Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
J Med Entomol ; 61(3): 726-732, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38372693

ABSTRACT

Culex panocossa, Dyar and Knab, an important enzootic vector of Venezuelan equine encephalitis virus subtype ID in Central and South America, was found to have invaded and become established in southern Florida in 2016. No information is currently available regarding the ecology of this invasive mosquito in the United States. Here, we use PCR-based blood meal analysis to investigate vertebrate host associations of Cx. panocossa from Florida to provide information necessary for determining the potential importance of this mosquito for arbovirus transmission in the United States. Culex panocossa fed mainly upon birds (49.5%) but took a substantial fraction of blood meals from mammals (33.3%) and reptiles (17.1%). By feeding upon amplifying hosts of Everglades virus (hispid cotton rat) and eastern equine encephalitis virus (wading birds) and humans, Cx. panocossa could act as a bridge vector for these pathogenic Alphaviruses in Florida, potentially resulting in increased human disease.


Subject(s)
Culex , Mosquito Vectors , Animals , Culex/virology , Culex/physiology , Florida , Mosquito Vectors/virology , Mosquito Vectors/physiology , Birds , Mammals , Reptiles , Arboviruses/physiology , Feeding Behavior , Arbovirus Infections/transmission , Humans , Encephalitis Virus, Eastern Equine/physiology , Female
2.
Viruses ; 16(2)2024 01 30.
Article in English | MEDLINE | ID: mdl-38399982

ABSTRACT

The Eastern Equine Encephalitis Virus (EEEV) is an emerging public health threat, with the number of reported cases in the US increasing in recent years. EEEV is a BSL3 pathogen, and the North American strain is a US Federal Select Agent (SA). These restrictions make experiments with EEEV difficult to perform, as high-tech equipment is often unavailable in BSL3 spaces and due to concerns about generating aerosols during manipulations. Therefore, a range of inactivation methods suitable for different downstream analysis methods are essential for advancing research on EEEV. We used heat, chemical, and ultraviolet (UV)-based methods for the inactivation of infected cells and supernatants infected with the non-select agent Madariaga virus (MADV). Although the MADV and EEEV strains are genetically distinct, differing by 8-11% at the amino acid level, they are expected to be similarly susceptible to various inactivation methods. We determined the following to be effective methods of inactivation: heat, TRIzol LS, 4% PFA, 10% formalin, and UV radiation for infected supernatants; TRIzol, 2.5% SDS with BME, 0.2% NP40, 4% PFA, and 10% formalin for infected cells. Our results have the potential to expand the types and complexity of experiments and analyses performed by EEEV researchers.


Subject(s)
Alphavirus , Encephalitis Virus, Eastern Equine , Encephalomyelitis, Equine , Phenols , Horses , Animals , Encephalitis Virus, Eastern Equine/physiology , Guanidines , Formaldehyde
3.
Cell ; 187(2): 360-374.e19, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38176410

ABSTRACT

The very-low-density lipoprotein receptor (VLDLR) comprises eight LDLR type A (LA) domains and supports entry of distantly related alphaviruses, including Eastern equine encephalitis virus (EEEV) and Semliki Forest virus (SFV). Here, by resolving multiple cryo-electron microscopy structures of EEEV-VLDLR complexes and performing mutagenesis and functional studies, we show that EEEV uses multiple sites (E1/E2 cleft and E2 A domain) to engage more than one LA domain simultaneously. However, no single LA domain is necessary or sufficient to support efficient EEEV infection. Whereas all EEEV strains show conservation of two VLDLR-binding sites, the EEEV PE-6 strain and a few other EEE complex members feature a single amino acid substitution that enables binding of LA domains to an additional site on the E2 B domain. These structural and functional analyses informed the design of a minimal VLDLR decoy receptor that neutralizes EEEV infection and protects mice from lethal challenge.


Subject(s)
Cryoelectron Microscopy , Encephalitis Virus, Eastern Equine , Encephalomyelitis, Equine , Receptors, LDL , Animals , Mice , Alphavirus/physiology , Encephalitis Virus, Eastern Equine/physiology , Encephalitis Virus, Eastern Equine/ultrastructure , Encephalomyelitis, Equine/metabolism , Horses , Protein Binding , Receptors, LDL/ultrastructure
4.
Viruses ; 14(2)2022 01 28.
Article in English | MEDLINE | ID: mdl-35215864

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is an Alphavirus in the Togaviridae family of positive-strand RNA viruses. The viral genome of positive-strand RNA viruses is infectious, as it produces infectious virus upon introduction into a cell. VEEV is a select agent and samples containing viral RNA are subject to additional regulations due to their infectious nature. Therefore, RNA isolated from cells infected with BSL-3 select agent strains of VEEV or other positive-strand viruses must be inactivated before removal from high-containment laboratories. In this study, we tested the inactivation of the viral genome after RNA fragmentation or cDNA synthesis, using the Trinidad Donkey and TC-83 strains of VEEV. We successfully inactivated VEEV genomic RNA utilizing these two protocols. Our cDNA synthesis method also inactivated the genomic RNA of eastern and western equine encephalitis viruses (EEEV and WEEV). We also tested whether the purified VEEV genomic RNA can produce infectious virions in the absence of transfection. Our result showed the inability of the viral genome to cause infection without being transfected into the cells. Overall, this work introduces RNA fragmentation and cDNA synthesis as reliable methods for the inactivation of samples containing the genomes of positive-strand RNA viruses.


Subject(s)
Encephalitis Virus, Venezuelan Equine/genetics , Genome, Viral , RNA, Viral , Virus Inactivation , Animals , Cells, Cultured , Chlorocebus aethiops , Cytopathogenic Effect, Viral , DNA, Complementary/biosynthesis , Encephalitis Virus, Eastern Equine/genetics , Encephalitis Virus, Eastern Equine/physiology , Encephalitis Virus, Venezuelan Equine/physiology , Encephalitis Virus, Western Equine/genetics , Encephalitis Virus, Western Equine/physiology , RNA, Viral/chemistry , RNA, Viral/physiology , Ribonucleases/metabolism , Vero Cells
5.
J Med Entomol ; 59(1): 1-13, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34734628

ABSTRACT

In the current review, we examine the regional history, ecology, and epidemiology of eastern equine encephalitis virus (EEEV) to investigate the major drivers of disease outbreaks in the northeastern United States. EEEV was first recognized as a public health threat during an outbreak in eastern Massachusetts in 1938, but historical evidence for equine epizootics date back to the 1800s. Since then, sporadic disease outbreaks have reoccurred in the Northeast with increasing frequency and northward expansion of human cases during the last 20 yr. Culiseta melanura (Coquillett) (Diptera: Culicidae) serves as the main enzootic vector that drives EEEV transmission among wild birds, but this mosquito species will occasionally feed on mammals. Several species have been implicated as bridge vectors to horses and humans, with Coquilletstidia perturbans (Walker) as a leading suspect based on its opportunistic feeding behavior, vector competence, and high infection rates during recent disease outbreaks. A diversity of bird species are reservoir competent, exposed to EEEV, and serve as hosts for Cs. melanura, with a few species, including the wood thrush (Hlocichia mustelina) and the American robin (Turdus migratorius), contributing disproportionately to virus transmission based on available evidence. The major factors responsible for the sustained resurgence of EEEV are considered and may be linked to regional landscape and climate changes that support higher mosquito densities and more intense virus transmission.


Subject(s)
Birds/virology , Disease Reservoirs/virology , Encephalitis Virus, Eastern Equine/physiology , Encephalomyelitis, Equine , Horse Diseases , Mosquito Vectors , Animals , Encephalomyelitis, Equine/epidemiology , Encephalomyelitis, Equine/transmission , Encephalomyelitis, Equine/veterinary , Encephalomyelitis, Equine/virology , Horse Diseases/epidemiology , Horse Diseases/transmission , Horse Diseases/virology , Horses , Humans , Mid-Atlantic Region/epidemiology , New England/epidemiology
6.
J Med Entomol ; 59(1): 41-48, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34734635

ABSTRACT

Eastern equine encephalitis virus (EEEV; family Togaviridae, genus Alphavirus) is a mosquito-borne pathogen found in eastern North America that causes severe disease in humans and horses. The mosquito Culiseta melanura (Coquillett) (Diptera: Culicidae) is the primary enzootic vector of EEEV throughout eastern North America while several mosquito species belonging to diverse genera serve as bridge vectors. The ecology of EEEV differs between northern and southern foci, with respect to phenology of outbreaks, important vertebrate hosts, and bridge vector species. Active transmission is limited to roughly half of the year in northern foci (New York, New Hampshire, Massachusetts, Connecticut), while year-round transmission occurs in the southeastern region (particularly Florida). Multiple phylogenetic analyses indicate that EEEV strains circulating in northern foci are likely transported from southern foci by migrating birds. Bird species that overwinter or migrate through Florida, are bitten by Cs. melanura in late spring, and arrive at northern breeding grounds in May are the most likely candidates to disperse EEEV northward. Available data indicate that common yellowthroat and green heron satisfy these criteria and could serve as virus dispersers. Understanding the factors that drive the phenology of Cs. melanura reproduction in the south and the timing of avian migration from southern foci could provide insight into how confluence of these biological phenomena shapes outbreaks of EEE throughout its range. This information could be used to develop models predicting the likelihood of outbreaks in a given year, allowing vector control districts to more efficiently marshal resources necessary to protect their stakeholders.


Subject(s)
Encephalitis Virus, Eastern Equine , Encephalomyelitis, Equine , Horse Diseases , Mosquito Vectors , Animals , Encephalitis Virus, Eastern Equine/physiology , Encephalomyelitis, Equine/epidemiology , Encephalomyelitis, Equine/transmission , Encephalomyelitis, Equine/veterinary , Encephalomyelitis, Equine/virology , Horse Diseases/epidemiology , Horse Diseases/transmission , Horse Diseases/virology , Horses , Southeastern United States/epidemiology , Tennessee
7.
J Med Entomol ; 58(6): 2385-2397, 2021 11 09.
Article in English | MEDLINE | ID: mdl-33893734

ABSTRACT

Eastern equine encephalitis virus (EEEV) is the most pathogenic arbovirus endemic to the United States. Studies have demonstrated Florida's role as a regional reservoir for the virus and its ability to support year-round transmission. Previous research has developed risk index models for mapping locations most at risk for EEEV transmission. We compared vector abundance, vector feeding behavior, potential host species, and fauna presence at high versus low-moderate risk sites during the winter and spring. Predicted high-risk sites had a significantly greater abundance of mosquitoes overall, including Culiseta melanura (Coquillett) (Diptera: Culicidae), the primary enzootic vector of EEEV. Twenty host species were identified from Cs. melanura bloodmeals, with the majority taken from avian species. Culiseta melanura largely fed upon the Northern Cardinal (Cardinalis cardinalis (Passeriformes: Cardinalidae)), which accounted for 20-24.4% of the bloodmeals obtained from this species in years 1 and 2, respectively. One EEEV-positive mosquito pool (Cs. melanura) and nine EEEV seropositive sentinel chickens were confirmed during winter-spring collections from high-risk sites; no seropositive chickens nor mosquito pools were found at the low-moderate risk sites. These results suggest that high-risk sites for EEEV activity are characterized by habitats that support populations of Cs. melanura and which may also provide ample opportunities to feed upon Northern Cardinals. The overall low level of mosquito populations during the winter also suggests that control of Cs. melanura populations in winter at high-risk sites may prove effective in reducing EEEV transmission during the peak summer season.


Subject(s)
Culicidae/physiology , Encephalitis Virus, Eastern Equine/physiology , Food Chain , Songbirds , Animals , Environment , Feeding Behavior , Florida , Seasons
8.
Cell Rep ; 25(11): 3136-3147.e5, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30540945

ABSTRACT

Alphaviruses are enveloped pathogens that cause arthritis and encephalitis. Here, we report a 4.4-Å cryoelectron microscopy (cryo-EM) structure of eastern equine encephalitis virus (EEEV), an alphavirus that causes fatal encephalitis in humans. Our analysis provides insights into viral entry into host cells. The envelope protein E2 showed a binding site for the cellular attachment factor heparan sulfate. The presence of a cryptic E2 glycan suggests how EEEV escapes surveillance by lectin-expressing myeloid lineage cells, which are sentinels of the immune system. A mechanism for nucleocapsid core release and disassembly upon viral entry was inferred based on pH changes and capsid dissociation from envelope proteins. The EEEV capsid structure showed a viral RNA genome binding site adjacent to a ribosome binding site for viral genome translation following genome release. Using five Fab-EEEV complexes derived from neutralizing antibodies, our investigation provides insights into EEEV host cell interactions and protective epitopes relevant to vaccine design.


Subject(s)
Antibodies, Viral/immunology , Cryoelectron Microscopy , Encephalitis Virus, Eastern Equine/physiology , Encephalitis Virus, Eastern Equine/ultrastructure , Neutralization Tests , Virus Assembly/physiology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Binding Sites , Capsid Proteins/chemistry , Capsid Proteins/ultrastructure , Cell Line, Tumor , Glycosylation , Heparitin Sulfate/metabolism , Humans , Integrins/metabolism , Models, Molecular , Protein Multimerization , Static Electricity
10.
PLoS Negl Trop Dis ; 12(8): e0006698, 2018 08.
Article in English | MEDLINE | ID: mdl-30118494

ABSTRACT

BACKGROUND: Eastern Equine Encephalitis (EEE) (Togaviridae, Alphavirus) is a highly pathogenic mosquito-borne arbovirus that circulates in an enzootic cycle involving Culiseta melanura mosquitoes and wild Passeriformes birds in freshwater swamp habitats. Recently, the northeastern United States has experienced an intensification of virus activity with increased human involvement and northward expansion into new regions. In addition to its principal role in enzootic transmission of EEE virus among avian hosts, recent studies on the blood-feeding behavior of Cs. melanura throughout its geographic range suggest that this mosquito may also be involved in epizootic / epidemic transmission to equines and humans in certain locales. Variations in blood feeding behavior may be a function of host availability, environmental factors, and/or underlying genetic differences among regional populations. Despite the importance of Cs. melanura in transmission and maintenance of EEE virus, the genetics of this species remains largely unexplored. METHODOLOGY AND PRINCIPLE FINDINGS: To investigate the occurrence of genetic variation in Cs. melanura, the genome of this mosquito vector was sequenced resulting in a draft genome assembly of 1.28 gigabases with a contig N50 of 93.36 kilobases. Populations of Cs. melanura from 10 EEE virus foci in the eastern North America were genotyped with double-digest RAD-seq. Following alignment of reads to the reference genome, variant calling, and filtering, 40,384 SNPs were retained for downstream analyses. Subsequent analyses revealed genetic differentiation between northern and southern populations of this mosquito species. Moreover, limited fine-scale population structure was detected throughout northeastern North America, suggesting local differentiation of populations but also a history of ancestral polymorphism or contemporary gene flow. Additionally, a genetically distinct cluster was identified predominantly at two northern sites. CONCLUSION AND SIGNIFICANCE: This study elucidates the first evidence of fine-scale population structure in Cs. melanura throughout its eastern range and detects evidence of gene flow between populations in northeastern North America. This investigation provides the groundwork for examining the consequences of genetic variations in the populations of this mosquito species that could influence vector-host interactions and the risk of human and equine infection with EEE virus.


Subject(s)
Culicidae/genetics , Encephalitis Virus, Eastern Equine/physiology , Mosquito Vectors/genetics , Animal Distribution , Animals , Cluster Analysis , Culicidae/virology , DNA/genetics , Female , Genetic Variation , Genome , Genotype , Mosquito Vectors/virology , Multivariate Analysis , Polymerase Chain Reaction , United States
11.
Parasit Vectors ; 10(1): 501, 2017 Oct 18.
Article in English | MEDLINE | ID: mdl-29047412

ABSTRACT

BACKGROUND: Eastern equine encephalitis virus (EEEV) is an expanding mosquito-borne threat to humans and domestic animal populations in the northeastern United States. Outbreaks of EEEV are challenging to predict due to spatial and temporal uncertainty in the abundance and viral infection of Cs. melanura, the principal enzootic vector. EEEV activity may be closely linked to wetlands because they provide essential habitat for mosquito vectors and avian reservoir hosts. However, wetlands are not homogeneous and can vary by vegetation, connectivity, size, and inundation patterns. Wetlands may also have different effects on EEEV transmission depending on the assessed spatial scale. We investigated associations between wetland characteristics and Cs. melanura abundance and infection with EEEV at multiple spatial scales in Connecticut, USA. RESULTS: Our findings indicate that wetland vegetative characteristics have strong associations with Cs. melanura abundance. Deciduous and evergreen forested wetlands were associated with higher Cs. melanura abundance, likely because these wetlands provide suitable subterranean habitat for Cs. melanura development. In contrast, Cs. melanura abundance was negatively associated with emergent and scrub/shrub wetlands, and wetland connectivity to streams. These relationships were generally strongest at broad spatial scales. Additionally, the relationships between wetland characteristics and EEEV infection in Cs. melanura were generally weak. However, Cs. melanura abundance was strongly associated with EEEV infection, suggesting that wetland-associated changes in abundance may be indirectly linked to EEEV infection in Cs. melanura. Finally, we found that wet hydrological conditions during the transmission season and during the fall/winter preceding the transmission season were associated with higher Cs. melanura abundance and EEEV infection, indicating that wet conditions are favorable for EEEV transmission. CONCLUSIONS: These results expand the broad-scale understanding of the effects of wetlands on EEEV transmission and help to reduce the spatial and temporal uncertainty associated with EEEV outbreaks.


Subject(s)
Culicidae/virology , Encephalitis Virus, Eastern Equine/isolation & purification , Encephalomyelitis, Eastern Equine/veterinary , Insect Vectors/virology , Animals , Birds , Disease Outbreaks/veterinary , Ecosystem , Encephalitis Virus, Eastern Equine/physiology , Encephalomyelitis, Eastern Equine/epidemiology , Encephalomyelitis, Eastern Equine/transmission , Encephalomyelitis, Eastern Equine/virology , Female , Horses , New England , Seasons
12.
Epidemiol Infect ; 145(14): 2940-2948, 2017 10.
Article in English | MEDLINE | ID: mdl-28956525

ABSTRACT

Periodic outbreaks of West Nile virus (WNV), Eastern equine encephalitis virus (EEEV) and to a lesser extent, California serogroup viruses (CSGV), have been reported in parts of Canada in the last decade. This study was designed to provide a broad assessment of arboviral activity in Quebec, Canada, by conducting serological surveys for these arboviruses in 196 horses, 1442 dogs and 485 humans. Sera were screened by a competitive enzyme linked immunosorbent assay and positive samples confirmed by plaque reduction neutralisation tests. The percentage of seropositive samples was 83·7%, 16·5%, 7·1% in horses, 18·8%, 0·6%, 0% in humans, 11·7%, 3·1%, 0% in adult dogs and 2·9%, 0·3%, 0% in juvenile dogs for CSGV, WNV and EEEV, respectively. Serological results in horses and dogs appeared to provide a meaningful assessment of risk to public health posed by multiple arboviruses.


Subject(s)
Arbovirus Infections/epidemiology , Arbovirus Infections/veterinary , Communicable Diseases, Emerging/epidemiology , Adult , Animals , Arbovirus Infections/virology , Arboviruses/physiology , Communicable Diseases, Emerging/virology , Dog Diseases/blood , Dog Diseases/epidemiology , Dog Diseases/virology , Dogs , Encephalitis Virus, California/physiology , Encephalitis Virus, Eastern Equine/physiology , Encephalitis, California/epidemiology , Encephalitis, California/virology , Encephalomyelitis, Equine/epidemiology , Encephalomyelitis, Equine/virology , Female , Horse Diseases/blood , Horse Diseases/epidemiology , Horse Diseases/virology , Horses , Humans , Male , Middle Aged , Public Health , Quebec/epidemiology , West Nile Fever/epidemiology , West Nile Fever/virology , West Nile virus/physiology
13.
J Med Entomol ; 54(5): 1365-1374, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28874017

ABSTRACT

Field studies were carried out in four Florida counties to investigate winter and spring ecology of host use by Culiseta melanura (Coquillet), the primary vector of eastern equine encephalomyelitis virus (EEEV) in North America. Bloodmeal analysis by PCR was used to identify 233 host bloodmeals, which mainly originated from birds (78.5%) and reptiles (17.2%), primarily Anolis spp. lizards. Across counties, the percentage of bloodmeals from reptiles (7-37% depending upon county) increased with increasing day length and temperature in the spring. Multiple logistic regression revealed that differences in reptile host use across collection sites were largely explained by differences in average day length and temperature on the day of collection, and is probably owing to environment-driven behavioral patterns of ectothermic animals. Although past studies have demonstrated reptile biting by epizootic vectors of EEEV, including Culex (Melanoconion) spp., this is the first study to demonstrate widespread and common feeding upon ectothermic hosts by Cs. melanura. This work suggests that reptiles, particularly anole lizards, play a role in the ecology of EEEV in Florida either as amplifying hosts or as noncompetent hosts which dilute vector feedings thereby suppressing transmission. Detailed laboratory studies investigating impacts of environmental variables (temperature and photoperiod) on EEEV competence of anoles are needed to assess whether these animals support virus amplification.


Subject(s)
Culicidae/physiology , Environment , Food Chain , Mosquito Vectors/physiology , Animals , Culicidae/virology , Encephalitis Virus, Eastern Equine/physiology , Feeding Behavior , Female , Florida , Lizards , Mosquito Vectors/virology , Population Dynamics , Seasons
14.
J Virol ; 91(14)2017 07 15.
Article in English | MEDLINE | ID: mdl-28468889

ABSTRACT

Eastern equine encephalitis virus (EEEV) is a representative member of the New World alphaviruses. It is pathogenic for a variety of vertebrate hosts, in which EEEV induces a highly debilitating disease, and the outcomes are frequently lethal. Despite a significant public health threat, the molecular mechanism of EEEV replication and interaction with hosts is poorly understood. Our previously published data and those of other teams have demonstrated that hypervariable domains (HVDs) of the alphavirus nsP3 protein interact with virus-specific host factors and play critical roles in assembly of viral replication complexes (vRCs). The most abundantly represented HVD-binding proteins are the FXR and G3BP family members. FXR proteins drive the assembly of vRCs of Venezuelan equine encephalitis virus (VEEV), and G3BPs were shown to function in vRC assembly in the replication of chikungunya and Sindbis viruses. Our new study demonstrates that EEEV exhibits a unique level of redundancy in the use of host factors in RNA replication. EEEV efficiently utilizes both the VEEV-specific FXR protein family and the Old World alphavirus-specific G3BP protein family. A lack of interaction with either FXRs or G3BPs does not affect vRC formation; however, removal of EEEV's ability to interact with both protein families has a deleterious effect on virus growth. Other identified EEEV nsP3 HVD-interacting host proteins are also capable of supporting EEEV replication, albeit with a dramatically lower efficiency. The ability to use a wide range of host factors with redundant functions in vRC assembly and function provides a plausible explanation for the efficient replication of EEEV and may contribute to its highly pathogenic phenotype.IMPORTANCE Eastern equine encephalitis virus (EEEV) is one of the most pathogenic New World alphaviruses. Despite the continuous public health threat, to date, the molecular mechanisms of its very efficient replication and high virulence are not sufficiently understood. The results of this new study demonstrate that North American EEEV exhibits a high level of redundancy in using host factors in replication complex assembly and virus replication. The hypervariable domain of the EEEV nsP3 protein interacts with all of the members of the FXR and G3BP protein families, and only a lack of interaction with both protein families strongly affects virus replication rates. Other identified HVD-binding factors are also involved in EEEV replication, but their roles are not as critical as those of FXRs and G3BPs. The new data present a plausible explanation for the exceptionally high replication rates of EEEV and suggest a new means of its attenuation and new targets for screening of antiviral drugs.


Subject(s)
Encephalitis Virus, Eastern Equine/physiology , Host-Pathogen Interactions , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Cell Line
15.
PLoS One ; 12(2): e0172309, 2017.
Article in English | MEDLINE | ID: mdl-28222120

ABSTRACT

Concurrent ingestion of microfilariae (MF) and arboviruses by mosquitoes can enhance mosquito transmission of virus compared to when virus is ingested alone. Within hours of being ingested, MF penetrate the mosquito midgut and introduce virus into mosquito hemocoel, creating a disseminated viral infection much sooner than normal. How virus is actually introduced is not known. In this report, we present experimental evidence that suggests that certain alphaviruses may adhere or otherwise associate with sheathed Brugia malayi MF in the blood of a dually-infected host and that the virus is carried into the mosquito hemocoel by the MF during their penetration of the mosquito midgut. The mechanism of MF enhancement may be more complex than simple leakage of viremic blood into the hemocoel during MF penetration. The affinity of arboviruses to adhere to or otherwise associate with MF may depend on the specific combination of the virus and MF involved in a dual host infection. This in turn may determine the relative importance that MF enhancement has within an arbovirus transmission system.


Subject(s)
Aedes/virology , Brugia malayi/physiology , Encephalitis Virus, Eastern Equine , Encephalitis Virus, Venezuelan Equine , Filariasis/transmission , Insect Vectors/virology , Intestines/parasitology , Intestines/virology , Microfilariae/physiology , Aedes/parasitology , Aedes/physiology , Animals , Biological Transport , Blood/parasitology , Blood/virology , Disease Susceptibility , Encephalitis Virus, Eastern Equine/physiology , Encephalitis Virus, Venezuelan Equine/physiology , Feeding Behavior , Filariasis/parasitology , Filariasis/virology , Gerbillinae , Insect Vectors/parasitology , Insect Vectors/physiology , Salivary Glands/virology , Species Specificity , Viremia/transmission , Viremia/virology , Virus Replication
16.
Parasit Vectors ; 9: 474, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27577939

ABSTRACT

BACKGROUND: Eastern equine encephalitis virus (EEEV) is a highly pathogenic mosquito-borne arbovirus, with active transmission foci in freshwater hardwood swamps in eastern North America, where enzootic transmission is maintained between the ornithophilic mosquito, Culiseta melanura, and wild passerine birds. The role of other locally abundant mosquito species in virus transmission and their associations with vertebrate hosts as sources of blood meals within these foci are largely unknown but are of importance in clarifying the dynamics of enzootic and epidemic/epizootic transmission. METHODS: Blood-engorged mosquitoes were collected from resting boxes at four established EEEV foci in Connecticut during 2010-2011. Mosquitoes were identified to species, and the identity of vertebrate hosts was determined based on mitochondrial cytochrome b and/or cytochrome c oxidase subunit I gene sequences of polymerase chain reaction products. RESULTS: The vertebrate hosts of 378 (50.3 % of engorged mosquitoes) specimens, representing 12 mosquito species, were identified. Culiseta morsitans (n = 54; 67.5 %), Culex restuans (n = 4; 66.7 %), and Cx. pipiens (n = 2; 100 %) acquired blood meals exclusively from avian hosts, whereas Aedes cinereus (n = 6; 66.7 %), Ae. canadensis (n = 2; 100 %), and Ae. stimulans (n = 1; 100 %) obtained blood meals solely from mammals. Species that fed opportunistically on both avian and mammalian hosts included: Ae. thibaulti (n = 21 avian, and n = 181 mammalian; 57.2 %), Anopheles punctipennis (n = 8 and n = 40; 44.0 %), An. quadrimaculatus (n = 1 and n = 23; 35.7 %), Coquillettidia perturbans (n = 3 and n = 3; 46.2 %) and Ae. abserratus (n = 1 and n = 2; 23.1 %). Culex territans obtained blood meals from avian and amphibian hosts (n = 18 and n = 5; 26.6 %). Mixed blood meals originating from both avian and mammalian hosts were identified in An. quadrimaculatus (n = 1), and Cx. territans (n = 2). CONCLUSIONS: Our findings indicate that wood thrush, tufted titmouse, and a few other avian species serve as hosts for mosquitoes, and likely contribute to amplification of EEEV. Our study supports the role of Cs. morsitans in enzootic transmission of EEEV among avian species. Culex territans will seek blood from multiple vertebrate classes, suggesting that this species may contribute to epizootic transmission of the virus. Our findings support roles for Cq. perturbans and An. quadrimaculatus as epidemic/epizootic vectors to humans, horses, and white-tailed deer. Despite its abundance, the potential of Ae. thibaulti to serve as a "bridge vector" for EEEV remains unclear in the absence of any definitive knowledge on its competency for the virus. The contribution of white-tailed deer to the dynamics of EEEV transmission is not fully understood, but findings indicate repeated exposure due to frequent feeding by vector competent mosquito species.


Subject(s)
Bird Diseases/virology , Culicidae/virology , Encephalitis Virus, Eastern Equine/physiology , Passeriformes/virology , Animals , Bird Diseases/epidemiology , Connecticut , Culicidae/classification , Mammals/blood , Passeriformes/blood
17.
J Virol Methods ; 234: 52-3, 2016 08.
Article in English | MEDLINE | ID: mdl-27079827

ABSTRACT

An examination using the routine rabies direct fluorescent antibody test was performed on rabies or Eastern equine encephalitis positive mammalian brain tissue to assess inactivation of the virus. Neither virus was inactivated with acetone fixation nor the routine test, thus laboratory employees should treat all samples as rabies and when appropriate Eastern equine encephalitis positive throughout the whole procedure.


Subject(s)
Encephalitis Virus, Eastern Equine/physiology , Encephalomyelitis, Eastern Equine/veterinary , Fluorescent Antibody Technique, Direct , Rabies virus/immunology , Rabies virus/physiology , Virus Inactivation , Acetone/chemistry , Acetone/pharmacology , Animals , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Brain/virology , Encephalitis Virus, Eastern Equine/immunology , Encephalomyelitis, Eastern Equine/diagnosis , Encephalomyelitis, Eastern Equine/immunology , Encephalomyelitis, Eastern Equine/virology , Histological Techniques/methods , Horses , Humans , Rabies/veterinary , Staining and Labeling/methods , Staining and Labeling/standards
18.
PLoS Negl Trop Dis ; 10(1): e0004347, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26751704

ABSTRACT

BACKGROUND: Eastern equine encephalitis (EEE) virus (Togaviridae, Alphavirus) is a highly pathogenic mosquito-borne zoonosis that is responsible for occasional outbreaks of severe disease in humans and equines, resulting in high mortality and neurological impairment in most survivors. In the past, human disease outbreaks in the northeastern U.S. have occurred intermittently with no apparent pattern; however, during the last decade we have witnessed recurring annual emergence where EEE virus activity had been historically rare, and expansion into northern New England where the virus had been previously unknown. In the northeastern U.S., EEE virus is maintained in an enzootic cycle involving the ornithophagic mosquito, Culiseta melanura, and wild passerine (perching) birds in freshwater hardwood swamps. However, the identity of key avian species that serve as principal virus reservoir and amplification hosts has not been established. The efficiency with which pathogen transmission occurs within an avian community is largely determined by the relative reservoir competence of each species and by ecological factors that influence contact rates between these avian hosts and mosquito vectors. METHODOLOGY AND PRINCIPLE FINDINGS: Contacts between vector mosquitoes and potential avian hosts may be directly quantified by analyzing the blood meal contents of field-collected specimens. We used PCR-based molecular methods and direct sequencing of the mitochondrial cytochrome b gene for profiling of blood meals in Cs. melanura, in an effort to quantify its feeding behavior on specific vertebrate hosts, and to infer epidemiologic implications in four historic EEE virus foci in the northeastern U.S. Avian point count surveys were conducted to determine spatiotemporal host community composition. Of 1,127 blood meals successfully identified to species level, >99% of blood meals were from 65 avian hosts in 27 families and 11 orders, and only seven were from mammalian hosts representing three species. We developed an empirically informed mathematical model for EEE virus transmission using Cs. melanura abundance and preferred and non-preferred avian hosts. To our knowledge this is the first mathematical model for EEE virus, a pathogen with many potential hosts, in the northeastern U.S. We measured strong feeding preferences for a number of avian species based on the proportion of mosquito blood meals identified from these bird species in relation to their observed frequencies. These included: American Robin, Tufted Titmouse, Common Grackle, Wood Thrush, Chipping Sparrow, Black-capped Chickadee, Northern Cardinal, and Warbling Vireo. We found that these bird species, most notably Wood Thrush, play a dominant role in supporting EEE virus amplification. It is also noteworthy that the competence of some of the aforementioned avian species for EEE virus has not been established. Our findings indicate that heterogeneity induced by mosquito host preference, is a key mediator of the epizootic transmission of vector-borne pathogens. CONCLUSION AND SIGNIFICANCE: Detailed knowledge of the vector-host interactions of mosquito populations in nature is essential for evaluating their vectorial capacity and for assessing the role of individual vertebrates as reservoir hosts involved in the maintenance and amplification of zoonotic agents of human diseases. Our study clarifies the host associations of Cs. melanura in four EEE virus foci in the northeastern U.S., identifies vector host preferences as the most important transmission parameter, and quantifies the contribution of preference-induced contact heterogeneity to enzootic transmission. Our study identifies Wood Thrush, American Robin and a few avian species that may serve as superspreaders of EEE virus. Our study elucidates spatiotemporal host species utilization by Cs. melanura in relation to avian host community. This research provides a basis to better understand the involvement of Cs. melanura and avian hosts in the transmission and ecology of EEE virus and the risk of human infection in virus foci.


Subject(s)
Birds/blood , Culicidae/physiology , Encephalitis Virus, Eastern Equine/physiology , Encephalomyelitis, Eastern Equine/veterinary , Insect Vectors/physiology , Animal Distribution , Animals , Birds/classification , Connecticut/epidemiology , Disease Reservoirs , Ecosystem , Encephalomyelitis, Eastern Equine/epidemiology , Encephalomyelitis, Eastern Equine/virology , Encephalomyelitis, Equine/epidemiology , Encephalomyelitis, Equine/veterinary , Encephalomyelitis, Equine/virology , Feeding Behavior , Female , Humans
19.
J Med Entomol ; 53(2): 473-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26659606

ABSTRACT

Field studies of the ecology of eastern equine encephalitis virus (EEEV; family Togaviridae, genus Alphavirus) in the southeastern United States have demonstrated that Culex erraticus (Dyar and Knab) is the most common mosquito at many enzootic sites and is often infected with the virus. However, the competence of Cx. erraticus for EEEV has not been explored in detail. Culex erraticus females were collected from the field and fed upon EEEV-infected chicks. The infected mosquitoes were provided honey for nutrition and to monitor for time to infectiveness. Of the mosquitoes that survived the 14-d postfeeding period, 89% were infected and 84% had evidence of a disseminated infection, though titers were generally low. EEEV was first detected in honey 6 d postinfection and was detected in samples collected from 94% of the mosquitoes with a disseminated infection overall. These data and others were then employed to estimate the relative vectorial capacity of Cx. erraticus at an EEEV enzootic site in Alabama. The vectorial capacity of Cx. erraticus at this site was 44% of Culiseta melanura (Coquillett), the accepted enzootic vector, suggesting Cx. erraticus may play a role in transmitting EEEV in areas where it is abundant and Cs. melanura rare.


Subject(s)
Culex/virology , Encephalitis Virus, Eastern Equine/physiology , Insect Vectors/virology , Animals , Female , Southeastern United States
20.
Parasit Vectors ; 8: 516, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26453283

ABSTRACT

BACKGROUND: Eastern equine encephalomyelitis virus (EEEV) causes a highly pathogenic zoonosis that circulates in an enzootic cycle involving the ornithophagic mosquito, Culiseta melanura, and wild passerine birds in freshwater hardwood swamps in the northeastern U.S. Epidemic/epizootic transmission to humans/equines typically occurs towards the end of the transmission season and is generally assumed to be mediated by locally abundant and contiguous mammalophagic "bridge vector" mosquitoes. METHODS: Engorged mosquitoes were collected using CDC light, resting box, and gravid traps during epidemic transmission of EEEV in 2012 in Addison and Rutland counties, Vermont. Mosquitoes were identified to species and blood meal analysis performed by sequencing mitochondrial cytochrome b gene polymerase chain reaction products. Infection status with EEEV in mosquitoes was determined using cell culture and RT-PCR assays, and all viral isolates were sequenced and compared to other EEEV strains by phylogenetic analysis. RESULTS: The host choices of 574 engorged mosquitoes were as follows: Cs. melanura (n = 331, 94.3 % avian-derived, 5.7 % mammalian-derived); Anopheles quadrimaculatus (n = 164, 3.0 % avian, 97.0 % mammalian); An. punctipennis (n = 56, 7.2 % avian, 92.8 % mammalian), Aedes vexans (n = 9, 22.2 % avian, 77.8 % mammalian); Culex pipiens s.l. n = 6, 100 % avian); Coquillettidia perturbans (n = 4, 25.0 % avian, 75.0 % mammalian); and Cs. morsitans (n = 4, 100 % avian). A seasonal shift in blood feeding by Cs. melanura from Green Heron towards other avian species was observed. EEEV was successfully isolated from blood-fed Cs. melanura and analyzed by phylogenetic analysis. Vermont strains from 2012 clustered with viral strains previously isolated in Virginia yet were genetically distinct from an earlier EEEV isolate from Vermont during 2011. CONCLUSIONS: Culiseta melanura acquired blood meals primarily from birds and focused feeding activity on several competent species capable of supporting EEEV transmission. Culiseta melanura also occasionally obtained blood meals from mammalian hosts including humans. This mosquito species serves as the primary vector of EEEV among wild bird species, but also is capable of occasionally contributing to epidemic/epizootic transmission of EEEV to humans/equines. Other mosquito species including Cq. perturbans that feed more opportunistically on both avian and mammalian hosts may be important in epidemic/epizootic transmission under certain conditions. Phylogenetic analyses suggest that EEEV was independently introduced into Vermont on at least two separate occasions.


Subject(s)
Culicidae/virology , Encephalitis Virus, Eastern Equine/physiology , Encephalomyelitis, Equine/veterinary , Encephalomyelitis, Equine/virology , Horse Diseases/virology , Insect Vectors/virology , Animals , Birds/classification , Birds/virology , Culicidae/classification , Culicidae/genetics , Culicidae/growth & development , Encephalitis Virus, Eastern Equine/classification , Encephalitis Virus, Eastern Equine/genetics , Encephalitis Virus, Eastern Equine/isolation & purification , Encephalomyelitis, Equine/transmission , Horses , Humans , Insect Vectors/classification , Insect Vectors/genetics , Insect Vectors/growth & development , Molecular Sequence Data , New England , Phylogeny
SELECTION OF CITATIONS
SEARCH DETAIL
...