Silva, E.; Santos, L.V.; Caiado, M.S.; Hastenreiter, L.S.N.; Fonseca, S.R.R.; Carbajal-de-la-Fuente, A.L.; Carvalho, M.G.; Pontes, E.G. The influence of larval density on triacylglycerol content in Aedes aegypti (Linnaeus) (Diptera: Culicidae). Arch. Insect Biochem. Physiol. 2021, 106, e21757. [Google Scholar] [CrossRef] [PubMed]
  93. Tose, L.V.; Weisbrod, C.R.; Michalkova, V.; Nouzova, M.; Noriega, F.G.; Fernandez-Lima, F. Following de novo triglyceride dynamics in ovaries of Aedes aegypti during the previtellogenic stage. Sci. Rep. 2021, 11, 9636. [Google Scholar] [CrossRef]
  94. Mensch, J.; Di Battista, C.; De Majo, M.S.; Campos, R.E.; Fischer, S. Increased size and energy reserves in diapausing eggs of temperate Aedes aegypti populations. J. Insect Physiol. 2021, 131, 104232. [Google Scholar] [CrossRef]
  95. Dou, X.; Chen, K.; Brown, M.R.; Strand, M.R. Multiple endocrine factors regulate nutrient mobilization and storage in Aedes aegypti during a gonadotrophic cycle. Insect Sci. 2022. Online Version of Record. [Google Scholar] [CrossRef]
  96. Wang, X.; Hou, Y.; Saha, T.T.; Pei, G.; Raikhel, A.S.; Zou, Z. Hormone and receptor interplay in the regulation of mosquito lipid metabolism. Proc. Natl. Acad. Sci. USA 2017, 114, E2709–E2718. [Google Scholar] [CrossRef]
  97. Morou, E.; Lirakis, M.; Pavlidi, N.; Zotti, M.; Nakagawa, Y.; Smagghe, G.; Vontas, J.; Swevers, L. A new dibenzoylhydrazine with insecticidal activity against Anopheles mosquito larvae. Pest Manag. Sci. 2013, 69, 827–833. [Google Scholar] [CrossRef]
  98. Childs, L.M.; Cai, F.Y.; Kakani, E.G.; Mitchell, S.N.; Paton, D.; Gabrieli, P.; Buckee, C.O.; Catteruccia, F. Disrupting Mosquito Reproduction and Parasite Development for Malaria Control. PLoS Pathog. 2016, 12, e1006060. [Google Scholar] [CrossRef]
  99. Price, D.P.; Schilkey, F.D.; Ulanov, A.; Hansen, I.A. Small mosquitoes, large implications: Crowding and starvation affects gene expression and nutrient accumulation in Aedes aegypti. Parasit. Vectors 2015, 8, 252. [Google Scholar] [CrossRef]
  100. Lyu, X.Y.; Wang, X.L.; Geng, D.Q.; Jiang, H.; Zou, Z. Juvenile hormone acts on male accessory gland function via regulating l-asparaginase expression and triacylglycerol mobilization in Aedes aegypti. Insect Sci. 2022. Online Version of Record. [Google Scholar] [CrossRef]
  101. Hou, Y.; Wang, X.L.; Saha, T.T.; Roy, S.; Zhao, B.; Raikhel, A.S.; Zou, Z. Temporal Coordination of Carbohydrate Metabolism during Mosquito Reproduction. PLoS Genet. 2015, 11, e1005309. [Google Scholar] [CrossRef] [PubMed]
  102. Wyatt, G.R.; Kale, G.F. The chemistry of insect hemolymph. II. Trehalose and other carbohydrates. J. Gen. Physiol. 1957, 40, 833–847. [Google Scholar] [CrossRef]
  103. Van Handel, E. The obese mosquito. J. Physiol. 1965, 181, 478–486. [Google Scholar] [CrossRef] [PubMed]
  104. Murphy, T.A.; Wyatt, G.R. Enzymatic regulation of trehalose and glycogen synthesis in the fat body of an insect. Nature 1964, 202, 1112–1113. [Google Scholar] [CrossRef] [PubMed]
  105. da Rocha Fernandes, M.; Martins, R.; Pessoa Costa, E.; Pacidônio, E.C.; Araujo de Abreu, L.; da Silva Vaz, I., Jr.; Moreira, L.A.; da Fonseca, R.N.; Logullo, C. The modulation of the symbiont/host interaction between Wolbachia pipientis and Aedes fluviatilis embryos by glycogen metabolism. PLoS ONE 2014, 9, e98966. [Google Scholar] [CrossRef]
  106. Vital, W.; Rezende, G.L.; Abreu, L.; Moraes, J.; Lemos, F.J.; Vaz Ida, S., Jr.; Logullo, C. Germ band retraction as a landmark in glucose metabolism during Aedes aegypti embryogenesis. BMC Dev. Biol. 2010, 10, 25. [Google Scholar] [CrossRef]
  107. Briegel, H.; Gut, T.; Lea, A.O. Sequential deposition of yolk components during oogenesis in an insect, Aedes aegypti (Diptera: Culicidae). J. Insect Physiol. 2003, 49, 249–260. [Google Scholar] [CrossRef]
  108. Naksathit, A.T.; Edman, J.D.; Scott, T.W. Partitioning of glycogen, lipid, and sugar in ovaries and body remnants of female Aedes aegypti (Diptera: Culicidae) fed human blood. J. Med. Entomol. 1999, 36, 18–22. [Google Scholar] [CrossRef]
  109. Elbein, A.D.; Pan, Y.T.; Pastuszak, I.; Carroll, D. New insights on trehalose: A multifunctional molecule. Glycobiology 2003, 13, 17R–27R. [Google Scholar] [CrossRef]
  110. Becker, A.; Schloder, P.; Steele, J.E.; Wegener, G. The regulation of trehalose metabolism in insects. Experientia 1996, 52, 433–439. [Google Scholar] [CrossRef]
  111. Katagiri, N.; Ando, O.; Yamashita, O. Reduction of glycogen in eggs of the silkworm, Bombyx mori, by use of a trehalase inhibitor, trehazolin, and diapause induction in glycogen-reduced eggs. J. Insect Physiol. 1998, 44, 1205–1212. [Google Scholar] [CrossRef] [PubMed]
  112. Liebl, M.; Nelius, V.; Kamp, G.; Ando, O.; Wegener, G. Fate and effects of the trehalase inhibitor trehazolin in the migratory locust (Locusta migratoria). J. Insect Physiol. 2010, 56, 567–574. [Google Scholar] [CrossRef] [PubMed]
  113. Tang, B.; Yang, M.; Shen, Q.; Xu, Y.; Wang, H.; Wang, S. Suppressing the activity of trehalase with validamycin disrupts the trehalose and chitin biosynthesis pathways in the rice brown planthopper, Nilaparvata lugens. Pestic. Biochem. Physiol. 2017, 137, 81–90. [Google Scholar] [CrossRef] [PubMed]
  114. Tatun, N.; Wangsantitham, O.; Tungjitwitayakul, J.; Sakurai, S. Trehalase activity in fungus-growing termite, Odontotermes feae (Isoptera: Termitideae) and inhibitory effect of validamycin. J. Econ. Entomol. 2014, 107, 1224–1232. [Google Scholar] [CrossRef]
  115. Thorat, L.J.; Gaikwad, S.M.; Nath, B.B. Trehalose as an indicator of desiccation stress in Drosophila melanogaster larvae: A potential marker of anhydrobiosis. Biochem. Biophys. Res. Commun. 2012, 419, 638–642. [Google Scholar] [CrossRef]
  116. Wegener, G.; Macho, C.; Schloder, P.; Kamp, G.; Ando, O. Long-term effects of the trehalase inhibitor trehazolin on trehalase activity in locust flight muscle. J. Exp. Biol. 2010, 213, 3852–3857. [Google Scholar] [CrossRef]
  117. Wegener, G.; Tschiedel, V.; Schloder, P.; Ando, O. The toxic and lethal effects of the trehalase inhibitor trehazolin in locusts are caused by hypoglycaemia. J. Exp. Biol. 2003, 206, 1233–1240. [Google Scholar] [CrossRef]
  118. Wolber, J.M.; Urbanek, B.L.; Meints, L.M.; Piligian, B.F.; Lopez-Casillas, I.C.; Zochowski, K.M.; Woodruff, P.J.; Swarts, B.M. The trehalose-specific transporter LpqY-SugABC is required for antimicrobial and anti-biofilm activity of trehalose analogues in Mycobacterium smegmatis. Carbohydr. Res. 2017, 450, 60–66. [Google Scholar] [CrossRef]
  119. Xia, Y.; Clarkson, J.M.; Charnley, A.K. Trehalose-hydrolysing enzymes of Metarhizium anisopliae and their role in pathogenesis of the tobacco hornworm, Manduca sexta. J. Invertebr. Pathol. 2002, 80, 139–147. [Google Scholar] [CrossRef]
  120. Zhang, L.; Qiu, L.Y.; Yang, H.L.; Wang, H.J.; Zhou, M.; Wang, S.G.; Tang, B. Study on the Effect of Wing Bud Chitin Metabolism and Its Developmental Network Genes in the Brown Planthopper, Nilaparvata lugens, by Knockdown of TRE Gene. Front. Physiol. 2017, 8, 750. [Google Scholar] [CrossRef]
  121. García, M.D.; Argüelles, J.C. Trehalase inhibition by validamycin A may be a promising target to design new fungicides and insecticides. Pest Manag. Sci. 2021, 77, 3832–3835. [Google Scholar] [CrossRef] [PubMed]
  122. Chen, J.; Zhang, D.; Yao, Q.; Zhang, J.; Dong, X.; Tian, H.; Zhang, W. Feeding-based RNA interference of a trehalose phosphate synthase gene in the brown planthopper, Nilaparvata lugens. Insect Mol. Biol. 2010, 19, 777–786. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, J.; Tang, B.; Chen, H.; Yao, Q.; Huang, X.; Zhang, D.; Zhang, W. Different functions of the insect soluble and membrane-bound trehalase genes in chitin biosynthesis revealed by RNA interference. PLoS ONE 2010, 5, e10133. [Google Scholar] [CrossRef]
  124. Kalera, K.; Stothard, A.I.; Woodruff, P.J.; Swarts, B.M. The role of chemoenzymatic synthesis in advancing trehalose analogues as tools for combatting bacterial pathogens. Chem. Commun. 2020, 56, 11528–11547. [Google Scholar] [CrossRef]
  125. Vaidyanathan, R.; Fleisher, A.E.; Minnick, S.L.; Simmons, K.A.; Scott, T.W. Nutritional stress affects mosquito survival and vector competence for West Nile virus. Vector Borne Zoonotic Dis. 2008, 8, 727–732. [Google Scholar] [CrossRef]
  126. Oringanje, C.; Delacruz, L.R.; Han, Y.; Luckhart, S.; Riehle, M.A. Overexpression of Activated AMPK in the Anopheles stephensi Midgut Impacts Mosquito Metabolism, Reproduction and Plasmodium Resistance. Genes 2021, 12, 119. [Google Scholar] [CrossRef]
  127. Liu, K.; Dong, Y.; Huang, Y.; Rasgon, J.L.; Agre, P. Impact of trehalose transporter knockdown on Anopheles gambiae stress adaptation and susceptibility to Plasmodium falciparum infection. Proc. Natl. Acad. Sci. USA 2013, 110, 17504–17509. [Google Scholar] [CrossRef]
  128. Somerville, A.G.T.; Gleave, K.; Jones, C.M.; Reimer, L.J. The consequences of Brugia malayi infection on the flight and energy resources of Aedes aegypti mosquitoes. Sci. Rep. 2019, 9, 18449. [Google Scholar] [CrossRef]
  129. Li, M.J.; Lan, C.J.; Gao, H.T.; Xing, D.; Gu, Z.Y.; Su, D.; Zhao, T.Y.; Yang, H.Y.; Li, C.X. Transcriptome analysis of Aedes aegypti Aag2 cells in response to dengue virus-2 infection. Parasit. Vectors 2020, 13, 421. [Google Scholar] [CrossRef]
  130. Lan, Q.; Massey, R.J. Subcellular localization of the mosquito sterol carrier protein-2 and sterol carrier protein-x. J. Lipid Res. 2004, 45, 1468–1474. [Google Scholar] [CrossRef]
