Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.537
Filtrar
1.
Viruses ; 16(6)2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38932175
2.
Nat Commun ; 15(1): 4906, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38851803

RESUMEN

Various low-density lipoprotein receptors (LPRs) have been identified as entry factors for alphaviruses, and structures of the corresponding virion-receptor complexes have been determined. Here, we analyze the similarities and differences in the receptor binding modes of multiple alphaviruses to understand their ability to infect a wide range of hosts. We further discuss the challenges associated with the development of broad-spectrum treatment strategies against a diverse range of alphaviruses.


Asunto(s)
Alphavirus , Antivirales , Receptores de LDL , Internalización del Virus , Animales , Humanos , Alphavirus/efectos de los fármacos , Alphavirus/fisiología , Alphavirus/genética , Infecciones por Alphavirus/tratamiento farmacológico , Infecciones por Alphavirus/virología , Antivirales/uso terapéutico , Antivirales/farmacología , Unión Proteica , Receptores de LDL/metabolismo , Receptores de LDL/genética , Receptores Virales/metabolismo , Receptores Virales/química , Virión/metabolismo , Internalización del Virus/efectos de los fármacos
3.
Appl Microbiol Biotechnol ; 108(1): 355, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38822832

RESUMEN

Getah virus (GETV) is a re-emerging mosquito-borne alphavirus that is highly pathogenic, mainly to pigs and horses. There are no vaccines or treatments available for GETV in swine in China. Therefore, the development of a simple, rapid, specific, and sensitive serological assay for GETV antibodies is essential for the prevention and control of GETV. Current antibody monitoring methods are time-consuming, expensive, and dependent on specialized instrumentation, and these features are not conducive to rapid detection in clinical samples. To address these problem, we developed immunochromatographic test strips (ICTS) using eukaryotically expressed soluble recombinant p62-E1 protein of GETV as a labelled antigen, which has good detection sensitivity and no cross-reactivity with other common porcine virus-positive sera. The ICTS is highly compatible with IFA and ELISA and can be stored for 1 month at 37 °C and for at least 3 months at room temperature. Hence, p62-E1-based ICTS is a rapid, accurate, and convenient method for rapid on-site detection of GETV antibodies. KEY POINTS: • We established a rapid antibody detection method that can monitor GETV infection • We developed colloidal gold test strips with high sensitivity and specificity • The development of colloidal gold test strips will aid in the field serologic detection of GETV.


Asunto(s)
Alphavirus , Anticuerpos Antivirales , Oro Coloide , Sensibilidad y Especificidad , Animales , Oro Coloide/química , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Alphavirus/inmunología , Porcinos , Cromatografía de Afinidad/métodos , Infecciones por Alphavirus/diagnóstico , Infecciones por Alphavirus/inmunología , Enfermedades de los Porcinos/diagnóstico , Enfermedades de los Porcinos/virología , Tiras Reactivas , China , Ensayo de Inmunoadsorción Enzimática/métodos
4.
Front Cell Infect Microbiol ; 14: 1388360, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38841111

RESUMEN

Background: Alphaviruses are a diverse group of pathogens that have garnered considerable attention due to their impact on human health. By investigating alphavirus receptors, researchers can elucidate viral entry mechanisms and gain important clues for the prevention and treatment of viral diseases. This study presents an in-depth analysis of the research progress made in the field of alphavirus receptors through bibliometric analysis. Methods: This study encompasses various aspects, including historical development, annual publication trends, author and cited-author analysis, institutional affiliations, global distribution of research contributions, reference analysis with strongest citation bursts, keyword analysis, and a detailed exploration of recent discoveries in alphavirus receptor research. Results: The results of this bibliometric analysis highlight key milestones in alphavirus receptor research, demonstrating the progression of knowledge in this field over time. Additionally, the analysis reveals current research hotspots and identifies emerging frontiers, which can guide future investigations and inspire novel therapeutic strategies. Conclusion: This study provides an overview of the state of the art in alphavirus receptor research, consolidating the existing knowledge and paving the way for further advancements. By shedding light on the significant developments and emerging areas of interest, this study serves as a valuable resource for researchers, clinicians, and policymakers engaged in combating alphavirus infections and improving public health.


Asunto(s)
Alphavirus , Bibliometría , Humanos , Receptores Virales/metabolismo , Animales , Internalización del Virus , Infecciones por Alphavirus/virología , Investigación Biomédica/tendencias
5.
Front Immunol ; 15: 1401086, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38903507

RESUMEN

The mitochondrial anti-viral signaling (MAVS) protein is an intermediary adaptor protein of retinoic acid-inducible gene-1 (RIG-I) like receptor (RLR) signaling, which activates the transcription factor interferon (IFN) regulatory factor 3 (IRF3) and NF-kB to produce type I IFNs. MAVS expression has been reported in different fish species, but few studies have shown its functional role in anti-viral responses to fish viruses. In this study, we used the transcription activator-like effector nuclease (TALEN) as a gene editing tool to disrupt the function of MAVS in Chinook salmon (Oncorhynchus tshawytscha) embryonic cells (CHSE) to understand its role in induction of interferon I responses to infections with the (+) RNA virus salmonid alphavirus subtype 3 (SAV-3), and the dsRNA virus infectious pancreatic necrosis virus (IPNV) infection. A MAVS-disrupted CHSE clone with a 7-aa polypeptide (GVFVSRV) deletion mutation at the N-terminal of the CARD domain infected with SAV-3 resulted in significantly lower expression of IRF3, IFNa, and ISGs and increased viral titer (1.5 log10) compared to wild-type. In contrast, the IPNV titer in MAVS-disrupted cells was not different from the wild-type. Furthermore, overexpression of salmon MAVS in MAVS-disrupted CHSE cells rescued the impaired type I IFN-mediated anti-viral effect against SAV-3.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Infecciones por Alphavirus , Alphavirus , Enfermedades de los Peces , Virus de la Necrosis Pancreática Infecciosa , Transducción de Señal , Replicación Viral , Animales , Virus de la Necrosis Pancreática Infecciosa/fisiología , Virus de la Necrosis Pancreática Infecciosa/inmunología , Alphavirus/inmunología , Alphavirus/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/virología , Salmón/virología , Salmón/inmunología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Proteínas de Peces/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Infecciones por Birnaviridae/inmunología , Infecciones por Birnaviridae/veterinaria , Infecciones por Birnaviridae/virología
6.
Expert Opin Ther Targets ; 28(5): 345-356, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38714500

RESUMEN

INTRODUCTION: Mayaro fever is an emerging viral disease that manifests as an acute febrile illness. The disease is self-limiting, however joint pain can persist for months leading to chronic arthralgia. There is no specific treatment available, which ultimately leads to socioeconomic losses in populations at risk as well as strains to the public health systems. AREAS COVERED: We reviewed the candidate treatments proposed for Mayaro virus (MAYV) infection and disease, including antiviral compounds targeting viral or host mechanisms, and pathways involved in disease development and pathogenicity. We assessed compound screening technologies and experimental infection models used in these studies and indicated the advantages and limitations of available technologies and intended therapeutic strategies. EXPERT OPINION: Although several compounds have been suggested as candidate treatments against MAYV infection, notably those with antiviral activity, most compounds were assessed only in vitro. Compounds rarely progress toin vivo or preclinical studies, and such difficulty may be associated with limited experimental models. MAYV biology is largely inferred from related alphaviruses and reflected by few studies focusing on target proteins or mechanisms of action for MAYV. Therapeutic strategies targeting pathogenic inflammatory responses have shown potential against MAYV-induced disease in vivo, which might reduce long-term sequelae.


Asunto(s)
Infecciones por Alphavirus , Antivirales , Descubrimiento de Drogas , Animales , Antivirales/farmacología , Humanos , Infecciones por Alphavirus/tratamiento farmacológico , Infecciones por Alphavirus/virología , Alphavirus , Artralgia/tratamiento farmacológico , Desarrollo de Medicamentos , Terapia Molecular Dirigida , Modelos Animales de Enfermedad
7.
mBio ; 15(6): e0042024, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38700353

RESUMEN

Chikungunya virus (CHIKV) is an enveloped, positive-sense RNA virus that has re-emerged to cause millions of human infections worldwide. In humans, acute CHIKV infection causes fever and severe muscle and joint pain. Chronic and debilitating arthritis and joint pain can persist for months to years. To date, there are no approved antivirals against CHIKV. Recently, the ribonucleoside analog 4'-fluorouridine (4'-FlU) was reported as a highly potent orally available inhibitor of SARS-CoV-2, respiratory syncytial virus, and influenza virus replication. In this study, we assessed 4'-FlU's potency and breadth of inhibition against a panel of alphaviruses including CHIKV, and found that it broadly suppressed alphavirus production in cell culture. 4'-FlU acted on the viral RNA replication step, and the first 4 hours post-infection were the critical time for its antiviral effect. In vitro replication assays identified nsP4 as the target of inhibition. In vivo, treatment with 4'-FlU reduced disease signs, inflammatory responses, and viral tissue burden in mouse models of CHIKV and Mayaro virus infection. Treatment initiated at 2 hours post-infection was most effective; however, treatment initiated as late as 24-48 hours post-infection produced measurable antiviral effects in the CHIKV mouse model. 4'-FlU showed effective oral delivery in our mouse model and resulted in the accumulation of both 4'-FlU and its bioactive triphosphate form in tissues relevant to arthritogenic alphavirus pathogenesis. Together, our data indicate that 4'-FlU inhibits CHIKV infection in vitro and in vivo and is a promising oral therapeutic candidate against CHIKV infection.IMPORTANCEAlphaviruses including chikungunya virus (CHIKV) are mosquito-borne positive-strand RNA viruses that can cause various diseases in humans. Although compounds that inhibit CHIKV and other alphaviruses have been identified in vitro, there are no licensed antivirals against CHIKV. Here, we investigated a ribonucleoside analog, 4'-fluorouridine (4'-FlU), and demonstrated that it inhibited infectious virus production by several alphaviruses in vitro and reduced virus burden in mouse models of CHIKV and Mayaro virus infection. Our studies also indicated that 4'-FlU treatment reduced CHIKV-induced footpad swelling and reduced the production of pro-inflammatory cytokines. Inhibition in the mouse model correlated with effective oral delivery of 4'-FlU and accumulation of both 4'-FlU and its bioactive form in relevant tissues. In summary, 4'-FlU exhibits potential as a novel anti-alphavirus agent targeting the replication of viral RNA.


Asunto(s)
Alphavirus , Antivirales , Virus Chikungunya , Replicación Viral , Replicación Viral/efectos de los fármacos , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Ratones , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/fisiología , Alphavirus/efectos de los fármacos , Alphavirus/fisiología , Uridina/análogos & derivados , Uridina/farmacología , Humanos , Fiebre Chikungunya/tratamiento farmacológico , Fiebre Chikungunya/virología , Modelos Animales de Enfermedad , Línea Celular , Chlorocebus aethiops , Femenino , Células Vero
8.
Viruses ; 16(5)2024 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-38793690

RESUMEN

The Mayaro virus (MAYV) is an arbovirus with emerging potential, though with a limited understanding of its epidemiology and evolution due to the lack of studies and surveillance. Here, we investigated 71 MAYV genome sequences from the Americas available at GenBank and characterized the phylogenetic relationship among virus strains. A phylogenetic analysis showed that sequences were grouped according to the genotypes L, D, and N. Genotype D sequences were closely related to sequences collected in adjacent years and from their respective countries, suggesting that isolates may have originated from circulating lineages. The coalescent analysis demonstrated similar results, indicating the continuous circulation of the virus between countries as well. An unidentified sequence from the USA was grouped with genotype D, suggesting the insertion of this genotype in the country. Furthermore, the recombination analysis detected homologous and three heterologous hybrids which presented an insertion into the nsP3 protein. Amino acid substitutions among sequences indicated selective pressure sites, suggesting viral adaptability. This also impacted the binding affinity between the E1-E2 protein complex and the Mxra8 receptor, associated with MAYV entry into human cells. These results provide information for a better understanding of genotypes circulating in the Americas.


Asunto(s)
Evolución Molecular , Variación Genética , Genoma Viral , Genotipo , Filogenia , Américas/epidemiología , Humanos , Alphavirus/genética , Alphavirus/clasificación , Alphavirus/aislamiento & purificación , Animales , Recombinación Genética , Infecciones por Alphavirus/virología , Infecciones por Alphavirus/epidemiología
9.
Dev Comp Immunol ; 157: 105193, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38729458

RESUMEN

The development and persistence of antibody secreting cells (ASC) after antigenic challenge remain inadequately understood in teleosts. In this study, intraperitoneal (ip) injection of Atlantic salmon (Salmo salar) with salmonid alphavirus (WtSAV3) increased the total ASC response, peaking 3-6 weeks post injection (wpi) locally in the peritoneal cavity (PerC) and in systemic lymphoid tissues, while at 13 wpi the response was only elevated in PerC. At the same time point a specific ASC response was induced by WtSAV3 in PerC and systemic tissues, with the highest frequency in PerC, suggesting a local role. Inactivated SAV (InSAV1) induced comparatively lower ASC responses in all sites, and specific serum antibodies were only induced by WtSAV3 and not by InSAV1. An InSAV1 boost did not increase these responses. Expression of immune marker genes implies a role for PerC adipose tissue in the PerC immune response. Overall, the study suggests the Atlantic salmon PerC as a secondary immune site and an ASC survival niche.


Asunto(s)
Infecciones por Alphavirus , Alphavirus , Anticuerpos Antivirales , Células Productoras de Anticuerpos , Enfermedades de los Peces , Cavidad Peritoneal , Salmo salar , Animales , Salmo salar/inmunología , Salmo salar/virología , Alphavirus/inmunología , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/veterinaria , Infecciones por Alphavirus/virología , Cavidad Peritoneal/citología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Células Productoras de Anticuerpos/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Inyecciones Intraperitoneales/veterinaria
10.
Parasit Vectors ; 17(1): 200, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704595

RESUMEN

BACKGROUND: Mayaro virus (MAYV) is an emerging alphavirus, primarily transmitted by the mosquito Haemagogus janthinomys in Central and South America. However, recent studies have shown that Aedes aegypti, Aedes albopictus and various Anopheles mosquitoes can also transmit the virus under laboratory conditions. MAYV causes sporadic outbreaks across the South American region, particularly in areas near forests. Recently, cases have been reported in European and North American travelers returning from endemic areas, raising concerns about potential introductions into new regions. This study aims to assess the vector competence of three potential vectors for MAYV present in Europe. METHODS: Aedes albopictus from Italy, Anopheles atroparvus from Spain and Culex pipiens biotype molestus from Belgium were exposed to MAYV and maintained under controlled environmental conditions. Saliva was collected through a salivation assay at 7 and 14 days post-infection (dpi), followed by vector dissection. Viral titers were determined using focus forming assays, and infection rates, dissemination rates, and transmission efficiency were calculated. RESULTS: Results indicate that Ae. albopictus and An. atroparvus from Italy and Spain, respectively, are competent vectors for MAYV, with transmission possible starting from 7 dpi under laboratory conditions. In contrast, Cx. pipiens bioform molestus was unable to support MAYV infection, indicating its inability to contribute to the transmission cycle. CONCLUSIONS: In the event of accidental MAYV introduction in European territories, autochthonous outbreaks could potentially be sustained by two European species: Ae. albopictus and An. atroparvus. Entomological surveillance should also consider certain Anopheles species when monitoring MAYV transmission.


Asunto(s)
Aedes , Infecciones por Alphavirus , Alphavirus , Culex , Mosquitos Vectores , Animales , Aedes/virología , Mosquitos Vectores/virología , Alphavirus/fisiología , Alphavirus/aislamiento & purificación , Culex/virología , Europa (Continente) , Infecciones por Alphavirus/transmisión , Infecciones por Alphavirus/virología , Saliva/virología , Anopheles/virología , España , Italia , Femenino , Bélgica
11.
Viruses ; 16(4)2024 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-38675846

RESUMEN

Replicating RNA, including self-amplifying RNA (saRNA) and trans-amplifying RNA (taRNA), holds great potential for advancing the next generation of RNA-based vaccines. Unlike in vitro transcribed mRNA found in most current RNA vaccines, saRNA or taRNA can be massively replicated within cells in the presence of RNA-amplifying enzymes known as replicases. We recently demonstrated that this property could enhance immune responses with minimal injected RNA amounts. In saRNA-based vaccines, replicase and antigens are encoded on the same mRNA molecule, resulting in very long RNA sequences, which poses significant challenges in production, delivery, and stability. In taRNA-based vaccines, these challenges can be overcome by splitting the replication system into two parts: one that encodes replicase and the other that encodes a short antigen-encoding RNA called transreplicon. Here, we review the identification and use of transreplicon RNA in alphavirus research, with a focus on the development of novel taRNA technology as a state-of-the art vaccine platform. Additionally, we discuss remaining challenges essential to the clinical application and highlight the potential benefits related to the unique properties of this future vaccine platform.


Asunto(s)
Alphavirus , ARN Viral , Desarrollo de Vacunas , Animales , Humanos , Alphavirus/genética , Alphavirus/inmunología , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/virología , ARN Viral/genética , Vacunas Virales/inmunología , Vacunas Virales/genética , Replicación Viral
13.
J Fish Dis ; 47(7): e13950, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38555528

RESUMEN

Disease interactions between farmed and wild populations have been poorly documented for most aquaculture species, in part due to the complexities to study this. Here, we tested 567 farmed Atlantic salmon escapees, captured in a Norwegian river during 2014-2018, for five viral infections that are prevalent in global salmonid aquaculture. Over 90% of the escapees were infected with one or more viruses. Overall prevalences were: 75.7% for piscine orthoreovirus (PRV-1), 43.6% for salmonid alphavirus (SAV), 31.2% for piscine myocarditis virus (PMCV), 1.2% for infectious pancreatic necrosis virus (IPNV) and 0.4% for salmon anaemia virus (ISAV). A significantly higher prevalence of PMCV infection was observed in immature compared to mature individuals. The prevalence of both SAV and PMCV infections was higher in fish determined by fatty acid profiling to be 'recent' as opposed to 'early' escapees that had been in the wild for a longer period of time. This is the first study to establish a time-series of viral infection status of escapees entering a river with a native salmon population. Our results demonstrate that farmed escapees represent a continuous source of infectious agents which could potentially be transmitted to wild fish populations.


Asunto(s)
Acuicultura , Enfermedades de los Peces , Ríos , Salmo salar , Animales , Enfermedades de los Peces/virología , Enfermedades de los Peces/epidemiología , Noruega/epidemiología , Prevalencia , Alphavirus/aislamiento & purificación , Alphavirus/fisiología , Infecciones por Alphavirus/veterinaria , Infecciones por Alphavirus/epidemiología , Infecciones por Alphavirus/virología
14.
Vaccine ; 42(10): 2695-2706, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38494412

RESUMEN

BACKGROUND: Three encephalitic alphaviruses-western, eastern, and Venezuelan equine encephalitis virus (WEEV, EEEV and VEEV)-can cause severe disease and have the potential to be used as biological weapons. There are no approved vaccines for human use. A novel multivalent MVA-BN-WEV vaccine encodes the envelope surface proteins of the 3 viruses and is thereby potentially able to protect against them all, as previously demonstrated in animal models. This first-in-human study assessed the safety, tolerability, and immunogenicity of MVA-BN-WEV vaccine in healthy adult participants. METHODS: Forty-five participants were enrolled into 3 dose groups (1 × 10E7 Inf.U, 1 × 10E8 Inf.U, and 2 × 10E8 Inf.U), received 2 doses 4 weeks apart, and were then monitored for 6 months. RESULTS: The safety profile of MVA-BN-WEV was acceptable at all administered doses, with incidence of local solicited AEs increased with increasing dose and no other clinically meaningful differences between dose groups. One SAE (Grade 2 pleural effusion) was reported in the lowest dose group and assessed as possibly related. No AEs resulted in death or led to withdrawal from the second vaccination or from the trial. The most common local solicited AE was injection site pain, and general solicited AEs were headache, fatigue, and myalgia. MVA-BN-WEV induced humoral immune responses; WEEV-, EEEV- and VEEV-specific neutralizing antibody responses peaked 2 weeks following the second vaccination, and the magnitude of these responses increased with dose escalation. The highest dose resulted in seroconversion of all (100 %) participants for WEEV and VEEV and 92.9 % for EEEV, 2 weeks following second vaccination, and durability was observed for 6 months. MVA-BN-WEV induced cellular immune responses to VEEV E1 and E2 (EEEV and WEEV not tested) and a dose effect for peptide pool E2. CONCLUSION: The study demonstrated that MVA-BN-WEV is well tolerated, induces immune responses, and is suitable for further development. CLINICAL TRIAL REGISTRY NUMBER: NCT04131595.


Asunto(s)
Alphavirus , Virus de la Encefalitis Equina Venezolana , Encefalomielitis Equina , Humanos , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Encefalomielitis Equina/prevención & control , Inmunogenicidad Vacunal , Virus Vaccinia
15.
Med Vet Entomol ; 38(2): 234-243, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38489505

RESUMEN

Mayaro virus (MAYV; Alphavirus: Togaviridae) is an emerging pathogen in Latin America, causing fever and polyarthritis. Sporadic outbreaks of MAYV have occurred in the region, with reported human cases being imported to Europe and North America. Although primarily a risk for those residing in the Amazon basin's tropical forests, recent reports highlight that urbanization would increase the risk of MAYV transmission in Latin America. Urban emergence depends on human susceptibility and the ability of mosquitos like Aedes aegypti  (Linnaeus, 1762) (Diptera: Culicidae) to transmit MAYV. Despite the absence of active MAYV transmission in Argentine, the risk of introduction is substantial due to human movement and the presence of Ae. aegypti in the region. This study aimed to evaluate the susceptibility of different Argentine Ae. aegypti populations to MAYV genotype L (MAYV-L) using dose-response assays and determine barriers to virus infection, dissemination and transmission. Immature mosquito stages were collected in Buenos Aires, Córdoba and Rosario cities. Female Ae. aegypti (F2) were orally infected by feeding on five concentrations of MAYV-L, ranging from 1.0 to 6.0 log10 PFU/mL. Abdomens, legs and saliva were analysed using viral plaque assays. Results revealed that MAYV-L between infection and dissemination were associated with viral doses rather than the population origin. Infection rates varied between 3% and 65%, with a 50% infectious dose >5.5 log10 PFU/mL. Dissemination occurred at 39%, with a 50% dissemination dose of ~6.0 log10 PFU/mL. Dissemination among infected mosquitoes ranged from 60% to 86%, and transmission from disseminated mosquitoes ranged from 11% to 20%. Argentine Ae. aegypti populations exhibited a need for higher viral doses of MAYV-L than those typically found in humans to become infected. In addition, only a small proportion of infected mosquitoes were capable of transmitting the virus. Understanding MAYV transmission in urban areas is crucial for public health interventions.


Asunto(s)
Aedes , Alphavirus , Mosquitos Vectores , Animales , Aedes/virología , Aedes/fisiología , Argentina , Mosquitos Vectores/virología , Mosquitos Vectores/fisiología , Alphavirus/fisiología , Femenino , Infecciones por Alphavirus/transmisión , Larva/virología , Larva/crecimiento & desarrollo
16.
Int J Biol Macromol ; 265(Pt 1): 130847, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38490381

RESUMEN

Getah virus (GETV) belongs to the Alphavirus genus in the Togaviridae family and is a zoonotic arbovirus causing disease in both humans and animals. The capsid protein (CP) of GETV regulates the viral core assembly, but the mechanism underlying this process is poorly understood. In this study, we demonstrate that CP undergoes liquid-liquid phase separation (LLPS) with the GETV genome RNA (gRNA) in vitro and forms cytoplasmic puncta in cells. Two regions of GETV gRNA (nucleotides 1-4000 and 5000-8000) enhance CP droplet formation in vitro and the lysine-rich Link region of CP is essential for its phase separation. CP(K/R) mutant with all lysines in the Link region replaced by arginines exhibits improved LLPS versus wild type (WT) CP, but CP(K/E) mutant with lysines substituted by glutamic acids virtually loses condensation ability. Consistently, recombinant virus mutant with CP(K/R) possesses significantly higher gRNA binding affinity, virion assembly efficiency and infectivity than the virus with WT-CP. Overall, our findings provide new insights into the understanding of GETV assembly and development of new antiviral drugs against alphaviruses.


Asunto(s)
Alphavirus , Animales , Humanos , Alphavirus/genética , Alphavirus/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , ARN Viral/genética , ARN Guía de Sistemas CRISPR-Cas , Genómica , Virión/genética
17.
Front Immunol ; 15: 1342816, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38515753

RESUMEN

Salmonid alphavirus (SAV) causes pancreas disease (PD), which negatively impacts farmed Atlantic salmon. In this study, fish were vaccinated with a DNA-PD vaccine (DNA-PD) and an oil-adjuvanted, inactivated whole virus PD vaccine (Oil-PD). Controls were two non-PD vaccinated groups. Fish were kept in one tank and challenged by cohabitation with SAV genotype 2 in seawater. Protection against infection and mortality was assessed for 84 days (Efficacy study). Nineteen days post challenge (dpc), subgroups of fish from all treatment groups were transferred to separate tanks and cohabited with naïve fish (Transmission study 1) or fish vaccinated with a homologous vaccine (Transmission study 2), to evaluate virus transmission for 26 days (47 dpc). Viremia, heart RT-qPCR and histopathological scoring of key organs affected by PD were used to measure infection levels. RT-droplet digital PCR quantified shedding of SAV into water for transmission studies. The Efficacy study showed that PD associated growth-loss was significantly lower and clearance of SAV2 RNA significantly higher in the PD-DNA group compared to the other groups. The PD-DNA group had milder lesions in the heart and muscle. Cumulative mortality post challenge was low and not different between groups, but the DNA-PD group had delayed time-to-death. In Transmission study 1, the lowest water levels of SAV RNA were measured in the tanks containing the DNA-PD group at 21 and 34 dpc. Despite this, and irrespective of the treatment group, SAV2 was effectively transmitted to the naïve fish during 26-day cohabitation. At 47 dpc, the SAV RNA concentrations in the water were lower in all tanks compared to 34 dpc. In Transmission study 2, none of the DNA-PD immunized cohabitants residing with DNA-PD-vaccinated, pre-challenged fish got infected. In contrast, Oil-PD immunized cohabitants residing with Oil-PD-vaccinated, pre-challenged fish, showed infection levels similar to the naïve cohabitants in Transmission study 1. The results demonstrate that the DNA-PD vaccine may curb the spread of SAV infection as the DNA-PD vaccinated, SAV2 exposed fish, did not spread the infection to cohabiting DNA-PD vaccinated fish. This signifies that herd immunity may be achieved by the DNA-PD vaccine, a valuable tool to control the PD epizootic in farmed Atlantic salmon.


Asunto(s)
Alphavirus , Enfermedades de los Peces , Enfermedades Pancreáticas , Salmo salar , Vacunas de ADN , Vacunas Virales , Animales , Enfermedades Pancreáticas/veterinaria , Enfermedades Pancreáticas/patología , ARN/genética , Agua , Páncreas/patología , ADN , Genotipo
18.
Rev Soc Bras Med Trop ; 57: e004032024, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38536999

RESUMEN

BACKGROUND: The riverine communities of the Amazon comprise different social groups that inhabit the rural areas on the banks of rivers and lakes. Residents usually travel by river to rural and urban areas and are then exposed to urbanized diseases such as those caused by arbovirus infection. In Brazil, emerging diseases such as dengue, Zika, chikungunya, and those caused by infection with Oropouche and Mayaro viruses necessitate epidemiological surveillance. This study was aimed at determining the frequency of positivity for immunoglobulin (Ig)G and IgM antibodies against Zika, chikungunya, and dengue viruses and performing molecular analyses to detect viral RNA for the Zika, chikungunya, dengue virus, Oropouche, and Mayaro viruses, in the same serum samples obtained from riverside populations. METHODS: This cross-sectional study was conducted in a riverside population in the Humaitá municipality of the Brazilian Amazon. More than 80% of the local population participated in this study. Entomological samples were collected to identify local mosquito vectors. RESULTS: Analysis of 205 human serological samples revealed IgG antibodies against the dengue virus in 85 individuals. No molecular positivity was observed in human samples. Entomological analyses revealed 3,187 Diptera species, with Mansonia being the most frequent genus. Aedes aegypti and Aedes albopictus were not detected in the two collections. CONCLUSIONS: IgG antibodies against the dengue virus were highly prevalent, suggesting previous exposure. The absence of the arbovirus vectors Aedes aegypti and Aedes albopictus in the samples supports the hypothesis that the infections recorded likely occurred outside the riverside communities investigated.


Asunto(s)
Aedes , Alphavirus , Infecciones por Arbovirus , Fiebre Chikungunya , Dengue , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Fiebre Chikungunya/epidemiología , Brasil/epidemiología , Estudios Transversales , Infecciones por Arbovirus/epidemiología , Mosquitos Vectores , Inmunoglobulina G
19.
Viruses ; 16(2)2024 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-38399982

RESUMEN

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


Asunto(s)
Alphavirus , Virus de la Encefalitis Equina del Este , Encefalomielitis Equina , Fenoles , Caballos , Animales , Virus de la Encefalitis Equina del Este/fisiología , Guanidinas , Formaldehído
20.
Viruses ; 16(2)2024 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-38399981

RESUMEN

Alphaviruses can replicate in arthropods and in many vertebrate species including humankind, but only in vertebrate cells do infections with these viruses result in a strong inhibition of host translation and transcription. Translation shutoff by alphaviruses is a multifactorial process that involves both host- and virus-induced mechanisms, and some of them are not completely understood. Alphavirus genomes contain cis-acting elements (RNA structures and dinucleotide composition) and encode protein activities that promote the translational and transcriptional resistance to type I IFN-induced antiviral effectors. Among them, IFIT1, ZAP and PKR have played a relevant role in alphavirus evolution, since they have promoted the emergence of multiple viral evasion mechanisms at the translational level. In this review, we will discuss how the adaptations of alphaviruses to vertebrate hosts likely involved the acquisition of new features in viral mRNAs and proteins to overcome the effect of type I IFN.


Asunto(s)
Alphavirus , Interferón Tipo I , Animales , Alphavirus/fisiología , Línea Celular , Interferón Tipo I/genética , Vertebrados , Tropismo , Antivirales/farmacología , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...