Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Metagenomic analysis of Aedes aegypti and Culex quinquefasciatus mosquitoes from Grenada, West Indies

  • Maria E. Ramos-Nino ,

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Software, Supervision, Writing – original draft, Writing – review & editing

    mramosni@sgu.edu (MERN); scheetha@sgu.edu (SC)

    Affiliation Department of Microbiology, Immunology, and Pharmacology, School of Medicine, St. George’s University, Grenada, West Indies

  • Daniel M. Fitzpatrick,

    Roles Conceptualization, Investigation, Methodology, Writing – review & editing

    Affiliation Department of Pathobiology, School of Veterinary Medicine, St. George’s University, Grenada, West Indies

  • Korin M. Eckstrom,

    Roles Data curation

    Affiliation University of Vermont Massively Parallel Sequencing Facility, Burlington, Vermont, United States of America

  • Scott Tighe,

    Roles Data curation

    Affiliation University of Vermont Massively Parallel Sequencing Facility, Burlington, Vermont, United States of America

  • Lindsey M. Hattaway,

    Roles Investigation

    Affiliation Department of Pathobiology, School of Veterinary Medicine, St. George’s University, Grenada, West Indies

  • Andy N. Hsueh,

    Roles Investigation

    Affiliation Department of Pathobiology, School of Veterinary Medicine, St. George’s University, Grenada, West Indies

  • Diana M. Stone,

    Roles Writing – review & editing

    Affiliation Department of Pathobiology, School of Veterinary Medicine, St. George’s University, Grenada, West Indies

  • Julie A. Dragon,

    Roles Data curation

    Affiliation University of Vermont Massively Parallel Sequencing Facility, Burlington, Vermont, United States of America

  • Sonia Cheetham

    Roles Conceptualization, Investigation, Writing – review & editing

    mramosni@sgu.edu (MERN); scheetha@sgu.edu (SC)

    Affiliation Department of Pathobiology, School of Veterinary Medicine, St. George’s University, Grenada, West Indies

Abstract

The mosquitoes Aedes aegypti (Linnaeus, 1762) (Diptera: Culicidae) and Culex quinquefasciatus Say, 1823 (Diptera: Culicidae) are two major vectors of arthropod-borne pathogens in Grenada, West Indies. As conventional vector control methods present many challenges, alternatives are urgently needed. Manipulation of mosquito microbiota is emerging as a field for the development of vector control strategies. Critical to this vector control approach is knowledge of the microbiota of these mosquitoes and finding candidate microorganisms that are common to the vectors with properties that could be used in microbiota modification studies. Results showed that bacteria genera including Asaia, Escherichia, Pantoea, Pseudomonas, and Serratia are common to both major arboviral vectors in Grenada and have previously been shown to be good candidates for transgenetic studies. Also, for the first time, the presence of Grenada mosquito rhabdovirus 1 is reported in C. quinquefasciatus.

Introduction

The mosquito species Aedes aegypti. and Culex quinquefasciatus are a public health concern due to their ability to be vectors of many arboviruses. Aedes aegypti, for example, transmits chikungunya virus, Zika virus, yellow fever virus, and dengue virus, one of the most rapidly spreading vector-borne pathogens in the world with 2.5 billion people at risk of infection and approximately 500,000 people developing severe dengue disease annually [1]. Culex quinquefasciatus, on the other hand, is capable of transmitting arboviruses like West Nile virus (WNV), the leading cause of mosquito-borne disease in the continental United States [2], as well as nematodes affecting human and animal health (lymphatic filariasis, dirofilariasis etc.) [35]. Currently, personal protective measures and control of mosquito populations are the only available strategies to prevent arboviral diseases because there are no therapeutic treatments for arboviruses and vaccines are limited. Organizations are constantly facing challenges with the use of conventional vector control methods because of sustainability, organizational complexity [6], and the rise of insecticide resistance [79].

Manipulation of mosquito microbiota is emerging as a promising method for the development of vector control strategies. Some of these strategies include: 1) The introduction of microorganisms that interfere with the pathogens within the vector. Examples of this strategy include the use of entomopathogenic fungi such as Beauveria bassiana and Beauveria brongniartii [10], and the alpha-proteobacteria Wolbachia [1113]. Wolbachia is a natural intracellular bacterial symbiont maternally transmitted to offspring that can induce cytoplasmic incompatibility, where mating between Wolbachia-infected males and uninfected females yields eggs that fail to develop. Also certain Wolbachia strains may cause a decrease in the vectorial capacity by interfering with vector competence or by shortening vector lifespan [14]. 2) Another vector control strategy is the use of symbiotic bacteria that are transformed to express effector molecules for use in paratransgenic approaches. For example, Metarhizium anisopliae has been genetically transformed to express the anti-Plasmodium effector molecules SM1 and scorpine, which are both reported to interfere with Plasmodium falciparum in Anopheles mosquitoes [15].

Aedes aegypti and C. quinquefasciatus are the two predominant anthroponotic mosquitoes in Grenada, West Indies, [16]. Aedes aegypti transmits dengue, Zika, and chikungunya locally. No major human pathogens transmitted by C. quinquefasciatus have been reported in Grenada, but the mosquito is capable of transmitting several arboviruses that occur in other Caribbean islands (e.g., WNV, St. Louis Encephalitis virus) or in neighboring mainland South American countries (e.g., Wuchereria bancrofti) [17]. The microbial composition of A. aegypti and C. quinquefasciatus mosquitoes has been previously studied [18,19,2027,2836]. However, several arthropod-borne diseases that are endemic to the mainland Americas are not believed to be established in Caribbean islands (e.g. Mayaro virus, Oropouche virus, Wuchereria) [17,3740]. While variation in climate and the makeup and behavior of local mosquito species may explain this in part, regional differences in local mosquito microbiota may also contribute to differences in their competence as vectors of pathogens of human and animal interest [6,33,41]. Metagenomic analyses have recently been used to identify novel arboviruses in Caribbean mosquitoes [19,42]; little else is known about these viruses. It is thus important to characterize the microbiome of mosquitoes on a regional level to identify heretofore unknown organisms, including agents that can be studied further for their effects on vector competence and their use in vector control studies. Through the use of metagenomics, this study will shed some light on: 1) the identification and determination of the relative estimated abundance (REA) of microbiota for these two arboviral vectors; 2) the identification of microbiota similarities and unique microbiota elements of these two mosquitos to provide potential targets for developing mosquito/arbovirus control strategies; and 3) the identification of microorganisms not yet known to occur in mosquitoes in Grenada.

Material and methods

Mosquito collection

Three hundred A. aegypti mosquitoes and 300 C. quinquefasciatus were randomly selected out of 1,152 A. aegypti and 3,000 C. quinquefasciatus collected between January 2018 and December 2018 from six semi-rural locations in St. George Parish (Fig 1), the most populated parish in Grenada (12°15'46'' N 61°36'15'' W). The six collection sites were within an approximately five square-mile area and were chosen for their proximity to the capital city (which contains the main seaport), the airport, most major marinas, and for their high density of people. Traps (Biogents Sentinel, Biogents, Regensburg, Germany) were baited with octenol and yeast-based carbon dioxide attractants, as previously described [43]. In brief, at each of the six sites, one trap was deployed twice weekly within three meters of a house and was collected after 24 hours. Mosquitoes were dispatched at −80°C and identified to species by morphological analysis. Identification keys in Darsie and Ward (2013) [44] were used to discriminate between species known to occur in Grenada based on the Walter Reed Biosystematics Unit (2019) [45]. Mosquitoes were placed in RNAlater® (Sigma Aldrich, St. Louis, Missouri, USA) after identification for later processing. Mosquito heads were removed before RNA extraction to prevent PCR inhibition [46]; wings and legs were also removed to reduce host (mosquito) RNA. No specific permissions were required for this study since it was carried out in private lands. The study did not involve endangered or protected species. No IACUC was required for the use of mosquitoes in this study.

thumbnail
Fig 1. Map of collection sites in St. George parish-Grenada.

Six sites were used during the year of collecting mosquitoes [47].

https://doi.org/10.1371/journal.pone.0231047.g001

Total RNA extraction and RNA-Seq

RNA extraction was performed in batches of 30 mosquitoes at a time (ten pools) using TRIzol (ThermoFisher, Carlsbad, California, USA). Invitrogen Phasemaker Tubes (ThermoFisher) were used for the phase separation. RNA was DNase-treated using TURBO DNA-free (ThermoFisher) and RNA quality evaluated utilizing an Agilent 2100 Bioanalyzer (Agilent, Santa Clara, California, USA) as previously described [48]. All sub-pools were pooled again for library construction, which was performed using the NuGEN Tecan universal RNA sequencing reagents as recommended by the manufacturer.

The metagenomic analysis flow used in this study can be found in Fig 2. Briefly, shotgun metagenomic sequencing was run using the Illumina HiSeq 1500 for deep sequencing. Raw fastq files were assessed for quality using Illumina FastQC version 0.11.8. Trimming and quality filtering of reads was performed using Atropos (https://omictools.com/atropos-tool), removing Illumina universal adaptors, reads with base calls below Q20, and reads with a length less than 35 bp. Additional host read removal was performed in silico using Bowtie2 (v. 2.3.4.3). Reads were mapped to their respective hosts, either the A. aegypti reference genome, assembly AaegL5, available at https://www.vectorbase.org/organisms/aedes-aegypti or the Cx. quinquefasciatus reference genome, assembly GCA_000209185.1 available at https://www.ebi.ac.uk/ena/data/view/GCA_000209185.1, using end-to-end read alignment. These non-mosquito reads were analyzed using CCmetagen (https://www.biorxiv.org/content/10.1101/641332v1) which uses the June 2019 version of the NCBI nt database excluding all taxids for environmental eukaryotes and prokaryotes, unclassified sequences, and artificial sequences in order to avoid misclassification based on contaminants. CCMetagen uses a weighted mapping approach based on the kma aligner in order to improve taxonomic classification of regions that are highly similar across microbial genomes. Abundance of the microorganisms, after metagenomic analysis, is expressed as the number of nucleotides mapped to the reference, normalized by the length of the reference in order to correct for differences in genome size. The relative estimated abundance (REA) reflects the percentage of the total abundance. Metagenomic data for Aedes aegypti and Culex quinquefasciatus is deposited in https://www.ncib.nlm.nih.gov/sra/PRJNA564787.

RT-PCR verification

Approximately 200 ng of total RNA per pool (ten sub-pools) was reverse transcribed using a High-Capacity cDNA Reverse Transcription Kit (ThermoFisher). RT-PCR conducted on the cDNA was produced using previously published specific primers (S1 Table). PCR amplicons of expected size were extracted from gels using the QIAquick Gel Extraction Kit (QIAgen, Hilden, Germany) following the manufacturer’s protocol. Amplicons were sent to the Molecular Cloning Lab, San Francisco, California (https://www.mclab.com/) for direct Sanger sequencing. Raw sequence data were manually edited using Chromas 2.6.5 software and then compared with the sequence database using the NIH’s Basic Local Alignment Search Tool (BLAST). Sequences of microorganisms were aligned with Clustal Omega (https://www.ebi.ac.uk/Tools/msa/clustalo/) to obtain a consensus sequence. Primers used in this project are included in S1 Table [4951].

Isolation of microorganisms and taxonomic assignment

Freshly collected A. aegypti and C. quinquefasciatus were surface cleaned and then dissected to obtain the salivary glands and midguts. Mosquitoes were serially rinsed in sterile phosphate-buffered saline (PBS), followed by ethanol (70%), and finally rinsed three times in PBS. Aliquots of 100 μl from the last PBS washes were plated on blood agar plates as control groups of the surface cleaning process. Mosquitoes were dissected under a microscope, and salivary glands and midguts were collected in sterile PBS and macerated with a pestle. An aliquot of 100 μl was transferred to blood agar plates and incubated at 28°C for 24–48 h, followed by DNA isolation of individual colonies using the Qiagen DNeasy Blood and Tissue kit. Universal bacterial 16s rRNA primers (see S1 Table) were used to amplify a 465 bp product [49] by PCR. Amplicons of the expected size were purified using the QIAquick Gel Extraction Kit (Qiagen) and Sanger-sequenced. Editing and assignment of a bacterial taxonomic hierarchy was done as described above. Isolates were stored as glycerol stocks at -80°C.

Results

The metagenomic analysis of both A. aegypti and C. quinquefasciatus in Grenada showed a microbiome composed primarily of bacteria (75.22% and 96.42% REA, respectively) (Table 1, S1A/Aedes Fig, S1B/Culex Fig).

thumbnail
Table 1. Aedes aegypti and Culex quinquefasciatus common microbiota in Grenada (Relative estimated abundance).

https://doi.org/10.1371/journal.pone.0231047.t001

The phylum Proteobacteria dominated the bacteria in both mosquitoes (81.28% REA in A. aegypti and 94.92% in C. quinquefasciatus) (Table 1, S2A1/Aedes Fig, S2B1/Culex Fig). Of the Proteobacteria, Escherichia (64.52% REA), a Gammaproteobacteria belonging to the Enterobacteriaceae family was predominant in Aedes. In Culex, the genera Wolbachia (62.79% REA), an Alphaproteobacteria belonging to the Anaplasmataceae family was the predominant bacteria, followed by Escherichia (11.15% REA) (S2A2–S2A4/Aedes Fig, S2B2–S2B4/Culex Fig).

Common bacteria found in both mosquitoes included the genera Escherichia, Actinomyces, Zymobacter, Spironema, Carnimonas, Acinetobacter, Halotalea, Pantoea, Leuconoctoc, Pseudomonas, Asaia, Stenotrophomonas, Xanthomonas, and Serratia. Actinomyces was the most abundant bacteria in both mosquitoes after Escherichia (Table 1). Unique to Culex were the genera Wolbachia, Arcobacter, Aeromonas, Burkholderia, Holospora, Salmonella, Erwinia, Anaplasma, and Fructobacillus. The bacteria genera Pseudoxanthomonas and Halomonas were unique to Aedes mosquitoes (S2A4/Aedes Fig,S2B4/Culex Fig).

The genus Asaia which was present in the metagenomic analysis of both Aedes and Culex, has unique features such as presence in organs of female and male mosquitoes and vertical and horizontal transmission. These characteristics lead to the possibility of introducing it as a robust candidate for vector control via paratransgenesis. Because it is vital to be able to culture bacteria for paratransgenic approaches, this bacterium was further tested for culturability and location in the vector [52]. Asaia was isolated by simple routine culture methods from both Aedes and Culex. Culture of the salivary glands and midguts of individual mosquitoes resulted in the isolation of Asaia from both the salivary glands and midguts of Aedes, and only from the guts of Culex.

A large number of reads were unclassified at the genera level for the fungi and the parasites. The fungi kingdom was only 0.19% and 0.1% REA for Aedes and Culex respectively. The family Sclerotiniaceae was common to both mosquitoes (7.75% REA in Aedes and 12.8% in Culex) and dominated in Culex, while the family Aspergillaceae (44.6%) was dominant in Aedes (S3A1–S3A2/Aedes Fig,S3B1 and S3B2/Culex Fig).

The parasites were also very rare with 0.08% in Aedes, and 1.86% REA in Culex with Trypanosomatidae as the dominant family in Culex and the Albuginaceae and Lecudinidae families dominant in Aedes (S4A1 and S4A2/Aedes Fig,S4B1 and S4B2/Culex Fig). Common to both mosquitoes was the genus Albugo.

Subsequent RT-PCR assays were performed on ten sub-pools of C. quinquefasciatus mosquitoes in order to confirm the presence of the trypanostomatids detected in the metagenomics analysis. All primers and references are listed in S1 Table. Amplicons of the correct size were produced in four of the ten sub-pool PCRs (S5 Fig). Three Sanger-sequenced amplicons had identical sequences to each other while the fourth failed to sequence. The best species-level match was Paratrypanosoma confusum, a recently characterized trypanostomatid that is likely insect-specific (identity: 99.7% to GenBank accession number KF963538.1) [53].

The dominant viruses in Aedes were described in previous publication [47]. Due to the limitations in the databases presently available, a large amount of reads for viruses from the metagenomic analysis remained un-classified, but for Culex, the Circoviridae (33.41% REA) family, particularly the genus Circovirus was the most dominant. The next most abundant family in Culex was Rhabdoviridae, dominated specifically by Grenada mosquito rhabdovirus 1. Finally, the third most abundant virus family for Culex was Flaviviridae dominated by Culex flavivirus (S6A1 and S6A2/Aedes Fig,S6B1 and S6B2/Culex Fig).

RT-PCR using hemi-nested pan-flavivirus primers produced amplicons of the expected size in nine of ten sub-pools (S1 Table, S7 Fig). Sanger sequencing of two of the amplicons confirmed infection with Culex flavivirus (identities: 98.8% and 99.2% to GenBank accession number MH719098.1), while the other seven amplicons produced overlapping reads, suggesting a mixed infection.

Two viruses with unknown families were also detected in Culex: Culex phasma-like virus and Terena virus.

Discussion

The understanding of an organism can no longer be assessed in isolation, but rather needs to be viewed as a complex that includes its community of associated microorganisms and their interactions [27]. With this in mind, our study goals were to identify and compare the microbiota of two important vectors of mosquito-borne pathogens in Grenada, Aedes aegypti and Culex quinquefasciatus. Studies on the role of microbial communities in the mosquito biology and pathogen interference has led to the development of new vector control approaches based on microbiota modifications [54]. It has also been shown that environmental factors influence the microbial composition of breeding sites and food resources (plant materials, water sources, blood) [23,27,55] that become part of the adult mosquitoes’ microbiome. Similarly, the microbiome for a given mosquito species can differ based upon geography and parental lineage [6,33,41]. Thus, it is important that regional studies are conducted, as microbiome region-specific vector control approaches may be required. Our results could lead to future studies on the use of these organisms in mosquito control projects in Grenada.

Our results were similar to several other studies [6,33,41], despite differences in techniques used for identification and characterization of microbiota. For instance, microbiota in Grenadian mosquitoes is dominated by bacteria, particularly those associated with the gut, an observation confirmed in other studies [56]. In Aedes aegypti adult mosquito for example, Proteobacteria, Bacteroides, Firmicutes, and Actinobacteria are the phyla that contain more than 99% of the total microbiota [29]. Among these, members of the families: Enterobacteriaceae, Erwiniaceae, Yersiniaceae, Acetobacteraceae, Enterococcacea, and Bacillaceae are the most-frequently described bacteria from the gut of adult Aedes spp. (reviewed in [6]).

Numerous efforts have been made to directly modify the mosquito genome in order to limit their ability to transmit pathogens, but there are biological and logistical limitations to overcome and public concerns about safety that must be addressed before these mosquitoes are introduced into the wild population universally [52,5760]. An alternative approach to manipulating the mosquito genome is the use of microbe symbionts of the mosquito. Paratransgenesis has emerged as a more suitable approach for vector control based on the use of symbiotic bacteria to deliver effector molecules to wild vectors [61]. An understanding of mosquito microbiota is critical for a paratransgenic system approach and includes: 1) the identification of microorganisms that are well established in mosquitoes, 2) are cultivable, 3) are amenable to genetic manipulation, and 4) can be transmitted to the next generation to propagate the desired traits [6165]. Furthermore, the chosen bacteria should be capable of colonizing a wide variety of mosquito species so that they can be deployed in different species, reducing the need of producing different transgenic mosquitoes for particular environment [52,60,66]. Among bacteria identified in this study, Pantoea, Pseudomonas, and Serratia have been put forward as potential candidates for paratransgenic modifications for vector control strategies [29]. Another candidate for paratransgenesis is Escherichia coli, which is not only one of the most abundant organisms in both Aedes and Culex from Grenada, but also are easy to genetically manipulate, and are culturable. Some studies have already used E. coli in paratransgenesis systems [60,67]. Unfortunately, it also has been shown that these constructs of E. coli disappear quickly from the midgut of some mosquitoes which may make them non-suitable for the use in paratransgenic interventions [60]. Some of these symbiotic bacteria may play a critical role in the mosquitoes’ survival [68], and hence, further studies are needed to determine the effect of these bacteria on the two very important populations of Aedes and Culex mosquitoes.

Other common bacteria found in our study, in the genus Asaia, have been found to have permanent association with mosquitoes and are able to quickly colonize tissue in several mosquito species including A. aegypti and mosquitoes from the C. pipiens complex [52]. Asaia bacteria can be cultured and genetically manipulated. Furthermore, they can be transmitted from males to females during mating [61,68,69].

A potential problem with the use of Asaia in mosquito control includes Asaia interference with the vertical transmission of Wolbachia [27], a bacteria of great importance in mosquito control. According to the literature, co-colonization of the mosquito with Wolbachia and Asaia, restricts Asaia to the gut [6,70]. However, Asaia is able to colonize reproductive organs and salivary glands in species uninfected by Wolbachia such as A. aegypti (reviewed in [27]). The exclusion pattern observed between Wolbachia and Asaia is also found in A. albopictus and C. quinquefasciatus naturally infected by both bacteria. [70]. Most studies focus on the microbiota of the gut because of its direct implications with mosquito vector biology [71], so we decided to confirm the location of Asaia in Aedes and Culex. Asaia reads were found in both Aedes and Culex mosquitoes using metagenomics. In agreement with published observations [70,72], Asaia from individual mosquitoes was cultured from all A. aegypti from both the salivary gland and gut, but only from the guts of C. quinquefasciatus. These observations suggest competition between the Wolbachia and Asaia for colonization outside the gut but confirm its presence in the guts of both mosquitoes. Therefore, Asaia may be a promising candidate for the control of Aedes and Culex, as reviewed in [6], and confirmed here. Further studies will be required to establish the suitability of Asaia as a candidate paratransgenetic organism.

Fungi can also be used in paratransgenic processes [73]. In our study, we found a common fungus belonging to the Sclerotinaceae family which was present in both mosquito populations. Many species in this family are plant pathogens which do not affect insects, and therefore make them unsuitable for paratransgenic techniques.

The presence of some bacteria found in this study, like Serratia (in both mosquito genera) as well as Wolbachia (in Culex) are known to provide protective effects from pathogen infections, particularly arboviruses [29,7478]. Reports on pathogen blocking by Wolbachia mainly reflect studies with dengue virus, but there is also evidence of pathogen blocking of other medically important, positive, single-stranded RNA viruses such as Zika, yellow fever, and chikungunya [7982].

Wolbachia is an intracellular endosymbiont naturally present in mosquitoes such as A. albopictus, C. pipiens, and C. quinquefaciatus, but is not thought to be present in Anopheles species or A. aegypti [83,84]. Recently, different Wolbachia strains were artificially introduced into A. aegypti (wMelPop‐CLA and wMel from Drosophila [85,86], wAlbB from A. albopictus [87], and wMelwAlbB [88]) where they formed stable infections. In Grenada, C. quinquefasciatus was found to have an abundant population of Wolbachia among their bacterial microbiota (62.79% REA) while the A. aegypti population was not found to carry Wolbachia. This is consistent with other studies in which A. aegypti is rarely, if ever, infected with Wolbachia naturally, whereas C. quinquefasciatus is infected throughout its range and in most individual mosquitoes tested [8991].

Serratia odorifera, a species of Serratia found in both Aedes and Culex in this study. Serratia secretes SmEnhancin, a protein that cleaves off membrane-bound mucins and weakens the peritrophic matrix favoring viral dissemination out of the mosquito midgut, and is thus able to enhance DENV-2 susceptibility in the mosquito [74,92]. Serratia-positive mosquitoes were obtained from DENV endemic regions, while Serratia-negative mosquitoes were caught in non-DENV-endemic regions supporting the hypothesis that microbiota composition may contribute to the observed differences in vector competence across A. aegypti populations [93].

A common parasite found in this study was Albugo, a common plant-pathogen known as white blister rust. Also, of interest is that Ascogregarina was among the parasites found in Aedes. Ascogregarina culicis is a common gregarine parasite of A. aegypti [94]. The sporozoites of these parasites invade the midgut epithelial cells and develop intracellularly and extracellularly in the gut to complete their life cycles. The midgut is also the primary site for virus replication in the vector mosquitoes. In previous studies it was found that Ascogregarina culicis may have an important role in the maintenance of chikungunya virus during the inter‐epidemic period [94].

The main virus that colonizes Aedes aegypti in Grenada is the insect-specific virus belonging to the Phasivirus genus, Phasi charoen-like virus [47]. In Culex, a very large number of unclassified viruses exist. Of those identified, the Circoviridae family was the most abundant, dominated by the animal virus Circovirus, whose natural hosts are pigeons, ducks, and pigs [9597].

The second most abundant viral family was Rhabdoviridae, specifically the Grenada mosquito rhabdovirus 1, a virus first described from a pool of female Deinocerites spp. mosquitoes collected in St. John Parish, Grenada, W.I. in March 2015 [42]. To our knowledge this is the first report of this virus in Culex spp. Recent publication by Shi et al. (2019) [98] reported a novel virus, the Guadeloupe Culex rhabdovirus in C. quinquefasciatus that has a 99.36% identity to the previously reported Grenada mosquito rhabdovirus 1 and the one reported in this study.

The third dominant virus was the insect-specific Culex flavivirus (CxFV). CxFV strains have been detected from different Culex sp. mosquitoes including C. pipiens, C. quinquefasciatus, C. coronator and others, in many parts of the world [99107]. Different strains of CxFV have been characterized in the literature [107]. CxFV in natural mosquito populations is maintained by vertical transmission [108].Though one study suggested a positive association between infection with CxFV and WNV [102], other studies found no effect of CxFV on WNV replication, infection, dissemination, or transmission in C. quinquefasciatus, as well as no significant correlation between CxFV and WNV infection rates throughout the United States [109,110].

Conclusions

Our study describes the composition of microbial communities in two common mosquito genera in Grenada, West Indies. that are involved in the transmission of human pathogens. We have identified few common bacteria among the two arboviral-vectors that may be used in mosquito control strategies. The results highlight the importance of these kind of studies in identifying targets for the development of alternative vector control approaches based on microbiota modification. Future studies are warranted to determine the impact of the organisms found here on the growth and vectorial capacity of the mosquitoes, as well as their feasibility for transgenesis or other mosquito control applications.

Supporting information

S1 Fig. Relative estimated abundance of microorganisms as determined by metagenomic analysis: A. Aedes aegypti and B. Culex quinquefasciatus.

https://doi.org/10.1371/journal.pone.0231047.s001

(TIF)

S2 Fig.

Relative estimated abundance of bacteria as determined by metagenomic analysis: A. Aedes aegypti (A1. By phyla A2. By class A3 by family, and A4. By genera) and B. Culex quinquefasciatus (B1. By phyla B2. By class B3 by family, and B4. By genera).

https://doi.org/10.1371/journal.pone.0231047.s002

(TIF)

S3 Fig.

Relative estimated abundance of fungi as determined by metagenomic analysis: A. Aedes aegypti (A1. By family, and A2. By genera) and B. Culex quinquefasciatus (B. By family).

https://doi.org/10.1371/journal.pone.0231047.s003

(TIF)

S4 Fig.

Relative estimated abundance of parasites as determined by metagenomic analysis: A. Aedes aegypti (A1. By family, and A2. By genera) and B. Culex quinquefasciatus (B1. By family, and B2. By genera).

https://doi.org/10.1371/journal.pone.0231047.s004

(TIF)

S5 Fig. Detection of Trypanosoma in mosquito pools by RT-PCR in Culex quinquefasciatus.

https://doi.org/10.1371/journal.pone.0231047.s005

(TIF)

S6 Fig.

Relative estimated abundance of viruses as determined by metagenomic analysis: A. Aedes aegypti (A1. By family, and A2. By genera) and B. Culex quinquefasciatus (B1. By family, and B2. By genera).

https://doi.org/10.1371/journal.pone.0231047.s006

(TIF)

S7 Fig. Detection of flavivirus(es) in mosquito pools by RT-PCR in Culex quinquefasciatus.

https://doi.org/10.1371/journal.pone.0231047.s007

(TIF)

S8 Fig. Uncropped S5 Fig (per journal requirements).

Top row of gel: First RT-PCR. Bottom row of gel: Second (nested) RT-PCR.

https://doi.org/10.1371/journal.pone.0231047.s008

(TIF)

S9 Fig. Uncropped S7 Fig (per journal requirements).

Top row of gel: Aedes aegypti flavivirus RT-PCR. Bottom row of gel: Culex quinquefasciatus flavivirus RT-PCR.

https://doi.org/10.1371/journal.pone.0231047.s009

(TIF)

References

  1. 1. WHO. Global strategy for dengue prevention and control 2012–2020. World Heal Organiszation. 2012. /entity/denguecontrol/9789241504034/en/index.html
  2. 2. CDC. West Nile Virus—Transmission. Centers Dis Control Prev. 2017.
  3. 3. Famakinde D Mosquitoes and the lymphatic filarial parasites: research trends and budding roadmaps to future disease eradication. Trop Med Infect Dis. 2018. pmid:30274403
  4. 4. Cancrini G, Scaramozzino P, Gabrielli S, Di Paolo M, Toma L, Romi R. Aedes albopictus and Culex pipiens implicated as natural vectors of Dirofilaria repens in central Italy. J Med Entomol. 2007. [1064:aaacpi]2.0.co;2
  5. 5. Cancrini G, Magi M, Gabrielli S, Arispici M, Tolari F, Dell’Omodarme M, et al. Natural vectors of dirofilariasis in rural and urban areas of the Tuscan region, central Italy. J Med Entomol. 2006.
  6. 6. Scolari F, Casiraghi M, Bonizzoni M. Aedes spp. and their microbiota: a review. Front Microbiol. 2019. pmid:31551973
  7. 7. Vontas J, Kioulos E, Pavlidi N, Morou E, della Torre A, Ranson H. Insecticide resistance in the major dengue vectors Aedes albopictus and Aedes aegypti. Pestic Biochem Physiol. 2012.
  8. 8. McCarroll L, Paton MG, Karunaratne SHPP, Jayasuryia HTR, Kalpage KSP, Hemingway J. Insecticides and mosquito-borne disease. Nature. 2000. pmid:11069167
  9. 9. Liu N. Insecticide resistance in mosquitoes: impact, mechanisms, and research directions. Annu Rev Entomol. 2015. pmid:25564745
  10. 10. Zimmermann G. Review on safety of the entomopathogenic fungi Beauveria bassiana and Beauveria brongniartii. Biocontrol Sci Techn. 2007.
  11. 11. Caragata EP, Walker T. Using bacteria to treat diseases. Expert Opin Biol Therpy. 2012. pmid:22500583
  12. 12. Hoffmann AA, Turelli M. Facilitating Wolbachia introductions into mosquito populations through insecticide-resistance selection. Proc R Soc B Biol Sci. 2013. pmid:23576788
  13. 13. Bourtzis K, Dobson SL, Xi Z, Rasgon JL, Calvitti M, Moreira LA, et al. Harnessing mosquito-Wolbachia symbiosis for vector and disease control. Acta Trop. 2014. pmid:24252486
  14. 14. Achee NL, Grieco JP, Vatandoost H, Seixas G, Pinto J, Ching-Ng L, et al. Alternative strategies for mosquito-borne arbovirus control. PLoS Negl Trop Dis. 2019. pmid:30605475
  15. 15. Fang W, Vega-Rodríguez J, Ghosh AK, Jacobs-Lorena M, Kang A, St. Leger RJ. Development of transgenic fungi that kill human malaria parasites in mosquitoes. Science, 2011. pmid:21350178
  16. 16. Fitzpatrick DM, Hattaway LM, Hsueh AN, Ramos-Nino ME, Cheetham SM. PCR-based bloodmeal analysis of Aedes aegypti and Culex quinquefasciatus (Diptera: Culicidae) in St. George Parish, Grenada. J Med Entomol. 2019; 1–6. pmid:30395250
  17. 17. Rawlins SC, Lammie P, Tiwari T, Pons P, Chadee DD, Oostburg BFJ, et al. Lymphatic filariasis in the Caribbean region: The opportunity for its elimination and certification. Rev Panam Salud Publica/Pan Am J Public Heal. 2000. pmid:10893972
  18. 18. Hegde S, Khanipov K, Albayrak L, Golovko G, Pimenova M, Saldaña MA, et al. Microbiome interaction networks and community structure from laboratory-reared and field-collected Aedes aegypti, Aedes albopictus, and Culex quinquefasciatus mosquito vectors. Front Microbiol. 2018. pmid:30250462
  19. 19. Shi C, Beller L, Deboutte W, Yinda KC, Delang L, Vega-Rúa A, et al. Stable distinct core eukaryotic viromes in different mosquito species from Guadeloupe, using single mosquito viral metagenomics. Microbiome. 2019. pmid:31462331
  20. 20. Audsley MD, Ye YH, McGraw EA. The microbiome composition of Aedes aegypti is not critical for Wolbachia-mediated inhibition of dengue virus. PLoS Negl Trop Dis. 2017. pmid:28267749
  21. 21. Boyles SM, Mavian CN, Finol E, Ukhanova M, Stephenson CJ, Hamerlinck G, et al. Under-the-radar dengue virus infections in natural populations of Aedes aegypti mosquitoes. bioRxiv. 2020.
  22. 22. Dickson LB, Ghozlane A, Volant S, Bouchier C, Ma L, Vega-Rúa A, et al. Diverse laboratory colonies of Aedes aegypti harbor the same adult midgut bacterial microbiome. Parasit Vectors. 2018. pmid:29587819
  23. 23. Duguma D, Hall MW, Rugman-Jones P, Stouthamer R, Terenius O, Neufeld JD, et al. Developmental succession of the microbiome of Culex mosquitoes. BMC Microbiol. 2015. pmid:26205080
  24. 24. Thongsripong P, Green A, Kittayapong P, Kapan D, Wilcox B, Bennett S. Mosquito Vector Diversity across Habitats in Central Thailand Endemic for Dengue and Other Arthropod-Borne Diseases. PLoS Negl Trop Dis. 2013. pmid:24205420
  25. 25. Short SM. Investigating variability in the gut microbiota of the dengue vector aedes aegypti. Am J Trop Med Hyg. 2013.
  26. 26. Ponnusamy L, Xu N, Stav G, Wesson DM, Schal C, Apperson CS. Diversity of bacterial communities in container habitats of mosquitoes. Microb Ecol. 2008. pmid:18373113
  27. 27. Guégan M, Zouache K, Démichel C, Minard G, Tran Van V, Potier P, et al. The mosquito holobiont: fresh insight into mosquito-microbiota interactions. Microbiome. 2018. pmid:29554951
  28. 28. Minard G, Mavingui P, Moro CV. Diversity and function of bacterial microbiota in the mosquito holobiont. Parasit Vectors. 2013. pmid:23688194
  29. 29. Mancini M V., Damiani C, Accoti A, Tallarita M, Nunzi E, Cappelli A, et al. Estimating bacteria diversity in different organs of nine species of mosquito by next generation sequencing. BMC Microbiol. 2018. pmid:30286722
  30. 30. Zhang X, Huang S, Jin T, Lin P, Huang Y, Wu C, et al. Discovery and high prevalence of Phasi Charoen-like virus in field-captured Aedes aegypti in South China. Virology. 2018. pmid:30077072
  31. 31. Wang S, Ghosh AK, Bongio N, Stebbings KA, Lampe DJ, Jacobs-Lorena M. Fighting malaria with engineered symbiotic bacteria from vector mosquitoes. Proc Natl Acad Sci U S A. 2012. pmid:22802646
  32. 32. Osei-Poku J, Mbogo CM, Palmer WJ, Jiggins FM. Deep sequencing reveals extensive variation in the gut microbiota of wild mosquitoes from Kenya. Mol Ecol. 2012. pmid:22988916
  33. 33. Muturi EJ, Lagos-Kutz D, Dunlap C, Ramirez JL, Rooney AP, Hartman GL, et al. Mosquito microbiota cluster by host sampling location. Parasit Vectors. 2018. pmid:30107817
  34. 34. Muturi EJ, Dunlap C, Ramirez JL, Rooney AP, Kim CH. Host blood-meal source has a strong impact on gut microbiota of Aedes aegypti. FEMS Microbiol Ecol. 2018. pmid:30357406
  35. 35. Tandina F, Almeras L, Koné AK, Doumbo OK, Raoult D, Parola P. Use of MALDI-TOF MS and culturomics to identify mosquitoes and their midgut microbiota. Parasit Vectors. 2016. pmid:27613238
  36. 36. Ceretti-Junior W, Christe R de O, Rizzo M, Strobel RC, Junior MO de M, Mello MHSH de, et al. Species composition and ecological aspects of immature mosquitoes (Diptera: Culicidae) in bromeliads in urban parks in the city of são paulo, brazil. J Arthropod Borne Dis. 2016.
  37. 37. Gibson KE, Fitzpatrick DM, Stone D, Noel TP, Macpherson CNL. Vector-borne diseases in the Caribbean: History and current status. CAB Reviews: Perspectives in Agriculture, Veterinary Science, Nutrition and Natural Resources. 2016.
  38. 38. Vasconcelos PFC, Calisher CH. Emergence of Human Arboviral Diseases in the Americas, 2000–2016. Vector-Borne and Zoonotic Dis. 2016. pmid:26991057
  39. 39. Sakkas H, Bozidis P, Franks A, Papadopoulou C. Oropouche fever: A review. Viruses. 2018. pmid:29617280
  40. 40. Romero-Alvarez D, Escobar LE. Oropouche fever, an emergent disease from the Americas. Microbes Infect. 2018. pmid:29247710
  41. 41. Muturi EJ, Ramirez JL, Rooney AP, Kim CH. Comparative analysis of gut microbiota of mosquito communities in central Illinois. PLoS Negl Trop Dis. 2017. pmid:28245239
  42. 42. Xu CL, Cantalupo PG, Sáenz-Robles MT, Baldwin A, Fitzpatrick D, Norris DE, et al. Draft genome sequence of a novel rhabdovirus isolated from Deinocerites mosquitoes. Genome Announc. 2018. pmid:29748415
  43. 43. Aldridge RL, Britch SC, Allan SA, Tsikolia M, Calix LC, Bernier UR, et al. Comparison of volatiles and mosquito capture efficacy for three carbohydrate sources in a yeast-fermentation CO 2 generator. J Am Mosq Control Assoc. 2017. pmid:28206863
  44. 44. Darsie Jr R, Ward R. Identification and geographical distribution of the mosquitoes of North America, north of Mexico, University Press of Florida, 2005. https://doi.org/10.1017/s0031182005228834
  45. 45. Gaffigan T, Wilkerson R, Pecor J, Stoffer J, Anderson T. Systematic catalog of Culicidae-Walter Reed Biosystematics Unit, 2015. Available: http://www.mosquitocatalog.org/default.aspx.
  46. 46. Beckmann JF, Fallon AM. Decapitation improves detection of Wolbachia pipientis (Rickettsiales: Anaplasmataceae) in Culex pipiens (Diptera: Culicidae) mosquitoes by the polymerase chain reaction. J Med Entomol. 2012. pmid:23025192
  47. 47. Ramos-Nino ME, Fitzpatrick DM, Tighe S, Eckstrom KM, Hattaway LM, Hsueh AN, et al. High prevalence of Phasi Charoen-like virus from wild-caught Aedes aegypti in Grenada, W. I. As revealed by metagenomic analysis. PLoS One. 2020. pmid:32004323
  48. 48. Sultan M, Amstislavskiy V, Risch T, Schuette M, Dökel S, Ralser M, et al. Influence of RNA extraction methods and library selection schemes on RNA-seq data. BMC Genomics. 2014. pmid:25113896
  49. 49. Cahill RJ, Tan S, Dougan G, O’Gaora P, Pickard D, Kennea N, et al. Universal DNA primers amplify bacterial DNA from human fetal membranes and link Fusobacterium nucleatum with prolonged preterm membrane rupture. Mol Hum Reprod. 2005. pmid:16254004
  50. 50. Sehgal RNM, Jones HI, Smith TB. Host specificity and incidence of Trypanosoma in some African rainforest birds: A molecular approach. Mol Ecol. 2001. pmid:11555273
  51. 51. Scaramozzino N, Crance JM, Jouan A, DeBriel DA, Stoll F, Garin D. Comparison of Flavivirus universal primer pairs and development of a rapid, highly sensitive heminested reverse transcription-PCR assay for detection of flaviviruses targeted to a conserved region of the NS5 gene sequences. J Clin Microbiol. 2001.
  52. 52. Wilke ABB, Marrelli MT. Paratransgenesis: A promising new strategy for mosquito vector control. Parasit Vectors. 2015. pmid:26104575
  53. 53. Flegontov P, Votýpka J, Skalický T, Logacheva MD, Penin AA, Tanifuji G, et al. Paratrypanosoma is a novel early-branching trypanosomatid. Curr Biol. 2013. pmid:24012313
  54. 54. Ricci I, Damiani C, Capone A, DeFreece C, Rossi P, Favia G. Mosquito/microbiota interactions: From complex relationships to biotechnological perspectives. Curr Opin Microbiol. 2012. pmid:22465193
  55. 55. Composition of Anopheles coluzzii and Anopheles gambiae microbiota from larval to adult stages. Infect Genet Evol. 2014. pmid:25283802
  56. 56. Dharne M, Patole M, Shouche YS. Microbiology of the insect gut: Tales from mosquitoes and bees. J Biosci. 2006. pmid:17006010
  57. 57. Ito J, Ghosh A, Moreira LA, Wimmer EA, Jacobs-Lorena M. Transgenic anopheline mosquitoes impaired in transmission of a malaria parasite. Nature. 2002. pmid:12024215
  58. 58. Alphey L. Re-engineering the sterile insect technique. Insect Biochem Mol Biol. 2002.
  59. 59. Riehle MA, Jacobs-Lorena M. Using bacteria to express and display anti-parasite molecules in mosquitoes: Current and future strategies. Insect Biochem Mol Biol. 2005. pmid:15894187
  60. 60. Riehle MA, Moreira CK, Lampe D, Lauzon C, Jacobs-Lorena M. Using bacteria to express and display anti-Plasmodium molecules in the mosquito midgut. Int J Parasitol. 2007. pmid:17224154
  61. 61. Beard C Ben, Cordon-Rosales C, Durvasula R V. Bacterial symbionts of the triatominae and their potential use in control of Chagas disease transmission. Annu Rev Entomol. 2002. pmid:11729071
  62. 62. Beard CB, Mason PW, Aksoy S, Tesh RB, Richards FF. Transformation of an insect symbiont and expression of a foreign gene in the Chagas’ disease vector Rhodnius prolixus. Am J Trop Med Hyg. 1992. pmid:1539755
  63. 63. Beard CB, O’Neill SL, Tesh RB, Richards FF, Aksoy S. Modification of arthropod vector competence via symbiotic bacteria. Parasitol Today. 1993.
  64. 64. Chavshin AR, Oshaghi MA, Vatandoost H, Pourmand MR, Raeisi A, Enayati AA, et al. Identification of bacterial microflora in the midgut of the larvae and adult of wild caught Anopheles stephensi: A step toward finding suitable paratransgenesis candidates. Acta Trop. 2012. pmid:22074685
  65. 65. Chavshin AR, Oshaghi MA, Vatandoost H, Pourmand MR, Raeisi A, Terenius O. Isolation and identification of culturable bacteria from wild Anopheles culicifacies, a first step in a paratransgenesis approach. Parasit Vectors. 2014. pmid:25189316
  66. 66. Sayler GS, Ripp S. Field applications of genetically engineered microorganisms for bioremediation processes. Curr Opin Biotechnol. 2000.
  67. 67. Yoshida S, Ioka D, Matsuoka H, Endo H, Ishii A. Bacteria expressing single-chain immunotoxin inhibit malaria parasite development in mosquitoes. Mol Biochem Parasitol. 2001.
  68. 68. Favia G, Ricci I, Damiani C, Raddadi N, Crotti E, Marzorati M, et al. Bacteria of the genus Asaia stably associate with Anopheles stephensi, an Asian malarial mosquito vector. Proc Natl Acad Sci U S A. 2007. pmid:17502606
  69. 69. De Freece C, Damiani C, Valzano M, D’Amelio S, Cappelli A, Ricci I, et al. Detection and isolation of the α-proteobacterium Asaia in Culex mosquitoes. Med Vet Entomol. 2014. pmid:25387864
  70. 70. Rossi P, Ricci I, Cappelli A, Damiani C, Ulissi U, Mancini MV, et al. Mutual exclusion of Asaia and Wolbachia in the reproductive organs of mosquito vectors. Parasit Vectors. 2015. pmid:25981386
  71. 71. Dennison NJ, Jupatanakul N, Dimopoulos G. The mosquito microbiota influences vector competence for human pathogens. Curr Opin Insect Sci. 2014. pmid:25584199
  72. 72. Crotti E, Damiani C, Pajoro M, Gonella E, Rizzi A, Ricci I, et al. Asaia, a versatile acetic acid bacterial symbiont, capable of cross-colonizing insects of phylogenetically distant genera and orders. Environ Microbiol. 2009. pmid:19735280
  73. 73. Pidiyar VJ, Jangid K, Patole MS, Shouche YS. Studies on cultured and uncultured microbiota of wild Culex quinquefasciatus mosquito midgut based on 16S ribosomal RNA gene analysis. Am J Trop Med Hyg. 2004.
  74. 74. Apte-Deshpande A, Paingankar M, Gokhale MD, Deobagkar DN. Serratia odorifera a midgut inhabitant of aedes aegypti mosquito enhances its susceptibility to dengue-2 virus. PLoS One. 2012. pmid:22848375
  75. 75. Ferguson NM, Kien DTH, Clapham H, Aguas R, Trung VT, Chau TNB, et al. Modeling the impact on virus transmission of Wolbachia-mediated blocking of dengue virus infection of Aedes aegypti. Sci Transl Med. 2015. pmid:25787763
  76. 76. Frentiu FD, Robinson J, Young PR, McGraw EA, O’Neill SL. Wolbachia-mediated resistance to dengue virus infection and death at the cellular level. PLoS One. 2010. pmid:20976219
  77. 77. Frentiu FD, Zakir T, Walker T, Popovici J, Pyke AT, van den Hurk A, et al. Limited Dengue virus replication in field-collected Aedes aegypti mosquitoes infected with Wolbachia. PLoS Negl Trop Dis. 2014. pmid:24587459
  78. 78. Terradas G, McGraw EA. Wolbachia-mediated virus blocking in the mosquito vector Aedes aegypti. Curr Opin Insect Sci. 2017. pmid:28805637
  79. 79. van den Hurk AF, Hall-Mendelin S, Pyke AT, Frentiu FD, McElroy K, Day A, et al. Impact of Wolbachia on infection with chikungunya and yellow fever viruses in the mosquito vector Aedes aegypti. PLoS Negl Trop Dis. 2012. pmid:23133693
  80. 80. Dutra HLC, Rocha MN, Dias FBS, Mansur SB, Caragata EP, Moreira LA. Wolbachia blocks currently circulating Zika virus isolates in brazilian Aedes aegypti mosquitoes. Cell Host Microbe. 2016. pmid:27156023
  81. 81. Caragata EP, Dutra HLC, O’Neill SL, Moreira LA. Zika control through the bacterium Wolbachia pipientis. Future Microbiol. 2016. pmid:27831765
  82. 82. O’Neill SL, Ryan PA, Turley AP, Wilson G, Retzki K, Iturbe-Ormaetxe I, et al. Scaled deployment of Wolbachia to protect the community from dengue and other Aedes transmitted arboviruses. Gates Open Res. 2019. pmid:30596205
  83. 83. Kittayapong P, Baisley KJ, Baimai V, O’Neill SL. Distribution and diversity of Wolbachia infections in southeast Asian mosquitoes (Diptera: Culicidae). J Med Entomol. 2008. [0340:dadowi]2.0.co;2
  84. 84. Amuzu HE, Tsyganov K, Koh C, Herbert RI, Powell DR, McGraw EA. Wolbachia enhances insect-specific flavivirus infection in Aedes aegypti mosquitoes. Ecol Evol. 2018. pmid:29938064
  85. 85. McMeniman CJ, Hughes GL, O’Neill SL. A Wolbachia symbiont in Aedes aegypti disrupts mosquito egg development to a greater extent when mosquitoes feed on nonhuman versus human blood. J Med Entomol. 2011. pmid:21337952
  86. 86. Walker T, Johnson PH, Moreira LA, Iturbe-Ormaetxe I, Frentiu FD, McMeniman CJ, et al. The wMel Wolbachia strain blocks dengue and invades caged Aedes aegypti populations. Nature. 2011. pmid:21866159
  87. 87. Xi Z. Generation of novel symbiosis in mosquitoes via embryo microinjection and characterization of the Wolbachia/host interaction. ProQuest Dissertations and Theses. 2005.
  88. 88. Joubert DA, O’Neill SL. Comparison of stable and transient Wolbachia infection models in Aedes aegypti to block dengue and West Nile viruses. PLoS Negl Trop Dis. 2017. pmid:28052065
  89. 89. Rasgon JL, Cornel AJ, Scott TW. Evolutionary history of a mosquito endosymbiont revealed through mitochondrial hitchhiking. Proc R Soc B Biol Sci. 2006. pmid:16769630
  90. 90. Dumas E, Atyame CM, Milesi P, Fonseca DM, Shaikevich E V., Unal S, et al. Population structure of Wolbachia and cytoplasmic introgression in a complex of mosquito species. BMC Evol Biol. 2013. pmid:24006922
  91. 91. Ross PA, Callahan AG, Yang Q, Jasper M, Arif MAK, Afizah AN, et al. An elusive endosymbiont: Does Wolbachia occur naturally in Aedes aegypti?. Ecol Evol. 2020. pmid:32076535
  92. 92. Wu P, Sun P, Nie K, Zhu Y, Shi M, Xiao C, et al. A gut commensal bacterium promotes mosquito permissiveness to arboviruses. Cell Host Microbe. 2019. pmid:30595552
  93. 93. Souza-Neto JA, Powell JR, Bonizzoni M. Aedes aegypti vector competence studies: A review. Infect Gen Evol. 2019. pmid:30465912
  94. 94. Mourya DT, Singh DK, Yadav P, Gokhale MD, Barde P V., Narayan NB, et al. Role of gregarine parasite Ascogregarina culicis (Apicomplexa: Lecudinidae) in the maintenance of Chikungunya virus in vector mosquito. J Eukaryot Microbiol. 2003. pmid:14563178
  95. 95. Marlier D, Vindevogel H. Viral infections in pigeons. Vet J. 2006. pmid:16772130
  96. 96. Zhang XX, Liu SN, Xie ZJ, Kong YB, Jiang SJ. Complete genome sequence analysis of duck circovirus strains from Cherry Valley duck. Virol Sin. 2012. pmid:22684469
  97. 97. Chae C. Porcine circovirus type 2 and its associated diseases in Korea. Virus Res. 2012. pmid:22027190
  98. 98. Shi C, Beller L, Deboutte W, Yinda KC, Delang L, Vega-Rúa A, et al. Stable distinct core eukaryotic viromes in different mosquito species from Guadeloupe, using single mosquito viral metagenomics. Microbiome. 2019. pmid:31462331
  99. 99. Miranda J, Mattar S, Gonzalez M, Hoyos-López R, Aleman A, Aponte J. First report of Culex flavivirus infection from Culex coronator (Diptera: Culicidae), Colombia. Virol J. 2019. pmid:30606229
  100. 100. Hoshino K, Isawa H, Tsuda Y, Yano K, Sasaki T, Yuda M, et al. Genetic characterization of a new insect flavivirus isolated from Culex pipiens mosquito in Japan. Virology. 2007. pmid:17070886
  101. 101. Goenaga S, Fabbri CM, García JB, Rondán JC, Gardenal N, Calderón GE, et al. New strains of Culex flavivirus isolated in Argentina. J Med Entomol. 2014. pmid:25118428
  102. 102. Newman CM, Krebs BL, Anderson TK, Hamer GL, Ruiz MO, Brawn JD, et al. Culex Flavivirus during West Nile Virus epidemic and interepidemic years in Chicago, United States. Vector-Borne Zoonotic Dis. 2017. pmid:28628366
  103. 103. Calzolari M, Zé-Zé L, Růžek D, Vázquez A, Jeffries C, Defilippo F, et al. Detection of mosquito-only flaviviruses in Europe. J Gen Virol. 2012.
  104. 104. Calzolari M, Zé-Zé L, Vázquez A, Sánchez Seco MP, Amaro F, Dottori M. Insect-specific flaviviruses, a worldwide widespread group of viruses only detected in insects. Infect Genet Evol. 2016. pmid:26235844
  105. 105. Morales-Betoulle ME, Pineda MLM, Sosa SM, Panella N, LóPEZ B MR, Cordón-Rosales C, et al. Culex flavivirus isolates from mosquitoes in Guatemala. J Med Entomol. 2008. [1187:cfifmi]2.0.co;2
  106. 106. Cook S, Moureau G, Harbach RE, Mukwaya L, Goodger K, Ssenfuka F, et al. Isolation of a novel species of flavivirus and a new strain of Culex flavivirus (Flaviviridae) from a natural mosquito population in Uganda. J Gen Virol. 2009.
  107. 107. Kim DY, Guzman H, Bueno R, Dennett JA, Auguste AJ, Carrington CVF, et al. Characterization of Culex flavivirus (Flaviviridae) strains isolated from mosquitoes in the United States and Trinidad. Virology. 2009. pmid:19193389
  108. 108. Bolling BG, Eisen L, Moore CG, Blair CD. Insect-specific flaviviruses from Culex mosquitoes in Colorado, with evidence of vertical transmission. Am J Trop Med Hyg. 2011.
  109. 109. Kent RJ, Crabtree MB, Miller BR. Transmission of West Nile virus by Culex quinquefasciatus say infected with Culex flavivirus izabal. PLoS Negl Trop Dis. 2010;4. pmid:20454569
  110. 110. Crockett RK, Burkhalter K, Mead D, Kelly R, Brown J, Varnado W, et al. Culex Flavivirus and West Nile Virus in Culex quinquefasciatus Populations in the Southeastern United States. J Med Entomol. 2012. pmid:22308785