Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.886
Filtrer
1.
PLoS Pathog ; 20(8): e1012409, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39146232

RÉSUMÉ

Flaviviridae is a family of positive-stranded RNA viruses, including human pathogens, such as Japanese encephalitis virus (JEV), dengue virus (DENV), Zika virus (ZIKV), and West Nile virus (WNV). Nuclear localization of the viral core protein is conserved among Flaviviridae, and this feature may be targeted for developing broad-ranging anti-flavivirus drugs. However, the mechanism of core protein translocation to the nucleus and the importance of nuclear translocation in the viral life cycle remain unknown. We aimed to identify the molecular mechanism underlying core protein nuclear translocation. We identified importin-7 (IPO7), an importin-ß family protein, as a nuclear carrier for Flaviviridae core proteins. Nuclear import assays revealed that core protein was transported into the nucleus via IPO7, whereas IPO7 deletion by CRISPR/Cas9 impaired their nuclear translocation. To understand the importance of core protein nuclear translocation, we evaluated the production of infectious virus or single-round-infectious-particles in wild-type or IPO7-deficient cells; both processes were significantly impaired in IPO7-deficient cells, whereas intracellular infectious virus levels were equivalent in wild-type and IPO7-deficient cells. These results suggest that IPO7-mediated nuclear translocation of core proteins is involved in the release of infectious virus particles of flaviviruses.


Sujet(s)
Transport nucléaire actif , Noyau de la cellule , Flavivirus , Humains , Flavivirus/métabolisme , Flavivirus/physiologie , Animaux , Noyau de la cellule/métabolisme , Noyau de la cellule/virologie , Réplication virale/physiologie , Protéines du core viral/métabolisme , Protéines du core viral/génétique , Caryophérines/métabolisme , Caryophérines/génétique , Infections à flavivirus/métabolisme , Infections à flavivirus/virologie , Chlorocebus aethiops , Cellules HEK293
2.
Parasitol Res ; 123(8): 304, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39162844

RÉSUMÉ

The family Cimicidae comprises ectoparasites feeding exclusively on the blood of endothermic animals. Cimicid swallow bugs specifically target swallow birds (Hirundinidae) and their nestlings in infested nests. Bugs of the genus Oeciacus are commonly found in mud nests of swallows and martins, while they rarely visit the homes of humans. Although-unlike other cimicid species-the house martin bug Oeciacus hirundinis has never been reported as a vector of zoonotic pathogens, its possible role in arbovirus circulation in continental Europe is unclear. Samples of O. hirundinis were therefore collected from abandoned house martin (Delichon urbicum) nests in southern Moravia (Czech Republic) during the 2021/2022 winter season and checked for alpha-, flavi- and bunyaviruses by RT-PCR. Of a total of 96 pools consisting of three adult bugs each, one pool tested positive for Usutu virus (USUV)-RNA. Phylogenetic analysis showed that the virus strain was closely related to Italian and some Central European strains and corresponded to USUV lineage 5. The detection of USUV in O. hirundinis during wintertime in the absence of swallows raises the question for a possible role of this avian ectoparasite in virus overwintering in Europe.


Sujet(s)
Cimicidae , Flavivirus , Phylogenèse , Saisons , Animaux , Cimicidae/virologie , Flavivirus/isolement et purification , Flavivirus/génétique , Flavivirus/classification , République tchèque , ARN viral/génétique , Maladies des oiseaux/parasitologie , Maladies des oiseaux/virologie
3.
Viruses ; 16(8)2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39205236

RÉSUMÉ

Flaviviruses comprise a large number of arthropod-borne viruses, some of which are associated with life-threatening diseases. Flavivirus infections are rising worldwide, mainly due to the proliferation and geographical expansion of their vectors. The main human pathogens are mosquito-borne flaviviruses, including dengue virus, Zika virus, and West Nile virus, but tick-borne flaviviruses are also emerging. As with any viral infection, the body's first line of defense against flavivirus infections is the innate immune defense, of which type I interferon is the armed wing. This cytokine exerts its antiviral activity by triggering the synthesis of hundreds of interferon-induced genes (ISGs), whose products can prevent infection. Among the ISGs that inhibit flavivirus replication, certain tripartite motif (TRIM) proteins have been identified. Although involved in other biological processes, TRIMs constitute a large family of antiviral proteins active on a wide range of viruses. Furthermore, whereas some TRIM proteins directly block viral replication, others are positive regulators of the IFN response. Therefore, viruses have developed strategies to evade or counteract TRIM proteins, and some even hijack certain TRIM proteins to their advantage. In this review, we summarize the current state of knowledge on the interactions between flaviviruses and TRIM proteins, covering both direct and indirect antiviral mechanisms.


Sujet(s)
Infections à flavivirus , Flavivirus , Réplication virale , Humains , Infections à flavivirus/virologie , Infections à flavivirus/immunologie , Flavivirus/physiologie , Flavivirus/immunologie , Animaux , Immunité innée , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Interactions hôte-pathogène/immunologie , Interféron de type I/immunologie , Interféron de type I/métabolisme
4.
Ann Parasitol ; 70(2): 55-71, 2024.
Article de Anglais | MEDLINE | ID: mdl-39097293

RÉSUMÉ

USUV in Europe is detected in vectors (mosquitoes) and has a reservoir in vertebrates. There are known fatal epidemics among birds, especially blackbirds. Currently, USUV also causes rare infections in humans. However, the emergence of clinical cases, including severe neurological symptoms, and the finding of seroprevalence in asymptomatic people (e.g. blood donors, forest workers), indicate that USUV, due to its neurotropism, may become a potential public health problem. Therefore, it is very important to monitor cases infections in humans, migratory and resident birds and other animals that may constitute a reservoir of the virus, but also detection of the virus in mosquitoes (vectors), including alien and invasive species, as well as the impact of climatic factors on the ability to spread the virus in the Europe. There is currently no evidence of virus transmission during transfusion or transplantation, but the potential risk of virus transmission from an asymptomatic blood donor to an mmunocompromised recipient must be considered. Although the occurrence of USUV in European countries is currently not a significant threat, surveillance and screening of blood donors for USUV should be carried out during the period of vector activity and during WNV epidemics, as well as in patients with symptoms of meningitis and encephalitis.


Sujet(s)
Flavivirus , Animaux , Humains , Culicidae/virologie , Infections à flavivirus/virologie , Vecteurs moustiques/virologie
5.
Parasit Vectors ; 17(1): 369, 2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39215365

RÉSUMÉ

BACKGROUND: Mosquito host feeding patterns are an important factor of the species-specific vector capacity determining pathogen transmission routes. Culex pipiens s.s./Cx. torrentium are competent vectors of several arboviruses, such as West Nile virus and Usutu virus. However, studies on host feeding patterns rarely differentiate the morphologically indistinguishable females. METHODS: We analyzed the host feeding attraction of Cx. pipiens and Cx. torrentium in host-choice studies for bird, mouse, and a human lure. In addition, we summarized published and unpublished data on host feeding patterns of field-collected specimens from Germany, Iran, and Moldova from 2012 to 2022, genetically identified as Cx. pipiens biotype pipiens, Cx. pipiens biotype molestus, Cx. pipiens hybrid biotype pipiens × molestus, and Cx. torrentium, and finally put the data in context with similar data found in a systematic literature search. RESULTS: In the host-choice experiments, we did not find a significant attraction to bird, mouse, and human lure for Cx. pipiens pipiens and Cx. torrentium. Hosts of 992 field-collected specimens were identified for Germany, Iran, and Moldova, with the majority determined as Cx. pipiens pipiens, increasing the data available from studies known from the literature by two-thirds. All four Culex pipiens s.s./Cx. torrentium taxa had fed with significant proportions on birds, humans, and nonhuman mammals. Merged with the data from the literature from 23 different studies showing a high prevalence of blood meals from birds, more than 50% of the blood meals of Cx. pipiens s.s. were identified as birds, while up to 39% were human and nonhuman mammalian hosts. Culex torrentium fed half on birds and half on mammals. However, there were considerable geographical differences in the host feeding patterns. CONCLUSIONS: In the light of these results, the clear characterization of the Cx. pipiens s.s./Cx. torrentium taxa as ornithophilic/-phagic or mammalophilic/-phagic needs to be reconsidered. Given their broad host ranges, all four Culex taxa could potentially serve as enzootic and bridge vectors.


Sujet(s)
Oiseaux , Culex , Comportement alimentaire , Vecteurs moustiques , Animaux , Culex/physiologie , Culex/virologie , Culex/classification , Souris , Humains , Vecteurs moustiques/physiologie , Vecteurs moustiques/virologie , Vecteurs moustiques/classification , Femelle , Allemagne , Iran , Spécificité d'hôte , Virus du Nil occidental/physiologie , Virus du Nil occidental/génétique , Flavivirus/génétique , Flavivirus/physiologie , Flavivirus/isolement et purification , Spécificité d'espèce
6.
Viruses ; 16(8)2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39205301

RÉSUMÉ

Tick-borne encephalitis virus (TBEV) is a neurotropic member of the genus Orthoflavivirus (former Flavivirus) and is of significant health concern in Europe and Asia. TBEV pathogenesis may occur directly via virus-induced damage to neurons or through immunopathology due to excessive inflammation. While primary cells isolated from the host can be used to study the immune response to TBEV, it is still unclear how well these reflect the immune response elicited in vivo. Here, we compared the transcriptional response to TBEV and the less pathogenic tick-borne flavivirus, Langat virus (LGTV), in primary monocultures of neurons, astrocytes and microglia in vitro, with the transcriptional response in vivo captured by single-nuclei RNA sequencing (snRNA-seq) of a whole mouse cortex. We detected similar transcriptional changes induced by both LGTV and TBEV infection in vitro, with the lower response to LGTV likely resulting from slower viral kinetics. Gene set enrichment analysis showed a stronger transcriptional response in vivo than in vitro for astrocytes and microglia, with a limited overlap mainly dominated by interferon signaling. Together, this adds to our understanding of neurotropic flavivirus pathogenesis and the strengths and limitations of available model systems.


Sujet(s)
Astrocytes , Virus de l'encéphalite à tiques (sous-groupe) , Encéphalites à tiques , Microglie , Neurones , Animaux , Astrocytes/virologie , Microglie/virologie , Virus de l'encéphalite à tiques (sous-groupe)/génétique , Virus de l'encéphalite à tiques (sous-groupe)/physiologie , Virus de l'encéphalite à tiques (sous-groupe)/pathogénicité , Souris , Neurones/virologie , Encéphalites à tiques/virologie , Infections à flavivirus/virologie , Infections à flavivirus/immunologie , Cellules cultivées , Flavivirus/physiologie , Flavivirus/génétique , Souris de lignée C57BL , Transcription génétique
7.
Sci Rep ; 14(1): 20095, 2024 08 29.
Article de Anglais | MEDLINE | ID: mdl-39209987

RÉSUMÉ

Usutu (USUV), West Nile (WNV), and Zika virus (ZIKV) are neurotropic arthropod-borne viruses (arboviruses) that cause severe neurological disease in humans. However, USUV-associated neurological disease is rare, suggesting a block in entry to or infection of the brain. We determined the replication, cell tropism and neurovirulence of these arboviruses in human brain tissue using a well-characterized human fetal organotypic brain slice culture model. Furthermore, we assessed the efficacy of interferon-ß and 2'C-methyl-cytidine, a synthetic nucleoside analogue, in restricting viral replication. All three arboviruses replicated within the brain slices, with WNV reaching the highest titers, and all primarily infected neuronal cells. USUV- and WNV-infected cells exhibited a shrunken morphology, not associated with detectable cell death. Pre-treatment with interferon-ß inhibited replication of all arboviruses, while 2'C-methyl-cytidine reduced only USUV and ZIKV titers. Collectively, USUV can infect human brain tissue, showing similarities in tropism and neurovirulence as WNV and ZIKV. These data suggest that a blockade to infection of the human brain may not be the explanation for the low clinical incidence of USUV-associated neurological disease. However, USUV replicated more slowly and to lower titers than WNV, which could help to explain the reduced severity of neurological disease resulting from USUV infection.