  131. Fu, Q.; Inankur, B.; Yin, J.; Striker, R.; Lan, Q. Sterol Carrier Protein 2, a Critical Host Factor for Dengue Virus Infection, Alters the Cholesterol Distribution in Mosquito Aag2 Cells. J. Med. Entomol. 2015, 52, 1124–1134. [Google Scholar] [CrossRef] [PubMed]
  132. da Silva, J.B.; Navarro, D.M.; da Silva, A.G.; Santos, G.K.; Dutra, K.A.; Moreira, D.R.; Ramos, M.N.; Espíndola, J.W.; de Oliveira, A.D.; Brondani, D.J.; et al. Thiosemicarbazones as Aedes aegypti larvicidal. Eur. J. Med. Chem. 2015, 100, 162–175. [Google Scholar] [CrossRef] [PubMed]
  133. Mounce, B.C.; Poirier, E.Z.; Passoni, G.; Simon-Loriere, E.; Cesaro, T.; Prot, M.; Stapleford, K.A.; Moratorio, G.; Sakuntabhai, A.; Levraud, J.P.; et al. Interferon-Induced Spermidine-Spermine Acetyltransferase and Polyamine Depletion Restrict Zika and Chikungunya Viruses. Cell Host Microbe 2016, 20, 167–177. [Google Scholar] [CrossRef] [PubMed]
  134. Danielson, N.D.; Collins, J.; Stothard, A.I.; Dong, Q.Q.; Kalera, K.; Woodruff, P.J.; DeBosch, B.J.; Britton, R.A.; Swarts, B.M. Degradation-resistant trehalose analogues block utilization of trehalose by hypervirulent Clostridioides difficile. Chem. Commun. 2019, 55, 5009–5012. [Google Scholar] [CrossRef]
Figure 1. Metabolism of dietary and stored nutrients is critical for each stage in the mosquito life cycle and provides an opportunity to design next-generation insecticides that have minimal off-target effects. Next-generation insecticides can be developed that inhibit uptake of nutrients, transport of nutrients, hormone activity, catabolic activity, and energy utilization pathways.
Figure 1. Metabolism of dietary and stored nutrients is critical for each stage in the mosquito life cycle and provides an opportunity to design next-generation insecticides that have minimal off-target effects. Next-generation insecticides can be developed that inhibit uptake of nutrients, transport of nutrients, hormone activity, catabolic activity, and energy utilization pathways.
Viruses 15 00469 g001
Figure 2. (A) Overview of major stored sugar pathways in mosquitoes, with a focus on trehalose metabolism. (B) Structures of trehalase inhibitors validamycin A and 5-ThioTre.
Figure 2. (A) Overview of major stored sugar pathways in mosquitoes, with a focus on trehalose metabolism. (B) Structures of trehalase inhibitors validamycin A and 5-ThioTre.
Viruses 15 00469 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Conway, M.J.; Haslitt, D.P.; Swarts, B.M. Targeting Aedes aegypti Metabolism with Next-Generation Insecticides. Viruses 2023, 15, 469. https://doi.org/10.3390/v15020469

AMA Style

Conway MJ, Haslitt DP, Swarts BM. Targeting Aedes aegypti Metabolism with Next-Generation Insecticides. Viruses. 2023; 15(2):469. https://doi.org/10.3390/v15020469

Chicago/Turabian Style

Conway, Michael J., Douglas P. Haslitt, and Benjamin M. Swarts. 2023. "Targeting Aedes aegypti Metabolism with Next-Generation Insecticides" Viruses 15, no. 2: 469. https://doi.org/10.3390/v15020469

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop