Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 991
Filter
1.
PLoS Negl Trop Dis ; 18(7): e0012172, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38985837

ABSTRACT

Usutu virus (USUV) is an emerging flavivirus that is maintained in an enzootic cycle with mosquitoes as vectors and birds as amplifying hosts. In Europe, the virus has caused mass mortality of wild birds, mainly among Common Blackbird (Turdus merula) populations. While mosquitoes are the primary vectors for USUV, Common Blackbirds and other avian species are exposed to other arthropod ectoparasites, such as ticks. It is unknown, however, if ticks can maintain and transmit USUV. We addressed this question using in vitro and in vivo experiments and field collected data. USUV replicated in IRE/CTVM19 Ixodes ricinus tick cells and in injected ticks. Moreover, I. ricinus nymphs acquired the virus via artificial membrane blood-feeding and maintained the virus for at least 70 days. Transstadial transmission of USUV from nymphs to adults was confirmed in 4.9% of the ticks. USUV disseminated from the midgut to the haemocoel, and was transmitted via the saliva of the tick during artificial membrane blood-feeding. We further explored the role of ticks by monitoring USUV in questing ticks and in ticks feeding on wild birds in the Netherlands between 2016 and 2019. In total, 622 wild birds and the Ixodes ticks they carried were tested for USUV RNA. Of these birds, 48 (7.7%) carried USUV-positive ticks. The presence of negative-sense USUV RNA in ticks, as confirmed via small RNA-sequencing, showed active virus replication. In contrast, we did not detect USUV in 15,381 questing ticks collected in 2017 and 2019. We conclude that I. ricinus can be infected with USUV and can transstadially and horizontally transmit USUV. However, in comparison to mosquito-borne transmission, the role of I. ricinus ticks in the epidemiology of USUV is expected to be minor.


Subject(s)
Bird Diseases , Flavivirus Infections , Flavivirus , Ixodes , Nymph , Animals , Ixodes/virology , Ixodes/physiology , Flavivirus/physiology , Flavivirus/genetics , Flavivirus Infections/transmission , Flavivirus Infections/veterinary , Flavivirus Infections/virology , Nymph/virology , Bird Diseases/virology , Bird Diseases/transmission , Birds/virology , Arachnid Vectors/virology , Arachnid Vectors/physiology , Netherlands , Female
2.
Parasit Vectors ; 17(1): 285, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956650

ABSTRACT

Usutu virus is an emerging pathogen transmitted by mosquitoes. Culex modestus mosquitoes are widespread in Europe, but their role in disease transmission is poorly understood. Recent data from a single infectious mosquito suggested that Culex modestus could be an unrecognized vector for Usutu virus. In this study, our aim was to corroborate this finding using a larger sample size. We collected immature Culex modestus from a reedbed pond in Flemish Brabant, Belgium, and reared them in the laboratory until the third generation. Adult females were then experimentally infected with Usutu virus in a blood meal and incubated at 25 °C for 14 days. The presence of Usutu virus in the saliva, head and body of each female was determined by plaque assay and quantitative real-time reverse-transcription polymerase chain reaction (qRT-PCR). The transmission efficiency was 54% (n = 15/28), confirming that Belgian Culex modestus can experimentally transmit Usutu virus.


Subject(s)
Culex , Flavivirus Infections , Flavivirus , Mosquito Vectors , Animals , Culex/virology , Female , Mosquito Vectors/virology , Flavivirus/genetics , Flavivirus/physiology , Belgium , Flavivirus Infections/transmission , Flavivirus Infections/virology , Saliva/virology
3.
Nat Commun ; 15(1): 5179, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898037

ABSTRACT

Viral genetic diversity presents significant challenges in developing antivirals with broad-spectrum activity and high barriers to resistance. Here we report development of proteolysis targeting chimeras (PROTACs) targeting the dengue virus envelope (E) protein through coupling of known E fusion inhibitors to ligands of the CRL4CRBN E3 ubiquitin ligase. The resulting small molecules block viral entry through inhibition of E-mediated membrane fusion and interfere with viral particle production by depleting intracellular E in infected Huh 7.5 cells. This activity is retained in the presence of point mutations previously shown to confer partial resistance to the parental inhibitors due to decreased inhibitor-binding. The E PROTACs also exhibit broadened spectrum of activity compared to the parental E inhibitors against a panel of mosquito-borne flaviviruses. These findings encourage further exploration of targeted protein degradation as a differentiated and potentially advantageous modality for development of broad-spectrum direct-acting antivirals.


Subject(s)
Antiviral Agents , Dengue Virus , Flavivirus , Proteolysis , Virus Internalization , Humans , Proteolysis/drug effects , Animals , Antiviral Agents/pharmacology , Flavivirus/drug effects , Flavivirus/genetics , Flavivirus/metabolism , Virus Internalization/drug effects , Dengue Virus/drug effects , Dengue Virus/physiology , Dengue Virus/genetics , Culicidae/virology , Ubiquitin-Protein Ligases/metabolism , Viral Envelope Proteins/metabolism , Cell Line
4.
Viruses ; 16(6)2024 May 30.
Article in English | MEDLINE | ID: mdl-38932175

ABSTRACT

Newly emerging viruses, primarily zoonotic or vector-borne, pose a persistent threat to public health and have led to outbreaks of global concern [...].


Subject(s)
Alphavirus Infections , Alphavirus , Flavivirus Infections , Flavivirus , Alphavirus/physiology , Alphavirus/genetics , Humans , Animals , Flavivirus/genetics , Flavivirus/physiology , Alphavirus Infections/virology , Alphavirus Infections/epidemiology , Flavivirus Infections/virology , Flavivirus Infections/epidemiology
5.
An Acad Bras Cienc ; 96(2): e20230452, 2024.
Article in English | MEDLINE | ID: mdl-38922274

ABSTRACT

The genus Flavivirus comprises approximately 80 different viruses. Phylogenetic relationships among its members indicate a clear ecological separation between those viruses transmitted by mosquitoes, ticks, with no known vector, and insect-specific Flaviviruses. The diversity and phylogenetic relationships among insect-specific flaviviruses circulating in the central and northern regions of Argentina were studied by performing molecular detection and characterization of the NS5 protein gene in mosquitoes collected in Córdoba, Chaco and Tucumán provinces. Overall, 68 out of 1776 pools were positive. CxFV, KRV and CFAV circulate in the 3 studied provinces. Several mosquito species (Aedes aegypti, Culex bidens, Cx. dolosus, Cx. interfor, Cx. quinquefasciatus, Cx. saltanensis, Haemagogus spegazzini) were found infected. A wide circulation of CxFV was observed in the central-northern region of Argentina. CxFV strains detected in our study clustered with strains circulating in Santa Fe and Buenos Aires provinces (Argentina), and other countries such as Indonesia, Mexico, Uganda and Taiwan. The presence of these viruses in mosquitoes could play an important role from the public health perspective, because it has been shown that previous CxFV infection can increase or block the infection of the mosquito by other pathogenic flaviviruses.


Subject(s)
Culicidae , Flavivirus , Mosquito Vectors , Phylogeny , Animals , Argentina , Flavivirus/classification , Flavivirus/genetics , Flavivirus/isolation & purification , Culicidae/virology , Culicidae/classification , Mosquito Vectors/virology , Mosquito Vectors/classification
6.
PLoS Negl Trop Dis ; 18(6): e0012295, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38935783

ABSTRACT

Usutu virus (USUV) is a zoonotic arbovirus infecting mainly wild birds. It is transmitted by ornithophilic mosquitoes, mainly of the genus Culex from birds to birds and to several vertebrate dead-end hosts. Several USUV lineages, differing in their virulence have emerged in the last decades and now co-circulate in Europe, impacting human populations. However, their relative transmission and effects on their mosquito vectors is still not known. We thus compared the vector competence and survival of Culex pipiens mosquitoes experimentally infected with two distinct USUV lineages, EU2 and EU3, that are known to differ in their virulence and replication in vertebrate hosts. Infection rate was variable among blood feeding assays but variations between EU2 and EU3 lineages were consistent suggesting that Culex pipiens was equally susceptible to infection by both lineages. However, EU3 viral load increased with viral titer in the blood meal while EU2 viral load was high at all titers which suggest a greater replication of EU2 than EU3 in mosquito. While their relative transmission efficiencies are similar, at least at low blood meal titer, positive correlation between transmission and blood meal titer was observed for EU3 only. Contrary to published results in vertebrates, EU3 induced a higher mortality to mosquitoes (i.e. virulence) than EU2 whatever the blood meal titer. Therefore, we found evidence of lineage-specific differences in vectorial capacity and virulence to both the vector and vertebrate host which lead to balanced propagation of both viral lineages. These results highlight the need to decipher the interactions between vectors, vertebrate hosts, and the diversity of arbovirus lineages to fully understand transmission dynamics.


Subject(s)
Culex , Flavivirus Infections , Flavivirus , Mosquito Vectors , Animals , Culex/virology , Mosquito Vectors/virology , Virulence , Flavivirus/pathogenicity , Flavivirus/genetics , Flavivirus/physiology , Flavivirus Infections/transmission , Flavivirus Infections/virology , Viral Load , Female , Humans , Virus Replication
7.
Nat Commun ; 15(1): 5426, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926343

ABSTRACT

Zika and dengue virus nonstructural protein 5 antagonism of STAT2, a critical interferon signaling transcription factor, to suppress the host interferon response is required for viremia and pathogenesis in a vertebrate host. This affects viral species tropism, as mouse STAT2 resistance renders only immunocompromised or humanized STAT2 mice infectable. Here, we explore how STAT2 evolution impacts antagonism. By measuring the susceptibility of 38 diverse STAT2 proteins, we demonstrate that resistance arose numerous times in mammalian evolution. In four species, resistance requires distinct sets of multiple amino acid changes that often individually disrupt STAT2 signaling. This reflects an evolutionary ridge where progressive resistance is balanced by the need to maintain STAT2 function. Furthermore, resistance may come with a fitness cost, as resistance that arose early in lemur evolution was subsequently lost in some lemur lineages. These findings underscore that while it is possible to evolve resistance to antagonism, complex evolutionary trajectories are required to avoid detrimental host fitness consequences.


Subject(s)
Evolution, Molecular , STAT2 Transcription Factor , Viral Nonstructural Proteins , STAT2 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , Animals , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Humans , Mice , Dengue Virus/genetics , Dengue Virus/physiology , Zika Virus/genetics , Flavivirus/genetics , Flavivirus/physiology , Phylogeny , Host-Pathogen Interactions/genetics
8.
RNA Biol ; 21(1): 14-30, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38797925

ABSTRACT

As positive-sense RNA viruses, the genomes of flaviviruses serve as the template for all stages of the viral life cycle, including translation, replication, and infectious particle production. Yet, they encode just 10 proteins, suggesting that the structure and dynamics of the viral RNA itself helps shepherd the viral genome through these stages. Herein, we highlight advances in our understanding of flavivirus RNA structural elements through the lens of their impact on the viral life cycle. We highlight how RNA structures impact translation, the switch from translation to replication, negative- and positive-strand RNA synthesis, and virion assembly. Consequently, we describe three major themes regarding the roles of RNA structure in flavivirus infections: 1) providing a layer of specificity; 2) increasing the functional capacity; and 3) providing a mechanism to support genome compaction. While the interactions described herein are specific to flaviviruses, these themes appear to extend more broadly across RNA viruses.


Subject(s)
Flavivirus , Genome, Viral , Nucleic Acid Conformation , RNA, Viral , Virus Replication , Flavivirus/genetics , Flavivirus/physiology , RNA, Viral/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , Humans , Flavivirus Infections/virology , Virus Assembly , Animals , Protein Biosynthesis
9.
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
11.
J Gen Virol ; 105(5)2024 May.
Article in English | MEDLINE | ID: mdl-38809251

ABSTRACT

Tick-borne orthoflaviviruses (TBFs) are classified into three conventional groups based on genetics and ecology: mammalian, seabird and probable-TBF group. Recently, a fourth basal group has been identified in Rhipicephalus ticks from Africa: Mpulungu flavivirus (MPFV) in Zambia and Ngoye virus (NGOV) in Senegal. Despite attempts, isolating these viruses in vertebrate and invertebrate cell lines or intracerebral injection of newborn mice with virus-containing homogenates has remained unsuccessful. In this study, we report the discovery of Xinyang flavivirus (XiFV) in Haemaphysalis flava ticks from Xìnyáng, Henan Province, China. Phylogenetic analysis shows that XiFV was most closely related to MPFV and NGOV, marking the first identification of this tick orthoflavivirus group in Asia. We developed a reverse transcriptase quantitative PCR assay to screen wild-collected ticks and egg clutches, with absolute infection rates of 20.75 % in adult females and 15.19 % in egg clutches, suggesting that XiFV could be potentially spread through transovarial transmission. To examine potential host range, dinucleotide composition analyses revealed that XiFV, MPFV and NGOV share a closer composition to classical insect-specific orthoflaviviruses than to vertebrate-infecting TBFs, suggesting that XiFV could be a tick-only orthoflavivirus. Additionally, both XiFV and MPFV lack a furin cleavage site in the prM protein, unlike other TBFs, suggesting these viruses might exist towards a biased immature particle state. To examine this, chimeric Binjari virus with XIFV-prME (bXiFV) was generated, purified and analysed by SDS-PAGE and negative-stain transmission electron microscopy, suggesting prototypical orthoflavivirus size (~50 nm) and bias towards uncleaved prM. In silico structural analyses of the 3'-untranslated regions show that XiFV forms up to five pseudo-knot-containing stem-loops and a prototypical orthoflavivirus dumbbell element, suggesting the potential for multiple exoribonuclease-resistant RNA structures.


Subject(s)
Flavivirus , Ixodidae , Phylogeny , Animals , Flavivirus/genetics , Flavivirus/classification , Flavivirus/isolation & purification , China , Ixodidae/virology , Female
12.
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
13.
Virology ; 595: 110084, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38692132

ABSTRACT

Duck Tembusu virus (DTMUV) belongs to the Flaviviridae family and mainly infects ducks. The genome of DTMUV is translated into a polyprotein, which is further cleaved into several protein by viral NS2B3 protease and host proteases. Crucially, the cleavage of the NS2A/2B precursor during this process is essential for the formation of replication complexes and viral packaging. Previous research has demonstrated that alanine mutations in NS2A/2B (P1P1' (AA)) result in an attenuated strain (rDTMUV-NS2A/2B-P1P1' (AA)) by disrupting NS2A/2B cleavage. In this study, we investigate the effects of the P1P1' (AA) mutation on the viral life cycle and explore compensatory mutations in rDTMUV-NS2A/2B-P1P1' (AA). Infected ducklings exhibit similar body weight gain and viral tissue loads to DTMUV-WT. Compensatory mutations E-M349E and P1(T) emerge, restoring proliferation levels to those of rDTMUV-WT. Specifically, E-M349E enhances viral packaging, while P1(T) reinstates NS2A/2B proteolysis in vitro. Thus, our findings reveal novel compensatory sites capable of restoring the attenuated DTMUV during polyprotein cleavage and packaging.


Subject(s)
Ducks , Flavivirus , Poultry Diseases , Viral Nonstructural Proteins , Virus Assembly , Virus Replication , Animals , Ducks/virology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Flavivirus/genetics , Flavivirus/physiology , Poultry Diseases/virology , Flavivirus Infections/virology , Mutation
14.
J Biosci ; 492024.
Article in English | MEDLINE | ID: mdl-38783793

ABSTRACT

A high level of disorder in many viral proteins is a direct consequence of their small genomes, which makes interaction with multiple binding partners a necessity for infection and pathogenicity. A segment of the flaviviral capsid protein (C), also known as the molecular recognition feature (MoRF), undergoes a disorder-toorder transition upon binding to several protein partners. To understand their role in pathogenesis, MoRFs were identified and their occurrence across different flaviviral capsids were studied. Despite lack of sequence similarities, docking studies of Cs with the host proteins indicate conserved interactions involving MoRFs across members of phylogenetic subclades. Additionally, it was observed from the protein-protein networks that some MoRFs preferentially bind proteins that are involved in specialized functions such as ribosome biogenesis. The findings point to the importance of MoRFs in the flaviviral life cycle, with important consequences for disease progression and suppression of the host immune system. Potentially, they might have impacted the way flaviviruses evolved to infect varied hosts using multiple vectors.


Subject(s)
Capsid Proteins , Flavivirus , Capsid Proteins/genetics , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Flavivirus/pathogenicity , Flavivirus/genetics , Flavivirus/physiology , Flavivirus/metabolism , Phylogeny , Humans , Protein Binding , Capsid/metabolism , Capsid/chemistry , Flavivirus Infections/virology , Flavivirus Infections/metabolism , Molecular Docking Simulation , Amino Acid Sequence
15.
Poult Sci ; 103(6): 103727, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38652953

ABSTRACT

Duck Tembusu virus (DTMUV) belongs to the Flaviviridae family and mainly infects ducks. Duck Tembusu virus genome encodes one polyprotein that undergoes cleavage to produce 10 proteins. Among these, NS4B, the largest transmembrane protein, plays a crucial role in the viral life cycle. In this study, we investigated the localization of NS4B and found that it is located in the endoplasmic reticulum, where it co-localizes with DTMUV dsRNA. Subsequently, we confirmed 5 different transmembrane domains of NS4B and discovered that only its transmembrane domain 3 (TMD3) can traverse ER membrane. Then mutations were introduced in the conserved amino acids of NS4B TMD3 of DTMUV replicon and infectious clone. The results showed that V111G, V117G, and I118G mutations enhanced viral RNA replication, while Q104A, T106A, A113L, M116A, H120A, Y121A, and A122G mutations reduced viral replication. Recombinant viruses with these mutations were rescued and studied in BHK21 cells. The findings demonstrated that A113L and H120A mutations led to higher viral titers than the wild-type strain, while Q104A, T106A, V111G, V117G, and Y121A mutations attenuated viral proliferation. Additionally, H120A, M116A, and A122G mutations enhanced viral proliferation. Furthermore, Q104A, T106A, V111G, M116A, V117G, Y121A, and A122G mutants showed reduced viral virulence to 10-d duck embryos. Animal experiments further indicated that all mutation viruses resulted in lower genome copy numbers in the spleen compared to the WT group 5 days postinfection. Our data provide insights into the topological model of DTMUV NS4B, highlighting the essential role of NS4B TMD3 in viral replication and proliferation.


Subject(s)
Ducks , Flavivirus , Viral Nonstructural Proteins , Virus Replication , Animals , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Flavivirus/physiology , Flavivirus/genetics , Poultry Diseases/virology , Flavivirus Infections/veterinary , Flavivirus Infections/virology , Mutation
16.
Proc Natl Acad Sci U S A ; 121(16): e2317978121, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38593069

ABSTRACT

Mosquito-borne flaviviruses such as dengue (DENV) and Zika (ZIKV) cause hundreds of millions of infections annually. The single-stranded RNA genome of flaviviruses is translated into a polyprotein, which is cleaved equally into individual functional proteins. While structural proteins are packaged into progeny virions and released, most of the nonstructural proteins remain intracellular and could become cytotoxic if accumulated over time. However, the mechanism by which nonstructural proteins are maintained at the levels optimal for cellular fitness and viral replication remains unknown. Here, we identified that the ubiquitin E3 ligase HRD1 is essential for flaviviruses infections in both mammalian hosts and mosquitoes. HRD1 directly interacts with flavivirus NS4A and ubiquitylates a conserved lysine residue for ER-associated degradation. This mechanism avoids excessive accumulation of NS4A, which otherwise interrupts the expression of processed flavivirus proteins in the ER. Furthermore, a small-molecule inhibitor of HRD1 named LS-102 effectively interrupts DENV2 infection in both mice and Aedes aegypti mosquitoes, and significantly disturbs DENV transmission from the infected hosts to mosquitoes owing to reduced viremia. Taken together, this study demonstrates that flaviviruses have evolved a sophisticated mechanism to exploit the ubiquitination system to balance the homeostasis of viral proteins for their own advantage and provides a potential therapeutic target to interrupt flavivirus infection and transmission.


Subject(s)
Aedes , Flavivirus Infections , Flavivirus , Zika Virus Infection , Zika Virus , Animals , Mice , Flavivirus/genetics , Zika Virus/genetics , Ubiquitin/metabolism , Ligases/metabolism , Viral Proteins/metabolism , Mammals
17.
Science ; 384(6693): eadn9524, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38669573

ABSTRACT

The commensal microbiota of the mosquito gut plays a complex role in determining the vector competence for arboviruses. In this study, we identified a bacterium from the gut of field Aedes albopictus mosquitoes named Rosenbergiella sp. YN46 (Rosenbergiella_YN46) that rendered mosquitoes refractory to infection with dengue and Zika viruses. Inoculation of 1.6 × 103 colony forming units (CFUs) of Rosenbergiella_YN46 into A. albopictus mosquitoes effectively prevents viral infection. Mechanistically, this bacterium secretes glucose dehydrogenase (RyGDH), which acidifies the gut lumen of fed mosquitoes, causing irreversible conformational changes in the flavivirus envelope protein that prevent viral entry into cells. In semifield conditions, Rosenbergiella_YN46 exhibits effective transstadial transmission in field mosquitoes, which blocks transmission of dengue virus by newly emerged adult mosquitoes. The prevalence of Rosenbergiella_YN46 is greater in mosquitoes from low-dengue areas (52.9 to ~91.7%) than in those from dengue-endemic regions (0 to ~6.7%). Rosenbergiella_YN46 may offer an effective and safe lead for flavivirus biocontrol.


Subject(s)
Aedes , Dengue Virus , Mosquito Vectors , Symbiosis , Zika Virus , Animals , Aedes/microbiology , Aedes/virology , Dengue Virus/physiology , Mosquito Vectors/virology , Mosquito Vectors/microbiology , Zika Virus/physiology , Dengue/transmission , Dengue/virology , Dengue/prevention & control , Gastrointestinal Microbiome , Acetobacteraceae/physiology , Female , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Flavivirus/physiology , Flavivirus/genetics , Zika Virus Infection/transmission , Zika Virus Infection/virology
18.
Proc Natl Acad Sci U S A ; 121(19): e2319400121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38687787

ABSTRACT

During their blood-feeding process, ticks are known to transmit various viruses to vertebrates, including humans. Recent viral metagenomic analyses using next-generation sequencing (NGS) have revealed that blood-feeding arthropods like ticks harbor a large diversity of viruses. However, many of these viruses have not been isolated or cultured, and their basic characteristics remain unknown. This study aimed to present the identification of a difficult-to-culture virus in ticks using NGS and to understand its epidemic dynamics using molecular biology techniques. During routine tick-borne virus surveillance in Japan, an unknown flaviviral sequence was detected via virome analysis of host-questing ticks. Similar viral sequences have been detected in the sera of sika deer and wild boars in Japan, and this virus was tentatively named the Saruyama virus (SAYAV). Because SAYAV did not propagate in any cultured cells tested, single-round infectious virus particles (SRIP) were generated based on its structural protein gene sequence utilizing a yellow fever virus-based replicon system to understand its nationwide endemic status. Seroepidemiological studies using SRIP as antigens have demonstrated the presence of neutralizing antibodies against SAYAV in sika deer and wild boar captured at several locations in Japan, suggesting that SAYAV is endemic throughout Japan. Phylogenetic analyses have revealed that SAYAV forms a sister clade with the Orthoflavivirus genus, which includes important mosquito- and tick-borne pathogenic viruses. This shows that SAYAV evolved into a lineage independent of the known orthoflaviviruses. This study demonstrates a unique approach for understanding the epidemiology of uncultured viruses by combining viral metagenomics and pseudoinfectious viral particles.


Subject(s)
Deer , Flavivirus , Metagenomics , Ticks , Animals , Metagenomics/methods , Japan/epidemiology , Deer/virology , Flavivirus/genetics , Flavivirus/isolation & purification , Flavivirus/classification , Ticks/virology , Phylogeny , Virome/genetics , Virion/genetics , Sus scrofa/virology , High-Throughput Nucleotide Sequencing , Humans , Seroepidemiologic Studies , Genome, Viral
19.
Viruses ; 16(4)2024 03 28.
Article in English | MEDLINE | ID: mdl-38675867

ABSTRACT

Extracellular vesicles (EVs) such as exosomes have been shown to play physiological roles in cell-to-cell communication by delivering various proteins and nucleic acids. In addition, several studies revealed that the EVs derived from the cells that are infected with certain viruses could transfer the full-length viral genomes, resulting in EVs-mediated virus propagation. However, the possibility cannot be excluded that the prepared EVs were contaminated with infectious viral particles. In this study, the cells that harbor subgenomic replicon derived from the Japanese encephalitis virus and dengue virus without producing any replication-competent viruses were employed as the EV donor. It was demonstrated that the EVs in the culture supernatants of those cells were able to transfer the replicon genome to other cells of various types. It was also shown that the EVs were incorporated by the recipient cells primarily through macropinocytosis after interaction with CD33 and Tim-1/Tim-4 on HeLa and K562 cells, respectively. Since the methods used in this study are free from contamination with infectious viral particles, it is unequivocally indicated that the flavivirus genome can be transferred by EVs from cell to cell, suggesting that this pathway, in addition to the classical receptor-mediated infection, may play some roles in the viral propagation and pathogenesis.


Subject(s)
Encephalitis Virus, Japanese , Extracellular Vesicles , Genome, Viral , Replicon , Viral Proteins , Extracellular Vesicles/virology , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Humans , Replicon/genetics , Encephalitis Virus, Japanese/genetics , Encephalitis Virus, Japanese/physiology , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Flavivirus/genetics , Flavivirus/physiology , Dengue Virus/genetics , Dengue Virus/physiology , HeLa Cells , K562 Cells , Animals , Cell Line , Subgenomic RNA
20.
Viruses ; 16(4)2024 04 12.
Article in English | MEDLINE | ID: mdl-38675940

ABSTRACT

West Nile Virus (WNV) and Usutu Virus (USUV) are both neurotropic mosquito-borne viruses belonging to the Flaviviridae family. These closely related viruses mainly follow an enzootic cycle involving mosquitoes as vectors and birds as amplifying hosts, but humans and other mammals can also be infected through mosquito bites. WNV was first identified in Uganda in 1937 and has since spread globally, notably in Europe, causing periodic outbreaks associated with severe cases of neuroinvasive diseases such as meningitis and encephalitis. USUV was initially isolated in 1959 in Swaziland and has also spread to Europe, primarily affecting birds and having a limited impact on human health. There has been a recent expansion of these viruses' geographic range in Europe, facilitated by factors such as climate change, leading to increased human exposure. While sharing similar biological traits, ecology, and epidemiology, there are significant distinctions in their pathogenicity and their impact on both human and animal health. While WNV has been more extensively studied and is a significant public health concern in many regions, USUV has recently been gaining attention due to its emergence in Europe and the diversity of its circulating lineages. Understanding the pathophysiology, ecology, and transmission dynamics of these viruses is important to the implementation of effective surveillance and control measures. This perspective provides a brief overview of the current situation of these two viruses in Europe and outlines the significant challenges that need to be addressed in the coming years.


Subject(s)
Birds , Flavivirus Infections , Flavivirus , West Nile Fever , West Nile virus , Europe/epidemiology , West Nile virus/genetics , West Nile virus/physiology , West Nile virus/isolation & purification , Animals , Humans , Flavivirus/classification , Flavivirus/genetics , Flavivirus/pathogenicity , Flavivirus/isolation & purification , Flavivirus/physiology , Flavivirus Infections/epidemiology , Flavivirus Infections/virology , Flavivirus Infections/transmission , Flavivirus Infections/veterinary , West Nile Fever/epidemiology , West Nile Fever/virology , West Nile Fever/transmission , Birds/virology , Culicidae/virology , Mosquito Vectors/virology , Disease Outbreaks
SELECTION OF CITATIONS
SEARCH DETAIL