Sujet(s)
Encéphale , Flavivirus , Réplication virale , Virus du Nil occidental , Virus Zika , Humains , Virus du Nil occidental/pathogénicité , Virus du Nil occidental/physiologie , Virus Zika/pathogénicité , Virus Zika/physiologie , Encéphale/virologie , Réplication virale/effets des médicaments et des substances chimiques , Flavivirus/pathogénicité , Flavivirus/physiologie , Flavivirus/effets des médicaments et des substances chimiques , Foetus/virologie , Interféron bêta/pharmacologie , Animaux , Virulence , Techniques de culture d'organes , Tropisme viral , Neurones/virologie , Infections à flavivirus/virologie , Infection par le virus Zika/virologie , Chlorocebus aethiops , Cellules Vero
8.
Virus Res ; 348: 199447, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39117146

RÉSUMÉ

One third of all emerging infectious diseases are vector-borne, with no licensed antiviral therapies available against any vector-borne viruses. Zika virus and Usutu virus are two emerging flaviviruses transmitted primarily by mosquitoes. These viruses modulate different host pathways, including the PI3K/AKT/mTOR pathway. Here, we report the effect on ZIKV and USUV replication of two AKT inhibitors, Miransertib (ARQ-092, allosteric inhibitor) and Capivasertib (AZD5363, competitive inhibitor) in different mammalian and mosquito cell lines. Miransertib showed a stronger inhibitory effect against ZIKV and USUV than Capivasertib in mammalian cells, while Capivasertib showed a stronger effect in mosquito cells. These findings indicate that AKT plays a conserved role in flavivirus infection, in both the vertebrate host and invertebrate vector. Nevertheless, the specific function of AKT may vary depending on the host species. These findings indicate that AKT may be playing a conserved role in flavivirus infection in both, the vertebrate host and the invertebrate vector. However, the specific function of AKT may vary depending on the host species. A better understanding of virus-host interactions is therefore required to develop new treatments to prevent human disease and new approaches to control transmission by insect vectors.


Sujet(s)
Infections à flavivirus , Flavivirus , Protéines proto-oncogènes c-akt , Réplication virale , Virus Zika , Animaux , Flavivirus/physiologie , Flavivirus/effets des médicaments et des substances chimiques , Flavivirus/génétique , Humains , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Lignée cellulaire , Virus Zika/physiologie , Virus Zika/effets des médicaments et des substances chimiques , Infections à flavivirus/virologie , Infections à flavivirus/transmission , Vertébrés/virologie , Antiviraux/pharmacologie , Vecteurs moustiques/virologie , Chlorocebus aethiops , Culicidae/virologie , Interactions hôte-pathogène
9.
Sci Rep ; 14(1): 19452, 2024 08 21.
Article de Anglais | MEDLINE | ID: mdl-39169115

RÉSUMÉ

Bagaza virus (BAGV) is a mosquito-borne flavivirus of the family Flaviviridae, genus Orthoflavivirus, Ntaya serocomplex. Like other viruses of the Ntaya and Japanese encephalitis serocomplexes, it is maintained in nature in transmission cycles involving viremic wild bird reservoirs and Culex spp. mosquitoes. The susceptibility of red-legged partridge, ring-necked pheasant, Himalayan monal and common wood pigeon is well known. Determining whether other species are susceptible to BAGV infection is fundamental to understanding the dynamics of disease transmission and maintenance. In September 2023, seven Eurasian magpies were found dead in a rural area in the Mértola district (southern Portugal) where a BAGV-positive cachectic red-legged partridge had been found two weeks earlier. BAGV had also been detected in several red-legged partridges in the same area in September 2021. Three of the magpies were tested for Bagaza virus, Usutu virus, West Nile virus, Avian influenza virus and Avian paramyxovirus serotype 1, and were positive for BAGV only. Sequencing data confirmed the specificity of the molecular detection. Our results indicate that BAGV is circulating in southern Portugal and confirm that Eurasian magpie is potential susceptible to BAGV infection. The inclusion of the abundant Eurasian magpie in the list of BAGV hosts raises awareness of the potential role of this species as as an amplifying host.


Sujet(s)
Flavivirus , Animaux , Portugal , Flavivirus/génétique , Flavivirus/isolement et purification , Phylogenèse , Maladies des oiseaux/virologie , Maladies des oiseaux/épidémiologie , Infections à flavivirus/virologie , Infections à flavivirus/médecine vétérinaire , Infections à flavivirus/transmission , Infections à flavivirus/épidémiologie
10.
Virol J ; 21(1): 163, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39044231

RÉSUMÉ

Usutu virus (USUV), an arbovirus from the Flaviviridae family, genus Flavivirus, has recently gained increasing attention because of its potential for emergence. After his discovery in South Africa, USUV spread to other African countries, then emerged in Europe where it was responsible for epizootics. The virus has recently been found in Asia. USUV infection in humans is considered to be most often asymptomatic or to cause mild clinical signs. However, a few cases of neurological complications such as encephalitis or meningo-encephalitis have been reported in both immunocompromised and immunocompetent patients. USUV natural life cycle involves Culex mosquitoes as its main vector, and multiple bird species as natural viral reservoirs or amplifying hosts, humans and horses can be incidental hosts. Phylogenetic studies carried out showed eight lineages, showing an increasing genetic diversity for USUV. This work describes the development and validation of a novel whole-genome amplicon-based sequencing approach to Usutu virus. This study was carried out on different strains from Senegal and Italy. The new approach showed good coverage using samples derived from several vertebrate hosts and may be valuable for Usutu virus genomic surveillance to better understand the dynamics of evolution and transmission of the virus.


Sujet(s)
Infections à flavivirus , Flavivirus , Génome viral , Phylogenèse , Flavivirus/génétique , Flavivirus/classification , Flavivirus/isolement et purification , Animaux , Infections à flavivirus/virologie , Infections à flavivirus/médecine vétérinaire , Humains , Sénégal , Italie , Oiseaux/virologie , ARN viral/génétique , Variation génétique , Culex/virologie , Séquençage du génome entier , Equus caballus/virologie
11.
Virologie (Montrouge) ; 28(3): 187-197, 2024 Jun 01.
Article de Français | MEDLINE | ID: mdl-38970340

RÉSUMÉ

Orthoflaviviruses are enveloped positive-sense RNA viruses comprising numerous human pathogens transmitted by hematophagous arthropods. This includes viruses such as dengue virus, Zika virus, and yellow fever virus. The viral nonstructural protein NS1 plays a central role in the pathogenesis and cycle of these viruses by acting in two different forms: associated with the plasma membrane (NS1m) or secreted outside the cell (NS1s). The versatility of NS1 is evident in its ability to modulate various aspects of the infectious process, from immune evasion to pathogenesis. As an intracellular protein, it disrupts many processes, interfering with signaling pathways and facilitating viral replication in concert with other viral proteins. As a secreted protein, NS1 actively participates in immune evasion, interfering with the host immune system, inhibiting the complement system, facilitating viral dissemination, and disrupting the integrity of endothelial barriers. This review primarily aims to address the role of NS1 in viral pathogenesis associated with orthoflaviviruses.


Sujet(s)
Protéines virales non structurales , Réplication virale , Protéines virales non structurales/métabolisme , Protéines virales non structurales/physiologie , Humains , Animaux , Infections à flavivirus/virologie , Échappement immunitaire , Flavivirus/physiologie , Flavivirus/pathogénicité , Virus Zika/physiologie , Virus Zika/pathogénicité , Virus de la dengue/physiologie
12.
J Mol Model ; 30(8): 295, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39083139

RÉSUMÉ

CONTEXT: Flaviviruses cause severe encephalitic or hemorrhagic diseases in humans. Its members, Kyasanur forest disease virus (KFDV) and Alkhumra hemorrhagic fever virus (ALKV), cause hemorrhagic fever and are prevalent in India and Saudi Arabia, respectively, while the tick-borne encephalitis virus (TBEV) causes a dangerous encephalitic infection in Europe and Asia. However, little information is available about the targets of immune responses for these deadly viruses. Here, we predict potential antigenic peptide epitopes of viral envelope protein for inducing a cell-mediated and humoral immune response. METHODS: Using the Immune Epitope Database and Analysis Resource (IEDB-AR), we identified 13 MHC-I and two MHC-II dominant conserved epitopes in KFDV and ALKV and six MHC-I and three MHC-II epitopes in TBEV envelope proteins. Parallelly, we also predicted B-cell linear and discontinuous envelope protein epitopes for these viruses. Interestingly, the epitopes are conserved in all three viral envelope proteins. Further, the discontinuous epitopes are structurally compared with the available DENV, ZIKV, WNV, TBEV, and LIV envelope protein antibody structures. Overall structural comparison analyses highlight (i) lateral ridge epitope in the ED-III domain of E protein, and (ii) envelope dimer epitope (EDE) could be targeted for developing potent vaccine candidates as well as therapeutic antibody production. Moreover, existing structural and biochemical functions of the same epitopes in homologous viruses are predicted to have a reduced antibody-dependent enhancement (ADE) effect on flaviviral infection.


Sujet(s)
Flavivirus , Flavivirus/immunologie , Humains , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/composition chimique , Biologie informatique , Séquence d'acides aminés , Déterminants antigéniques des lymphocytes B/immunologie , Déterminants antigéniques des lymphocytes B/composition chimique , Similitude de séquences d'acides aminés , Épitopes/immunologie , Épitopes/composition chimique , Modèles moléculaires , Virus de l'encéphalite à tiques (sous-groupe)/immunologie
13.
PLoS Negl Trop Dis ; 18(7): e0012172, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38985837

RÉSUMÉ

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.


Sujet(s)
Maladies des oiseaux , Infections à flavivirus , Flavivirus , Ixodes , Nymphe , Animaux , Ixodes/virologie , Ixodes/physiologie , Flavivirus/physiologie , Flavivirus/génétique , Infections à flavivirus/transmission , Infections à flavivirus/médecine vétérinaire , Infections à flavivirus/virologie , Nymphe/virologie , Maladies des oiseaux/virologie , Maladies des oiseaux/transmission , Oiseaux/virologie , Vecteurs arachnides/virologie , Vecteurs arachnides/physiologie , Pays-Bas , Femelle
14.
Parasit Vectors ; 17(1): 285, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956650

RÉSUMÉ

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.


Sujet(s)
Culex , Infections à flavivirus , Flavivirus , Vecteurs moustiques , Animaux , Culex/virologie , Femelle , Vecteurs moustiques/virologie , Flavivirus/génétique , Flavivirus/physiologie , Belgique , Infections à flavivirus/transmission , Infections à flavivirus/virologie , Salive/virologie
15.
Sci Adv ; 10(27): eadl1888, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38959313

RÉSUMÉ

We present structures of three immature tick-borne encephalitis virus (TBEV) isolates. Our atomic models of the major viral components, the E and prM proteins, indicate that the pr domains of prM have a critical role in holding the heterohexameric prM3E3 spikes in a metastable conformation. Destabilization of the prM furin-sensitive loop at acidic pH facilitates its processing. The prM topology and domain assignment in TBEV is similar to the mosquito-borne Binjari virus, but is in contrast to other immature flavivirus models. These results support that prM cleavage, the collapse of E protein ectodomains onto the virion surface, the large movement of the membrane domains of both E and M, and the release of the pr fragment from the particle render the virus mature and infectious. Our work favors the collapse model of flavivirus maturation warranting further studies of immature flaviviruses to determine the sequence of events and mechanistic details driving flavivirus maturation.


Sujet(s)
Virus de l'encéphalite à tiques (sous-groupe) , Protéines de l'enveloppe virale , Virus de l'encéphalite à tiques (sous-groupe)/physiologie , Protéines de l'enveloppe virale/composition chimique , Protéines de l'enveloppe virale/métabolisme , Modèles moléculaires , Flavivirus/physiologie , Animaux , Virion , Encéphalites à tiques/virologie , Humains
16.
Acta Trop ; 258: 107330, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39043331

RÉSUMÉ

Usutu virus (USUV) is a mosquito-borne flavivirus originating from Africa, that belongs to the Japanese encephalitis virus (JEV) complex. In nature, USUV involves Culex spp. mosquitoes acting as vectors and birds as amplifying hosts. The virus has recently spread in Europe and is considered an emerging human pathogen. This is the first research study performed in Greece revealing the presence and circulation of USUV in Culex spp. mosquito populations. Out of the 1,500 mosquito pools tested with real-time RT-PCR, four (Roesch et al., 2019) were positive for USUV. All four pools were collected from the region of Central Macedonia, Northern Greece.


Sujet(s)
Culex , Flavivirus , Vecteurs moustiques , Animaux , Culex/virologie , Grèce , Flavivirus/génétique , Flavivirus/isolement et purification , Flavivirus/classification , Vecteurs moustiques/virologie , Réaction de polymérisation en chaine en temps réel , Infections à flavivirus/virologie , Infections à flavivirus/médecine vétérinaire , Infections à flavivirus/transmission , ARN viral/génétique , ARN viral/isolement et purification
17.
Virus Res ; 347: 199431, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38969013

RÉSUMÉ

Usutu virus (USUV) is an emerging flavivirus that can infect birds and mammals. In humans, in severe cases, it may cause neuroinvasive disease. The innate immune system, and in particular the interferon response, functions as the important first line of defense against invading pathogens such as USUV. Many, if not all, viruses have developed mechanisms to suppress and/or evade the interferon response in order to facilitate their replication. The ability of USUV to antagonize the interferon response has so far remained largely unexplored. Using dual-luciferase reporter assays we observed that multiple of the USUV nonstructural (NS) proteins were involved in suppressing IFN-ß production and signaling. In particular NS4A was very effective at suppressing IFN-ß production. We found that NS4A interacted with the mitochondrial antiviral signaling protein (MAVS) and thereby blocked its interaction with melanoma differentiation-associated protein 5 (MDA5), resulting in reduced IFN-ß production. The TM1 domain of NS4A was found to be essential for binding to MAVS. By screening a panel of flavivirus NS4A proteins we found that the interaction of NS4A with MAVS is conserved among flaviviruses. The increased understanding of the role of NS4A in flavivirus immune evasion could aid the development of vaccines and therapeutic strategies.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Flavivirus , Hélicase IFIH1 inductrice de l'interféron , Interféron bêta , Transduction du signal , Protéines virales non structurales , Protéines virales non structurales/métabolisme , Protéines virales non structurales/génétique , Protéines virales non structurales/immunologie , Humains , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/immunologie , Flavivirus/immunologie , Flavivirus/génétique , Flavivirus/physiologie , Interféron bêta/génétique , Interféron bêta/immunologie , Interféron bêta/métabolisme , Hélicase IFIH1 inductrice de l'interféron/génétique , Hélicase IFIH1 inductrice de l'interféron/métabolisme , Hélicase IFIH1 inductrice de l'interféron/immunologie , Cellules HEK293 , Échappement immunitaire , Infections à flavivirus/immunologie , Infections à flavivirus/virologie , Interactions hôte-pathogène/immunologie , Liaison aux protéines , Immunité innée , Animaux
18.
Poult Sci ; 103(9): 104005, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39053372

RÉSUMÉ

Annexin A2 (ANXA2) is a multifaceted protein implicated in various stages of viral infections, particularly in envelope virus replication through mechanisms such as endocytosis and exocytosis. This study delves into the characterization and functional dynamics of duck ANXA2 (duANXA2). We successfully cloned the full-length coding sequence of duANXA2 and conducted a detailed structural analysis. The open reading frame (ORF) of duANXA2 is 1020 bp, encoding 339 amino acids and featuring 4 conserved domains. Phylogenetic tree analysis indicates that duANXA2 is most closely related to Gallus gallus, with significantly lesser homology to fish species. We evaluated the tissue-specific expression of duANXA2 in healthy ducks, noting its ubiquitous presence but varying expression levels across different organs, with notably high expression in the esophagus and immune organs. Upon infecting duck embryo fibroblast (DEF) cells with the duck Tembusu virus (DTMUV), a flavivirus causing ducks substantial mortality and a dramatic decline in egg production, we observed a pronounced upregulation of duANXA2. Functional assays demonstrated that overexpression of duANXA2 in DEF cells augments DTMUV replication, while its interference markedly reduces DTMUV replication. These findings underscore the role of duANXA2 as a facilitator of DTMUV replication, presenting it as a potential target for therapeutic intervention in managing DTMUV infections.


Sujet(s)
Annexine A2 , Protéines aviaires , Canards , Flavivirus , Phylogenèse , Maladies de la volaille , Réplication virale , Animaux , Canards/génétique , Annexine A2/génétique , Annexine A2/métabolisme , Maladies de la volaille/virologie , Maladies de la volaille/génétique , Flavivirus/physiologie , Flavivirus/génétique , Protéines aviaires/génétique , Protéines aviaires/métabolisme , Protéines aviaires/composition chimique , Clonage moléculaire , Infections à flavivirus/médecine vétérinaire , Infections à flavivirus/virologie , Infections à flavivirus/génétique , Séquence d'acides aminés , Alignement de séquences/médecine vétérinaire
19.
Trends Microbiol ; 32(8): 725-727, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38853121

RÉSUMÉ

The transmission of flaviviruses, such as dengue virus (DENV) and Zika virus (ZIKV), poses a significant threat to global public health. Zhang et al. recently showed that Rosenbergiella sp. YN46 (Rosenbergiella_YN46), a bacterium from the mosquito gut, inhibits flavivirus transmission and thus offers a potential biocontrol strategy with broad public health implications.


Sujet(s)
Flavivirus , Animaux , Flavivirus/physiologie , Humains , Infections à flavivirus/transmission , Infections à flavivirus/virologie , Infections à flavivirus/prévention et contrôle , Virus Zika/physiologie , Culicidae/microbiologie , Culicidae/virologie , Virus de la dengue/physiologie , Microbiome gastro-intestinal/physiologie , Vecteurs moustiques/virologie , Vecteurs moustiques/microbiologie
20.
Virus Res ; 347: 199422, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38901564

RÉSUMÉ

Autophagy is a lysosomal degradative pathway, which regulates the homeostasis of eukaryotic cells. This pathway can degrade misfolded or aggregated proteins, clear damaged organelles, and eliminate intracellular pathogens, including viruses, bacteria, and parasites. But, not all types of viruses are eliminated by autophagy. Flaviviruses (e.g., Yellow fever, Japanese encephalitis, Hepatitis C, Dengue, Zika, and West Nile viruses) are single-stranded and enveloped RNA viruses, and transmitted to humans primarily through the bites of arthropods, leading to severe and widespread illnesses. Like the coronavirus SARS-CoV-II, flaviviruses hijack autophagy for their infection and escape from host immune clearance. Thus, it is possible to control these viral infections by inhibiting autophagy. In this review, we summarize recent research progresses on hijacking of autophagy by flaviviruses and discuss the feasibility of antiviral therapies using autophagy inhibitors.


Sujet(s)
Autophagie , Infections à flavivirus , Flavivirus , Humains , Flavivirus/physiologie , Flavivirus/pathogénicité , Infections à flavivirus/virologie , Animaux , Antiviraux/usage thérapeutique , Antiviraux/pharmacologie , Interactions hôte-pathogène
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE