Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.728
Filtrar
1.
RNA Biol ; 21(1): 14-30, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38797925

RESUMEN

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.


Asunto(s)
Flavivirus , Genoma Viral , Conformación de Ácido Nucleico , ARN Viral , Replicación Viral , Flavivirus/genética , Flavivirus/fisiología , ARN Viral/metabolismo , ARN Viral/química , ARN Viral/genética , Humanos , Infecciones por Flavivirus/virología , Ensamble de Virus , Animales , Biosíntesis de Proteínas
2.
J Gen Virol ; 105(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38809251

RESUMEN

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.


Asunto(s)
Flavivirus , Ixodidae , Filogenia , Animales , Flavivirus/genética , Flavivirus/clasificación , Flavivirus/aislamiento & purificación , China , Ixodidae/virología , Femenino
3.
Virology ; 595: 110084, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38692132

RESUMEN

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.


Asunto(s)
Patos , Flavivirus , Enfermedades de las Aves de Corral , Proteínas no Estructurales Virales , Ensamble de Virus , Replicación Viral , Animales , Patos/virología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Flavivirus/genética , Flavivirus/fisiología , Enfermedades de las Aves de Corral/virología , Infecciones por Flavivirus/virología , Mutación
4.
Euro Surveill ; 29(20)2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38757289

RESUMEN

Aedes albopictus collected in 2023 in the greater Paris area (Île-de-France) were experimentally able to transmit five arboviruses: West Nile virus from 3 days post-infection (dpi), chikungunya virus and Usutu virus from 7 dpi, dengue virus and Zika virus from 21 dpi. Given the growing number of imported dengue cases reported in early 2024 in France, surveillance of Ae. albopictus should be reinforced during the Paris Olympic Games in July, when many international visitors including from endemic countries are expected.


Asunto(s)
Aedes , Virus Chikungunya , Virus del Dengue , Virus Zika , Animales , Aedes/virología , Humanos , Virus Zika/aislamiento & purificación , Virus del Dengue/aislamiento & purificación , Virus Chikungunya/aislamiento & purificación , Paris , Mosquitos Vectores/virología , Virus del Nilo Occidental/aislamiento & purificación , Arbovirus/aislamiento & purificación , Infecciones por Arbovirus/transmisión , Flavivirus/aislamiento & purificación , Francia , Dengue/transmisión , Dengue/epidemiología , Infección por el Virus Zika/transmisión
5.
Viruses ; 16(5)2024 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-38793692

RESUMEN

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.


Asunto(s)
Patos , Infecciones por Flavivirus , Flavivirus , Enfermedades de las Aves de Corral , Animales , Patos/virología , Flavivirus/patogenicidad , Flavivirus/inmunología , Flavivirus/genética , Infecciones por Flavivirus/veterinaria , Infecciones por Flavivirus/virología , Infecciones por Flavivirus/transmisión , Genoma Viral , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/transmisión , Vacunas Virales/inmunología , Agricultores , Anticuerpos Antivirales/sangre , Humanos
6.
J Biosci ; 492024.
Artículo en Inglés | MEDLINE | ID: mdl-38783793

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside , Flavivirus , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química , Flavivirus/patogenicidad , Flavivirus/genética , Flavivirus/fisiología , Flavivirus/metabolismo , Filogenia , Humanos , Unión Proteica , Cápside/metabolismo , Cápside/química , Infecciones por Flavivirus/virología , Infecciones por Flavivirus/metabolismo , Simulación del Acoplamiento Molecular , Secuencia de Aminoácidos
7.
Sci Adv ; 10(18): eadm8275, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38691607

RESUMEN

Flaviviruses encode a conserved, membrane-associated nonstructural protein 1 (NS1) with replication and immune evasion functions. The current knowledge of secreted NS1 (sNS1) oligomers is based on several low-resolution structures, thus hindering the development of drugs and vaccines against flaviviruses. Here, we revealed that recombinant sNS1 from flaviviruses exists in a dynamic equilibrium of dimer-tetramer-hexamer states. Two DENV4 hexameric NS1 structures and several tetrameric NS1 structures from multiple flaviviruses were solved at atomic resolution by cryo-EM. The stacking of the tetrameric NS1 and hexameric NS1 is facilitated by the hydrophobic ß-roll and connector domains. Additionally, a triacylglycerol molecule located within the central cavity may play a role in stabilizing the hexamer. Based on differentiated interactions between the dimeric NS1, two distinct hexamer models (head-to-head and side-to-side hexamer) and the step-by-step assembly mechanisms of NS1 dimer into hexamer were proposed. We believe that our study sheds light on the understanding of the NS1 oligomerization and contributes to NS1-based therapies.


Asunto(s)
Microscopía por Crioelectrón , Flavivirus , Modelos Moleculares , Multimerización de Proteína , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Flavivirus/metabolismo , Flavivirus/química , Conformación Proteica
8.
Vet Microbiol ; 292: 110071, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38574695

RESUMEN

Duck Tembusu virus (DTMUV) is a newly emerging pathogen that causes massive economic losses to the poultry industry in China and neighbouring countries. Vimentin, an intermediate filament protein, has been demonstrated to be involved in viral replication during infection. However, the specific role of vimentin in DTMUV replication has not been determined. In this study, we found that overexpression of vimentin in BHK-21 cells can inhibit DTMUV replication. Moreover, DTMUV replication was enhanced after vimentin expression was reduced in BHK-21 cells via small interfering RNA (siRNA). Further research indicated that DTMUV infection had no effect on the transcription or expression of vimentin. However, we found that DTMUV infection induced vimentin rearrangement, and the rearrangement of vimentin was subsequently confirmed to negatively modulate viral replication through the use of a vimentin network disrupting agent. Vimentin rearrangement is closely associated with its phosphorylation. Our experiments revealed that the phosphorylation of vimentin at Ser56 was promoted in the early stage of DTMUV infection. In addition, by inhibiting the phosphorylation of vimentin at Ser56 with a CDK5 inhibitor, vimentin rearrangement was suppressed, and DTMUV replication was significantly enhanced. These results indicated that DTMUV infection induced vimentin phosphorylation and rearrangement through CDK5, resulting in the inhibition of DTMUV replication. In summary, our study reveals a role for vimentin as a negative factor in the process of DTMUV replication, which helps to elucidate the function of cellular proteins in regulating DTMUV replication.


Asunto(s)
Infecciones por Flavivirus , Flavivirus , Enfermedades de las Aves de Corral , Animales , Patos , Vimentina/genética , Flavivirus/fisiología , Infecciones por Flavivirus/veterinaria , Replicación Viral
9.
Science ; 384(6693): eadn9524, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38669573

RESUMEN

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.


Asunto(s)
Aedes , Virus del Dengue , Mosquitos Vectores , Simbiosis , Virus Zika , Animales , Aedes/microbiología , Aedes/virología , Virus del Dengue/fisiología , Mosquitos Vectores/virología , Mosquitos Vectores/microbiología , Virus Zika/fisiología , Dengue/transmisión , Dengue/virología , Dengue/prevención & control , Microbioma Gastrointestinal , Acetobacteraceae/fisiología , Femenino , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/genética , Flavivirus/fisiología , Flavivirus/genética , Infección por el Virus Zika/transmisión , Infección por el Virus Zika/virología
10.
Proc Natl Acad Sci U S A ; 121(16): e2317978121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38593069

RESUMEN

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.


Asunto(s)
Aedes , Infecciones por Flavivirus , Flavivirus , Infección por el Virus Zika , Virus Zika , Animales , Ratones , Flavivirus/genética , Virus Zika/genética , Ubiquitina/metabolismo , Ligasas/metabolismo , Proteínas Virales/metabolismo , Mamíferos
11.
Viral Immunol ; 37(3): 167-175, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38574259

RESUMEN

Zika virus (ZIKV) is an emerging flavivirus associated with several neurological diseases such as Guillain-Barré syndrome in adults and microcephaly in newborn children. Its distribution and mode of transmission (via Aedes aegypti and Aedes albopictus mosquitoes) collectively cause ZIKV to be a serious concern for global health. High genetic homology of flaviviruses and shared ecology is a hurdle for accurate detection. Distinguishing infections caused by different viruses based on serological recognition can be misleading as many anti-flavivirus monoclonal antibodies (mAbs) discovered to date are highly cross-reactive, especially those against the envelope (E) protein. To provide more specific research tools, we produced ZIKV E directed hybridoma cell lines and characterized two highly ZIKV-specific mAb clones (mAbs A11 and A42) against several members of the Flavivirus genus. Epitope mapping of mAb A11 revealed glycan loop specificity in Domain I of the ZIKV E protein. The development of two highly specific mAbs targeting the surface fusion protein of ZIKV presents a significant advancement in research capabilities as these can be employed as essential tools to enhance our understanding of ZIKV identification on infected cells ex vivo or in culture.


Asunto(s)
Aedes , Flavivirus , Infección por el Virus Zika , Virus Zika , Animales , Recién Nacido , Humanos , Proteínas del Envoltorio Viral , Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales
12.
Antiviral Res ; 226: 105878, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38582134

RESUMEN

Flaviviruses can cause severe illness in humans. Effective and safe vaccines are available for some species; however, for many flaviviruses disease prevention or specific treatments remain unavailable. The viral replication cycle depends on the proteolytic activity of the NS2B-NS3 protease, which releases functional viral proteins from a non-functional polyprotein precursor, rendering the protease a promising drug target. In this study, we characterised recombinant NS2B-NS3 proteases from ten flaviviruses including three unreported proteases from the Usutu, Kyasanur forest disease and Powassan viruses. All protease constructs comprise a covalent Gly4-Ser-Gly4 linker connecting the NS3 serine protease domain with its cofactor NS2B. We conducted a comprehensive cleavage site analysis revealing areas of high conversion. While all proteases were active in enzymatic assays, we noted a 1000-fold difference in catalytic efficiency across proteases from different flaviviruses. Two bicyclic peptide inhibitors displayed anti-pan-flaviviral protease activity with inhibition constants ranging from 10 to 1000 nM.


Asunto(s)
Antivirales , Flavivirus , Serina Endopeptidasas , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Flavivirus/efectos de los fármacos , Flavivirus/enzimología , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/química , Antivirales/farmacología , Antivirales/química , Humanos , ARN Helicasas/metabolismo , ARN Helicasas/química , ARN Helicasas/genética , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/química , Proteasas Virales , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-box
13.
Theor Popul Biol ; 157: 118-128, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38626854

RESUMEN

Infectious disease agents can influence each other's dynamics in shared host populations. We consider such influence for two mosquito-borne infections where one pathogen is endemic at the time that a second pathogen invades. We regard a setting where the vector has a bias towards biting host individuals infected with the endemic pathogen and where there is a cost to co-infected hosts. As a motivating case study, we regard Plasmodium spp., that cause avian malaria, as the endemic pathogen, and Usutu virus (USUV) as the invading pathogen. Hosts with malaria attract more mosquitoes compared to susceptible hosts, a phenomenon named vector bias. The possible trade-off between the vector-bias effect and the co-infection mortality is studied using a compartmental epidemic model. We focus first on the basic reproduction number R0 for Usutu virus invading into a malaria-endemic population, and then explore the long-term dynamics of both pathogens once Usutu virus has become established. We find that the vector bias facilitates the introduction of malaria into a susceptible population, as well as the introduction of Usutu in a malaria-endemic population. In the long term, however, both a vector bias and co-infection mortality lead to a decrease in the number of individuals infected with either pathogen, suggesting that avian malaria is unlikely to be a promoter of Usutu invasion. This proposed approach is general and allows for new insights into other negative associations between endemic and invading vector-borne pathogens.


Asunto(s)
Aves , Flavivirus , Plasmodium , Animales , Aves/virología , Aves/parasitología , Plasmodium/patogenicidad , Flavivirus/patogenicidad , Coinfección/virología , Malaria Aviar , Enfermedades Endémicas , Infecciones por Flavivirus/virología , Mosquitos Vectores/virología , Mosquitos Vectores/parasitología , Malaria
14.
Proc Natl Acad Sci U S A ; 121(19): e2319400121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38687787

RESUMEN

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.


Asunto(s)
Ciervos , Flavivirus , Metagenómica , Garrapatas , Animales , Metagenómica/métodos , Japón/epidemiología , Ciervos/virología , Flavivirus/genética , Flavivirus/aislamiento & purificación , Flavivirus/clasificación , Garrapatas/virología , Filogenia , Viroma/genética , Virión/genética , Sus scrofa/virología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Estudios Seroepidemiológicos , Genoma Viral
15.
Viruses ; 16(4)2024 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-38675867

RESUMEN

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.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie) , Vesículas Extracelulares , Genoma Viral , Replicón , Proteínas Virales , Vesículas Extracelulares/virología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/genética , Humanos , Replicón/genética , Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/fisiología , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral , Flavivirus/genética , Flavivirus/fisiología , Virus del Dengue/genética , Virus del Dengue/fisiología , Células HeLa , Células K562 , Animales , Línea Celular , ARN Subgenómico
16.
Viruses ; 16(4)2024 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-38675940

RESUMEN

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.


Asunto(s)
Aves , Infecciones por Flavivirus , Flavivirus , Fiebre del Nilo Occidental , Virus del Nilo Occidental , Europa (Continente)/epidemiología , Virus del Nilo Occidental/genética , Virus del Nilo Occidental/fisiología , Virus del Nilo Occidental/aislamiento & purificación , Animales , Humanos , Flavivirus/clasificación , Flavivirus/genética , Flavivirus/patogenicidad , Flavivirus/aislamiento & purificación , Flavivirus/fisiología , Infecciones por Flavivirus/epidemiología , Infecciones por Flavivirus/virología , Infecciones por Flavivirus/transmisión , Infecciones por Flavivirus/veterinaria , Fiebre del Nilo Occidental/epidemiología , Fiebre del Nilo Occidental/virología , Fiebre del Nilo Occidental/transmisión , Aves/virología , Culicidae/virología , Mosquitos Vectores/virología , Brotes de Enfermedades
17.
Poult Sci ; 103(6): 103727, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38652953

RESUMEN

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.


Asunto(s)
Patos , Flavivirus , Proteínas no Estructurales Virales , Replicación Viral , Animales , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Flavivirus/fisiología , Flavivirus/genética , Enfermedades de las Aves de Corral/virología , Infecciones por Flavivirus/veterinaria , Infecciones por Flavivirus/virología , Mutación
18.
ACS Biomater Sci Eng ; 10(4): 2041-2061, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38526408

RESUMEN

In tropical and developing countries, mosquito-borne diseases by flaviviruses pose a serious threat to public health. Early detection is critical for preventing their spread, but conventional methods are time-consuming and require skilled technicians. Biosensors have been developed to address this issue, but cross-reactivity with other flaviviruses remains a challenge. Peptides are essentially biomaterials used in diagnostics that allow virological and serological techniques to identify flavivirus selectively. This biomaterial originated as a small protein consisting of two to 50 amino acid chains. They offer flexibility in chemical modification and can be easily synthesized and applied to living cells in the engineering process. Peptides could potentially be developed as robust, low-cost, sensitive, and selective receptors for detecting flaviviruses. However, modification and selection of the receptor agents are crucial to determine the effectiveness of binding between the targets and the receptors. This paper addresses two potential peptide nucleic acids (PNAs) and affinity peptides that can detect flavivirus from another target-based biosensor as well as the potential peptide behaviors of flaviviruses. The PNAs detect flaviviruses based on the nucleotide base sequence of the target's virological profile via Watson-Crick base pairing, while the affinity peptides sense the epitope or immunological profile of the targets. Recent developments in the functionalization of peptides for flavivirus biosensors are explored in this Review by division into electrochemical, optical, and other detection methods.


Asunto(s)
Flavivirus , Ácidos Nucleicos de Péptidos , Animales , Flavivirus/química , Péptidos/química
19.
PLoS Negl Trop Dis ; 18(3): e0011862, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38527081

RESUMEN

African populations of the mosquito Aedes aegypti are usually considered less susceptible to infection by human-pathogenic flaviviruses than globally invasive populations found outside Africa. Although this contrast has been well documented for Zika virus (ZIKV), it is unclear to what extent it is true for dengue virus (DENV), the most prevalent flavivirus of humans. Addressing this question is complicated by substantial genetic diversity among DENV strains, most notably in the form of four genetic types (DENV1 to DENV4), that can lead to genetically specific interactions with mosquito populations. Here, we carried out a survey of DENV susceptibility using a panel of seven field-derived Ae. aegypti colonies from across the African range of the species and a colony from Guadeloupe, French West Indies as non-African reference. We found considerable variation in the ability of African Ae. aegypti populations to acquire and replicate a panel of six DENV strains spanning the four DENV types. Although African Ae. aegypti populations were generally less susceptible than the reference non-African population from Guadeloupe, in several instances some African populations were equally or more susceptible than the Guadeloupe population. Moreover, the relative level of susceptibility between African mosquito populations depended on the DENV strain, indicating genetically specific interactions. We conclude that unlike ZIKV susceptibility, there is no clear-cut dichotomy in DENV susceptibility between African and non-African Ae. aegypti. DENV susceptibility of African Ae. aegypti populations is highly heterogeneous and largely governed by the specific pairing of mosquito population and DENV strain.


Asunto(s)
Aedes , Virus del Dengue , Dengue , Flavivirus , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Virus del Dengue/genética , Virus Zika/genética , Aedes/genética , Mosquitos Vectores/genética , Dengue/epidemiología
20.
Virol Sin ; 39(2): 228-234, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38461965

RESUMEN

Guaico Culex virus (GCXV) is a newly identified segmented Jingmenvirus from Culex spp. mosquitoes in Central and South America. The genome of GCXV is composed of four or five single-stranded positive RNA segments. However, the infection kinetics and transmission capability of GCXV in mosquitoes remain unknown. In this study, we used reverse genetics to rescue two GCXVs (4S and 5S) that contained four and five RNA segments, respectively, in C6/36 â€‹cells. Further in vitro characterization revealed that the two GCXVs exhibited comparable replication kinetics, protein expression and viral titers. Importantly, GCXV RNAs were detected in the bodies, salivary glands, midguts and ovaries of Culex quinquefasciatus at 4-10 days after oral infection. In addition, two GCXVs can colonize Cx. quinquefasciatus eggs, resulting in positive rates of 15%-35% for the second gonotrophic cycle. In conclusion, our results demonstrated that GCXVs with four or five RNA segments can be detected in Cx. quinquefasciatus eggs during the first and second gonotrophic cycles after oral infection.


Asunto(s)
Culex , Mosquitos Vectores , ARN Viral , Replicación Viral , Animales , Culex/virología , Mosquitos Vectores/virología , ARN Viral/genética , Femenino , Línea Celular , Flavivirus/genética , Flavivirus/fisiología , Flavivirus/aislamiento & purificación , Cinética , Carga Viral , Genoma Viral , Glándulas Salivales/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA