Next Article in Journal
Advances in Engineering Circular RNA Vaccines
Previous Article in Journal
Baculovirus-Assisted Production of Bartonella bacilliformis Proteins: A Potential Strategy for Improving Serological Diagnosis of Carrion’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mosquito Gut Microbiota: A Review

1
Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai 200025, China
2
Digestive Disease Hospital of Shandong First Medical University, Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining 272000, China
3
World Health Organization Collaborating Centre for Tropical Diseases, Shanghai 200025, China
*
Authors to whom correspondence should be addressed.
Pathogens 2024, 13(8), 691; https://doi.org/10.3390/pathogens13080691
Submission received: 29 June 2024 / Revised: 29 July 2024 / Accepted: 6 August 2024 / Published: 15 August 2024

Abstract

:
Mosquitoes are vectors of many important human diseases. The prolonged and widespread use of insecticides has led to the development of mosquito resistance to these insecticides. The gut microbiota is considered the master of host development and physiology; it influences mosquito biology, disease pathogen transmission, and resistance to insecticides. Understanding the role and mechanisms of mosquito gut microbiota in mosquito insecticide resistance is useful for developing new strategies for tackling mosquito insecticide resistance. We searched online databases, including PubMed, MEDLINE, SciELO, Web of Science, and the Chinese Science Citation Database. We searched all terms, including microbiota and mosquitoes, or any specific genera or species of mosquitoes. We reviewed the relationships between microbiota and mosquito growth, development, survival, reproduction, and disease pathogen transmission, as well as the interactions between microbiota and mosquito insecticide resistance. Overall, 429 studies were included in this review after filtering 8139 search results. Mosquito gut microbiota show a complex community structure with rich species diversity, dynamic changes in the species composition over time (season) and across space (environmental setting), and variation among mosquito species and mosquito developmental stages (larval vs. adult). The community composition of the microbiota plays profound roles in mosquito development, survival, and reproduction. There was a reciprocal interaction between the mosquito midgut microbiota and virus infection in mosquitoes. Wolbachia, Asaia, and Serratia are the three most studied bacteria that influence disease pathogen transmission. The insecticide resistance or exposure led to the enrichment or reduction in certain microorganisms in the resistant mosquitoes while enhancing the abundance of other microorganisms in insect-susceptible mosquitoes, and they involved many different species/genera/families of microorganisms. Conversely, microbiota can promote insecticide resistance in their hosts by isolating and degrading insecticidal compounds or altering the expression of host genes and metabolic detoxification enzymes. Currently, knowledge is scarce about the community structure of mosquito gut microbiota and its functionality in relation to mosquito pathogen transmission and insecticide resistance. The new multi-omics techniques should be adopted to find the links among environment, mosquito, and host and bring mosquito microbiota studies to the next level.

1. Introduction

Microbiotas, including bacteria, archaea, protists, fungi, and viruses, are a range of microorganisms that may be commensal, mutualistic, or pathogenic and are found in and on other organisms. Human microbiome study started in the 17th century [1]. The human gut microbiota, especially its relationship with human health, is probably the best studied microbiota among all living organisms [1,2,3]. Bacteria are the largest and, to date, the most studied component of the human microbiota [4], as supported by the development of many types of antibiotics [5]. The gut microbiota is considered the master of host development and physiology [6,7,8]. The gut microbiota affects nutrient absorption, immunity development, direct defense against pathogens, and many other roles [9,10,11,12,13]. In the view of drug metabolism mediated by the gut microbiota, it includes the modification of the chemical structure of drugs by microbial metabolite enzymes and/or affecting the expression of host metabolizing enzymes such as cytochrome P450 [14]. These studies are parallel to the discovery of the metabolizing enzyme cytochrome P450 in insects, especially agricultural pests and human disease vectors such as mosquitoes [15,16,17], promoting the study of the mechanism of insecticide/pesticide resistance in insects. Nonetheless, research in microbiota is an area with rapid development, with the new techniques of multi-omics (genomics, metagenomics, proteomics, transcriptomics, metabolomics, and fluxomics) at the forefront [18].
In addition to humans and other vertebrates, insects also have complex and diverse gut microbiota [19,20,21,22]. Insect gut bacterial diversity is determined by environmental habitat, diet, developmental stage, and phylogeny of the host [23,24,25]. The gut microbiota of insects not only contribute to nutrition, protection from parasites and pathogens, modulation of immune responses, and communication [22], but also affect insect growth, development, survival, and fitness [22,26]. Particularly, there is a strong link between insect gut microbiota and insecticide resistance [27,28,29,30,31,32], mainly through insecticide degradation [32,33,34] and metabolic detoxification [31,35,36,37,38].
The mosquito (Culicidae family) gut microbiota study has increasingly gained attention [38,39,40]. Mosquitoes are vectors of many human and zoonotic diseases, including Anopheles, which transmits malaria parasites (the deadliest of all tropical infectious diseases); Aedes, which transmits dengue virus (DENV), Chikungunya virus (CHIKV), yellow fever virus, and Zika virus (ZIKV); and Culex, which transmits West Nile virus (WNV), filarial parasites, and different kinds of human and zoonotic encephalitis viruses (e.g., Japanese encephalitis virus, eastern equine encephalitis virus, La Crosse encephalitis virus, St. Louis encephalitis virus) [41,42,43]. Currently, no effective treatment or vaccine is available for many diseases caused by these pathogens, such as dengue and Chikungunya fevers, except for a small number of diseases such as malaria (drug treatment) and yellow fever (vaccine), leaving vector controls as the primary option for prophylaxis. Recently, many studies have linked these infections and diseases to the mosquito gut microbiota [39,40,44,45,46,47,48]. However, the interaction between disease transmission and mosquito gut microbiota is dependent on mosquito species and specific diseases. For example, in Aedes mosquitoes, most of the focus was on the preventive effect of the bacteria genus Wolbachia on Aedes-borne diseases such as dengue fever and Zika [47,49,50]. In WNV-infected Culex mosquitoes, the results were variable regarding the directionality of this relationship, although these studies suggested that bacteria of the genera Serratia and Enterobacter contribute to WNV development [39]. Studies of Culex mosquitoes in Asia show complex interactions between Japanese encephalitis virus and other virus infections in the mosquito gut [51,52]. However, studies on Plasmodium-Anopheles gut microbiota interactions are relatively limited in the literature [53], although it was reported that the microbiota of both the mosquito and the human host play important roles in Plasmodium parasite transmission, malaria progression, and clearance of Plasmodium infection [54].
Insecticides have been widely used since the 1950s in the form of indoor residual spraying (IRS) [55], ultra-low volume (ULV) outdoor spray [56], aerial spraying [57], and insecticide-treated nets [58], not only for mosquito control for public health, but also for agricultural pest control, which leads to the development of extensive resistance to all five classes of insecticides (pyrethroid, organochloride, carbamate, phosphorothioate, and pyrrole) in mosquitoes [17,59]. Mechanisms of mosquito insecticide resistance include knockdown resistance (kdr) caused by target site gene mutations [60], metabolic resistance caused by the elevated expression of esterases, monooxygenases P450 and glutathione S-transferases enzymes [61,62,63,64,65], and reduced insecticide penetration due to cuticle thickening and modification [66]. In addition, multiple resistance has become increasingly common in different mosquitoes [67,68]. Currently, the potential link between insect gut microbiota and insecticide resistance has been gradually revealed [16,69,70,71,72,73,74], especially host detoxification ability, which is currently the point of interest [16,35,75,76,77].
The aim of this study is to review mosquito gut microbiota, with special emphasis on the role of gut microbiota in preventing disease transmission and the interplay with insecticide resistance. The implication of mosquito microbiota in disease control has been reviewed previously [18,38], therefore it was not included in this review. In addition, this review focused on mosquito-transmitted pathogens that cause human diseases rather than zoonotic diseases.

2. Methods

2.1. Protocols

For this review, mosquito microbiota are defined as studies that involve both mosquitos and microbiota. General studies of individual microorganisms, such as molecular detections of an individual microorganism (for example, Wolbachia), were mentioned in this study, but they were either summarized in a special way (see the “Study Selection 2.4” below) or not considered in the general summary. However, studies to confirm the functionality of gut microorganisms were considered in this review. Field studies of Wolbachia as a disease control agency were excluded from this review.
The primary focuses of this review are (1) the interactions among the environment, mosquito microbiota, and disease transmission; (2) the key microorganisms that reside in mosquito bodies that are important for disease or vector control; and (3) the interactions between the microbiota and mosquito insecticide resistance.

2.2. Search Strategy

The search terms consisted of the 11 searches specified below. Although some of the searches overlap, we found that all were necessary.
  • (Mosquito microbiota) or (mosquito microbiome);
  • (Aedes aegypti microbiota);
  • (Aedes albopictus microbiota);
  • (Culex microbiota);
  • (Anopheles microbiota);
  • (Aedes microbiota);
  • (Armigeres microbiota);
  • (Haemagogus microbiota);
The following searches for special bacteria are based on a preliminary summary of published studies, which shows that these commonly occurring bacteria affect the development, survival, and insecticide resistance of mosquitoes, and pathogen infections in mosquitoes.
  • (Mosquito Wolbachia);
  • (Mosquito Asaia);
  • (Mosquito Serratia).
Wolbachia, Asaia, and Serratia are in fact the three most studied bacteria associated with the three classes of the deadliest mosquito-borne human pathogens, i.e., Aedes-borne dengue, Chikungunya, and Zika viruses, Anopheles-borne Plasmodium malaria parasites, and Culex-borne encephalitis virus (both human and zoonotic).
Databases searched include PubMed, MEDLINE, SciELO, Web of Science, and Chinese Science Citation Database (CSCD). SciELO focuses on Spanish- and Portuguese-language journals. CSCD includes all journals hosted by Chinese institutions. The date of the final search was 31 July 2023.

2.3. Study Eligibility Criteria

All studies involving microbiota and mosquitoes were included in the initial selection (Figure 1), including both larval and adult mosquitoes of any species. Studies including the following experiments were included: (1) general exploration of microbiota in mosquito larvae and adults; (2) the effect of microbiota on mosquito development, survival, and reproduction; (3) the effect of microbiota on pathogen transmission; and (4) the interactions between mosquito microbiota and mosquito larval or adult resistance to insecticides. However, pure semi-field or field experiments or trials involving the assessment of intervention efficacy or effectiveness under natural conditions, for example, field work evaluating the effectiveness of Wolbachia infection on Aedes population reduction, were included in the general review but not included in the final study selection.
  • Inclusion criteria:
Mosquito larval and adult laboratory experiments or analysis of the community structure of the microbiota;
Mosquito larval and adult laboratory experiments to confirm the functionality of specific microorganisms isolated from mosquito microbiota;
Mosquito larval and adult laboratory experiments that compare differences in microbiota community structures between mosquitoes collected from different sites or between mosquito species;
Mosquito larval and adult laboratory experiments or molecular analyses that study the interactions between mosquito insecticide resistance and microbiota or microorganisms isolated from mosquito microbiota.
  • Exclusion criteria:
Pure field or semi-field experiments under natural or semi-natural conditions to evaluate the efficacy or effectiveness of Wolbachia or other microorganisms in controlling mosquito populations or pathogen transmission;
Studies that purely focus on methodology development, pathogen or disease transmission, isolation, or confirmation of the existence of a single microorganism;
Studies that were conducted under unconventional conditions or with unconventional standards.

2.4. Study Selection

Studies were selected manually by checking their results, with exceptions for pure Wolbachia, Asaia, and Serratia studies (Figure 1). If a study included the words microbiota and mosquito in the title and abstract, the study was included in this review for further analysis. Data generated from studies under unconventional conditions or with unconventional standards were excluded from this review. For the special bacteria Wolbachia, Asaia, and Serratia and mosquito interactions, such as how these bacteria affect mosquito infection of pathogens (e.g., Wolbachia preventing dengue virus infection in Ae. aegypti), only up to five published articles for each bacteria were included in this review since most of these studies used similar methods and yielded very similar results.
The data were manually extracted. The outcomes include interactions between microbiota and mosquito development, survival, pathogen infection, and insecticide resistance. Although qualitatively summarized results were presented in this review, we did not conduct meta-analyses, especially quantitative summaries of published studies, as a broad range of results was presented in the results.

3. Results

3.1. Description of Search Results

The multiple combined search terms yielded a total of 893 records from regular searches and 7246 from special searches. After the removal of duplicates (n = 178) and articles with non-relevant contents (n = 92), 623 records from the regular search were deemed eligible for full-text screening (Figure 1). Among these, 100 records were not related to microbiota. Additional records were removed, including 59 review or opinion articles that are not closely related to microbiota and mosquitoes and 50 focused on studies of individual bacteria/viruses alone with mosquitoes (Figure 1). For the special search, only 15 articles were included in the final review. Overall, 429 studies were included in this review (Figure 1).

3.2. Mosquito Gut Microbiota: Environment, Disease Transmission, and Mosquito Development

The mosquito gut microbiota shows a complex community structure with rich species diversity, and dynamic changes in the species composition over time and across space [78,79,80,81,82,83]. For example, 18 phyla, 138 families, and 337 genera of microorganisms were identified from the larval gut of Anopheles gambiae sensu lato in naturally collected samples in Kenya, while only 42 families of microorganisms were found in the gut of a 3-day-old sugar-fed adult [79]. Meanwhile, the larval gut bacterial microbiota (dominant family Chlorophyta, 17%) has a similar structure as that in its habitat (dominant family Chlorophyta, 16%), while the diversity reduced as the larvae developed into pupa (dominant family Aeromonas, 31%) [79]. Furthermore, gut bacterial diversity and dominant species changed dramatically in adult mosquitos after sugar feeding (7-day-old, dominant family Elizabethkingia, 62%) and blood feeding (7-day-old, dominant family Elizabethkingia, 84%), indicating a lowered diversity [79]. A similar dynamic in gut microbiota community structure along with the growth stages was also reported in Aedes albopictus [78]. Interestingly, in a laboratory strain of Ae. albopictus fed with brewer’s yeast Saccharomyces cerevisiae cells, Wolbachia accounted for a tiny proportion of bacterial reads at the larval stage (7% at day 3), but it reached 80% of the total bacterial reads in 17-day-old adults [78]. For Culex mosquitoes, although the species of microorganisms and the community structure in the gut microbiota are very different from the above two mosquitoes, a general decreasing trend in the number of observed species and diversity was also observed over time [84]. In addition, substantial change was found in the diversity and relative abundance of different families (species, genera) of microbiota over seasons, regardless of mosquito species [84,85].
The spatial heterogeneity in the mosquito larval gut microbiota community is likely associated with their living environment. For example, significant differences were found in the gut microbial composition among different species of field-collected mosquitoes (Ae. albopictus, Aedes galloisi, Culex pallidothorax, Culex pipiens, Culex gelidus, and Armigeres subalbatus) at the same collection location and substantial variations in the gut microbiota among individuals of the same mosquito species collected at different geographical locations (eight sites in Hainan, China) [80]. However, similar bacterial diversity and evenness were found among mosquito species across the four genera, indicating that the mosquito midgut plays an important role in regulating the colonization and assembly of bacterial communities [80,86,87].
The microbiota has also been implicated in pathogen infections in mosquito vectors. It was reported that there was a complex microbiota community structure in the midgut of Aedes aegypti mosquitoes, in which there was a reciprocal interaction between the mosquito midgut microbiota and dengue virus infection, i.e., a marked decrease in susceptibility to dengue virus infection was found when Ae. aegypti was infected with the bacteria species Proteus sp. and Paenibacillus sp.; conversely, the dengue virus infection influenced the microbial load in the mosquito midgut [88]. In contrast, gut infection with Serratia marcescens in Ae. aegypti could facilitate dengue virus infection [89]. Moreover, Anopheles stephensi mosquitoes were more susceptible to Plasmodium berghei infection when their native microbiota was cleared with antibiotics compared to the untreated mosquitoes [90], indicating that bacteria in the midgut may help to prevent P. berghei infection. Similarly, a strong correlation between the abundance of bacterial Enterobacteriaceae and Plasmodium falciparum infection was found in An. gambiae mosquitoes [86]. In fact, P. falciparum development in the mosquito gut was affected by many bacterial isolates (e.g., Escherichia coli, Serratia marcescens, and Pseudomonas stutzeri) in challenge experiments [91].
Mosquito gut microbiota play profound roles in mosquito growth and development, survival, and reproduction [92,93,94,95]. For example, the Ae. aegypti mosquito’s survivorship and fecundity were reduced after midgut microbial clearance using antibiotic treatment [45]. Similarly, adult Anopheles arabiensis mosquito longevity could be increased after their midgut was treated with the broad-spectrum bactericidal antibiotic gentamycin, the Gram-positive narrow-spectrum antibiotic vancomycin, and the Gram-negative narrow-spectrum antibiotic streptomycin, while longevity was significantly reduced under treatment with the broad-spectrum bacteriostatic antibiotic erythromycins [93], indicating the differential influence of different bacteria in the mosquito gut and their complex balance in supporting mosquito survival. Furthermore, An. stepehsni adult longevity, fecundity, and infection with malaria parasites would be reduced after their midguts were treated with tetracycline [96]. Unfortunately, the exact microorganisms responsible for these changes were unknown in the above studies. Bacillus and Staphylococcus in the midgut were tested to be essential for the normal and high fecundity of Culex pipiens [97], and Wolbachia (wPip) could contribute to the higher longevity and improved survivorship of Culex quinquefasciatus, but with lower reproductive fitness [98,99]. In addition, the antibiotic-treated An. gambiae adult mosquitoes had a reduced lifespan with a median survival of 14 days, while the untreated mosquitoes survived up to a median of 22 days, whereas an increase in egg hatchability and larval development were observed after the reintroduction of Enterobacter cloacae and Serratia marcescens bacteria in the mosquito [100].

3.3. Key Bacteria in Mosquito Gut Microbiota: Importance for Preventing Disease Transmission

As illustrated earlier, bacteria are the most studied component of the gut microbiota. Being part of the microbiota community, bacterial symbionts are abundant in the mosquito gut (both larval and adult) and influence mosquito development, survival, and reproduction [83,93,96,101,102]. The community structure of bacterial endosymbionts has been examined in many mosquito species, including Ae. aegypti [103,104,105], Ae. albopictus [104,105], Ae. galloisi [80], Aedes triseriatus [106,107], Aedes japonicus [107,108], An. gambiae s.l. [104,109,110,111,112], Anopheles funestus [109,110], An. stephensi [104,113], Ar. subalbatus [80], Cx. gelidus [80,114], Cx. pallidothorax [80], Cx. pipiens [80,106,108,115], Cx. quinquefasciatus [116], Culex restuans [115], Culex tritaeniorhynchus [114], Mansonia annulifera [114], and Psorophora columbiae [106]. The bacterial community from mosquito gut is usually dominated by a few (4–5) genera regardless of mosquito species, although many species of bacteria have been found from mosquito gut [108,112,116]. For example, the most abundant genera were Enterobacter (32.8%) and Aeromonas (29.8%), followed by Pseudomonas (11.8%), Acinetobacter (5.9%), and Thorsellia (2.2%) [112] identified from the gut of An. gambiae. Similarly, four bacterial genera were the most abundant in gut from Cx. pipiens and Ae. japonicus, with the most prevalent genera being Sphingomonas and Rahnella, respectively [108]. Here, we focused the review on the three bacteria that may interfere with the transmission of these three pathogens.
The most well-known mosquito endosymbiont bacterial genus is probably the intracellular bacteria of Wolbachia, which infects a high proportion of insects and is possibly the most common reproductive parasite in the biosphere [117,118,119]. It has been commonly detected in different species of mosquitoes, including natural populations of Aedes, Culex, Mansonia, and Armigeres mosquitoes all over the world [120,121,122,123], and it is the predominant bacterial genus in many Aedes and Culex mosquitoes [78,80,115,124,125], but natural infection of Wolbachia in Anopheles mosquitoes is uncommon [126,127,128]. Wolbachia is of special interest due to its unique biological and pathogenetic characteristics that induce reproductive manipulation phenotypes, including parthenogenesis, feminization, cytoplasmic incompatibility, and male-killing, which increase the endosymbiont’s reproductive success [121,122,123,129]. Thus, it acts as a biocontrol agent against arbovirus vectors [130,131,132]. Although only Wolbachia-infected females can pass the infection on to their offspring, Wolbachia bacteria maximize their transmission by significantly altering the reproductive capacity of their hosts through male killing [133], feminization [134], parthenogenesis [135], and cytoplasmic incompatibility, respectively [125,126,136].
Biological control using Wolbachia is based on two management strategies, i.e., population suppression using the incompatible insect technique (IIT) and population replacement using anti-virus mosquito strains. Cytoplasmic incompatibility has been proposed as a tool to suppress mosquito populations and decrease the arbovirus burden in humans [132,137,138,139]. Population suppression occurs when males infected with Wolbachia are released into the environment to reproduce with Wolbachia-free females, which leads to cytoplasmic incompatibility between gametes and thus cannot produce viable offspring [49,50,140]. The continued release of infected males over time reduces the target mosquito population at a given site [141,142]. This strategy has been effectively used in different countries for Aedes mosquito control [141,142,143]. In the replacement strategy, females infected with a specific strain of Wolbachia can reduce the replication of arthropod-borne viruses (arboviruses) such as DENV, ZIKV, CHIKV, and WNV [49,50,99,144].
Secondly, Asaia is a versatile acetic acid bacterial symbiont capable of cross-colonizing insects of phylogenetically distant genera and orders of agricultural insects and mosquitoes [145,146,147]. It is commonly found in Anopheles mosquitoes, including An. gambuae, An. stephensi, and An. funestus, as well as Monsonia mosquitoes, and is the dominant bacterial symbiont [148,149,150]. For example, it was found that the bacterial genus Asaia predominated the adult internal and cuticle surface microbiota in Anopheles albimanus, making up at least 70% of the taxa in each microbial niche across all collection sites [66]. Moreover, Asaia has also been detected in Aedes, Culex, and other mosquito species [124,147,148]. Interestingly, co-infection of Wolbachia and Asaia in mosquitoes is uncommon, although it has been detected in uncommon Aedes mosquito species other than Ae. aegypti and Ae. albopictus [147,151], and in some other mosquitoes such as Culex and Haemagogus species [124,151,152]. Meanwhile, the abundance of Asaia is usually very low in these co-infected mosquitoes compared to the accompanied mosquitoes [124,147,151]. In fact, it was reported that the presence of one of the symbionts could prevent the establishment of the second one in some mosquito species, and there were microbial niche differences between Wolbachia and Asaia, i.e., a reciprocal negative interference in terms of the colonization of the gonads [104]. The mutual exclusion or competition between Asaia and Wolbachia may help explain the inability of Wolbachia to colonize the female reproductive organs of Anopheles mosquitoes, inhibit its vertical transmission, and explain the absence of Wolbachia infection in Ae. aegypti and in the majority of natural populations of Anopheles mosquitoes [104]. Furthermore, this co-exclusion pattern between Wolbachia and Asaia was also found in Ae. albopictus and Cx. quinquefasciatus naturally, and Asaia is able to colonize reproductive organs and salivary glands in species uninfected with Wolbachia such as An. gambiae, An. Stephensi, and Ae. aegypti [81,104,151,153]. More importantly, it was found that the infection of Asaia in Anopheles mosquiotes could inhibit Plasmodium infections [154,155], which renders its potential as a paratransgenic weapon against malaria [156,157].
Thirdly, Serratia is another important genus of mosquito gut bacteria, which is a genus of Gram-negative, facultatively anaerobic, rod-shaped bacteria of the family Enterobacteriaceae. It has been found in many mosquito species, including Ae. aegypti, Culex, Anopheles, and Armigeres mosquitoes, and has been linked to mosquito development and survival, insecticide resistance, and malaria infections [80,100,150,158,159,160]. Moreover, it can be genetically engineered to prevent malaria parasites in mosquitoes [161,162].

3.4. Mosquito Gut Microbiota Mediating Insecticide Resistance

The impacts of insecticides on mosquito microbiota have been studied on most mosquitoes, including Ae. aegypti [47,73,75,163], Ae. albopictus [164,165], An. albimanus [166], An. arabiensis [93], An. coluzzi [150], Anopheles gambiae sensu stricto [149], An. stephensi [28], Cx. pipiens [167], and Cx. quinquefasciatus [74] (Table 1). Most focused on adult mosquitoes and chemical insecticides, with a lesser focus on larval and microbial larvicide Bacillus thuringiensis var. israelensis (Bti). In all cases, insecticide resistance or exposure leads to the enrichment or reduction in certain microorganisms in resistant mosquitoes while enhancing the abundance of other microorganisms in insect-susceptible mosquitoes, and they involve many different species/genera/families of microorganisms [28,74,168] (Table 1). For example, the genera Coprococcus and Ruminococcus (class Clostridia) [169], Bilophila (class Deltaproteobacteria), Enterobacter (class Gammaproteobacteria), Porphyromonas (class Bacteroidia), Bifidobacterium (class Actinobacteria) [168], Weissella (class Bacilli), and Delftia (class Betaproteobacteria) were enriched in the resistant group of Ae. aegypti [47] (Table 1). Whereas the species of Bacteroides faecichinchillae was significantly decreased in the midguts of resistant Ae. aegypti and Akkermansia muciniphila was increased in insecticide-susceptible mosquitoes [47,163] (Table 1). However, resistance development is usually accompanied by reduced diversity in microorganisms [69]. In addition, these changes in microbiota community structure and predominant species or genera of microorganisms vary among mosquito species and among different studies of the same species at different sites. For example, An. albimanus resistant to fenitrothion, an organophosphorus insecticide, enriched the abundance of Bacillus and Klebsiella pneumoniae, while the resistance to pyrethroids increased the abundance of Pseudomonas fragi and Pantoea agglomerans [69,166,167] (Table 1).
Conversely, there are effects of gut microbiota on insecticide resistance. Microbiota can promote insecticide resistance in their hosts by isolating and degrading insecticidal compounds or altering the expression of host genes [31]. Due to the tedious nature of the infection process, only the bacteria Wolbachia, Streptococcus pyrogenes, Escherichia coli, Serratia oryzae, and Acinetobacter junii [93,164,165,170,171,172], and the fungi Metarhizium anisopliae or Beauveria bassiana [173,174,175] have been used in the microorganism challenge studies [173,174,175,176] (Table 2), but the results are inconsistent. Scathes et al. found that adding cultured gut bacteria isolated from mosquito larvae to antibiotic-cleared larval food can significantly reduce Ae. aegypti larval mortality against propoxur and naled larvicides [177]. Wang et al. further found that the survival of Serratia oryzae-enriched Ae. albopictus mosquitos significantly increased under deltamethrin, and metabolic detoxification enzymes esterase, P450, and GST were also increased in S. oryzae-enriched mosquitoes [165]. Meanwhile, carboxylesterase activity was detected in S. oryzae, which can degrade deltamethrin in vitro, and the degradation efficiency was positively correlated with time and bacterial amount [165] (Table 2). Inversely, it was found that supplementation of the bacteria Streptococcus pyrogenes or Escherichia coli could increase insecticide tolerance to the insecticides deltamethrin and malathion but decrease tolerance under antibiotic treatment via sugar feeding [93]. However, it was found that α-esterase activity was decreased in both resistant and susceptible females after antibiotic (gentamicin, streptomycin, and vancomycin) or bacteria enrichment (heat-killed S. pyrogenes or live E. coli) treatment, while GST and P450 enzyme activities remained unchanged [93] (Table 2). These unusual results may be caused by other resistance mechanisms. More controversial is the case of fungi Metarhizium anisopliae or Beauveria bassiana [93,173,174,175,178,179]. Clearly, mosquitoes will be killed by infection with both fungi, regardless of insecticide resistance levels [175,179]. In fact, Me. anisopliae infection can lead to very high (up to 97%) mortality in Ae. aegypti, An. Stephensi, and Cx. quinquefasciatus larvae [176,179]. On the other hand, the activities of acetylcholinesterase (AChE), glutathione S-transferase (GST), esterase (EST), acid phosphatases (ACP), and alkaline phosphatases (ALP) were increased in the chlorpyrifos-selected Cx. quinquefasciatus mosquitoes but suppressed when exposed to Me. anisopliae or Be. bassiana [173] (Table 2). Therefore, whether the anti-mosquito effects of the combination of these fungi and insecticides are additive or synergistic warrants further investigation.
In addition, the species composition of the gut microbiota has been linked to insect susceptibility to insecticides [31,180]. Although many insecticide-degrading microorganisms have been reported from soil samples or agricultural insects [18,35,76,181,182,183], few species have been reported from mosquitoes [69] (Table 3). We listed some insecticide-degrading microorganisms found in mosquito guts in Table 3; however, the function was only confirmed in one study [69].

3.5. Perspectives

So far, most mosquito gut microbiota studies are more descriptive and exploratory and lack in-depth analysis. Future studies on the mosquito gut microbiota and its functions should better utilize advanced techniques such as state-of-the-art multi-omics tools [18] (Figure 2). Exploring the community ecology of mosquito gut microbiota is necessary; however, we need to further investigate the function of the community structure or the function of the major symbiont microorganisms, and the environment—mosquito gut microbiota—host interactions. These studies can target DNA, RNA, proteins, biochemicals, and complex mixing levels (Figure 2). At the core of all studies is in vitro and in vivo laboratory identification and functional confirmation (Figure 2). Genomics (DNA) and transcriptomics (RNA) can be used to identify gut microbiota community composition and structure; proteomics can be used to examine protein expression levels; metabolomics can be used to assess the metabolites associated with certain functions, such as P450 enzyme levels versus insecticide resistance intensity; and metagenomics can be used to investigate the genetics of multiple sources of samples [18]. Most of these techniques have been used in different studies but not in an integral minor [18,31,184].
There are many unsolved issues in environment–microbiota–mosquito (host) interactions. We proposed four major areas for future studies of the mosquito microbiota (Figure 3). Some of these questions can be answered directly. For example, differences in the microbiota community at different mosquito stages can be solved through simple DNA/RNA sequencing (Figure 3, Question 1). However, what causes these differences may need further in vitro or in vivo experiments to confirm. Why do disease agents such as viruses co-exist with mosquitoes but cause human disease (Figure 3, Question 2)? Do disease-agent-carrying mosquitoes have different microbiota communities than disease-agent-free mosquitoes? In other words, does the microbiota support mosquitos coexisting with these disease agents? Studies have found that mosquito larvae and adults have different levels of tolerance to the same insecticide [185,186]. Does this have anything to do mosquito microbiota community structure (Figure 3, Question 3)? Mosquito gut microbial clearance and reintroduction studies find that the existence of gut microbiota affects mosquito metabolic enzyme levels. What is the pathway for this effect (Figure 3, Question 4)? We know that some bacteria can produce metabolic enzymes [187]. Does the mosquito microbiota directly release microbial enzymes to metabolize insecticides? Or do microorganisms influence the host receptors and signaling pathways through microbial metabolites (Figure 3, Question 4)? Many of these questions may be answered by the integration of multiple techniques; this may be the most robust way to move science forward.

3.6. Concluding Remarks

Compared to the extensive research that has been accomplished with microbiota in agricultural insects [76,183], research in mosquito microbiota needs a major catch-up. Although microbiota certainly affect mosquito resistance to insecticides, evidence is scarce after reviewing the existing literature. Insecticide selections enrich certain microorganisms in mosquito microbiota, but the specific microorganisms selected depend on the type of insecticides and mosquito species. Based on experiments involving the removal/addition of individual microorganisms, it seems that insecticide resistance in mosquitoes is likely supported by a combination of multiple microorganisms. Here are some of the areas that need in-depth investigations, e.g., diversity of symbiotic microbiota and their role in mosquito physiology and ecology, functions of mosquito gut microbiota at either individual microorganism or group/community levels, molecular mechanisms of insecticide resistance by insect gut microbiota using multi-omics approaches, the role of gut microbiota in the biodegradation of insecticides, and microbial metabolic pathways related to insecticide resistance. Mosquitoes may go through different physiological and/or genetic changes, such as gene mutations, to defend against insecticides; meanwhile, changes in mosquito microbiota, either actively or passively, may contribute to the mosquito’s defense system in a complementary or supplementary way. The extent of these roles is still unclear and awaits further study. Understanding the relationships between mosquito gut microbiota and mosquito insecticide resistance and the mechanisms by which the microbiota affects mosquito insecticide resistance is useful for developing new strategies for tackling insecticide resistance.

Author Contributions

Data acquisition, H.L., X.H., C.Z. and Y.Z. Data sorting and analysis, H.L. Funding acquisition, H.L., J.Y., J.C. and M.G. Draft writing, H.L. Writing—review and editing, H.L., X.H., J.Y., J.C. and M.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Taishan Scholars Project of Shandong Province (No. tsqn202312373 to H.L.); the National Natural Science Foundation of China (81702034 o H.L.); the Open Project of NHC Key Laboratory of Parasite and Vector Biology (NHCKFKT2021-02 to H.L. and J.C.); the Ministry of Education Industry-University Cooperative Education Project (No. 230726153807227 to H.L.); the Innovation Project of Shandong Academy of Medical Sciences; and the Three-Year Initiative Plan for Strengthening Public Health System Construction in Shanghai (2023–2025) Principal Investigator Project (No. GWVI-11.2-XD34 to J.Y.); Science Foundation of Shandong First Medical University (Shandong Academy of Medical Sciences) (202201-041 to D.H.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data generated or analyzed during this study are included in this published article and its additional files.

Acknowledgments

We sincerely thank the anonymous reviewers and editors for their constructive comments and suggestions to make it a better paper.

Conflicts of Interest

The authors declare that they have no competing interests.

References

  1. Gotschlich, E.C.; Colbert, R.A.; Gill, T. Methods in microbiome research: Past, present, and future. Best Pract. Res. Clin. Rheumatol. 2019, 33, 101498. [Google Scholar] [CrossRef] [PubMed]
  2. Human Microbiome Project C. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef] [PubMed]
  3. Lloyd-Price, J.; Mahurkar, A.; Rahnavard, G.; Crabtree, J.; Orvis, J.; Hall, A.B.; Brady, A.; Creasy, H.H.; McCracken, C.; Giglio, M.G.; et al. Strains, functions and dynamics in the expanded Human Microbiome Project. Nature 2017, 550, 61–66. [Google Scholar] [CrossRef] [PubMed]
  4. Beaugerie, L.; Petit, J.C. Microbial-gut interactions in health and disease. Antibiotic-associated diarrhoea. Best. Pract. Res. Clin. Gastroenterol. 2004, 18, 337–352. [Google Scholar] [CrossRef]
  5. Baran, A.; Kwiatkowska, A.; Potocki, L. Antibiotics and Bacterial Resistance-A short story of an endless arms race. Int. J. Mol. Sci. 2023, 24, 5777. [Google Scholar] [CrossRef] [PubMed]
  6. Durack, J.; Lynch, S.V. The gut microbiome: Relationships with disease and opportunities for therapy. J. Exp. Med. 2019, 216, 20–40. [Google Scholar] [CrossRef] [PubMed]
  7. Integrative HMPRNC. The Integrative Human Microbiome Project: Dynamic analysis of microbiome-host omics profiles during periods of human health and disease. Cell Host Microbe 2014, 16, 276–289. [Google Scholar] [CrossRef] [PubMed]
  8. Sommer, F.; Backhed, F. The gut microbiota—Masters of host development and physiology. Nat. Rev. Microbiol. 2013, 11, 227–238. [Google Scholar] [CrossRef] [PubMed]
  9. Gibson, G.R.; Roberfroid, M.B. Dietary modulation of the human colonic microbiota: Introducing the concept of prebiotics. J. Nutr. 1995, 125, 1401–1412. [Google Scholar] [CrossRef]
  10. Cho, I.; Blaser, M.J. The human microbiome: At the interface of health and disease. Nat. Rev. Genet. 2012, 13, 260–270. [Google Scholar] [CrossRef]
  11. Koppel, N.; Maini Rekdal, V.; Balskus, E.P. Chemical transformation of xenobiotics by the human gut microbiota. Science 2017, 356, 6344. [Google Scholar] [CrossRef] [PubMed]
  12. Cao, H.; Xu, M.; Dong, W.; Deng, B.; Wang, S.; Zhang, Y.; Wang, S.; Luo, S.; Wang, W.; Qi, Y.; et al. Secondary bile acid-induced dysbiosis promotes intestinal carcinogenesis. Int. J. Cancer 2017, 140, 2545–2556. [Google Scholar] [CrossRef]
  13. Dinan, T.G.; Cryan, J.F. The impact of gut microbiota on brain and behaviour: Implications for psychiatry. Curr. Opin. Clin. Nutr. Metab. Care 2015, 18, 552–558. [Google Scholar] [CrossRef]
  14. Spanogiannopoulos, P.; Bess, E.N.; Carmody, R.N.; Turnbaugh, P.J. The microbial pharmacists within us: A metagenomic view of xenobiotic metabolism. Nat. Rev. Microbiol. 2016, 14, 273–287. [Google Scholar] [CrossRef] [PubMed]
  15. Salam, L.B.; Obayori, O.S.; Ilori, M.O.; Amund, O.O. Deciphering the cytochrome P450 genes in the microbiome of a chronically polluted soil with history of agricultural activities. Bull. Natl. Res. Cent. 2022, 46, 256. [Google Scholar] [CrossRef]
  16. Sun, Z.; Liu, Y.; Xu, H.; Yan, C. Genome-wide identification of P450 genes in chironomid Propsilocerus akamusi reveals candidate genes involved in gut microbiotamediated detoxification of chlorpyrifos. Insects 2022, 13, 765. [Google Scholar] [CrossRef]
  17. Yunta, C.; Ooi, J.M.F.; Oladepo, F.; Grafanaki, S.; Pergantis, S.A.; Tsakireli, D.; Ismail, H.M.; Paine, M.J.I. Chlorfenapyr metabolism by mosquito P450s associated with pyrethroid resistance identifies potential activation markers. Sci. Rep. 2023, 13, 14124. [Google Scholar]
  18. Malla, M.A.; Dubey, A.; Raj, A.; Kumar, A.; Upadhyay, N.; Yadav, S. Emerging frontiers in microbe-mediated pesticide remediation: Unveiling role of omics and In silico approaches in engineered environment. Environ. Pollut. 2022, 299, 118851. [Google Scholar] [CrossRef]
  19. Brune, A. Symbiotic digestion of lignocellulose in termite guts. Nat. Rev. Microbiol. 2014, 12, 168–180. [Google Scholar] [CrossRef]
  20. Mikaelyan, A.; Dietrich, C.; Kohler, T.; Poulsen, M.; Sillam-Dusses, D.; Brune, A. Diet is the primary determinant of bacterial community structure in the guts of higher termites. Mol. Ecol. 2015, 24, 5284–5295. [Google Scholar] [CrossRef]
  21. Engel, P.; Moran, N.A. The gut microbiota of insects—Diversity in structure and function. FEMS Microbiol. Rev. 2013, 37, 699–735. [Google Scholar] [CrossRef] [PubMed]
  22. Schmidt, K.; Engel, P. Mechanisms underlying gut microbiota-host interactions in insects. J. Exp. Biol. 2021, 224, jeb207696. [Google Scholar] [CrossRef] [PubMed]
  23. Yun, J.H.; Roh, S.W.; Whon, T.W.; Jung, M.J.; Kim, M.S.; Park, D.S.; Yoon, C.; Nam, Y.D.; Kim, Y.J.; Choi, J.H.; et al. Insect gut bacterial diversity determined by environmental habitat, diet, developmental stage, and phylogeny of host. Appl. Environ. Microbiol. 2014, 80, 5254–5264. [Google Scholar] [CrossRef] [PubMed]
  24. Guo, D.; Ge, J.; Tang, Z.; Tian, B.; Li, W.; Li, C.; Xu, L.; Luo, J. Dynamic gut microbiota of Apolygus lucorum across different life stages reveals potential pathogenic bacteria for facilitating the pest management. Microb. Ecol. 2023, 87, 9. [Google Scholar] [CrossRef] [PubMed]
  25. Awad, W.A.; Mann, E.; Dzieciol, M.; Hess, C.; Schmitz-Esser, S.; Wagner, M.; Hess, M. Age-related differences in the luminal and mucosa-associated gut microbiome of broiler chickens and shifts associated with Campylobacter jejuni Infection. Front. Cell. Infect. Microbiol. 2016, 6, 154. [Google Scholar] [CrossRef]
  26. Ishigami, K.; Jang, S.; Itoh, H.; Kikuchi, Y. Obligate gut symbiotic association with caballeronia in the mulberry seed bug Paradieuches dissimilis (Lygaeoidea: Rhyparochromidae). Microb. Ecol. 2023, 86, 1307–1318. [Google Scholar] [CrossRef]
  27. Kikuchi, Y.; Hayatsu, M.; Hosokawa, T.; Nagayama, A.; Tago, K.; Fukatsu, T. Symbiont-mediated insecticide resistance. Proc. Natl. Acad. Sci. USA 2012, 109, 8618–8622. [Google Scholar] [CrossRef]
  28. Soltani, A.; Vatandoost, H.; Oshaghi, M.A.; Enayati, A.A.; Chavshin, A.R. The role of midgut symbiotic bacteria in resistance of Anopheles stephensi (Diptera: Culicidae) to organophosphate insecticides. Pathog. Glob. Health 2017, 111, 289–296. [Google Scholar] [CrossRef]
  29. Gressel, J. Microbiome facilitated pest resistance: Potential problems and uses. Pest Manag. Sci. 2018, 74, 511–515. [Google Scholar] [CrossRef]
  30. Chaitra, H.S.; Kalia, V.K. Gut symbionts: Hidden players of pesticide resistance in insects. Indian. J. Entomol. 2022, 84, 997–1002. [Google Scholar]
  31. Sato, Y.; Jang, S.; Takeshita, K.; Itoh, H.; Koike, H.; Tago, K.; Hayatsu, M.; Hori, T.; Kikuchi, Y. Insecticide resistance by a host-symbiont reciprocal detoxification. Nat. Commun. 2021, 12, 6432. [Google Scholar] [CrossRef] [PubMed]
  32. Ishigami, K.; Jang, S.; Itoh, H.; Kikuchi, Y. Insecticide resistance governed by gut symbiosis in a rice pest, Cletus punctiger, under laboratory conditions. Biol. Lett. 2021, 17, 20200780. [Google Scholar] [CrossRef]
  33. Ahmad, S.; Ahmad, H.W.; Bhatt, P. Microbial adaptation and impact into the pesticide’s degradation. Arch. Microbiol. 2022, 204, 288. [Google Scholar] [CrossRef]
  34. Itoh, H.; Hori, T.; Sato, Y.; Nagayama, A.; Tago, K.; Hayatsu, M.; Kikuchi, Y. Infection dynamics of insecticide-degrading symbionts from soil to insects in response to insecticide spraying. ISME J. 2018, 12, 909–920. [Google Scholar] [CrossRef]
  35. Jaffar, S.; Ahmad, S.; Lu, Y. Contribution of insect gut microbiota and their associated enzymes in insect physiology and biodegradation of pesticides. Front. Microbiol. 2022, 13, 979383. [Google Scholar] [CrossRef]
  36. Zhang, Y.; Cai, T.; Yuan, M.; Li, Z.; Jin, R.; Ren, Z.; Qin, Y.; Yu, C.; Cai, Y.; Shu, R.; et al. Microbiome variation correlates with the insecticide susceptibility in different geographic strains of a significant agricultural pest, Nilaparvata lugens. NPJ Biofilms Microbiomes 2023, 9, 2. [Google Scholar] [CrossRef]
  37. Daisley, B.A.; Trinder, M.; McDowell, T.W.; Collins, S.L.; Sumarah, M.W.; Reid, G. Microbiota-mediated modulation of organophosphate insecticide toxicity by species-dependent interactions with Lactobacilli in a Drosophila melanogaster insect model. Appl. Environ. Microbiol. 2018, 84, e02820-17. [Google Scholar] [CrossRef]
  38. Gao, H.; Cui, C.; Wang, L.; Jacobs-Lorena, M.; Wang, S. Mosquito microbiota and implications for disease control. Trends Parasitol. 2020, 36, 98–111. [Google Scholar] [CrossRef] [PubMed]
  39. Garrigos, M.; Garrido, M.; Panisse, G.; Veiga, J.; Martinez-de la Puente, J. Interactions between West Nile Virus and the microbiota of Culex pipiens vectors: A literature review. Pathogens 2023, 12, 1287. [Google Scholar] [CrossRef] [PubMed]
  40. Wang, J.; Gao, L.; Aksoy, S. Microbiota in disease-transmitting vectors. Nat. Rev. Microbiol. 2023, 21, 604–618. [Google Scholar] [CrossRef]
  41. Lee, H.; Halverson, S.; Ezinwa, N. Mosquito-borne diseases. Prim. Care 2018, 45, 393–407. [Google Scholar] [CrossRef] [PubMed]
  42. Tolle, M.A. Mosquito-borne diseases. Curr. Probl. Pediatr. Adolesc. Health Care 2009, 39, 97–140. [Google Scholar] [CrossRef]
  43. WHO. Mosquito-Borne Diseases Geneva; WHO: Geneva, Switzerland, 2020.
  44. Song, X.; Zhong, Z.; Gao, L.; Weiss, B.L.; Wang, J. Metabolic interactions between disease-transmitting vectors and their microbiota. Trends Parasitol. 2022, 38, 697–708. [Google Scholar] [CrossRef] [PubMed]
  45. Hill, C.L.; Sharma, A.; Shouche, Y.; Severson, D.W. Dynamics of midgut microflora and dengue virus impact on life history traits in Aedes aegypti. Acta Trop. 2014, 140, 151–157. [Google Scholar] [CrossRef]
  46. Rodpai, R.; Boonroumkaew, P.; Sadaow, L.; Sanpool, O.; Janwan, P.; Thanchomnang, T.; Intapan, P.M.; Maleewong, W. Microbiome composition and microbial community structure in mosquito vectors Aedes aegypti and Aedes albopictus in Northeastern Thailand, a dengue-endemic area. Insects 2023, 14, 184. [Google Scholar] [CrossRef] [PubMed]
  47. Arevalo-Cortes, A.; Damania, A.; Granada, Y.; Zuluaga, S.; Mejia, R.; Triana-Chavez, O. Association of midgut bacteria and their metabolic pathways with Zika infection and insecticide resistance in colombian Aedes aegypti populations. Viruses 2022, 14, 2197. [Google Scholar] [CrossRef] [PubMed]
  48. Djihinto, O.Y.; Medjigbodo, A.A.; Gangbadja, A.R.A.; Saizonou, H.M.; Lagnika, H.O.; Nanmede, D.; Djossou, L.; Bohounton, R.; Sovegnon, P.M.; Fanou, M.J.; et al. Malaria-transmitting vectors microbiota: Overview and interactions with Anopheles mosquito biology. Front. Microbiol. 2022, 13, 891573. [Google Scholar] [CrossRef]
  49. 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]
  50. Nazni, W.A.; Hoffmann, A.A.; NoorAfizah, A.; Cheong, Y.L.; Mancini, M.V.; Golding, N.; Kamarul, G.M.R.; Arif, M.A.K.; Thohir, H.; NurSyamimi, H.; et al. Establishment of Wolbachia strain wAlbB in malaysian populations of Aedes aegypti for dengue control. Curr. Biol. 2019, 29, 4241–4248.e5. [Google Scholar] [CrossRef]
  51. Sanborn, M.A.; Wuertz, K.M.; Kim, H.C.; Yang, Y.; Li, T.; Pollett, S.D.; Jarman, R.G.; Berry, I.M.; Klein, T.A.; Hang, J. Metagenomic analysis reveals Culex mosquito virome diversity and Japanese encephalitis genotype V. in the Republic of Korea. Mol. Ecol. 2021, 30, 5470–5487. [Google Scholar] [CrossRef]
  52. Faizah, A.N.; Kobayashi, D.; Isawa, H.; Amoa-Bosompem, M.; Murota, K.; Higa, Y.; Futami, K.; Shimada, S.; Kim, K.S.; Itokawa, K.; et al. Deciphering the virome of Culex vishnui subgroup mosquitoes, the major vectors of Japanese Encephalitis, in Japan. Viruses 2020, 12, 264. [Google Scholar] [CrossRef] [PubMed]
  53. Demirci, M.; Yildiz Zeyrek, F. Malaria and gut microbiota: Microbial interactions in the host. Mikrobiyol. Bul. 2022, 56, 763–775. [Google Scholar] [PubMed]
  54. Omondi, Z.N.; Caner, A. An overview on the impact of microbiota on malaria transmission and severity: Plasmodium-vector-host axis. Acta Parasitol. 2022, 67, 1471–1486. [Google Scholar] [CrossRef] [PubMed]
  55. Muirhead-Thomson, R.C. DDT and gammexane as residual insecticides against Anopheles gambiae in African houses. Trans. R. Soc. Trop. Med. Hyg. 1950, 43, 401–412. [Google Scholar] [CrossRef] [PubMed]
  56. Farooq, M.; Cilek, J.E.; Sumners, E.; Briley, A.K.C.; Weston, J.; Richardson, A.G.; Lindroth, E.J. Potential of outdoor ultra-low-volume aerosol and thermal fog tosuppress the dengue vector, Aedes aegypti, inside dwellings. J. Am. Mosq. Control Assoc. 2020, 36, 189–196. [Google Scholar] [CrossRef] [PubMed]
  57. Ruktanonchai, D.J.; Stonecipher, S.; Lindsey, N.; McAllister, J.; Pillai, S.K.; Horiuchi, K.; Delorey, M.; Biggerstaff, B.J.; Sidwa, T.; Zoretic, J.; et al. Effect of aerial insecticide spraying on West Nile virus disease—North-central Texas, 2012. Am. J. Trop. Med. Hyg. 2014, 91, 240–245. [Google Scholar] [CrossRef]
  58. Gleave, K.; Lissenden, N.; Richardson, M.; Ranson, H. Piperonyl butoxide (PBO) combined with pyrethroids in long-lasting insecticidal nets (LLINs) to prevent malaria in Africa. Cochrane Database Syst. Rev. 2017, 11, CD012776. [Google Scholar] [CrossRef]
  59. WHO. Global Report on Insecticide Resistance in Malaria Vectors: 2010–2016; WHO: Geneva, Switzerland, 2018.
  60. Al Nazawi, A.M.; Aqili, J.; Alzahrani, M.; McCall, P.J.; Weetman, D. Combined target site (kdr) mutations play a primary role in highly pyrethroid resistant phenotypes of Aedes aegypti from Saudi Arabia. Parasit. Vectors 2017, 10, 161. [Google Scholar] [CrossRef]
  61. Ochomo, E.; Bayoh, M.N.; Brogdon, W.G.; Gimnig, J.E.; Ouma, C.; Vulule, J.M.; Walker, E.D. Pyrethroid resistance in Anopheles gambiae s.s. and Anopheles arabiensis in western Kenya: Phenotypic, metabolic and target site characterizations of three populations. Med. Vet. Entomol. 2013, 27, 156–164. [Google Scholar] [CrossRef]
  62. Muthusamy, R.; Shivakumar, M.S. Involvement of metabolic resistance and F1534C kdr mutation in the pyrethroid resistance mechanisms of Aedes aegypti in India. Acta Trop. 2015, 148, 137–141. [Google Scholar] [CrossRef]
  63. Adedeji, E.O.; Ogunlana, O.O.; Fatumo, S.; Beder, T.; Ajamma, Y.; Koenig, R.; Adebiyi, E. Anopheles metabolic proteins in malaria transmission, prevention and control: A review. Parasit. Vectors 2020, 13, 465. [Google Scholar] [CrossRef] [PubMed]
  64. Rahman, R.U.; Souza, B.; Uddin, I.; Carrara, L.; Brito, L.P.; Costa, M.M.; Mahmood, M.A.; Khan, S.; Lima, J.B.P.; Martins, A.J. Insecticide resistance and underlying targets-site and metabolic mechanisms in Aedes aegypti and Aedes albopictus from Lahore, Pakistan. Sci. Rep. 2021, 11, 4555. [Google Scholar] [CrossRef] [PubMed]
  65. Zhou, G.; Li, Y.; Jeang, B.; Wang, X.; Cummings, R.F.; Zhong, D.; Yan, G. Emerging mosquito resistance to piperonyl butoxide-synergized pyrethroid insecticide and its mechanism. J. Med. Entomol. 2022, 59, 638–647. [Google Scholar] [CrossRef] [PubMed]
  66. Dada, N.; Benedict, A.C.; López, F.; Lol, J.C.; Sheth, M.; Dzuris, N.; Padilla, N.; Lenhart, A. Comprehensive characterization of internal and cuticle surface microbiota of laboratory-reared F(1) Anopheles albimanus originating from different sites. Malar. J. 2021, 20, 414. [Google Scholar] [CrossRef] [PubMed]
  67. Keita, M.; Sogoba, N.; Kane, F.; Traore, B.; Zeukeng, F.; Coulibaly, B.; Sodio, A.B.; Traore, S.F.; Djouaka, R.; Doumbia, S. Multiple resistance mechanisms to pyrethroids insecticides in Anopheles gambiae sensu lato population from mali, West Africa. J. Infect. Dis. 2021, 223, S81–S90. [Google Scholar] [CrossRef] [PubMed]
  68. Li, Y.; Zhou, G.; Zhong, D.; Wang, X.; Hemming-Schroeder, E.; David, R.E.; Lee, M.C.; Zhong, S.; Yi, G.; Liu, Z.; et al. Widespread multiple insecticide resistance in the major dengue vector Aedes albopictus in Hainan Province, China. Pest Manag. Sci. 2021, 77, 1945–1953. [Google Scholar] [CrossRef] [PubMed]
  69. Dada, N.; Sheth, M.; Liebman, K.; Pinto, J.; Lenhart, A. Whole metagenome sequencing reveals links between mosquito microbiota and insecticide resistance in malaria vectors. Sci. Rep. 2018, 8, 2084. [Google Scholar] [CrossRef] [PubMed]
  70. Xia, X.; Sun, B.; Gurr, G.M.; Vasseur, L.; Xue, M.; You, M. Gut microbiota mediate insecticide resistance in the diamondback moth, Plutella xylostella (L.). Front. Microbiol. 2018, 9, 25. [Google Scholar] [CrossRef] [PubMed]
  71. Yuan, X.; Pan, Z.; Jin, C.; Ni, Y.; Fu, Z.; Jin, Y. Gut microbiota: An underestimated and unintended recipient for pesticide-induced toxicity. Chemosphere 2019, 227, 425–434. [Google Scholar] [CrossRef]
  72. Wang, S.; Wen, Q.; Qin, Y.; Xia, Q.; Shen, C.; Song, S. Gut microbiota and host cytochrome P450 characteristics in the pseudo germ-free model: Co-contributors to a diverse metabolic landscape. Gut Pathog. 2023, 15, 15. [Google Scholar] [CrossRef]
  73. Gomez-Govea, M.A.; Ramirez-Ahuja, M.L.; Contreras-Perera, Y.; Jimenez-Camacho, A.J.; Ruiz-Ayma, G.; Villanueva-Segura, O.K.; Trujillo-Rodriguez, G.J.; Delgado-Enciso, I.; Martinez-Fierro, M.L.; Manrique-Saide, P.; et al. Suppression of midgut microbiota impact pyrethroid susceptibility in Aedes aegypti. Front. Microbiol. 2022, 13, 761459. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, Y.T.; Shen, R.X.; Xing, D.; Zhao, C.P.; Gao, H.T.; Wu, J.H.; Zhang, N.; Zhang, H.D.; Chen, Y.; Zhao, T.Y.; et al. Metagenome sequencing reveals the midgut microbiota makeup of Culex pipiens quinquefasciatus and its possible relationship with insecticide resistance. Front. Microbiol. 2021, 12, 625539. [Google Scholar] [CrossRef] [PubMed]
  75. Muturi, E.J.; Dunlap, C.; Smartt, C.T.; Shin, D. Resistance to permethrin alters the gut microbiota of Aedes aegypti. Sci. Rep. 2021, 11, 14406. [Google Scholar] [CrossRef] [PubMed]
  76. Siddiqui, J.A.; Khan, M.M.; Bamisile, B.S.; Hafeez, M.; Qasim, M.; Rasheed, M.T.; Rasheed, M.A.; Ahmad, S.; Shahid, M.I.; Xu, Y. Role of Insect Gut Microbiota in Pesticide Degradation: A Review. Front. Microbiol. 2022, 13, 870462. [Google Scholar] [CrossRef] [PubMed]
  77. Giambo, F.; Teodoro, M.; Costa, C.; Fenga, C. Toxicology and microbiota: How do pesticides influence gut microbiota? A review. Int. J. Environ. Res. Public Health 2021, 18, 5510. [Google Scholar] [CrossRef] [PubMed]
  78. Wang, X.; Liu, T.; Wu, Y.; Zhong, D.; Zhou, G.; Su, X.; Xu, J.; Sotero, C.F.; Sadruddin, A.A.; Wu, K.; et al. Bacterial microbiota assemblage in Aedes albopictus mosquitoes and its impacts on larval development. Mol. Ecol. 2018, 27, 2972–2985. [Google Scholar] [CrossRef]
  79. Wang, Y.; Gilbreath, T.M.; Kukutla, P., 3rd; Yan, G.; Xu, J. Dynamic gut microbiome across life history of the malaria mosquito Anopheles gambiae in Kenya. PLoS ONE 2011, 6, e24767. [Google Scholar] [CrossRef] [PubMed]
  80. Kang, X.; Wang, Y.; Li, S.; Sun, X.; Lu, X.; Rajaofera, M.J.N.; Lu, Y.; Kang, L.; Zheng, A.; Zou, Z.; et al. Comparative analysis of the gut microbiota of adult mosquitoes from eight locations in Hainan, China. Front. Cell. Infect. Microbiol. 2020, 10, 596750. [Google Scholar] [CrossRef]
  81. Guegan, M.; Zouache, K.; Demichel, C.; Minard, G.; Tran Van, V.; Potier, P.; Mavingui, P.; Valiente Moro, C. The mosquito holobiont: Fresh insight into mosquito-microbiota interactions. Microbiome 2018, 6, 49. [Google Scholar] [CrossRef]
  82. Singh, A.; Allam, M.; Kwenda, S.; Khumalo, Z.T.H.; Ismail, A.; Oliver, S.V. The dynamic gut microbiota of zoophilic members of the Anopheles gambiae complex (Diptera: Culicidae). Sci. Rep. 2022, 12, 1495. [Google Scholar] [CrossRef]
  83. Ngo, C.T.; Aujoulat, F.; Veas, F.; Jumas-Bilak, E.; Manguin, S. Bacterial diversity associated with wild caught Anopheles mosquitoes from Dak Nong Province, Vietnam using culture and DNA fingerprint. PLoS ONE 2015, 10, e0118634. [Google Scholar] [CrossRef] [PubMed]
  84. Didion, E.M.; Doyle, M.; Benoit, J.B. Bacterial Communities of Lab and Field Northern House Mosquitoes (Diptera: Culicidae) Throughout Diapause. J. Med. Entomol. 2022, 59, 648–658. [Google Scholar] [CrossRef] [PubMed]
  85. Akorli, J.; Gendrin, M.; Pels, N.A.; Yeboah-Manu, D.; Christophides, G.K.; Wilson, M.D. Seasonality and locality affect the diversity of Anopheles gambiae and Anopheles coluzzii midgut microbiota from Ghana. PLoS ONE 2016, 11, e0157529. [Google Scholar] [CrossRef] [PubMed]
  86. Boissiere, A.; Tchioffo, M.T.; Bachar, D.; Abate, L.; Marie, A.; Nsango, S.E.; Shahbazkia, H.R.; Awono-Ambene, P.H.; Levashina, E.A.; Christen, R.; et al. Midgut microbiota of the malaria mosquito vector Anopheles gambiae and interactions with Plasmodium falciparum infection. PLoS Pathog. 2012, 8, e1002742. [Google Scholar] [CrossRef] [PubMed]
  87. Seabourn, P.; Spafford, H.; Yoneishi, N.; Medeiros, M. The Aedes albopictus (Diptera: Culicidae) microbiome varies spatially and with Ascogregarine infection. PLoS Negl. Trop. Dis. 2020, 14, e0008615. [Google Scholar] [CrossRef] [PubMed]
  88. Ramirez, J.L.; Souza-Neto, J.; Torres Cosme, R.; Rovira, J.; Ortiz, A.; Pascale, J.M.; Dimopoulos, G. Reciprocal tripartite interactions between the Aedes aegypti midgut microbiota, innate immune system and dengue virus influences vector competence. PLoS Negl. Trop. Dis. 2012, 6, e1561. [Google Scholar] [CrossRef] [PubMed]
  89. Wu, P.; Sun, P.; Nie, K.; Zhu, Y.; Shi, M.; Xiao, C.; Liu, H.; Liu, Q.; Zhao, T.; Chen, X.; et al. A gut commensal bacterium promotes mosquito permissiveness to arboviruses. Cell Host Microbe 2019, 25, 101–112.e5. [Google Scholar] [CrossRef] [PubMed]
  90. Kalappa, D.M.; Subramani, P.A.; Basavanna, S.K.; Ghosh, S.K.; Sundaramurthy, V.; Uragayala, S.; Tiwari, S.; Anvikar, A.R.; Valecha, N. Influence of midgut microbiota in Anopheles stephensi on Plasmodium berghei infections. Malar. J. 2018, 17, 385. [Google Scholar] [CrossRef] [PubMed]
  91. Tchioffo, M.T.; Boissiere, A.; Churcher, T.S.; Abate, L.; Gimonneau, G.; Nsango, S.E.; Awono-Ambene, P.H.; Christen, R.; Berry, A.; Morlais, I. Modulation of malaria infection in Anopheles gambiae mosquitoes exposed to natural midgut bacteria. PLoS ONE 2013, 8, e81663. [Google Scholar] [CrossRef]
  92. Xiao, X.; Yang, L.; Pang, X.; Zhang, R.; Zhu, Y.; Wang, P.; Gao, G.; Cheng, G. A Mesh-Duox pathway regulates homeostasis in the insect gut. Nat. Microbiol. 2017, 2, 17020. [Google Scholar] [CrossRef]
  93. Barnard, K.; Jeanrenaud, A.; Brooke, B.D.; Oliver, S.V. The contribution of gut bacteria to insecticide resistance and the life histories of the major malaria vector Anopheles arabiensis (Diptera: Culicidae). Sci. Rep. 2019, 9, 9117. [Google Scholar] [CrossRef] [PubMed]
  94. Chabanol, E.; Behrends, V.; Prevot, G.; Christophides, G.K.; Gendrin, M. Antibiotic treatment in Anopheles coluzzii affects carbon and nitrogen metabolism. Pathogens 2020, 9, 679. [Google Scholar] [CrossRef]
  95. Romoli, O.; Schonbeck, J.C.; Hapfelmeier, S.; Gendrin, M. Production of germ-free mosquitoes via transient colonisation allows stage-specific investigation of host-microbiota interactions. Nat. Commun. 2021, 12, 942. [Google Scholar] [CrossRef] [PubMed]
  96. Sharma, A.; Dhayal, D.; Singh, O.P.; Adak, T.; Bhatnagar, R.K. Gut microbes influence fitness and malaria transmission potential of Asian malaria vector Anopheles stephensi. Acta Trop. 2013, 128, 41–47. [Google Scholar] [CrossRef] [PubMed]
  97. Fouda, M.A.; Hassan, M.I.; Al-Daly, A.G.; Hammad, K.M. Effect of midgut bacteria of Culex pipiens L. on digestion and reproduction. J. Egypt. Soc. Parasitol. 2001, 31, 767–780. [Google Scholar]
  98. Almeida, F.; Moura, A.S.; Cardoso, A.F.; Winter, C.E.; Bijovsky, A.T.; Suesdek, L. Effects of Wolbachia on fitness of Culex quinquefasciatus (Diptera; Culicidae). Infect. Genet. Evol. 2011, 11, 2138–2143. [Google Scholar] [CrossRef]
  99. Alomar, A.A.; Perez-Ramos, D.W.; Kim, D.; Kendziorski, N.L.; Eastmond, B.H.; Alto, B.W.; Caragata, E.P. Native Wolbachia infection and larval competition stress shape fitness and West Nile virus infection in Culex quinquefasciatus mosquitoes. Front. Microbiol. 2023, 14, 1138476. [Google Scholar] [CrossRef]
  100. Ezemuoka, L.C.; Akorli, E.A.; Aboagye-Antwi, F.; Akorli, J. Mosquito midgut Enterobacter cloacae and Serratia marcescens affect the fitness of adult female Anopheles gambiae s.l. PLoS ONE 2020, 15, e0238931. [Google Scholar] [CrossRef]
  101. Mitraka, E.; Stathopoulos, S.; Siden-Kiamos, I.; Christophides, G.K.; Louis, C. Asaia accelerates larval development of Anopheles gambiae. Pathog. Glob. Health 2013, 107, 305–311. [Google Scholar] [CrossRef]
  102. Fouda, M.A.; Hassan, M.I.; Hammad, K.M.; Hasaballah, A.I. Effects of midgut bacteria and two protease inhibitors on the reproductive potential and midgut enzymes of Culex pipiens infected with Wuchereria bancrofti. J. Egypt. Soc. Parasitol. 2013, 43, 537–546. [Google Scholar] [CrossRef]
  103. Gusmao, D.S.; Santos, A.V.; Marini, D.C.; Bacci, M., Jr.; Berbert-Molina, M.A.; Lemos, F.J. Culture-dependent and culture-independent characterization of microorganisms associated with Aedes aegypti (Diptera: Culicidae) (L.) and dynamics of bacterial colonization in the midgut. Acta Trop. 2010, 115, 275–281. [Google Scholar] [CrossRef] [PubMed]
  104. Rossi, P.; Ricci, I.; Cappelli, A.; Damiani, C.; Ulissi, U.; Mancini, M.V.; Valzano, M.; Capone, A.; Epis, S.; Crotti, E.; et al. Mutual exclusion of Asaia and Wolbachia in the reproductive organs of mosquito vectors. Parasit. Vectors 2015, 8, 278. [Google Scholar] [CrossRef]
  105. Ranasinghe, K.; Gunathilaka, N.; Amarasinghe, D.; Rodrigo, W.; Udayanga, L. Diversity of midgut bacteria in larvae and females of Aedes aegypti and Aedes albopictus from Gampaha District, Sri Lanka. Parasit. Vectors 2021, 14, 433. [Google Scholar] [CrossRef] [PubMed]
  106. Demaio, J.; Pumpuni, C.B.; Kent, M.; Beier, J.C. The midgut bacterial flora of wild Aedes triseriatus, Culex pipiens, and Psorophora columbiae mosquitoes. Am. J. Trop. Med. Hyg. 1996, 54, 219–223. [Google Scholar] [CrossRef]
  107. Kim, C.H.; Lampman, R.L.; Muturi, E.J. Bacterial communities and midgut microbiota associated with mosquito populations from Waste Tires in East-Central Illinois. J. Med. Entomol. 2015, 52, 63–75. [Google Scholar] [CrossRef] [PubMed]
  108. Zotzmann, S.; Steinbrink, A.; Schleich, K.; Frantzmann, F.; Xoumpholphakdy, C.; Spaeth, M.; Moro, C.V.; Mavingui, P.; Klimpel, S. Bacterial diversity of cosmopolitan Culex pipiens and invasive Aedes japonicus from Germany. Parasitol. Res. 2017, 116, 1899–1906. [Google Scholar] [CrossRef] [PubMed]
  109. Straif, S.C.; Mbogo, C.N.; Toure, A.M.; Walker, E.D.; Kaufman, M.; Toure, Y.T.; Beier, J.C. Midgut bacteria in Anopheles gambiae and An. funestus (Diptera: Culicidae) from Kenya and Mali. J. Med. Entomol. 1998, 35, 222–226. [Google Scholar] [CrossRef] [PubMed]
  110. Lindh, J.M.; Terenius, O.; Faye, I. 16S rRNA gene-based identification of midgut bacteria from field-caught Anopheles gambiae sensu lato and A. funestus mosquitoes reveals new species related to known insect symbionts. Appl. Environ. Microbiol. 2005, 71, 7217–7223. [Google Scholar] [CrossRef]
  111. Buck, M.; Nilsson, L.K.; Brunius, C.; Dabire, R.K.; Hopkins, R.; Terenius, O. Bacterial associations reveal spatial population dynamics in Anopheles gambiae mosquitoes. Sci. Rep. 2016, 6, 22806. [Google Scholar] [CrossRef]
  112. Zoure, A.A.; Sare, A.R.; Yameogo, F.; Somda, Z.; Massart, S.; Badolo, A.; Francis, F. Bacterial communities associated with the midgut microbiota of wild Anopheles gambiae complex in Burkina Faso. Mol. Biol. Rep. 2020, 47, 211–224. [Google Scholar] [CrossRef] [PubMed]
  113. Favia, G.; Ricci, I.; Damiani, C.; Raddadi, N.; Crotti, E.; Marzorati, M.; Rizzi, A.; Urso, R.; Brusetti, L.; Borin, S.; et al. Bacteria of the genus Asaia stably associate with Anopheles stephensi, an Asian malarial mosquito vector. Proc. Natl. Acad. Sci. USA 2007, 104, 9047–9051. [Google Scholar] [CrossRef] [PubMed]
  114. Gunathilaka, N.; Ranasinghe, K.; Amarasinghe, D.; Rodrigo, W.; Mallawarachchi, H.; Chandrasena, N. Molecular characterization of culturable aerobic bacteria in the midgut of field-caught Culex tritaeniorhynchus, Culex gelidus, and Mansonia annulifera Mosquitoes in the Gampaha District of Sri Lanka. Biomed. Res. Int. 2020, 2020, 8732473. [Google Scholar] [CrossRef] [PubMed]
  115. Muturi, E.J.; Kim, C.H.; Bara, J.; Bach, E.M.; Siddappaji, M.H. Culex pipiens and Culex restuans mosquitoes harbor distinct microbiota dominated by few bacterial taxa. Parasit. Vectors 2016, 9, 18. [Google Scholar] [CrossRef] [PubMed]
  116. Chandel, K.; Mendki, M.J.; Parikh, R.Y.; Kulkarni, G.; Tikar, S.N.; Sukumaran, D.; Prakash, S.; Parashar, B.D.; Shouche, Y.S.; Veer, V. Midgut microbial community of Culex quinquefasciatus mosquito populations from India. PLoS ONE 2013, 8, e80453. [Google Scholar] [CrossRef] [PubMed]
  117. Duron, O.; Bouchon, D.; Boutin, S.; Bellamy, L.; Zhou, L.; Engelstadter, J.; Hurst, G.D. The diversity of reproductive parasites among arthropods: Wolbachia do not walk alone. BMC Biol. 2008, 6, 27. [Google Scholar] [CrossRef] [PubMed]
  118. Taylor, M.J.; Bordenstein, S.R.; Slatko, B. Microbe Profile: Wolbachia: A sex selector, a viral protector and a target to treat filarial nematodes. Microbiology 2018, 164, 1345–1347. [Google Scholar] [CrossRef] [PubMed]
  119. Kozek, W.J.; Rao, R.U. The Discovery of Wolbachia in Arthropods and Nematodes—A Historical Perspective; Karger Publishers: Basel, Switzerland, 2007; pp. 1–14. [Google Scholar]
  120. Li, Y.; Sun, Y.; Zou, J.; Zhong, D.; Liu, R.; Zhu, C.; Li, W.; Zhou, Y.; Cui, L.; Zhou, G.; et al. Characterizing the Wolbachia infection in field-collected Culicidae mosquitoes from Hainan Province, China. Parasit. Vectors 2023, 16, 128. [Google Scholar] [CrossRef]
  121. Wong, M.L.; Liew, J.W.K.; Wong, W.K.; Pramasivan, S.; Mohamed Hassan, N.; Wan Sulaiman, W.Y.; Jeyaprakasam, N.K.; Leong, C.S.; Low, V.L.; Vythilingam, I. Natural Wolbachia infection in field-collected Anopheles and other mosquito species from Malaysia. Parasit. Vectors 2020, 13, 414. [Google Scholar] [CrossRef] [PubMed]
  122. Nugapola, N.; De Silva, W.; Karunaratne, S. Distribution and phylogeny of Wolbachia strains in wild mosquito populations in Sri Lanka. Parasit. Vectors 2017, 10, 230. [Google Scholar] [CrossRef] [PubMed]
  123. Ding, H.; Yeo, H.; Puniamoorthy, N. Wolbachia infection in wild mosquitoes (Diptera: Culicidae): Implications for transmission modes and host-endosymbiont associations in Singapore. Parasit. Vectors 2020, 13, 612. [Google Scholar] [CrossRef]
  124. da Silva, H.; Oliveira, T.M.P.; Sallum, M.A.M. Bacterial community diversity and bacterial interaction network in eight mosquito species. Genes 2022, 13, 2052. [Google Scholar] [CrossRef] [PubMed]
  125. Flores, G.A.M.; Lopez, R.P.; Cerrudo, C.S.; Perotti, M.A.; Consolo, V.F.; Beron, C.M. Wolbachia dominance influences the Culex quinquefasciatus microbiota. Sci. Rep. 2023, 13, 18980. [Google Scholar] [CrossRef] [PubMed]
  126. Chrostek, E.; Gerth, M. Is Anopheles gambiae a natural host of Wolbachia? mBio 2019, 10, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  127. Jeffries, C.L.; Lawrence, G.G.; Golovko, G.; Kristan, M.; Orsborne, J.; Spence, K.; Hurn, E.; Bandibabone, J.; Tantely, L.M.; Raharimalala, F.N.; et al. Novel Wolbachia strains in Anopheles malaria vectors from Sub-Saharan Africa. Wellcome Open Res. 2018, 3, 113. [Google Scholar] [CrossRef] [PubMed]
  128. Walker, T.; Quek, S.; Jeffries, C.L.; Bandibabone, J.; Dhokiya, V.; Bamou, R.; Kristan, M.; Messenger, L.A.; Gidley, A.; Hornett, E.A.; et al. Stable high-density and maternally inherited Wolbachia infections in Anopheles moucheti and Anopheles demeilloni mosquitoes. Curr. Biol. 2021, 31, 2310–2320.e5. [Google Scholar] [CrossRef] [PubMed]
  129. Moretti, R.; Calvitti, M. Male mating performance and cytoplasmic incompatibility in a wPip Wolbachia trans-infected line of Aedes albopictus (Stegomyia albopicta). Med. Vet. Entomol. 2013, 27, 377–386. [Google Scholar] [CrossRef]
  130. Araujo, N.J.S.; Macedo, M.J.F.; de Morais, L.P.; da Cunha, F.A.B.; de Matos, Y.; de Almeida, R.S.; Braga, M.; Coutinho, H.D.M. Control of arboviruses vectors using biological control by Wolbachia pipientis: A short review. Arch. Microbiol. 2022, 204, 376. [Google Scholar] [CrossRef]
  131. Mohanty, I.; Rath, A.; Mahapatra, N.; Hazra, R.K. Wolbachia: A biological control strategy against arboviral diseases. J. Vector Borne Dis. 2016, 53, 199–207. [Google Scholar]
  132. Iturbe-Ormaetxe, I.; Walker, T.; O’Neill, S.L. Wolbachia and the biological control of mosquito-borne disease. EMBO Rep. 2011, 12, 508–518. [Google Scholar] [CrossRef]
  133. Hurst, G.D.D.; Jiggins, F.M.; Hinrich Graf von der Schulenburg, J.; Bertrand, D.; West, S.A.; Goriacheva, I.I.; Zakharov, I.A.; Werren, J.H.; Stouthamer, R.; Majerus, M.E.N. Male–killing Wolbachia in two species of insect. Proc. R. Soc. Lond. B Biol. Sci. 1999, 266, 735–740. [Google Scholar] [CrossRef]
  134. Fujii, Y.; Kageyama, D.; Hoshizaki, S.; Ishikawa, H.; Sasaki, T. Transfection of Wolbachia in Lepidoptera: The feminizer of the adzuki bean borer Ostrinia scapulalis causes male killing in the Mediterranean flour moth Ephestia kuehniella. Proc. Biol. Sci. 2001, 268, 855–859. [Google Scholar] [CrossRef]
  135. Knight, J. Meet the Herod bug. Nature 2001, 412, 12–14. [Google Scholar] [CrossRef]
  136. Yen, J.H.; Barr, A.R. The etiological agent of cytoplasmic incompatibility in Culex pipiens. J. Invertebr. Pathol. 1973, 22, 242–250. [Google Scholar] [CrossRef]
  137. 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]
  138. Laven, H. Eradication of Culex pipiens fatigans through cytoplasmic incompatibility. Nature 1967, 216, 383–384. [Google Scholar] [CrossRef]
  139. Yen, P.S.; Failloux, A.B. A Review: Wolbachia-Based Population Replacement for Mosquito Control Shares Common Points with Genetically Modified Control Approaches. Pathogens 2020, 9, 404. [Google Scholar] [CrossRef]
  140. Yen, J.H.; Barr, A.R. New hypothesis of the cause of cytoplasmic incompatibility in Culex pipiens L. Nature 1971, 232, 657–658. [Google Scholar] [CrossRef]
  141. Zheng, X.; Zhang, D.; Li, Y.; Yang, C.; Wu, Y.; Liang, X.; Liang, Y.; Pan, X.; Hu, L.; Sun, Q.; et al. Incompatible and sterile insect techniques combined eliminate mosquitoes. Nature 2019, 572, 56–61. [Google Scholar] [CrossRef]
  142. Crawford, J.E.; Clarke, D.W.; Criswell, V.; Desnoyer, M.; Cornel, D.; Deegan, B.; Gong, K.; Hopkins, K.C.; Howell, P.; Hyde, J.S.; et al. Efficient production of male Wolbachia-infected Aedes aegypti mosquitoes enables large-scale suppression of wild populations. Nat. Biotechnol. 2020, 38, 482–492. [Google Scholar] [CrossRef]
  143. Marris, E. Bacteria could be key to freeing South Pacific of mosquitoes. Nature 2017, 548, 17–18. [Google Scholar] [CrossRef]
  144. Souza-Neto, J.A.; Powell, J.R.; Bonizzoni, M. Aedes aegypti vector competence studies: A review. Infect. Genet. Evol. 2019, 67, 191–209. [Google Scholar] [CrossRef]
  145. Li, F.; Hua, H.; Ali, A.; Hou, M. Characterization of a bacterial symbiont Asaia sp. in the white-backed planthopper, Sogatella furcifera, and its effects on host fitness. Front. Microbiol. 2019, 10, 2179. [Google Scholar] [CrossRef] [PubMed]
  146. Crotti, E.; Damiani, C.; Pajoro, M.; Gonella, E.; Rizzi, A.; Ricci, I.; Negri, I.; Scuppa, P.; Rossi, P.; Ballarini, P.; et al. Asaia, a versatile acetic acid bacterial symbiont, capable of cross-colonizing insects of phylogenetically distant genera and orders. Environ. Microbiol. 2009, 11, 3252–3264. [Google Scholar] [CrossRef] [PubMed]
  147. Mercant Osuna, A.; Gidley, A.; Mayi, M.P.A.; Bamou, R.; Dhokiya, V.; Antonio-Nkondjio, C.; Jeffries, C.L.; Walker, T. Diverse novel Wolbachia bacteria strains and genera-specific co-infections with Asaia bacteria in Culicine mosquitoes from ecologically diverse regions of Cameroon. Wellcome Open Res. 2023, 8, 267. [Google Scholar] [CrossRef]
  148. Chouaia, B.; Rossi, P.; Montagna, M.; Ricci, I.; Crotti, E.; Damiani, C.; Epis, S.; Faye, I.; Sagnon, N.; Alma, A.; et al. Molecular evidence for multiple infections as revealed by typing of Asaia bacterial symbionts of four mosquito species. Appl. Environ. Microbiol. 2010, 76, 7444–7450. [Google Scholar] [CrossRef] [PubMed]
  149. Omoke, D.; Kipsum, M.; Otieno, S.; Esalimba, E.; Sheth, M.; Lenhart, A.; Njeru, E.M.; Ochomo, E.; Dada, N. Western Kenyan Anopheles gambiae showing intense permethrin resistance harbour distinct microbiota. Malar. J. 2021, 20, 77. [Google Scholar] [CrossRef] [PubMed]
  150. Pelloquin, B.; Kristan, M.; Edi, C.; Meiwald, A.; Clark, E.; Jeffries, C.L.; Walker, T.; Dada, N.; Messenger, L.A. Overabundance of Asaia and Serratia bacteria is associated with deltamethrin insecticide susceptibility in Anopheles coluzzii from Agboville, Cote d’Ivoire. Microbiol. Spectr. 2021, 9, e0015721. [Google Scholar] [CrossRef] [PubMed]
  151. Jeffries, C.L.; Tantely, L.M.; Raharimalala, F.N.; Hurn, E.; Boyer, S.; Walker, T. Diverse novel resident Wolbachia strains in Culicine mosquitoes from Madagascar. Sci. Rep. 2018, 8, 17456. [Google Scholar] [CrossRef] [PubMed]
  152. Schrieke, H.; Maignien, L.; Constancias, F.; Trigodet, F.; Chakloute, S.; Rakotoarivony, I.; Marie, A.; L’Ambert, G.; Makoundou, P.; Pages, N.; et al. The mosquito microbiome includes habitat-specific but rare symbionts. Comput. Struct. Biotechnol. J. 2022, 20, 410–420. [Google Scholar] [CrossRef]
  153. Hughes, G.L.; Dodson, B.L.; Johnson, R.M.; Murdock, C.C.; Tsujimoto, H.; Suzuki, Y.; Patt, A.A.; Cui, L.; Nossa, C.W.; Barry, R.M.; et al. Native microbiome impedes vertical transmission of Wolbachia in Anopheles mosquitoes. Proc. Natl. Acad. Sci. USA 2014, 111, 12498–12503. [Google Scholar] [CrossRef]
  154. Capone, A.; Ricci, I.; Damiani, C.; Mosca, M.; Rossi, P.; Scuppa, P.; Crotti, E.; Epis, S.; Angeletti, M.; Valzano, M.; et al. Interactions between Asaia, Plasmodium and Anopheles: New insights into mosquito symbiosis and implications in Malaria Symbiotic Control. Parasites Vectors 2013, 6, 182. [Google Scholar] [CrossRef]
  155. Grogan, C.; Bennett, M.; Moore, S.; Lampe, D. Novel Asaia bogorensis Signal Sequences for Plasmodium Inhibition in Anopheles stephensi. Front. Microbiol. 2021, 12, 633667. [Google Scholar] [CrossRef]
  156. Favia, G.; Ricci, I.; Marzorati, M.; Negri, I.; Alma, A.; Sacchi, L.; Bandi, C.; Daffonchio, D. Bacteria of the genus Asaia: A potential paratransgenic weapon against malaria. Adv. Exp. Med. Biol. 2008, 627, 49–59. [Google Scholar]
  157. Damiani, C.; Ricci, I.; Crotti, E.; Rossi, P.; Rizzi, A.; Scuppa, P.; Capone, A.; Ulissi, U.; Epis, S.; Genchi, M.; et al. Mosquito-bacteria symbiosis: The case of Anopheles gambiae and Asaia. Microb. Ecol. 2010, 60, 644–654. [Google Scholar] [CrossRef]
  158. Kozlova, E.V.; Hegde, S.; Roundy, C.M.; Golovko, G.; Saldana, M.A.; Hart, C.E.; Anderson, E.R.; Hornett, E.A.; Khanipov, K.; Popov, V.L.; et al. Microbial interactions in the mosquito gut determine Serratia colonization and blood-feeding propensity. ISME J. 2021, 15, 93–108. [Google Scholar] [CrossRef]
  159. Bai, L.; Wang, L.; Vega-Rodriguez, J.; Wang, G.; Wang, S. A gut symbiotic bacterium Serratia marcescens renders mosquito resistance to Plasmodium infection through activation of mosquito immune responses. Front. Microbiol. 2019, 10, 1580. [Google Scholar] [CrossRef]
  160. Dinparast Djadid, N.; Jazayeri, H.; Raz, A.; Favia, G.; Ricci, I.; Zakeri, S. Identification of the midgut microbiota of An. stephensi and An. maculipennis for their application as a paratransgenic tool against malaria. PLoS ONE 2011, 6, e28484. [Google Scholar] [CrossRef]
  161. Wang, S.; Dos-Santos, A.L.A.; Huang, W.; Liu, K.C.; Oshaghi, M.A.; Wei, G.; Agre, P.; Jacobs-Lorena, M. Driving mosquito refractoriness to Plasmodium falciparum with engineered symbiotic bacteria. Science 2017, 357, 1399–1402. [Google Scholar] [CrossRef]
  162. Gao, H.; Bai, L.; Jiang, Y.; Huang, W.; Wang, L.; Li, S.; Zhu, G.; Wang, D.; Huang, Z.; Li, X.; et al. A natural symbiotic bacterium drives mosquito refractoriness to Plasmodium infection via secretion of an antimalarial lipase. Nat. Microbiol. 2021, 6, 806–817. [Google Scholar] [CrossRef]
  163. Arevalo-Cortes, A.; Mejia-Jaramillo, A.M.; Granada, Y.; Coatsworth, H.; Lowenberger, C.; Triana-Chavez, O. The midgut microbiota of colombian Aedes aegypti populations with different levels of resistance to the insecticide lambda-cyhalothrin. Insects 2020, 11, 584. [Google Scholar] [CrossRef]
  164. Wang, H.; Zhang, C.; Cheng, P.; Wang, Y.; Liu, H.; Wang, H.; Wang, H.; Gong, M. Differences in the intestinal microbiota between insecticide-resistant and -sensitive Aedes albopictus based on full-length 16S rRNA sequencing. Microbiologyopen 2021, 10, e1177. [Google Scholar] [CrossRef]
  165. Wang, H.; Liu, H.; Peng, H.; Wang, Y.; Zhang, C.; Guo, X.; Wang, H.; Liu, L.; Lv, W.; Cheng, P.; et al. A symbiotic gut bacterium enhances Aedes albopictus resistance to insecticide. PLoS Negl. Trop. Dis. 2022, 16, e0010208. [Google Scholar] [CrossRef]
  166. Dada, N.; Lol, J.C.; Benedict, A.C.; Lopez, F.; Sheth, M.; Dzuris, N.; Padilla, N.; Lenhart, A. Pyrethroid exposure alters internal and cuticle surface bacterial communities in Anopheles albimanus. ISME J. 2019, 13, 2447–2464. [Google Scholar] [CrossRef]
  167. Berticat, C.; Rousset, F.; Raymond, M.; Berthomieu, A.; Weill, M. High Wolbachia density in insecticide-resistant mosquitoes. Proc. Biol. Sci. 2002, 269, 1413–1416. [Google Scholar] [CrossRef]
  168. Zhang, R.; Liu, W.; Zhang, Q.; Zhang, X.; Zhang, Z. Microbiota and transcriptome changes of Culex pipiens pallens larvae exposed to Bacillus thuringiensis israelensis. Sci. Rep. 2021, 11, 20241. [Google Scholar] [CrossRef]
  169. Receveur, J.P.; Pechal, J.L.; Benbow, M.E.; Donato, G.; Rainey, T.; Wallace, J.R. Changes in larval mosquito microbiota reveal non-target effects of insecticide treatments in hurricane-created habitats. Microb. Ecol. 2018, 76, 719–728. [Google Scholar] [CrossRef]
  170. Endersby, N.M.; Hoffmann, A.A. Effect of Wolbachia on insecticide susceptibility in lines of Aedes aegypti. Bull. Entomol. Res. 2013, 103, 269–277. [Google Scholar] [CrossRef]
  171. Duron, O.; Labbé, P.; Berticat, C.; Rousset, F.; Guillot, S.; Raymond, M.; Weill, M. High Wolbachia density correlates with cost ofinfection for insecticide resistant Culex Pipiens mosquitoes. Evolution 2006, 60, 303. [Google Scholar]
  172. Shemshadian, A.; Vatandoost, H.; Oshaghi, M.A.; Abai, M.R.; Djadid, N.D.; Karimian, F. Relationship between Wolbachia infection in Culex quinquefasciatus and its resistance to insecticide. Heliyon 2021, 7, e06749. [Google Scholar] [CrossRef]
  173. Ismail, H.M.; Freed, S.; Naeem, A.; Malik, S.; Ali, N. The effect of entomopathogenic fungi on enzymatic activity in chlorpyrifos-resistant mosquitoes, Culex quinquefasciatus (Diptera: Culicidae). J. Med. Entomol. 2020, 57, 204–213. [Google Scholar] [CrossRef]
  174. Farenhorst, M.; Mouatcho, J.C.; Kikankie, C.K.; Brooke, B.D.; Hunt, R.H.; Thomas, M.B.; Koekemoer, L.L.; Knols, B.G.; Coetzee, M. Fungal infection counters insecticide resistance in African malaria mosquitoes. Proc. Natl. Acad. Sci. USA 2009, 106, 17443–17447. [Google Scholar] [CrossRef]
  175. Howard, A.F.; Koenraadt, C.J.; Farenhorst, M.; Knols, B.G.; Takken, W. Pyrethroid resistance in Anopheles gambiae leads to increased susceptibility to the entomopathogenic fungi Metarhizium anisopliae and Beauveria bassiana. Malar. J. 2010, 9, 168. [Google Scholar] [CrossRef]
  176. Patil, C.D.; Borase, H.P.; Salunke, B.K.; Patil, S.V. Alteration in Bacillus thuringiensis toxicity by curing gut flora: Novel approach for mosquito resistance management. Parasitol. Res. 2013, 112, 3283–3288. [Google Scholar] [CrossRef]
  177. Scates, S.S.; O’Neal, S.T.; Anderson, T.D. Bacteria-mediated modification of insecticide toxicity in the yellow fever mosquito, Aedes aegypti. Pestic. Biochem. Physiol. 2019, 161, 77–85. [Google Scholar] [CrossRef]
  178. Kikankie, C.K.; Brooke, B.D.; Knols, B.G.; Koekemoer, L.L.; Farenhorst, M.; Hunt, R.H.; Thomas, M.B.; Coetzee, M. The infectivity of the entomopathogenic fungus Beauveria bassiana to insecticide-resistant and susceptible Anopheles arabiensis mosquitoes at two different temperatures. Malar. J. 2010, 9, 71. [Google Scholar] [CrossRef] [PubMed]
  179. Vivekanandhan, P.; Swathy, K.; Kalaimurugan, D.; Ramachandran, M.; Yuvaraj, A.; Kumar, A.N.; Manikandan, A.T.; Poovarasan, N.; Shivakumar, M.S.; Kweka, E.J. Larvicidal toxicity of Metarhizium anisopliae metabolites against three mosquito species and non-targeting organisms. PLoS ONE 2020, 15, e0232172. [Google Scholar] [CrossRef]
  180. Xia, Z.; Lei, L.; Zhang, H.Y.; Wei, H.L. Characterization of the ModABC Molybdate Transport System of Pseudomonas putida in Nicotine Degradation. Front. Microbiol. 2018, 9, 3030. [Google Scholar]
  181. Bhatt, P.; Huang, Y.; Zhan, H.; Chen, S. Insight Into Microbial Applications for the Biodegradation of Pyrethroid Insecticides. Front. Microbiol. 2019, 10, 1778. [Google Scholar] [CrossRef]
  182. Bhatt, P.; Rene, E.R.; Huang, Y.; Lin, Z.; Pang, S.; Zhang, W.; Chen, S. Systems biology analysis of pyrethroid biodegradation in bacteria and its effect on the cellular environment of pests and humans. J. Environ. Chem. Eng. 2021, 9, 106582. [Google Scholar] [CrossRef]
  183. Bose, S.; Kumar, P.S.; Vo, D.-V.N.; Rajamohan, N.; Saravanan, R. Microbial degradation of recalcitrant pesticides: A review. Environ. Chem. Lett. 2021, 19, 3209–3228. [Google Scholar] [CrossRef]
  184. Singh, A.K.; Bilal, M.; Iqbal, H.M.N.; Raj, A. Trends in predictive biodegradation for sustainable mitigation of environmental pollutants: Recent progress and future outlook. Sci. Total Environ. 2021, 770, 144561. [Google Scholar] [CrossRef]
  185. Kumar, S.; Thomas, A.; Sahgal, A.; Verma, A.; Samuel, T.; Pillai, M.K. Effect of the synergist, piperonyl butoxide, on the development of deltamethrin resistance in yellow fever mosquito, Aedes aegypti L. (Diptera: Culicidae). Arch. Insect Biochem. Physiol. 2002, 50, 1–8. [Google Scholar] [CrossRef]
  186. Kumar, S.; Thomas, A.; Sahgal, A.; Verma, A.; Samuel, T.; Pillai, M.K. Variations in the insecticide-resistance spectrum of Anopheles stephensi after selection with deltamethrin or a deltamethrin-piperonyl-butoxide combination. Ann. Trop. Med. Parasitol. 2004, 98, 861–871. [Google Scholar] [CrossRef] [PubMed]
  187. Dempsey, J.L.; Cui, J.Y. Microbiome is a functional modifier of P450 drug metabolism. Curr. Pharmacol. Rep. 2019, 5, 481–490. [Google Scholar] [CrossRef]
Figure 1. Flowchart of the article search and screening process.
Figure 1. Flowchart of the article search and screening process.
Pathogens 13 00691 g001
Figure 2. Environments involved in mosquito development and tools and methods for the analysis of mosquito microbiota.
Figure 2. Environments involved in mosquito development and tools and methods for the analysis of mosquito microbiota.
Pathogens 13 00691 g002
Figure 3. Some of the key areas for mosquito microbiota studies.
Figure 3. Some of the key areas for mosquito microbiota studies.
Pathogens 13 00691 g003
Table 1. Impact of insecticide resistance on microbiota.
Table 1. Impact of insecticide resistance on microbiota.
Mosquito SpeciesInsecticideMosquito StageDescription of FindingsReference
Ae. aegyptiPermethrin selectionAdultCutibacterium, Corynebacterium, Citricoccus, Leucobacter, Acinetobacter, Dietzia, and Anaerococcus spp. were more abundant in the selected strain.[75]
Ae. aegyptiLambda-cyhalothrinAdultGenera of Coprococcus, Ruminococcus, Bilophila, Enterobacter, Porphyromonas, Bifidobacterium, Weissella, and Delftia were enriched in the resistant group. Bacteria Bacteroides faecichinchillae decreased significantly in resistant midguts.[47]
Ae. aegyptiLambda-cyhalothrinAdultThe presence of Pseudomonas viridiflava is associated with pyrethroid degradation. Parabacteroides, Megasphaera, Akkermansia, Lardizabala, Ruminococcus, and Coprococcus genera were enriched in susceptible mosquitoes.[163]
Ae. aegyptiPermethrin, deltamethrin exposureAdultAfter exposure to permethrin, the most abundant bacterial species were Pantoea agglomerans and Pseudomonas azotoformans-fluorescens-synxantha. Elizabethkingia meningoseptica and Ps. azotoformans-fluorescens-synxantha were the most abundant after exposure to deltamethrin.[73]
Ae. albopictusDeltamethrinAdultAbundance of Serratia oryzae was significantly higher in the resistant strain.[165]
Ae. albopictusDeltamethrinAdultAcinetobacter junii and Se. oryzae significantly increased after deltamethrin treatment.[164]
Ae. stimulansMethopreneLarvalIncreased abundances of Clostridium spp. and Lysinibacillus spp. [169]
An. albimanusFenitrothion AdultResistance selection enriches bacterial taxa, reduces diversity, and significantly increases Bacillus and Klebsiella pneumoniae.[69]
An. albimanusAlphacypermethrin or permethrinAdultThe abundance of Pseudomonas fragi and Pa. agglomerans increased with pyrethroid exposure.[166]
An. arabiensisDeltamethrin and malathionAdultSusceptible mosquitoes showed greater gut bacterial diversity than resistant mosquitoes.[93]
An. coluzziiDeltamethrinAdultOchrobactrum, Lysinibacillus, and Stenotrophomonas genera were significantly enriched in resistant mosquitoes; Asaia and Serratia dominated the susceptible individuals.[150]
An. gambiae s.s.PermethrinAdultSphingobacterium, Lysinibacillus, Streptococcus, and Rubrobacter were associated with resistant mosquitoes; Myxococcus was associated with susceptible mosquitoes.[149]
An. stephensiTemephos selection RR > 10LarvalResistant strain with 4 dominant genera, i.e., Pseudomonas, Aeromonas, Exiguobacterium, and Microbacterium[28]
Cx. pipiensOrganophosphateAdultResistant mosquitoes with higher loads of Wolbachia.[167]
Cx. pipiens pallensBti exposureLarvalThe predominant bacteria changed from Actinobacteria to Firmicutes, and the abundance of Actinobacteria was gradually reduced with an increase in the concentration of Bti. At the genus level, Bacillus replaced Microbacterium as the predominant genus.[168]
Cx. quinquefasciatusDeltamethrinAdultAt the genus level, Aeromonas, Morganella, Elizabethkingia, Enterobacter, Cedecea, and Thorsellia showed significant differences between strains. At the species level, Bacillus cereus, Enterobacter cloacae s.l., Streptomyces sp., Pseudomonas sp., and Wolbachia were more abundant in the resistant strains.[74]
Bti: Bacillus thuringiensis var. israelensis; s.s.: sensu stricto; s.l.: sensu lato.
Table 2. Impact of microbiota on insecticide resistance.
Table 2. Impact of microbiota on insecticide resistance.
Mosquito SpeciesInsecticideMosquito StageDescription of FindingsReference
Ae. aegyptiBifenthrin, Bti, temephos, methopreneAdult, larvalInfection with Wolbachia has no effect on susceptibility to insecticides.[170]
Ae. aegyptiPropoxur, naledLarvalBroad-spectrum antibiotic treatment of larvae decreases the metabolic detoxification of propoxur and naled. Adding cultured gut bacteria isolated from mosquito larvae reduces larval mortality.[177]
Ae. albopictusDeltamethrinAdultCultured bacteria Serratia oryzae and Acinetobacter junii promote resistance.[164]
Ae. albopictusDeltamethrinAdultThe survival of Se. oryzae-enriched mosquitoes significantly increased. Three metabolic detoxification enzymes in Se. oryzae-enriched mosquitoes increased. Carboxylesterase activity was detected in Se. oryzae. Se. oryzae can degrade deltamethrin in vitro; degradation efficiency was positively correlated with time and bacterial amount.[165]
An. arabiensisDeltamethrin, malathionAdultResistance mosquitoes have lower gut bacterial diversity. Supplementation of bacterial St. pyrogenes or Escherichia coli increased insecticide tolerance. Antibiotic supplementation via sugar decreased tolerance to the insecticides deltamethrin and malathion. Both R/S females had decreased α-esterase activity after gentamicin, streptomycin, vancomycin, heat-killed St. pyrogenes, or live E. coli treatment. GST, P450, and β-esterase changes are inconsistent.[93]
An. gambiae s.s.PyrethroidAdultResistance leads to increased mortality by the fungi Beauveria bassiana and Metarhizium anisopliae infection.[175]
An. gambiae s.s., An. funestus, An. arabiensisPyrethroids, organochlorines, carbamatesAdultResistant mosquitoes preinfected with Be. bassiana or Me. anisopliae showed a significant increase in mortality after insecticide exposure.[174]
An. stephensiBtLarvalCommensal microbes in the midgut are capable of degrading insecticidal Bt proteins, decreasing larval susceptibility to Bt. Antibiotic treatment increased mortality and reached 100% mortality at a concentration of 110 μg/mL of an antibiotic mixture of penicillin, streptomycin, and erythromycin.[176]
An. stephensiTemephos selection RR > 10LarvalAdding symbiotic bacteria collected from the breeding place can boost the activity of α-esterase and GST enzymes.[28]
Cx. pipiensChlorpyrifos, propoxurLarvalInfection with Wolbachia has no effect on resistance to chlorpyrifos and propoxur.[171]
Cx. quinquefasciatusDDTAdultInfection with Wolbachia increases susceptibility to DDT.[172]
Cx. quinquefasciatusChlorpyrifos selectionLarvalActivities of acetylcholinesterase (AChE), glutathione S-transferase (GST), esterase (EST), acid phosphatases (ACP), and alkaline phosphatases (ALP) increased in the chlorpyrifos-selected (Chlor-SEL) population. Activities of all enzymes were suppressed when exposed to Me. anisopliae or Be. Bassiana.[173]
Table 3. Members of the mosquito microbiota that degrade insecticide resistance.
Table 3. Members of the mosquito microbiota that degrade insecticide resistance.
Mosquito SpeciesInsecticideInsecticide Degradation SymbiontsReference
Ae. aegyptiLambda-cyhalothrinPseudomonas viridiflava[163]
An. albimanusFenitrothionBacillus cereus and Acinetobacter baumannii[69]
An. albimanusPermethrin, AlphacypermethrinPantoea agglomerans[166]
An. coluzziiDeltamethrinOchrobactrum, Lysinibacillus, and Stenotrophomonas genera[150]
An. gambiaePermethrinSphingobacterium, Lysinibacillus, and Streptococcus genera[149]
An. stephensiTemephosPseudomonas sp., Aeromonas sp., Exiguobacterium sp., and Microbacterium sp.[28]
Multiple speciesPyrethroidsBacteria and fungi: Bacillus spp., Raoultella ornithinolytica, Pseudomonas fluorescens, Brevibacterium sp., Acinetobacter sp., Aspergillus sp., Candida sp., Trichoderma sp., and Candia spp.[181]
† Identified and insecticide degradation function confirmed by the corresponding study.
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

Liu, H.; Yin, J.; Huang, X.; Zang, C.; Zhang, Y.; Cao, J.; Gong, M. Mosquito Gut Microbiota: A Review. Pathogens 2024, 13, 691. https://doi.org/10.3390/pathogens13080691

AMA Style

Liu H, Yin J, Huang X, Zang C, Zhang Y, Cao J, Gong M. Mosquito Gut Microbiota: A Review. Pathogens. 2024; 13(8):691. https://doi.org/10.3390/pathogens13080691

Chicago/Turabian Style

Liu, Hongmei, Jianhai Yin, Xiaodan Huang, Chuanhui Zang, Ye Zhang, Jianping Cao, and Maoqing Gong. 2024. "Mosquito Gut Microbiota: A Review" Pathogens 13, no. 8: 691. https://doi.org/10.3390/pathogens13080691

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop