Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.014
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(2): 360-374.e19, 2024 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-38176410

RESUMEN

The very-low-density lipoprotein receptor (VLDLR) comprises eight LDLR type A (LA) domains and supports entry of distantly related alphaviruses, including Eastern equine encephalitis virus (EEEV) and Semliki Forest virus (SFV). Here, by resolving multiple cryo-electron microscopy structures of EEEV-VLDLR complexes and performing mutagenesis and functional studies, we show that EEEV uses multiple sites (E1/E2 cleft and E2 A domain) to engage more than one LA domain simultaneously. However, no single LA domain is necessary or sufficient to support efficient EEEV infection. Whereas all EEEV strains show conservation of two VLDLR-binding sites, the EEEV PE-6 strain and a few other EEE complex members feature a single amino acid substitution that enables binding of LA domains to an additional site on the E2 B domain. These structural and functional analyses informed the design of a minimal VLDLR decoy receptor that neutralizes EEEV infection and protects mice from lethal challenge.


Asunto(s)
Microscopía por Crioelectrón , Virus de la Encefalitis Equina del Este , Encefalomielitis Equina , Receptores de LDL , Animales , Ratones , Alphavirus/fisiología , Virus de la Encefalitis Equina del Este/fisiología , Virus de la Encefalitis Equina del Este/ultraestructura , Encefalomielitis Equina/metabolismo , Caballos , Unión Proteica , Receptores de LDL/ultraestructura
2.
Cell ; 184(17): 4430-4446.e22, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34416147

RESUMEN

Alphaviruses cause severe arthritogenic or encephalitic disease. The E1 structural glycoprotein is highly conserved in these viruses and mediates viral fusion with host cells. However, the role of antibody responses to the E1 protein in immunity is poorly understood. We isolated E1-specific human monoclonal antibodies (mAbs) with diverse patterns of recognition for alphaviruses (ranging from Eastern equine encephalitis virus [EEEV]-specific to alphavirus cross-reactive) from survivors of natural EEEV infection. Antibody binding patterns and epitope mapping experiments identified differences in E1 reactivity based on exposure of epitopes on the glycoprotein through pH-dependent mechanisms or presentation on the cell surface prior to virus egress. Therapeutic efficacy in vivo of these mAbs corresponded with potency of virus egress inhibition in vitro and did not require Fc-mediated effector functions for treatment against subcutaneous EEEV challenge. These studies reveal the molecular basis for broad and protective antibody responses to alphavirus E1 proteins.


Asunto(s)
Alphavirus/inmunología , Anticuerpos Antivirales/inmunología , Reacciones Cruzadas/inmunología , Proteínas Virales/inmunología , Liberación del Virus/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Neutralizantes/inmunología , Antígenos Virales/inmunología , Línea Celular , Virus Chikungunya/inmunología , Virus de la Encefalitis Equina del Este/inmunología , Encefalomielitis Equina/inmunología , Encefalomielitis Equina/virología , Mapeo Epitopo , Femenino , Caballos , Humanos , Concentración de Iones de Hidrógeno , Articulaciones/patología , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Unión Proteica , ARN Viral/metabolismo , Receptores Fc/metabolismo , Temperatura , Virión/metabolismo , Internalización del Virus
3.
Cell ; 183(7): 1884-1900.e23, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33301709

RESUMEN

Eastern equine encephalitis virus (EEEV) is one of the most virulent viruses endemic to North America. No licensed vaccines or antiviral therapeutics are available to combat this infection, which has recently shown an increase in human cases. Here, we characterize human monoclonal antibodies (mAbs) isolated from a survivor of natural EEEV infection with potent (<20 pM) inhibitory activity of EEEV. Cryo-electron microscopy reconstructions of two highly neutralizing mAbs, EEEV-33 and EEEV-143, were solved in complex with chimeric Sindbis/EEEV virions to 7.2 Å and 8.3 Å, respectively. The mAbs recognize two distinct antigenic sites that are critical for inhibiting viral entry into cells. EEEV-33 and EEEV-143 protect against disease following stringent lethal aerosol challenge of mice with highly pathogenic EEEV. These studies provide insight into the molecular basis for the neutralizing human antibody response against EEEV and can facilitate development of vaccines and candidate antibody therapeutics.


Asunto(s)
Aerosoles/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Virus de la Encefalitis Equina del Este/inmunología , Encefalomielitis Equina/inmunología , Encefalomielitis Equina/prevención & control , Adulto , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Neutralizantes/inmunología , Antígenos Virales/inmunología , Microscopía por Crioelectrón , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina del Este/ultraestructura , Encefalomielitis Equina/virología , Epítopos/química , Femenino , Glicoproteínas/inmunología , Humanos , Ratones , Modelos Moleculares , Mutagénesis/genética , Pruebas de Neutralización , Unión Proteica , Dominios Proteicos , Proteínas Recombinantes/inmunología , Virus Sindbis/inmunología , Virión/inmunología , Virión/ultraestructura , Internalización del Virus
4.
Proc Natl Acad Sci U S A ; 120(13): e2213690120, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36961925

RESUMEN

Selection and development of monoclonal antibody (mAb) therapeutics against pathogenic viruses depends on certain functional characteristics. Neutralization potency, or the half-maximal inhibitory concentration (IC50) values, is an important characteristic of candidate therapeutic antibodies. Structural insights into the bases of neutralization potency differences between antiviral neutralizing mAbs are lacking. In this report, we present cryo-electron microscopy (EM) reconstructions of three anti-Eastern equine encephalitis virus (EEEV) neutralizing human mAbs targeting overlapping epitopes on the E2 protein, with greater than 20-fold differences in their respective IC50 values. From our structural and biophysical analyses, we identify several constraints that contribute to the observed differences in the neutralization potencies. Cryo-EM reconstructions of EEEV in complex with these Fab fragments reveal structural constraints that dictate intravirion or intervirion cross-linking of glycoprotein spikes by their IgG counterparts as a mechanism of neutralization. Additionally, we describe critical features for the recognition of EEEV by these mAbs including the epitope-paratope interaction surface, occupancy, and kinetic differences in on-rate for binding to the E2 protein. Each constraint contributes to the extent of EEEV inhibition for blockade of virus entry, fusion, and/or egress. These findings provide structural and biophysical insights into the differences in mechanism and neutralization potencies of these antibodies, which help inform rational design principles for candidate vaccines and therapeutic antibodies for all icosahedral viruses.


Asunto(s)
Virus de la Encefalitis Equina del Este , Encefalomielitis Equina , Humanos , Caballos , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Microscopía por Crioelectrón , Epítopos , Anticuerpos Monoclonales , Pruebas de Neutralización
5.
Proc Natl Acad Sci U S A ; 117(16): 8890-8899, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32245806

RESUMEN

Eastern equine encephalitis virus (EEEV), a mosquito-borne icosahedral alphavirus found mainly in North America, causes human and equine neurotropic infections. EEEV neurovirulence is influenced by the interaction of the viral envelope protein E2 with heparan sulfate (HS) proteoglycans from the host's plasma membrane during virus entry. Here, we present a 5.8-Å cryoelectron microscopy (cryo-EM) structure of EEEV complexed with the HS analog heparin. "Peripheral" HS binding sites were found to be associated with the base of each of the E2 glycoproteins that form the 60 quasi-threefold spikes (q3) and the 20 sites associated with the icosahedral threefold axes (i3). In addition, there is one HS site at the vertex of each q3 and i3 spike (the "axial" sites). Both the axial and peripheral sites are surrounded by basic residues, suggesting an electrostatic mechanism for HS binding. These residues are highly conserved among EEEV strains, and therefore a change in these residues might be linked to EEEV neurovirulence.


Asunto(s)
Diseño de Fármacos , Virus de la Encefalitis Equina del Este/ultraestructura , Encefalomielitis Equina/tratamiento farmacológico , Proteoglicanos de Heparán Sulfato/metabolismo , Heparina/ultraestructura , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Sitios de Unión/efectos de los fármacos , Línea Celular , Sulfatos de Condroitina/farmacología , Microscopía por Crioelectrón , Virus de la Encefalitis Equina del Este/metabolismo , Encefalomielitis Equina/virología , Proteoglicanos de Heparán Sulfato/análogos & derivados , Heparina/metabolismo , Humanos , Mesocricetus , Estructura Molecular , Relación Estructura-Actividad , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/ultraestructura , Acoplamiento Viral/efectos de los fármacos
6.
PLoS Pathog ; 16(2): e1008102, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32027727

RESUMEN

Understanding the circumstances under which arboviruses emerge is critical for the development of targeted control and prevention strategies. This is highlighted by the emergence of chikungunya and Zika viruses in the New World. However, to comprehensively understand the ways in which viruses emerge and persist, factors influencing reductions in virus activity must also be understood. Western equine encephalitis virus (WEEV), which declined during the late 20th century in apparent enzootic circulation as well as equine and human disease incidence, provides a unique case study on how reductions in virus activity can be understood by studying evolutionary trends and mechanisms. Previously, we showed using phylogenetics that during this period of decline, six amino acid residues appeared to be positively selected. To assess more directly the effect of these mutations, we utilized reverse genetics and competition fitness assays in the enzootic host and vector (house sparrows and Culex tarsalis mosquitoes). We observed that the mutations contemporary with reductions in WEEV circulation and disease that were non-conserved with respect to amino acid properties had a positive effect on enzootic fitness. We also assessed the effects of these mutations on virulence in the Syrian-Golden hamster model in relation to a general trend of increased virulence in older isolates. However, no change effect on virulence was observed based on these mutations. Thus, while WEEV apparently underwent positive selection for infection of enzootic hosts, residues associated with mammalian virulence were likely eliminated from the population by genetic drift or negative selection. These findings suggest that ecologic factors rather than fitness for natural transmission likely caused decreased levels of enzootic WEEV circulation during the late 20th century.


Asunto(s)
Virus de la Encefalitis Equina del Oeste/genética , Encefalomielitis Equina/genética , Flujo Genético , Selección Genética , Animales , Culex/inmunología , Culex/virología , Virus de la Encefalitis Equina del Oeste/inmunología , Virus de la Encefalitis Equina del Oeste/patogenicidad , Encefalomielitis Equina/inmunología , Encefalomielitis Equina/patología , Encefalomielitis Equina/transmisión , Humanos , Mesocricetus , Mosquitos Vectores/inmunología , Mosquitos Vectores/virología , Gorriones/inmunología , Gorriones/virología
7.
Emerg Infect Dis ; 27(4): 1173-1176, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33754982

RESUMEN

During the 2019 Eastern equine encephalitis virus (EEEV) outbreak in Michigan, two 2-month old Mexican wolf pups experienced neurologic signs, lymphohistiocytic neutrophilic meningoencephalitis with neuronal necrosis and neuronophagia, and acute death. We identified EEEV by reverse transcription real-time PCR and in situ hybridization. Vector mosquitoes were trapped at the zoo.


Asunto(s)
Virus de la Encefalitis Equina del Este , Encefalomielitis Equina Oriental , Encefalomielitis Equina , Lobos , Animales , Caballos , Michigan , Mosquitos Vectores
8.
J Virol ; 94(17)2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32581106

RESUMEN

Eastern equine encephalitis virus (EEEV) is the most pathogenic member of the Alphavirus genus in the Togaviridae family. This virus continues to circulate in the New World and has a potential for deliberate use as a bioweapon. Despite the public health threat, to date no attenuated EEEV variants have been applied as live EEEV vaccines. Our previous studies demonstrated the critical function of the hypervariable domain (HVD) in EEEV nsP3 for the assembly of viral replication complexes (vRCs). EEEV HVD contains short linear motifs that recruit host proteins required for vRC formation and function. In this study, we developed a set of EEEV mutants that contained combinations of deletions in nsP3 HVD and clustered mutations in capsid protein, and tested the effects of these modifications on EEEV infection in vivo These mutations had cumulative negative effects on viral ability to induce meningoencephalitis. The deletions of two critical motifs, which interact with the members of cellular FXR and G3BP protein families, made EEEV cease to be neurovirulent. The additional clustered mutations in capsid protein, which affect its ability to induce transcriptional shutoff, diminished EEEV's ability to develop viremia. Most notably, despite the inability to induce detectable disease, the designed EEEV mutants remained highly immunogenic and, after a single dose, protected mice against subsequent infection with wild-type (wt) EEEV. Thus, alterations of interactions of EEEV HVD and likely HVDs of other alphaviruses with host factors represent an important direction for development of highly attenuated viruses that can be applied as live vaccines.IMPORTANCE Hypervariable domains (HVDs) of alphavirus nsP3 proteins recruit host proteins into viral replication complexes. The sets of HVD-binding host factors are specific for each alphavirus, and we have previously identified those specific for EEEV. The results of this study demonstrate that the deletions of the binding sites of the G3BP and FXR protein families in the nsP3 HVD of EEEV make the virus avirulent for mice. Mutations in the nuclear localization signal in EEEV capsid protein have an additional negative effect on viral replication in vivo Despite the inability to cause a detectable disease, the double HVD and triple HVD/capsid mutants induce high levels of neutralizing antibodies. Single immunization protects mice against infection with the highly pathogenic North American strain of EEEV. High safety, the inability to revert to wild-type phenotype, and high immunogenicity make the designed mutants attractive vaccine candidates for EEEV infection.


Asunto(s)
Virus de la Encefalitis Equina del Este/inmunología , Vacunas Atenuadas/inmunología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/inmunología , Factores de Virulencia/inmunología , Animales , Anticuerpos Neutralizantes , Sitios de Unión , Proteínas de la Cápside/genética , Línea Celular , Virus de la Encefalitis Equina del Este/genética , Virus de la Encefalitis Equina del Este/patogenicidad , Encefalomielitis Equina/inmunología , Encefalomielitis Equina/prevención & control , Ratones , Mutación , Proteínas no Estructurales Virales/genética , Virulencia/genética , Virulencia/inmunología , Factores de Virulencia/genética , Replicación Viral
9.
PLoS Pathog ; 15(10): e1007867, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31658290

RESUMEN

Eastern equine encephalitis virus (EEEV), a mosquito-borne RNA virus, is one of the most acutely virulent viruses endemic to the Americas, causing between 30% and 70% mortality in symptomatic human cases. A major factor in the virulence of EEEV is the presence of four binding sites for the hematopoietic cell-specific microRNA, miR-142-3p, in the 3' untranslated region (3' UTR) of the virus. Three of the sites are "canonical" with all 7 seed sequence residues complimentary to miR-142-3p while one is "non-canonical" and has a seed sequence mismatch. Interaction of the EEEV genome with miR-142-3p limits virus replication in myeloid cells and suppresses the systemic innate immune response, greatly exacerbating EEEV neurovirulence. The presence of the miRNA binding sequences is also required for efficient EEEV replication in mosquitoes and, therefore, essential for transmission of the virus. In the current studies, we have examined the role of each binding site by point mutagenesis of the seed sequences in all combinations of sites followed by infection of mammalian myeloid cells, mosquito cells and mice. The resulting data indicate that both canonical and non-canonical sites contribute to cell infection and animal virulence, however, surprisingly, all sites are rapidly deleted from EEEV genomes shortly after infection of myeloid cells or mice. Finally, we show that the virulence of a related encephalitis virus, western equine encephalitis virus, is also dependent upon miR-142-3p binding sites.


Asunto(s)
Regiones no Traducidas 3'/genética , Virus de la Encefalitis Equina del Este/genética , Virus de la Encefalitis Equina del Oeste/genética , MicroARNs/genética , Replicación Viral/genética , Aedes , Animales , Sitios de Unión/genética , Línea Celular , Cricetinae , Virus de la Encefalitis Equina del Este/inmunología , Virus de la Encefalitis Equina del Este/patogenicidad , Virus de la Encefalitis Equina del Oeste/inmunología , Virus de la Encefalitis Equina del Oeste/patogenicidad , Encefalomielitis Equina/inmunología , Encefalomielitis Equina/virología , Femenino , Inmunidad Innata/inmunología , Células L , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Células RAW 264.7 , Virulencia/genética
10.
Clin Infect Dis ; 69(3): 450-458, 2019 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-30371754

RESUMEN

BACKGROUND: In fall 2017, 3 solid organ transplant (SOT) recipients from a common donor developed encephalitis within 1 week of transplantation, prompting suspicion of transplant-transmitted infection. Eastern equine encephalitis virus (EEEV) infection was identified during testing of endomyocardial tissue from the heart recipient. METHODS: We reviewed medical records of the organ donor and transplant recipients and tested serum, whole blood, cerebrospinal fluid, and tissue from the donor and recipients for evidence of EEEV infection by multiple assays. We investigated blood transfusion as a possible source of organ donor infection by testing remaining components and serum specimens from blood donors. We reviewed data from the pretransplant organ donor evaluation and local EEEV surveillance. RESULTS: We found laboratory evidence of recent EEEV infection in all organ recipients and the common donor. Serum collected from the organ donor upon hospital admission tested negative, but subsequent samples obtained prior to organ recovery were positive for EEEV RNA. There was no evidence of EEEV infection among donors of the 8 blood products transfused into the organ donor or in products derived from these donations. Veterinary and mosquito surveillance showed recent EEEV activity in counties nearby the organ donor's county of residence. Neuroinvasive EEEV infection directly contributed to the death of 1 organ recipient and likely contributed to death in another. CONCLUSIONS: Our investigation demonstrated EEEV transmission through SOT. Mosquito-borne transmission of EEEV to the organ donor was the likely source of infection. Clinicians should be aware of EEEV as a cause of transplant-associated encephalitis.


Asunto(s)
Encefalomielitis Equina/transmisión , Donantes de Tejidos , Receptores de Trasplantes/estadística & datos numéricos , Trasplante/efectos adversos , Adulto , Animales , Culicidae/virología , Virus de la Encefalitis Equina del Este , Encefalomielitis Equina/sangre , Resultado Fatal , Femenino , Trasplante de Corazón/efectos adversos , Humanos , Trasplante de Hígado/efectos adversos , Trasplante de Pulmón/efectos adversos , Registros Médicos , Persona de Mediana Edad
11.
J Virol ; 92(12)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29618651

RESUMEN

Eastern equine encephalitis virus (EEEV) has a high case-fatality rate in horses and humans, and Florida has been hypothesized to be the source of EEEV epidemics for the northeastern United States. To test this hypothesis, we sequenced complete genomes of 433 EEEV strains collected within the United States from 1934 to 2014. Phylogenetic analysis suggested EEEV evolves relatively slowly and that transmission is enzootic in Florida, characterized by higher genetic diversity and long-term local persistence. In contrast, EEEV strains in New York and Massachusetts were characterized by lower genetic diversity, multiple introductions, and shorter local persistence. Our phylogeographic analysis supported a source-sink model in which Florida is the major source of EEEV compared to the other localities sampled. In sum, this study revealed the complex epidemiological dynamics of EEEV in different geographic regions in the United States and provided general insights into the evolution and transmission of other avian mosquito-borne viruses in this region.IMPORTANCE Eastern equine encephalitis virus (EEEV) infections are severe in horses and humans on the east coast of the United States with a >90% mortality rate in horses, an ∼33% mortality rate in humans, and significant brain damage in most human survivors. However, little is known about the evolutionary characteristics of EEEV due to the lack of genome sequences. By generating large collection of publicly available complete genome sequences, this study comprehensively determined the evolution of the virus, described the epidemiological dynamics of EEEV in different states in the United States, and identified Florida as one of the major sources. These results may have important implications for the control and prevention of other mosquito-borne viruses in the Americas.


Asunto(s)
Virus de la Encefalitis Equina del Este/clasificación , Encefalomielitis Equina/transmisión , Secuenciación Completa del Genoma/métodos , Animales , Virus de la Encefalitis Equina del Este/genética , Encefalomielitis Equina/epidemiología , Florida/epidemiología , Variación Genética , Tamaño del Genoma , Genoma Viral , Secuenciación de Nucleótidos de Alto Rendimiento , Caballos , Massachusetts/epidemiología , New York/epidemiología , Filogenia , Filogeografía
12.
Microb Pathog ; 132: 80-86, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31029717

RESUMEN

Madariaga Virus (MADV) is an emergent Alphavirus of the eastern equine encephalitis virus (EEEV) strain complex causing epizootic epidemics. In this study the genetic diversity and the transmission dynamics of Madariaga virus has been investigated by Bayesian phylogenetics and phylodynamic analysis. A database of 32 sequences of MADV group structural polyprotein were downloaded from GenBank, aligned manually edited by Bioedit Software. ModelTest v. 3.7 was used to select the simplest evolutionary model that adequately fitted the sequence data. Neighbor-joining tree was generated using MEGA7. The phylogenetic signal of the dataset was tested by the likelihood mapping analysis. The Bayesian phylogenetic tree was built using BEAST. Selective pressure analysis revealed one positive selection site. The phylogenetic trees showed two main clusters. In particular, Lineage II showed an epizootic infection in monkeys and Lineage III, including 2 main clusters (IIIa and IIIB), revealing an epizootic infection in humans in Haiti and an epizootic infection in humans in Venezuela during the 2016, respectively. The Bayesian maximum clade credibility tree and the time of the most common recent ancestor estimates, showed that the root of the tree dated back to the year 346 with the probable origin in Brazil. Gene flow analysis revealed viral exchanges between different neighbor countries of South America. In conclusion, Bayesian phylogenetic and phylodynamic represent useful tools to follow the transmission dynamic of emergent pathogens to prevent new epidemics spreading worldwide.


Asunto(s)
Virus de la Encefalitis Equina del Este/genética , Virus de la Encefalitis Equina del Este/patogenicidad , Encefalomielitis Equina/epidemiología , Encefalomielitis Equina/transmisión , Encefalomielitis Equina/virología , Filogenia , Infecciones por Alphavirus , Animales , Secuencia de Bases , Teorema de Bayes , Brasil , Virus de la Encefalitis Equina del Este/clasificación , Epidemias , Evolución Molecular , Flujo Génico , Variación Genética , Haití , Haplorrinos , Humanos , ARN Viral/genética , Alineación de Secuencia , América del Sur , Venezuela
13.
Mem Inst Oswaldo Cruz ; 114: e180332, 2019 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-30672980

RESUMEN

BACKGROUND: Serological evidence of West Nile virus (WNV) infection has been reported in different regions of Brazil from equine and human hosts but the virus had never been isolated in the country. OBJECTIVES: We sought to identify the viral etiology of equine encephalitis in Espírito Santo state. METHODS: We performed viral culture in C6/36 cells, molecular detection of WNV genome, histopathology and immunohistochemistry from horse cerebral tissue. We also carried out sequencing, phylogenetic analysis and molecular clock. FINDINGS: Histopathologic analysis from horse cerebral tissue showed injury related to encephalitis and WNV infection was confirmed by immunohistochemistry. The virus was detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR) from brain tissue and subsequently isolated in C6/36 cells. WNV full-length genome was sequenced showing the isolated strain belongs to lineage 1a. The molecular clock indicated that Brazilian WNV strain share the same common ancestor that were circulating in US during 2002-2005. MAIN CONCLUSIONS: Here we report the first isolation of WNV in Brazil from a horse with neurologic disease, which was clustered into lineage 1a with others US WNV strains isolated in beginning of 2000's decade.


Asunto(s)
Encefalomielitis Equina/veterinaria , Enfermedades de los Caballos/virología , Fiebre del Nilo Occidental/veterinaria , Virus del Nilo Occidental/genética , Animales , Brasil , Encefalomielitis Equina/virología , Enfermedades de los Caballos/diagnóstico , Caballos , Inmunohistoquímica , Masculino , Filogeografía , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Fiebre del Nilo Occidental/diagnóstico , Virus del Nilo Occidental/aislamiento & purificación
14.
J Virol ; 91(8)2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28148802

RESUMEN

The demonstrated clinical efficacy of a recombinant vesicular stomatitis virus (rVSV) vaccine vector has stimulated the investigation of additional serologically distinct Vesiculovirus vectors as therapeutic and/or prophylactic vaccine vectors to combat emerging viral diseases. Among these viral threats are the encephalitic alphaviruses Venezuelan equine encephalitis virus (VEEV) and Eastern equine encephalitis virus (EEEV), which have demonstrated potential for natural disease outbreaks, yet no licensed vaccines are available in the event of an epidemic. Here we report the rescue of recombinant Isfahan virus (rISFV) from genomic cDNA as a potential new vaccine vector platform. The rISFV genome was modified to attenuate virulence and express the VEEV and EEEV E2/E1 surface glycoproteins as vaccine antigens. A single dose of the rISFV vaccine vectors elicited neutralizing antibody responses and protected mice from lethal VEEV and EEEV challenges at 1 month postvaccination as well as lethal VEEV challenge at 8 months postvaccination. A mixture of rISFV vectors expressing the VEEV and EEEV E2/E1 glycoproteins also provided durable, single-dose protection from lethal VEEV and EEEV challenges, demonstrating the potential for a multivalent vaccine formulation. These findings were paralleled in studies with an attenuated form of rVSV expressing the VEEV E2/E1 glycoproteins. Both the rVSV and rISFV vectors were attenuated by using an approach that has demonstrated safety in human trials of an rVSV/HIV-1 vaccine. Vaccines based on either of these vaccine vector platforms may present a safe and effective approach to prevent alphavirus-induced disease in humans.IMPORTANCE This work introduces rISFV as a novel vaccine vector platform that is serologically distinct and phylogenetically distant from VSV. The rISFV vector has been attenuated by an approach used for an rVSV vector that has demonstrated safety in clinical studies. The vaccine potential of the rISFV vector was investigated in a well-established alphavirus disease model. The findings indicate the feasibility of producing a safe, efficacious, multivalent vaccine against the encephalitic alphaviruses VEEV and EEEV, both of which can cause fatal disease. This work also demonstrates the efficacy of an attenuated rVSV vector that has already demonstrated safety and immunogenicity in multiple HIV-1 phase I clinical studies. The absence of serological cross-reactivity between rVSV and rISFV and their phylogenetic divergence within the Vesiculovirus genus indicate potential for two stand-alone vaccine vector platforms that could be used to target multiple bacterial and/or viral agents in successive immunization campaigns or as heterologous prime-boost agents.


Asunto(s)
Portadores de Fármacos , Virus de la Encefalitis Equina del Este/inmunología , Virus de la Encefalitis Equina Venezolana/inmunología , Encefalomielitis Equina/prevención & control , Vesiculovirus/genética , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina del Este/genética , Virus de la Encefalitis Equina Venezolana/genética , Glicoproteínas/genética , Glicoproteínas/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Análisis de Supervivencia , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/genética
15.
J Vet Pharmacol Ther ; 41(2): 184-194, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28833212

RESUMEN

Triazines are relatively new antiprotozoal drugs that have successfully controlled coccidiosis and equine protozoal myeloencephalitis. These drugs have favorably treated other protozoal diseases such as neosporosis and toxoplasmosis. In this article, we discuss the pharmacological characteristics of five triazines, toltrazuril, ponazuril, clazuril, diclazuril, and nitromezuril which are used in veterinary medicine to control protozoal diseases which include coccidiosis, equine protozoal myeloencephalitis, neosporosis, and toxoplasmosis.


Asunto(s)
Antiprotozoarios/uso terapéutico , Infecciones Protozoarias en Animales/tratamiento farmacológico , Triazinas/uso terapéutico , Acetonitrilos/uso terapéutico , Animales , Coccidiosis/tratamiento farmacológico , Coccidiosis/veterinaria , Encefalomielitis Equina/tratamiento farmacológico , Encefalomielitis Equina/parasitología , Encefalomielitis Equina/veterinaria , Caballos , Nitrilos/uso terapéutico , Toxoplasmosis Animal/tratamiento farmacológico
17.
Epidemiol Infect ; 145(14): 2940-2948, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28956525

RESUMEN

Periodic outbreaks of West Nile virus (WNV), Eastern equine encephalitis virus (EEEV) and to a lesser extent, California serogroup viruses (CSGV), have been reported in parts of Canada in the last decade. This study was designed to provide a broad assessment of arboviral activity in Quebec, Canada, by conducting serological surveys for these arboviruses in 196 horses, 1442 dogs and 485 humans. Sera were screened by a competitive enzyme linked immunosorbent assay and positive samples confirmed by plaque reduction neutralisation tests. The percentage of seropositive samples was 83·7%, 16·5%, 7·1% in horses, 18·8%, 0·6%, 0% in humans, 11·7%, 3·1%, 0% in adult dogs and 2·9%, 0·3%, 0% in juvenile dogs for CSGV, WNV and EEEV, respectively. Serological results in horses and dogs appeared to provide a meaningful assessment of risk to public health posed by multiple arboviruses.


Asunto(s)
Infecciones por Arbovirus/epidemiología , Infecciones por Arbovirus/veterinaria , Enfermedades Transmisibles Emergentes/epidemiología , Adulto , Animales , Infecciones por Arbovirus/virología , Arbovirus/fisiología , Enfermedades Transmisibles Emergentes/virología , Enfermedades de los Perros/sangre , Enfermedades de los Perros/epidemiología , Enfermedades de los Perros/virología , Perros , Virus de la Encefalitis de California/fisiología , Virus de la Encefalitis Equina del Este/fisiología , Encefalitis de California/epidemiología , Encefalitis de California/virología , Encefalomielitis Equina/epidemiología , Encefalomielitis Equina/virología , Femenino , Enfermedades de los Caballos/sangre , Enfermedades de los Caballos/epidemiología , Enfermedades de los Caballos/virología , Caballos , Humanos , Masculino , Persona de Mediana Edad , Salud Pública , Quebec/epidemiología , Fiebre del Nilo Occidental/epidemiología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/fisiología
19.
J Neuroinflammation ; 13(1): 196, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27562117

RESUMEN

BACKGROUND: Alphaviruses can cause fatal encephalitis in humans. Natural infections occur via the bite of infected mosquitos, but aerosol transmissibility makes some of these viruses potential bioterrorism agents. Central nervous system (CNS) host responses contribute to alphavirus pathogenesis in experimental models and are logical therapeutic targets. We investigated whether reactive oxygen species (ROS) generated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) activity within the CNS contributes to fatal alphavirus encephalitis in mice. METHODS: Infected animals were treated systemically with the angiotensin receptor-blocking drug, telmisartan, given its ability to cross the blood-brain barrier, selectively block type-1 angiotensin receptors (AT1R), and inhibit Nox-derived ROS production in vascular smooth muscle and other extraneural tissues. Clinical, virological, biochemical, and histopathological outcomes were followed over time. RESULTS: The importance of the angiotensin II (Ang II)/AT1R axis in disease pathogenesis was confirmed by demonstrating increased Ang II levels in the CNS following infection, enhanced disease survival when CNS Ang II production was suppressed, increased AT1R expression on microglia and tissue-infiltrating myeloid cells, and enhanced disease survival in AT1R-deficient mice compared to wild-type (WT) controls. Systemic administration of telmisartan protected WT mice from lethal encephalitis caused by two different alphaviruses in a dose-dependent manner without altering virus replication or exerting any anti-inflammatory effects in the CNS. Infection triggered up-regulation of multiple Nox subunits in the CNS, while drug treatment inhibited local Nox activity, ROS production, and oxidative neuronal damage. Telmisartan proved ineffective in Nox-deficient mice, demonstrating that this enzyme is its main target in this experimental setting. CONCLUSIONS: Nox-derived ROS, likely arising from CNS myeloid cells triggered by AT1R signaling, are pathogenic during fatal alphavirus encephalitis in mice. Systemically administered telmisartan at non-hypotensive doses targets Nox activity in the CNS to exert a neuroprotective effect. Disruption of this pathway may have broader implications for the treatment of related infections as well as for other CNS diseases driven by oxidative injury.


Asunto(s)
Sistema Nervioso Central/patología , Encefalomielitis Equina/patología , Células Mieloides/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal/fisiología , ATPasas Asociadas con Actividades Celulares Diversas , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Bencimidazoles/farmacología , Benzoatos/farmacología , Receptor 1 de Quimiocinas CX3C , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/virología , ADN Helicasas/genética , ADN Helicasas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Equina/tratamiento farmacológico , Encefalomielitis Equina/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/clasificación , Células Mieloides/ultraestructura , Células Mieloides/virología , Neuronas/patología , Neuronas/ultraestructura , Consumo de Oxígeno/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Telmisartán
20.
J Virol ; 88(16): 9260-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24899192

RESUMEN

UNLABELLED: Western equine encephalitis virus (WEEV) is an arbovirus from the genus Alphavirus, family Togaviridae, which circulates in North America between birds and mosquitoes, occasionally causing disease in humans and equids. In recent decades, human infection has decreased dramatically; the last documented human case in North America occurred in 1994, and the virus has not been detected in mosquito pools since 2008. Because limited information exists regarding the evolution of WEEV, we analyzed the genomic sequences of 33 low-passage-number strains with diverse geographic and temporal distributions and performed comprehensive phylogenetic analyses. Our results indicated that WEEV is a highly conserved alphavirus with only approximately 5% divergence in its most variable genes. We confirmed the presence of the previously determined group A and B lineages and further resolved group B into three sublineages. We also observed an increase in relative genetic diversity during the mid-20th century, which correlates with the emergence and cocirculation of several group B sublineages. The estimated WEEV population size dropped in the 1990s, with only the group B3 lineage being sampled in the past 20 years. Structural mapping showed that the majority of substitutions in the envelope glycoproteins occurred at the E2-E2 interface. We hypothesize that an event occurred in the mid-20th century that resulted in the increased genetic diversity of WEEV in North America, followed by genetic constriction due to either competitive displacement by the B3 sublineage or stochastic events resulting from a population decline. IMPORTANCE: Western equine encephalitis virus (WEEV) has caused several epidemics that resulted in the deaths of thousands of humans and hundreds of thousands of equids during the past century. During recent decades, human infection decreased drastically and the virus has not been found in mosquito pools since 2008. Because limited information exists regarding the evolution of WEEV, we analyzed 33 complete genome sequences and conducted comprehensive phylogenetic analyses. We confirmed the presence of two major lineages, one of which diverged into three sublineages. Currently, only one of those sublineages is found circulating in nature. Understanding the evolution of WEEV over the past century provides a unique opportunity to observe an arbovirus that is in decline and to better understand what factors can cause said decline.


Asunto(s)
Evolución Biológica , Virus de la Encefalitis Equina del Oeste/genética , Genoma Viral/genética , Animales , Encefalomielitis Equina/virología , Variación Genética/genética , Caballos , América del Norte , Filogenia , Análisis de Secuencia/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA