Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 617
Filter
1.
Viruses ; 16(6)2024 May 24.
Article in English | MEDLINE | ID: mdl-38932129

ABSTRACT

The complete lack of yellow fever virus (YFV) in Asia, and the lack of urban YFV transmission in South America, despite the abundance of the peridomestic mosquito vector Aedes (Stegomyia.) aegypti is an enigma. An immunologically naïve population of over 2 billion resides in Asia, with most regions infested with the urban YF vector. One hypothesis for the lack of Asian YF, and absence of urban YF in the Americas for over 80 years, is that prior immunity to related flaviviruses like dengue (DENV) or Zika virus (ZIKV) modulates YFV infection and transmission dynamics. Here we utilized an interferon α/ß receptor knock-out mouse model to determine the role of pre-existing dengue-2 (DENV-2) and Zika virus (ZIKV) immunity in YF virus infection, and to determine mechanisms of cross-protection. We utilized African and Brazilian YF strains and found that DENV-2 and ZIKV immunity significantly suppresses YFV viremia in mice, but may or may not protect relative to disease outcomes. Cross-protection appears to be mediated mainly by humoral immune responses. These studies underscore the importance of re-assessing the risks associated with YF outbreak while accounting for prior immunity from flaviviruses that are endemic.


Subject(s)
Cross Protection , Dengue Virus , Disease Models, Animal , Mice, Knockout , Receptor, Interferon alpha-beta , Yellow Fever , Yellow fever virus , Zika Virus Infection , Zika Virus , Animals , Yellow Fever/immunology , Yellow Fever/prevention & control , Yellow Fever/virology , Mice , Cross Protection/immunology , Yellow fever virus/immunology , Zika Virus/immunology , Zika Virus Infection/immunology , Zika Virus Infection/prevention & control , Zika Virus Infection/virology , Dengue Virus/immunology , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/deficiency , Antibodies, Viral/immunology , Antibodies, Viral/blood , Flavivirus/immunology , Aedes/virology , Aedes/immunology , Dengue/immunology , Dengue/prevention & control , Dengue/virology , Female , Viremia/immunology , Mosquito Vectors/virology , Mosquito Vectors/immunology , Flavivirus Infections/immunology , Flavivirus Infections/prevention & control , Flavivirus Infections/virology , Mice, Inbred C57BL
2.
Viruses ; 16(5)2024 05 20.
Article in English | MEDLINE | ID: mdl-38793692

ABSTRACT

Duck Tembusu Virus (DTMUV) is a pathogen of the Flaviviridae family that causes infections in poultry, leading to significant economic losses in the duck farming industry in recent years. Ducks infected with this virus exhibit clinical symptoms such as decreased egg production and neurological disorders, along with serious consequences such as ovarian hemorrhage, organ enlargement, and necrosis. Variations in morbidity and mortality rates exist across different age groups of ducks. It is worth noting that DTMUV is not limited to ducks alone; it can also spread to other poultry such as chickens and geese, and antibodies related to DTMUV have even been found in duck farm workers, suggesting a potential risk of zoonotic transmission. This article provides a detailed overview of DTMUV research, delving into its genomic characteristics, vaccines, and the interplay with host immune responses. These in-depth research findings contribute to a more comprehensive understanding of the virus's transmission mechanism and pathogenic process, offering crucial scientific support for epidemic prevention and control.


Subject(s)
Ducks , Flavivirus Infections , Flavivirus , Poultry Diseases , Animals , Ducks/virology , Flavivirus/pathogenicity , Flavivirus/immunology , Flavivirus/genetics , Flavivirus Infections/veterinary , Flavivirus Infections/virology , Flavivirus Infections/transmission , Genome, Viral , Poultry Diseases/virology , Poultry Diseases/transmission , Viral Vaccines/immunology , Farmers , Antibodies, Viral/blood , Humans
3.
Antiviral Res ; 227: 105915, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38777094

ABSTRACT

The genus of flavivirus includes many mosquito-borne human pathogens, such as Zika (ZIKV) and the four serotypes of dengue (DENV1-4) viruses, that affect billions of people as evidenced by epidemics and endemicity in many countries and regions in the world. Among the 10 viral proteins encoded by the viral genome, the nonstructural protein 1 (NS1) is the only secreted protein and has been used as a diagnostic biomarker. NS1 has also been an attractive target for its biotherapeutic potential as a vaccine antigen. This review focuses on the recent advances in the structural landscape of the secreted NS1 (sNS1) and its complex with monoclonal antibodies (mAbs). NS1 forms an obligatory dimer, and upon secretion, it has been reported to be hexametric (trimeric dimers) that could dissociate and bind to the epithelial cell membrane. However, high-resolution structural information has been missing about the high-order oligomeric states of sNS1. Several cryoEM studies have since shown that DENV and ZIKV recombinant sNS1 (rsNS1) are in dynamic equilibrium of dimer-tetramer-hexamer states, with tetramer being the predominant form. It was recently revealed that infection-derived sNS1 (isNS1) forms a complex of the NS1 dimer partially embedded in a High-Density Lipoprotein (HDL) particle. Structures of NS1 in complexes with mAbs have also been reported which shed light on their protective roles during infection. The biological significance of the diversity of NS1 oligomeric states remains to be further studied, to inform future research on flaviviral pathogenesis and the development of therapeutics and vaccines. Given the polymorphism of flavivirus NS1 across sample types with variations in antigenicity, we propose a nomenclature to accurately define NS1 based on the localization and origin.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Flavivirus , Viral Nonstructural Proteins , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Humans , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Viral/immunology , Flavivirus/immunology , Flavivirus/chemistry , Flavivirus/genetics , Animals , Zika Virus/immunology , Zika Virus/genetics , Zika Virus/chemistry , Dengue Virus/immunology , Dengue Virus/genetics , Dengue Virus/chemistry , Protein Multimerization , Protein Conformation
4.
Cell Rep ; 43(6): 114298, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38819991

ABSTRACT

Flaviviruses such as dengue virus (DENV), Zika virus (ZIKV), and yellow fever virus (YFV) are spread by mosquitoes and cause human disease and mortality in tropical areas. In contrast, Powassan virus (POWV), which causes severe neurologic illness, is a flavivirus transmitted by ticks in temperate regions of the Northern hemisphere. We find serologic neutralizing activity against POWV in individuals living in Mexico and Brazil. Monoclonal antibodies P002 and P003, which were derived from a resident of Mexico (where POWV is not reported), neutralize POWV lineage I by recognizing an epitope on the virus envelope domain III (EDIII) that is shared with a broad range of tick- and mosquito-borne flaviviruses. Our findings raise the possibility that POWV, or a flavivirus closely related to it, infects humans in the tropics.


Subject(s)
Antibodies, Neutralizing , Humans , Brazil , Antibodies, Neutralizing/immunology , Mexico , Antibodies, Viral/immunology , Animals , Encephalitis Viruses, Tick-Borne/immunology , Flavivirus/immunology , Epitopes/immunology , Antibodies, Monoclonal/immunology , Ticks/virology , Ticks/immunology , Female , Male
5.
Trends Microbiol ; 32(7): 678-696, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38135616

ABSTRACT

In recent decades, the presence of flaviviruses of concern for human health in Europe has drastically increased,exacerbated by the effects of climate change - which has allowed the vectors of these viruses to expand into new territories. Co-circulation of West Nile virus (WNV), Usutu virus (USUV), and tick-borne encephalitis virus (TBEV) represents a threat to the European continent, and this is further complicated by the difficulty of obtaining an early and discriminating diagnosis of infection. Moreover, the possibility of introducing non-endemic pathogens, such as Japanese encephalitis virus (JEV), further complicates accurate diagnosis. Current flavivirus diagnosis is based mainly on RT-PCR and detection of virus-specific antibodies. Yet, both techniques suffer from limitations, and the development of new assays that can provide an early, rapid, low-cost, and discriminating diagnosis of viral infection is warranted. In the pursuit of ideal diagnostic assays, flavivirus non-structural protein 1 (NS1) serves as an excellent target for developing diagnostic assays based on both the antigen itself and the antibodies produced against it. This review describes the potential of such NS1-based diagnostic methods, focusing on the application of flaviviruses that co-circulate in Europe.


Subject(s)
Biomarkers , Flavivirus Infections , Flavivirus , Viral Nonstructural Proteins , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/genetics , Flavivirus/isolation & purification , Flavivirus/genetics , Flavivirus/immunology , Flavivirus Infections/diagnosis , Flavivirus Infections/virology , Humans , Animals , Biomarkers/analysis , Antibodies, Viral/immunology , Europe , Arthropods/virology
6.
Viruses ; 14(2)2022 02 08.
Article in English | MEDLINE | ID: mdl-35215941

ABSTRACT

Flavivirus outbreaks require fast and reliable diagnostics that can be easily adapted to newly emerging and re-emerging flaviviruses. Due to the serological cross-reactivity among flavivirus antibodies, neutralization tests (NT) are considered the gold standard for sero-diagnostics. Here, we first established wild-type single-round infectious virus replicon particles (VRPs) by packaging a yellow fever virus (YFV) replicon expressing Gaussia luciferase (Gluc) with YFV structural proteins in trans using a double subgenomic Sindbis virus (SINV) replicon. The latter expressed the YFV envelope proteins prME via the first SINV subgenomic promoter and the capsid protein via a second subgenomic SINV promoter. VRPs were produced upon co-electroporation of replicon and packaging RNA. Introduction of single restriction enzyme sites in the packaging construct flanking the prME sequence easily allowed to exchange the prME moiety resulting in chimeric VRPs that have the surface proteins of other flaviviruses including dengue virus 1--4, Zika virus, West Nile virus, and tick-borne encephalitis virus. Besides comparing the YF-VRP based NT assay to a YF reporter virus NT assay, we analyzed the neutralization efficiencies of different human anti-flavivirus sera or a monoclonal antibody against all established VRPs. The assays were performed in a 96-well high-throughput format setting with Gluc as readout in comparison to classical plaque reduction NTs indicating that the VRP-based NT assays are suitable for high-throughput analyses of neutralizing flavivirus antibodies.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Flavivirus/immunology , High-Throughput Screening Assays/methods , Cross Reactions , Flavivirus/classification , Flavivirus/genetics , Flavivirus/physiology , Genes, Reporter , Luciferases/genetics , Luciferases/metabolism , Neutralization Tests , Replicon , Sindbis Virus/genetics , Sindbis Virus/immunology , Sindbis Virus/physiology , Virion/genetics , Virion/immunology , Virion/physiology , Yellow fever virus/genetics , Yellow fever virus/immunology , Yellow fever virus/physiology
7.
Viruses ; 14(2)2022 02 21.
Article in English | MEDLINE | ID: mdl-35216028

ABSTRACT

Emerging and re-emerging mosquito-borne viral diseases impose a significant burden on global public health. The most common mosquito-borne viruses causing recent epidemics include flaviviruses in the family Flaviviridae, including Dengue virus (DENV), Zika virus (ZIKV), Japanese encephalitis virus (JEV) and West Nile virus (WNV) and Togaviridae viruses, such as chikungunya virus (CHIKV). Several factors may have contributed to the recent re-emergence and spread of mosquito-borne viral diseases. Among these important causes are the evolution of mosquito-borne viruses and the genetic mutations that make them more adaptive and virulent, leading to widespread epidemics. RNA viruses tend to acquire genetic diversity due to error-prone RNA-dependent RNA polymerases, thus promoting high mutation rates that support adaptation to environmental changes or host immunity. In this review, we discuss recent findings on the adaptive evolution of mosquito-borne viruses and their impact on viral infectivity, pathogenicity, vector fitness, transmissibility, epidemic potential and disease emergence.


Subject(s)
Culicidae/virology , Flavivirus/physiology , Mosquito Vectors/virology , Vector Borne Diseases/virology , Animals , Biological Evolution , Culicidae/immunology , Epidemics , Flavivirus/classification , Flavivirus/immunology , Humans , Virus Replication
9.
Front Immunol ; 13: 829433, 2022.
Article in English | MEDLINE | ID: mdl-35154151

ABSTRACT

The innate immune system is the host's first line of defense against the invasion of pathogens including flavivirus. The programmed cell death controlled by genes plays an irreplaceable role in resisting pathogen invasion and preventing pathogen infection. However, the inflammatory cell death, which can trigger the overflow of a large number of pro-inflammatory cytokines and cell contents, will initiate a severe inflammatory response. In this review, we summarized the current understanding of the innate immune response, inflammatory cell death pathway and cytokine secretion regulation during Dengue virus, West Nile virus, Zika virus, Japanese encephalitis virus and other flavivirus infections. We also discussed the impact of these flavivirus and viral proteins on these biological processes. This not only provides a scientific basis for elucidating the pathogenesis of flavivirus, but also lays the foundation for the development of effective antiviral therapies.


Subject(s)
Flavivirus/immunology , Flavivirus/physiology , Animals , Cell Death , Cytokines/metabolism , Flavivirus/classification , Host-Pathogen Interactions , Humans , Immunity, Innate , Inflammasomes/metabolism
10.
Viruses ; 14(1)2022 01 06.
Article in English | MEDLINE | ID: mdl-35062297

ABSTRACT

Arboviruses have two ecological transmission cycles: sylvatic and urban. For some, the sylvatic cycle has not been thoroughly described in America. To study the role of wildlife in a putative sylvatic cycle, we sampled free-ranging bats and birds in two arbovirus endemic locations and analyzed them using molecular, serological, and histological methods. No current infection was detected, and no significant arbovirus-associated histological changes were observed. Neutralizing antibodies were detected against selected arboviruses. In bats, positivity in 34.95% for DENV-1, 16.26% for DENV-2, 5.69% for DENV-3, 4.87% for DENV-4, 2.43% for WNV, 4.87% for SLEV, 0.81% for YFV, 7.31% for EEEV, and 0.81% for VEEV was found. Antibodies against ZIKV were not detected. In birds, PRNT results were positive against WNV in 0.80%, SLEV in 5.64%, EEEV in 8.4%, and VEEV in 5.63%. An additional retrospective PRNT analysis was performed using bat samples from three additional DENV endemic sites resulting in a 3.27% prevalence for WNV and 1.63% for SLEV. Interestingly, one sample resulted unequivocally WNV positive confirmed by serum titration. These results suggest that free-ranging bats and birds are exposed to not currently reported hyperendemic-human infecting Flavivirus and Alphavirus; however, their role as reservoirs or hosts is still undetermined.


Subject(s)
Alphavirus/immunology , Animals, Wild/immunology , Antibodies, Viral/blood , Birds/immunology , Chiroptera/immunology , Flavivirus/immunology , Seroepidemiologic Studies , Alphavirus Infections/epidemiology , Alphavirus Infections/veterinary , Animals , Antibodies, Neutralizing/blood , Bird Diseases/epidemiology , Costa Rica/epidemiology , Dengue Virus/immunology , Disease Reservoirs , Female , Flavivirus Infections/epidemiology , Flavivirus Infections/veterinary , Humans , Male , Neutralization Tests , Prevalence
11.
Viruses ; 13(12)2021 11 23.
Article in English | MEDLINE | ID: mdl-34960621

ABSTRACT

West Nile virus (WNV) and Usutu virus (USUV) are mosquito-borne flaviviruses that can cause neuroinvasive disease in humans. WNV and USUV circulate in both Africa and Europe and are closely related. Due to antigenic similarity, WNV-specific antibodies and USUV-specific antibodies have the potential to bind heterologous viruses; however, it is unclear whether this interaction may offer protection against infection. To investigate how prior WNV exposure would influence USUV infection, we used an attenuated WNV vaccine that contains the surface proteins of WNV in the backbone of a dengue virus 2 vaccine strain and protects against WNV disease. We hypothesized that vaccination with this attenuated WNV vaccine would protect against USUV infection. Neutralizing responses against WNV and USUV were measured in vitro using sera following vaccination. Sera from vaccinated CD-1 and Ifnar1-/- mice cross-neutralized with WNV and USUV. All mice were then subsequently challenged with an African or European USUV strain. In CD-1 mice, there was no difference in USUV titers between vaccinated and mock-vaccinated mice. However, in the Ifnar1-/- model, vaccinated mice had significantly higher survival rates and significantly lower USUV viremia compared to mock-vaccinated mice. Our results indicate that exposure to an attenuated form of WNV protects against severe USUV disease in mice and elicits a neutralizing response to both WNV and USUV. Future studies will investigate the immune mechanisms responsible for the protection against USUV infection induced by WNV vaccination, providing critical insight that will be essential for USUV and WNV vaccine development.


Subject(s)
Flavivirus Infections/prevention & control , Flavivirus/immunology , West Nile Virus Vaccines/administration & dosage , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Female , Male , Mice , Mice, Knockout , Vaccination
12.
Viruses ; 13(12)2021 11 30.
Article in English | MEDLINE | ID: mdl-34960673

ABSTRACT

The surveillance for West Nile virus (WNV) in Catalonia (northeastern Spain) has consistently detected flaviviruses not identified as WNV. With the aim of characterizing the flaviviruses circulating in Catalonia, serum samples from birds and horses collected between 2010 and 2019 and positive by panflavivirus competition ELISA (cELISA) were analyzed by microneutralization test (MNT) against different flaviviruses. A third of the samples tested were inconclusive by MNT, highlighting the limitations of current diagnostic techniques. Our results evidenced the widespread circulation of flaviviruses, in particular WNV, but also Usutu virus (USUV), and suggest that chicken and horses could serve as sentinels for both viruses. In several regions, WNV and USUV overlapped, but no significant geographical aggregation was observed. Bagaza virus (BAGV) was not detected in birds, while positivity to tick-borne encephalitis virus (TBEV) was sporadically detected in horses although no endemic foci were observed. So far, no human infections by WNV, USUV, or TBEV have been reported in Catalonia. However, these zoonotic flaviviruses need to be kept under surveillance, ideally within a One Health framework.


Subject(s)
Bird Diseases/epidemiology , Flavivirus Infections/veterinary , Flavivirus/physiology , Horse Diseases/epidemiology , Animals , Antibodies, Viral/blood , Bird Diseases/blood , Bird Diseases/virology , Birds , Enzyme-Linked Immunosorbent Assay/veterinary , Flavivirus/genetics , Flavivirus/immunology , Flavivirus/isolation & purification , Flavivirus Infections/blood , Flavivirus Infections/epidemiology , Flavivirus Infections/virology , Horse Diseases/blood , Horse Diseases/virology , Horses , Seroepidemiologic Studies , Spain/epidemiology
13.
Vet Microbiol ; 263: 109281, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34785476

ABSTRACT

Melanoma differentiation associated factor 5 (MDA5), which belongs to the retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) family, has been proved to be a key pattern recognition receptor of innate antiviral signaling in duck, which plays an important role in anti-Tembusu virus (TMUV) infection. However, laboratory of genetics and physiology 2 (LGP2), the third member of RLRs family, the regulatory function on antiviral innate immunity of MDA5 is currently unclear. In this study, we investigated the subcellular localization of duck LGP2 (duLGP2) and confirmed that it is an important regulator of the duMDA5-mediated host innate antiviral immune response. The present experimental data demonstrate that the overexpression of duLGP2 inhibits duMDA5 downstream transcriptional factor (IRF-7, IFN-ß, and NF-κB) promoter activity, and duMDA5-mediated type I IFNs and ISGs expression were significantly suppressed by duLGP2 regardless of viral infection in vitro. The inhibition of duLGP2 on the antiviral activity of duMDA5 ultimately leads to an increase in viral replication. However, the overexpression of duLGP2 promotes expression of mitochondrial antiviral-signaling protein (MAVS) and duMDA5-mediated proinflammatory cytokines. This study provides a new rationale support for the duLGP2 regulates duMDA5-mediated anti-viral immune signaling pathway theory in duck.


Subject(s)
Ducks , Flavivirus Infections , Immunity, Innate , Interferon-Induced Helicase, IFIH1 , RNA Helicases , Animals , Antiviral Agents , Flavivirus/immunology , Flavivirus Infections/immunology , Flavivirus Infections/veterinary , Immunity, Innate/genetics , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/immunology , RNA Helicases/metabolism
14.
Viruses ; 13(11)2021 10 20.
Article in English | MEDLINE | ID: mdl-34834923

ABSTRACT

Mosquito-borne viruses of the Flavivirus genus (Flaviviridae family) pose an ongoing threat to global public health. For example, dengue, Japanese encephalitis, West Nile, yellow fever, and Zika viruses are transmitted by infected mosquitoes and cause severe and fatal diseases in humans. The means by which mosquito-borne flaviviruses establish persistent infection in mosquitoes and cause disease in humans are complex and depend upon a myriad of virus-host interactions, such as those of the innate immune system, which are the main focus of our review. This review also covers the different strategies utilized by mosquito-borne flaviviruses to antagonize the innate immune response in humans and mosquitoes. Given the lack of antiviral therapeutics for mosquito-borne flaviviruses, improving our understanding of these virus-immune interactions could lead to new antiviral therapies and strategies for developing refractory vectors incapable of transmitting these viruses, and can also provide insights into determinants of viral tropism that influence virus emergence into new species.


Subject(s)
Culicidae/immunology , Flavivirus Infections/immunology , Flavivirus Infections/veterinary , Flavivirus/immunology , Persistent Infection/immunology , Persistent Infection/veterinary , Animals , Culicidae/physiology , Culicidae/virology , Flavivirus/genetics , Flavivirus/physiology , Flavivirus Infections/transmission , Flavivirus Infections/virology , Humans , Immunity, Innate , Mosquito Vectors/immunology , Mosquito Vectors/physiology , Mosquito Vectors/virology , Persistent Infection/virology
15.
Vet Microbiol ; 263: 109262, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34715462

ABSTRACT

West Nile virus (WNV) and Usutu virus (USUV) are the two most widespread mosquito-borne flaviviruses in Europe causing severe neuroinvasive disease in humans. Here, following standardization of the murine model with wild type (wt) viruses, we engineered WNV and USUV genome by reverse genetics. A recombinant virus carrying the 5' UTR of WNV within the USUV genome backbone (r-USUV5'-UTR WNV) was rescued; when administered to mice this virus did not cause signs or disease as wt USUV suggesting that 5' UTR of a marked neurotropic parental WNV was not per se a virulence factor. Interestingly, a chimeric virus carrying the envelope (E) protein of USUV in the WNV genome backbone (r-WNVE-USUV) showed an attenuated profile in mice compared to wt WNV but significantly more virulent than wt USUV. Moreover, except when tested against serum samples originating from a live WNV infection, r-WNVE-USUV showed an identical antigenic profile to wt USUV confirming that E is also the major immunodominant protein of USUV.


Subject(s)
Flavivirus , West Nile Fever , West Nile virus , 5' Untranslated Regions , Animals , Flavivirus/genetics , Flavivirus/immunology , Genome, Viral , Mice , Virulence , West Nile Fever/prevention & control , West Nile Fever/veterinary , West Nile Fever/virology , West Nile virus/genetics , West Nile virus/pathogenicity
16.
Viruses ; 13(9)2021 09 19.
Article in English | MEDLINE | ID: mdl-34578454

ABSTRACT

The emergence of West Nile virus (WNV) and Usutu virus (USUV) in addition to the autochthonous tick-borne encephalitis virus (TBEV) in Europe causes rising concern for public and animal health. The first equine case of West Nile neuroinvasive disease in Austria was diagnosed in 2016. As a consequence, a cross-sectional seroprevalence study was conducted in 2017, including 348 equids from eastern Austria. Serum samples reactive by ELISA for either flavivirus immunoglobulin G or M were further analyzed with the plaque reduction neutralization test (PRNT-80) to identify the specific etiologic agent. Neutralizing antibody prevalences excluding vaccinated equids were found to be 5.3% for WNV, 15.5% for TBEV, 0% for USUV, and 1.2% for WNV from autochthonous origin. Additionally, reverse transcription quantitative polymerase chain reaction (RT-qPCR) was performed to detect WNV nucleic acid in horse sera and was found to be negative in all cases. Risk factor analysis did not identify any factors significantly associated with seropositivity.


Subject(s)
Antibodies, Viral/blood , Endemic Diseases/veterinary , Equidae/virology , Flavivirus Infections/veterinary , Flavivirus/immunology , Horse Diseases/epidemiology , Animals , Austria/epidemiology , Cross-Sectional Studies , Encephalitis Viruses, Tick-Borne/immunology , Encephalitis, Tick-Borne/epidemiology , Encephalitis, Tick-Borne/veterinary , Female , Flavivirus Infections/epidemiology , Horses , Male , Risk Factors , Seroepidemiologic Studies , West Nile Fever/epidemiology , West Nile Fever/veterinary , West Nile virus/immunology
17.
Front Immunol ; 12: 711517, 2021.
Article in English | MEDLINE | ID: mdl-34335626

ABSTRACT

Duck Tembusu virus (DTMUV) is an emerging pathogenic flavivirus that has caused a substantial drop in egg production and severe neurological disorders in domestic waterfowl. Several studies have revealed that viral proteins encoded by DTMUV antagonize host IFN-mediated antiviral responses to facilitate virus replication. However, the role of host gene expression regulated by DTMUV in innate immune evasion remains largely unknown. Here, we utilized a stable isotope labeling with amino acids in cell culture (SILAC)-based proteomics analysis of DTMUV-infected duck embryo fibroblasts (DEFs) to comprehensively investigate host proteins involved in DTMUV replication and innate immune response. A total of 250 differentially expressed proteins were identified from 2697 quantified cellular proteins, among which duck interferon-induced protein 35 (duIFI35) was dramatically up-regulated due to DTMUV infection in DEFs. Next, we demonstrated that duIFI35 expression promoted DTMUV replication and impaired Sendai virus-induced IFN-ß production. Moreover, duIFI35 was able to impede duck RIG-I (duRIG-I)-induced IFN-ß promoter activity, rather than IFN-ß transcription mediated by MDA5, MAVS, TBK1, IKKϵ, and IRF7. Importantly, we found that because of the specific interaction with duIFI35, the capacity of duRIG-I to recognize double-stranded RNA was significantly impaired, resulting in the decline of duRIG-I-induced IFN-ß production. Taken together, our data revealed that duIFI35 expression stimulated by DTMUV infection disrupted duRIG-I-mediated host antiviral response, elucidating a distinct function of duIFI35 from human IFI35, by which DTMUV escapes host innate immune response, and providing information for the design of antiviral drug.


Subject(s)
Ducks/virology , Flavivirus Infections/veterinary , Flavivirus/physiology , Gene Expression Regulation, Viral , Immune Evasion , Intracellular Signaling Peptides and Proteins/physiology , Poultry Diseases/virology , Animals , Cell Line , Ducks/embryology , Fibroblasts/metabolism , Fibroblasts/virology , Flavivirus/immunology , Flavivirus Infections/immunology , Flavivirus Infections/metabolism , Flavivirus Infections/virology , Genes, Reporter , HEK293 Cells , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Interferon-beta/biosynthesis , Interferon-beta/genetics , Poly I-C/metabolism , Poultry Diseases/immunology , Poultry Diseases/metabolism , Proteomics/methods , RNA Interference , RNA, Double-Stranded/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Tandem Mass Spectrometry
18.
Viruses ; 13(7)2021 07 20.
Article in English | MEDLINE | ID: mdl-34372622

ABSTRACT

The mosquito-borne flaviviruses USUV and WNV are known to co-circulate in large parts of Europe. Both are a public health concern, and USUV has been the cause of epizootics in both wild and domestic birds, and neurological cases in humans in Europe. Here, we explore the susceptibility of magpies to experimental USUV infection, and how previous exposure to USUV would affect infection with WNV. None of the magpies exposed to USUV showed clinical signs, viremia, or detectable neutralizing antibodies. After challenge with a neurovirulent WNV strain, neither viremia, viral titer of WNV in vascular feathers, nor neutralizing antibody titers of previously USUV-exposed magpies differed significantly with respect to magpies that had not previously been exposed to USUV. However, 75% (6/8) of the USUV-exposed birds survived, while only 22.2% (2/9) of those not previously exposed to USUV survived. WNV antigen labeling by immunohistochemistry in tissues was less evident and more restricted in magpies exposed to USUV prior to challenge with WNV. Our data indicate that previous exposure to USUV partially protects magpies against a lethal challenge with WNV, while it does not prevent viremia and direct transmission, although the mechanism is unclear. These results are relevant for flavivirus ecology and contention.


Subject(s)
Cross Protection/immunology , Disease Transmission, Infectious/veterinary , Flavivirus Infections/veterinary , Flavivirus/immunology , Passeriformes/virology , West Nile Fever/transmission , West Nile Fever/veterinary , West Nile virus/immunology , Animals , Antibodies, Viral/blood , Bird Diseases/virology , Flavivirus Infections/immunology , Spain , West Nile Fever/prevention & control
19.
Front Immunol ; 12: 694959, 2021.
Article in English | MEDLINE | ID: mdl-34421904

ABSTRACT

Avian Tembusu virus (TMUV) is a novel flavivirus causing severe egg drop and fatal encephalitis in avian in Asia. In the present study, we screened the structural and functional requirements of TMUV capsid protein (CP) for viral morphogenesis using reverse genetics methods in combination with replicon packaging assays. TMUV-CP showed dramatic functional and structural flexibility, and even though 44 residues were removed from the N-terminus, it was still capable of packaging replicon RNA; in addition, 33 residues were deleted from the C-terminus (containing nearly the entire α4-helix), and infectious particles were still produced, although α4-α4' is supposedly vital for CP dimerization and nucleocapsid formation. We further analyzed two mutants (ΔC20-43 and ΔC64-96 viruses) with relatively large deletions that still replicated well in BHK-21 cells. Our data indicate that internal deletions within CP impaired viral replication or assembly, resulting in attenuated virus proliferation in cells and attenuated virulence in duck embryos, and these deletion mutations are quite stable in cell culture. An in vivo assay indicated that both ΔC20-43 virus and ΔC64-96 virus were highly attenuated in ducklings but still immunogenic. Single-dose immunization with ΔC20-43 virus or ΔC64-96 virus could protect ducklings from a lethal challenge with good antigen clearance. Together, our data shed light on replication/assembly defective TMUV with internal deletions in CP and provide an effective approach to attenuate viral virulence in live vaccines without changing the antigen composition.


Subject(s)
Capsid Proteins/genetics , Flavivirus Infections/prevention & control , Flavivirus/genetics , Poultry Diseases/prevention & control , Sequence Deletion , Viral Vaccines/genetics , Virus Assembly/genetics , Virus Replication/genetics , Animals , Capsid Proteins/immunology , Cell Line , Cricetinae , Ducks , Flavivirus/growth & development , Flavivirus/immunology , Flavivirus/pathogenicity , Flavivirus Infections/immunology , Flavivirus Infections/virology , Immunogenicity, Vaccine , Poultry Diseases/immunology , Poultry Diseases/virology , Vaccination , Vaccines, Live, Unattenuated/administration & dosage , Vaccines, Live, Unattenuated/genetics , Vaccines, Live, Unattenuated/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Virulence
20.
mSphere ; 6(4): e0033921, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34259560

ABSTRACT

Thailand is a hyperendemic country for flavivirus infections in Southeast Asia. Although the reporting system for flavivirus surveillance in Thailand is well established, syndromic surveillance tends to underestimate the true epidemiological status of flaviviruses due to the majority of infections being asymptomatic. To accurately understand the prevalence of flaviviruses in endemic regions, we performed neutralization tests against multiple flaviviruses using 147 serum samples from healthy donors collected from four distinct regions in Thailand. Single-round infectious particles (SRIP) for six flaviviruses, dengue virus types 1 to 4 (DENV-1 to -4), Japanese encephalitis virus (JEV), and Zika virus (ZIKV), were used as antigens for developing a safe, high-throughput neutralization assay. Titers of neutralizing antibodies (NAbs) against the six flaviviruses revealed that DENV-1 and DENV-2, followed by ZIKV were the predominant circulating flaviviruses in a total of four regions, whereas the prevalence of NAbs against JEV varied among regions. Although the seroprevalence of ZIKV was low relative to that of DENV-1 and DENV-2, the findings strongly suggested that ZIKV has been circulating at a sustained level in Thailand since before 2012. These findings not only demonstrated the application of an SRIP-neutralization test in a serological study, but also elucidated the circulation and distribution trends of different flaviviruses in Thailand. IMPORTANCE Neutralization tests are the most reliable assay for flavivirus antibody detection; however, these assays are not suitable for high-throughput processing due to their time-consuming and labor-intensive nature. In this study, we developed single-round infectious particles (SRIPs) with a luciferase gene for dengue virus types 1 to 4, Japanese encephalitis virus, and Zika virus for use in a safe, high-throughput neutralization assay. We performed neutralization tests against multiple flaviviruses using 147 serum samples that were collected from healthy donors residing in four distinct regions of Thailand in 2011 to 2012. The assay was useful for surveys of flavivirus seroprevalence. The data revealed that dengue virus type 1 (DENV-1) and DENV-2 were the predominant circulating flaviviruses in Thailand and that Zika virus has been circulating at a sustained level in Thailand since before 2012.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Flavivirus Infections/epidemiology , Flavivirus Infections/immunology , Flavivirus/immunology , Zika Virus Infection/epidemiology , Zika Virus/immunology , Adolescent , Adult , Child , Cross Reactions/immunology , Dengue Virus/classification , Dengue Virus/immunology , Female , Flavivirus/classification , High-Throughput Screening Assays/methods , Humans , Male , Middle Aged , Neutralization Tests/methods , Seroepidemiologic Studies , Thailand/epidemiology , Young Adult , Zika Virus Infection/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...