Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Infect Dis ; 229(1): 43-53, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-37368353

RESUMEN

West Nile virus (WNV), an arthropod-borne flavivirus, can cause severe symptoms, including encephalitis, and death, posing a threat to public health and the economy. However, there is still no approved treatment or vaccine available for humans. Here, we developed a novel vaccine platform based on a classical insect-specific flavivirus (cISF) YN15-283-02, which was derived from Culicoides. The cISF-WNV chimera was constructed by replacing prME structural genes of the infectious YN15-283-02 cDNA clone with those of WNV and successfully rescued in Aedes albopictus cells. cISF-WNV was nonreplicable in vertebrate cells and nonpathogenic in type I interferon receptor (IFNAR)-deficient mice. A single-dose immunization of cISF-WNV elicited considerable Th1-biased antibody responses in C57BL/6 mice, which was sufficient to offer complete protection against lethal WNV challenge with no symptoms. Our studies demonstrated the potential of the insect-specific cISF-WNV as a prophylactic vaccine candidate to prevent infection with WNV.


Asunto(s)
Aedes , Flavivirus , Vacunas , Fiebre del Nilo Occidental , Virus del Nilo Occidental , Animales , Ratones , Humanos , Virus del Nilo Occidental/genética , Flavivirus/genética , Fiebre del Nilo Occidental/prevención & control , Anticuerpos Antivirales , Ratones Endogámicos C57BL
2.
J Virol ; 96(6): e0148021, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35107379

RESUMEN

In our previous study, we found that a new type of Chikungunya virus particle with a complete capsid deletion (ΔC-CHIKV) is still infectious in BHK-21 cells and demonstrated its potential as a live attenuated vaccine candidate. However, the low yield as well as the disability to propagate in vaccine production cell line Vero of ΔC-CHIKV are not practical for commercial vaccine development. In this study, we not only achieved the successful propagation of the viral particle in Vero cells, but significantly improved its yield through construction of a chimeric VEEV-ΔC-CHIKV and extensive passage in Vero cells. Mechanistically, high production of VEEV-ΔC-CHIKV is due to the improvement of viral RNA packaging efficiency conferred by adaptive mutations, especially those in envelope proteins. Similar to ΔC-CHIKV, the passaged VEEV-ΔC-CHIKV is safe, immunogenic, and efficacious, which protects mice from CHIKV challenge after only one shot of immunization. Our study demonstrates that the utilization of infectious capsidless viral particle of CHIKV as a vaccine candidate is a practical strategy for the development of alphavirus vaccine. IMPORTANCE Chikungunya virus (CHIKV) is one of important emerging alphaviruses. Currently, there are no licensed vaccines against CHIKV infection. We have previously found a new type of Chikungunya virus particle with a complete capsid deletion (ΔC-CHIKV) as a live attenuated vaccine candidate that is not suitable for commercial vaccine development with the low viral titer production. In this study, we significantly improved its production through construction of a chimeric VEEV-ΔC-CHIKV. Our results proved the utilization of infectious capsidless viral particle of CHIKV as a safe and practical vaccine candidate.


Asunto(s)
Fiebre Chikungunya , Virus Chikungunya , Vacunas Virales , Cultivo de Virus , Animales , Proteínas de la Cápside/genética , Fiebre Chikungunya/prevención & control , Virus Chikungunya/genética , Chlorocebus aethiops , Ratones , Desarrollo de Vacunas , Vacunas Atenuadas/genética , Células Vero , Vacunas Virales/genética , Cultivo de Virus/métodos
3.
Phys Rev Lett ; 131(8): 080401, 2023 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-37683167

RESUMEN

Quantum simulation of different exotic topological phases of quantum matter on a noisy intermediate-scale quantum (NISQ) processor is attracting growing interest. Here, we develop a one-dimensional 43-qubit superconducting quantum processor, named Chuang-tzu, to simulate and characterize emergent topological states. By engineering diagonal Aubry-André-Harper (AAH) models, we experimentally demonstrate the Hofstadter butterfly energy spectrum. Using Floquet engineering, we verify the existence of the topological zero modes in the commensurate off-diagonal AAH models, which have never been experimentally realized before. Remarkably, the qubit number over 40 in our quantum processor is large enough to capture the substantial topological features of a quantum system from its complex band structure, including Dirac points, the energy gap's closing, the difference between even and odd number of sites, and the distinction between edge and bulk states. Our results establish a versatile hybrid quantum simulation approach to exploring quantum topological systems in the NISQ era.

4.
Nucleic Acids Res ; 48(3): 1392-1405, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31863580

RESUMEN

The enterovirus 71 (EV71) 3Dpol is an RNA-dependent RNA polymerase (RdRP) that plays the central role in the viral genome replication, and is an important target in antiviral studies. Here, we report a crystal structure of EV71 3Dpol elongation complex (EC) at 1.8 Å resolution. The structure reveals that the 5'-end guanosine of the downstream RNA template interacts with a fingers domain pocket, with the base sandwiched by H44 and R277 side chains through hydrophobic stacking interactions, and these interactions are still maintained after one in-crystal translocation event induced by nucleotide incorporation, implying that the pocket could regulate the functional properties of the polymerase by interacting with RNA. When mutated, residue R277 showed an impact on virus proliferation in virological studies with residue H44 having a synergistic effect. In vitro biochemical data further suggest that mutations at these two sites affect RNA binding, EC stability, but not polymerase catalytic rate (kcat) and apparent NTP affinity (KM,NTP). We propose that, although rarely captured by crystallography, similar surface pocket interaction with nucleobase may commonly exist in nucleic acid motor enzymes to facilitate their processivity. Potential applications in antiviral drug and vaccine development are also discussed.


Asunto(s)
Enterovirus Humano A/ultraestructura , Complejos Multiproteicos/ultraestructura , Conformación Proteica , ARN Polimerasa Dependiente del ARN/ultraestructura , Antivirales/química , Sitios de Unión , Cristalografía por Rayos X , Enterovirus Humano A/química , Enterovirus Humano A/genética , Genoma Viral , Humanos , Modelos Moleculares , Complejos Multiproteicos/química , Nucleótidos/química , ARN Viral/química , ARN Viral/ultraestructura , ARN Polimerasa Dependiente del ARN/química , Replicación Viral/genética
5.
J Gen Virol ; 102(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33956592

RESUMEN

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) virus, which is highly pathogenic and classified as a biosafety level 3 (BSL-3) agent, has greatly threatened global health and efficacious antivirals are urgently needed. The high requirement of facilities to manipulate the live virus has limited the development of antiviral study. Here, we constructed a reporter replicon of SARS-CoV-2, which can be handled in a BSL-2 laboratory. The Renilla luciferase activity effectively reflected the transcription and replication levels of the replicon genome. We identified the suitability of the replicon in antiviral screening using the known inhibitors, and thus established the replicon-based high-throughput screening (HTS) assay for SARS-CoV-2. The application of the HTS assay was further validated using a few hit natural compounds, which were screened out in a SARS-CoV-2 induced cytopathic-effect-based HTS assay in our previous study. This replicon-based HTS assay will be a safe platform for SARS-CoV-2 antiviral screening in a BSL-2 laboratory without the live virus.


Asunto(s)
Antivirales/farmacología , Evaluación Preclínica de Medicamentos/métodos , Replicón/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Animales , Chlorocebus aethiops , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Replicón/genética , SARS-CoV-2/genética , Células Vero , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
6.
J Virol ; 94(5)2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31826997

RESUMEN

Mosquito-borne flaviviruses, which include many important human pathogens, such as West Nile virus (WNV), dengue virus (DENV), and Zika virus (ZIKV), have caused numerous emerging epidemics in recent years. Details of the viral genome functions necessary for effective viral replication in mosquito and vertebrate hosts remain obscure. Here, using ZIKV as a model, we found that the conserved "downstream of AUG region" (DAR), which is known to be an essential element for genome cyclization, is involved in viral replication in a host-specific manner. Mutational analysis of the DAR element showed that a single-nucleotide mismatch between the 5' DAR and the 3' DAR had little effect on ZIKV replication in mammalian cells but dramatically impaired viral propagation in mosquito cells. The revertant viruses passaged in mosquito cells generated compensatory mutations restoring the base pairing of the DAR, further confirming the importance of the complementarity of the DAR in mosquito cells. We demonstrate that a single-nucleotide mutation in the DAR is sufficient to destroy long-range RNA interaction of the ZIKV genome and affects de novo RNA synthesis at 28°C instead of 37°C, resulting in the different replication efficiencies of the mutant viruses in mosquito and mammalian cells. Our results reveal a novel function of the circular form of the flavivirus genome in host-specific viral replication, providing new ideas to further explore the functions of the viral genome during host adaptation.IMPORTANCE Flaviviruses naturally cycle between the mosquito vector and vertebrate hosts. The disparate hosts provide selective pressures that drive virus genome evolution to maintain efficient replication during host alteration. Host adaptation may occur at different stages of the viral life cycle, since host-specific viral protein processing and virion conformations have been reported in the individual hosts. However, the viral determinants and the underlying mechanisms associated with host-specific functions remain obscure. In this study, using Zika virus, we found that the DAR-mediated genome cyclization regulates viral replication differently and is under different selection pressures in mammalian and mosquito cells. A more constrained complementarity of the DAR is required in mosquito cells than in mammalian cells. Since the DAR element is stably maintained among mosquito-borne flaviviruses, our findings could provide new information for understanding the role of flavivirus genome cyclization in viral adaptation and RNA evolution in the two hosts.


Asunto(s)
Flavivirus/genética , Especificidad del Huésped/fisiología , Replicación Viral/fisiología , Virus Zika/genética , Animales , Secuencia de Bases , Chlorocebus aethiops , Culicidae/virología , Ciclización , Evolución Molecular , Genoma Viral , Humanos , Mutación , ARN Viral/genética , Células Vero , Virus Zika/crecimiento & desarrollo , Infección por el Virus Zika/genética
7.
J Virol ; 94(6)2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31896596

RESUMEN

Mosquito-borne flaviviruses consist of a positive-sense genome RNA flanked by the untranslated regions (UTRs). There is a panel of highly complex RNA structures in the UTRs with critical functions. For instance, Xrn1-resistant RNAs (xrRNAs) halt Xrn1 digestion, leading to the production of subgenomic flaviviral RNA (sfRNA). Conserved short direct repeats (DRs), also known as conserved sequences (CS) and repeated conserved sequences (RCS), have been identified as being among the RNA elements locating downstream of xrRNAs, but their biological function remains unknown. In this study, we revealed that the specific DRs are involved in the production of specific sfRNAs in both mammalian and mosquito cells. Biochemical assays and structural remodeling demonstrate that the base pairings in the stem of these DRs control sfRNA formation by maintaining the binding affinity of the corresponding xrRNAs to Xrn1. On the basis of these findings, we propose that DRs functions like a bracket holding the Xrn1-xrRNA complex for sfRNA formation.IMPORTANCE Flaviviruses include many important human pathogens. The production of subgenomic flaviviral RNAs (sfRNAs) is important for viral pathogenicity as a common feature of flaviviruses. sfRNAs are formed through the incomplete degradation of viral genomic RNA by the cytoplasmic 5'-3' exoribonuclease Xrn1 halted at the Xrn1-resistant RNA (xrRNA) structures within the 3'-UTR. The 3'-UTRs of the flavivirus genome also contain distinct short direct repeats (DRs), such as RCS3, CS3, RCS2, and CS2. However, the biological functions of these ancient primary DR sequences remain largely unknown. Here, we found that DR sequences are involved in sfRNA formation and viral virulence and provide novel targets for the rational design of live attenuated flavivirus vaccine.


Asunto(s)
Regiones no Traducidas 3'/fisiología , Flavivirus/metabolismo , Genoma Viral/fisiología , Conformación de Ácido Nucleico , ARN Viral/biosíntesis , Secuencias Repetidas en Tándem/fisiología , Células A549 , Animales , Chlorocebus aethiops , Cricetinae , Culicidae/metabolismo , Culicidae/virología , Flavivirus/genética , Humanos , ARN Viral/genética , Células Vero
8.
J Virol ; 93(17)2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31189715

RESUMEN

We previously produced a replication-defective West Nile virus (WNV) lacking NS1 (WNV-ΔNS1) that could propagate at low levels (105 infectious units [IU]/ml) in a 293T cell line expressing wild-type (WT) NS1. This finding indicates the potential of developing WNV-ΔNS1 as a noninfectious vaccine. To explore this idea, we developed an NS1-expressing Vero cell line (VeroNS1) that significantly improved the yield of WNV-ΔNS1 (108 IU/ml). We evaluated the safety and efficacy of WNV-ΔNS1 in mice. WNV-ΔNS1 appeared to be safe, as no replicative virus was found in naive Vero cells after continuous culturing of WNV-ΔNS1 in VeroNS1 cells for 15 rounds. WNV-ΔNS1 was noninfectious in mice, even when IFNAR-/- mice were administered a high dose of WNV-ΔNS1. Vaccination with a single dose of WNV-ΔNS1 protected mice from a highly lethal challenge with WT WNV. The antibody response against WNV correlated well with the protection of vaccinated mice. Our study demonstrates the potential of the NS1 trans complementation system as a new platform for flavivirus vaccine development.IMPORTANCE Many flaviviruses are significant human pathogens that frequently cause outbreaks and epidemics around the world. Development of novel vaccine platforms against these pathogens is a public health priority. Using WNV as a model, we developed a new vaccine platform for flaviviruses. WNV containing a NS1 deletion (WNV-ΔNS1) could be efficiently trans complemented in Vero cells that constitutively expressed WT NS1 protein. A single-dose immunization with WNV-ΔNS1 elicited robust immune responses in mice. The immunized animals were fully protected against pathogenic WNV infection. No adverse effects related to the WNV-ΔNS1 vaccination were observed. The results have demonstrated the potential of the NS1 complementation system as an alternative platform for flavivirus vaccine development, especially for highly pathogenic flaviviruses.


Asunto(s)
Receptor de Interferón alfa y beta/genética , Eliminación de Secuencia , Proteínas no Estructurales Virales/genética , Vacunas Virales/administración & dosificación , Fiebre del Nilo Occidental/prevención & control , Virus del Nilo Occidental/inmunología , Animales , Anticuerpos Antivirales/metabolismo , Chlorocebus aethiops , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Inmunización , Ratones , Células Vero , Vacunas Virales/inmunología , Replicación Viral , Fiebre del Nilo Occidental/genética , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/genética
9.
J Virol ; 93(15)2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31092567

RESUMEN

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that causes epidemics of debilitating disease worldwide. Currently, there are no licensed vaccines or antivirals available against CHIKV infection. In this study, we generated a novel live attenuated vaccine (LAV) candidate for CHIKV with a complete deficiency of capsid (ΔC-CHIKV). It could propagate in BHK-21 cells, and had antigenic properties similar to those of native CHIKV. Vaccination of either immunocompromised IFNAR-/- mice or immunocompetent C57BL/6 mice with a single dose of ΔC-CHIKV conferred complete protection upon challenge with wild-type (WT) CHIKV. Taken together, this vaccine candidate appeared to be safe and efficacious, representing a novel strategy for CHIKV vaccine design.IMPORTANCE Currently, there is no licensed vaccine against CHIKV infection. An ideal CHIKV vaccine should generate an optimal balance between efficacy and safety. Live attenuated vaccines that can elicit strong immune responses often involve a trade-off of reduced safety. Here, a novel live attenuated vaccine candidate for CHIKV lacking the entire capsid gene, ΔC-CHIKV, was developed. It was demonstrated to be genetically stable, highly attenuated, immunogenic, and able to confer complete protection against lethal CHIKV challenge after a single dose of immunization. Such an infectious vaccine candidate devoid of capsid provides a novel strategy for the development of a live attenuated CHIKV vaccine.


Asunto(s)
Proteínas de la Cápside/genética , Fiebre Chikungunya/prevención & control , Virus Chikungunya/genética , Virus Chikungunya/inmunología , Eliminación de Secuencia , Vacunas Virales/inmunología , Animales , Línea Celular , Fiebre Chikungunya/inmunología , Cricetinae , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Noqueados , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/aislamiento & purificación , Vacunas Virales/genética , Vacunas Virales/aislamiento & purificación
11.
J Virol ; 91(18)2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28659477

RESUMEN

West Nile virus (WNV) is a mosquito-borne flavivirus that causes epidemics of encephalitis and viscerotropic disease worldwide. This virus has spread rapidly and has posed a significant public health threat since the outbreak in New York City in 1999. The interferon (IFN)-mediated antiviral response represents an important component of virus-host interactions and plays an essential role in regulating viral replication. Previous studies have suggested that multifunctional nonstructural proteins encoded by flaviviruses antagonize the host IFN response via various means in order to establish efficient viral replication. In this study, we demonstrated that the nonstructural protein 1 (NS1) of WNV antagonizes IFN-ß production, most likely through suppression of retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) activation. In a dual-luciferase reporter assay, WNV NS1 significantly inhibited the activation of the IFN-ß promoter after Sendai virus infection or poly(I·C) treatment. NS1 also suppressed the activation of the IFN-ß promoter when it was stimulated by interferon regulatory factor 3 (IRF3)/5D or its upstream molecules in the RLR signaling pathway. Furthermore, NS1 blocked the phosphorylation and nuclear translocation of IRF3 upon stimulation by various inducers. Mechanistically, WNV NS1 targets RIG-I and melanoma differentiation-associated gene 5 (MDA5) by interacting with them and subsequently causing their degradation by the proteasome. Furthermore, WNV NS1 inhibits the K63-linked polyubiquitination of RIG-I, thereby inhibiting the activation of downstream sensors in the RLR signaling pathway. Taken together, our results reveal a novel mechanism by which WNV NS1 interferes with the host antiviral response.IMPORTANCE WNV Nile virus (WNV) has received increased attention since its introduction to the United States. However, the pathogenesis of this virus is poorly understood. This study demonstrated that the nonstructural protein 1 (NS1) of WNV antagonizes the induction of interferon beta (IFN-ß) by interacting with and degrading retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5), which are crucial viral sensors in the host innate immune system. Further experiments suggested that NS1-mediated inhibition of the RIG-I-like receptor (RLR) signaling pathway involves inhibition of RIG-I K63-linked polyubiquitination and that the proteasome plays a role in RIG-I degradation. This study provides new insights into the regulation of WNV NS1 in the RLR signaling pathway and reveals a novel mechanism by which WNV evades the host innate immune response. The novel findings may guide us to discover new therapeutic targets and develop effective vaccines for WNV infections.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Interacciones Huésped-Patógeno , Evasión Inmune , Helicasa Inducida por Interferón IFIH1/metabolismo , Interferón beta/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Virus del Nilo Occidental/patogenicidad , Animales , Línea Celular , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Receptores Inmunológicos , Virus del Nilo Occidental/inmunología
12.
J Virol ; 91(12)2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28404849

RESUMEN

Zika virus (ZIKV) is an emerging arbovirus belonging to the genus Flavivirus of the family Flaviviridae During replication processes, flavivirus manipulates host cell systems to facilitate its replication, while the host cells activate antiviral responses. Identification of host proteins involved in the flavivirus replication process may lead to the discovery of antiviral targets. The mosquitoes Aedes aegypti and Aedes albopictus are epidemiologically important vectors for ZIKV, and effective restrictions of ZIKV replication in mosquitoes will be vital in controlling the spread of virus. In this study, an iTRAQ-based quantitative proteomic analysis of ZIKV-infected Aedes albopictus C6/36 cells was performed to investigate host proteins involved in the ZIKV infection process. A total of 3,544 host proteins were quantified, with 200 being differentially regulated, among which CHCHD2 can be upregulated by ZIKV infection in both mosquito C6/36 and human HeLa cells. Our further study indicated that CHCHD2 can promote ZIKV replication and inhibit beta interferon (IFN-ß) production in HeLa cells, suggesting that ZIKV infection may upregulate CHCHD2 to inhibit IFN-I production and thus promote virus replication. Bioinformatics analysis of regulated host proteins highlighted several ZIKV infection-regulated biological processes. Further study indicated that the ubiquitin proteasome system (UPS) plays roles in the ZIKV entry process and that an FDA-approved inhibitor of the 20S proteasome, bortezomib, can inhibit ZIKV infection in vivo Our study illustrated how host cells respond to ZIKV infection and also provided a candidate drug for the control of ZIKV infection in mosquitoes and treatment of ZIKV infection in patients.IMPORTANCE ZIKV infection poses great threats to human health, and there is no FDA-approved drug available for the treatment of ZIKV infection. During replication, ZIKV manipulates host cell systems to facilitate its replication, while host cells activate antiviral responses. Identification of host proteins involved in the ZIKV replication process may lead to the discovery of antiviral targets. In this study, the first quantitative proteomic analysis of ZIKV-infected cells was performed to investigate host proteins involved in the ZIKV replication process. Bioinformatics analysis highlighted several ZIKV infection-regulated biological processes. Further study indicated that the ubiquitin proteasome system (UPS) plays roles in the ZIKV entry process and that an FDA-approved inhibitor of the UPS, bortezomib, can inhibit ZIKV infection in vivo Our study not only illustrated how host cells respond to ZIKV infection but also provided a candidate drug for the control of ZIKV infection in mosquitoes and treatment of ZIKV infection in patients.


Asunto(s)
Aedes/virología , Interacciones Huésped-Patógeno/genética , Proteínas de Insectos/metabolismo , Mosquitos Vectores/virología , Proteómica/métodos , Virus Zika/fisiología , Aedes/citología , Aedes/efectos de los fármacos , Aedes/fisiología , Animales , Bortezomib/administración & dosificación , Bortezomib/uso terapéutico , Chlorocebus aethiops , Biología Computacional , Células HeLa , Humanos , Proteínas de Insectos/genética , Interferón beta/antagonistas & inhibidores , Ratones , Complejo de la Endopetidasa Proteasomal/genética , Células Vero , Internalización del Virus , Replicación Viral/efectos de los fármacos , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/virología
13.
J Med Virol ; 90(3): 389-396, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28980717

RESUMEN

Zika virus (ZIKV) and chikungunya virus (CHIKV) are important human pathogens and mosquito-borne arboviruses, which have resembling history, common vectors, circulating regions, and indistinguishable clinical symptoms. Wide geographical range that is suitable for ZIKV and CHIKV transmission underlines the concern about the impact of epidemic and endemic infections on burden of public health. In the present study, a highly sensitive and specific one-step multiplex real-time RT-PCR assay was developed and evaluated for simultaneous detection and quantification of ZIKV and CHIKV. The single reaction assay employs two pairs of primers and two TaqMan probes that differentiate ZIKV and CHIKV infections. The entire viral genomic RNA in vitro transcribed from full-length infectious clones were used to generate the standard curves for absolute quantification in subsequent tests. The detection limit of the one-step multiplex assay was 1 and 0.5 PFU for infectious ZIKV and CHIKV, respectively. The assessment of specificity indicated this assay is highly specific to targeted viruses showing no amplification of a variety of other flaviviruses. Our assay was able to detect geographically separated and phylogenetically diverse strains of ZIKV and CHIKV. On the applicability of monitoring viral multiplication in cells and testing clinical samples, the one-step multiplex assay provided efficient and accurate determination. The one-step multiplex real-time RT-PCR assay offers a valuable tool for detection of ZIKV and CHIKV and potentially contributes to general surveillance and clinical treatment.


Asunto(s)
Virus Chikungunya/genética , Reacción en Cadena de la Polimerasa Multiplex/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Virus Zika/genética , Fiebre Chikungunya/diagnóstico , Virus Chikungunya/aislamiento & purificación , Cartilla de ADN/genética , Humanos , Límite de Detección , Técnicas de Diagnóstico Molecular , Sensibilidad y Especificidad , Virus Zika/aislamiento & purificación , Infección por el Virus Zika/diagnóstico
14.
J Gen Virol ; 98(7): 1739-1743, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28703698

RESUMEN

In this study, an in vitro ligation method was developed to assemble a full-length infectious cDNA clone of the Zika virus (ZIKV). Four contiguous cDNA subclones covering the complete ZIKV genome were constructed with unique BglI restriction sites at the ends of each fragment. The BglI restriction sites only allow in vitro ligation to happen between interconnecting restriction sites from adjacent cDNA fragments, resulting in an intact full-length cDNA of ZIKV. RNA transcripts derived from the full-length cDNA were infectious. The recombinant virus replicated as efficiently as the wild-type virus with similar growth kinetics and plaque morphologies in Vero and C6/36 cells. Both viruses were inhibited by NITD008 treatment. This in vitro ligation method will facilitate manipulation of the viral genome through genetic modifications of four separated subclones of ZIKV for the rapid and rational development of candidate vaccines and viral replication study.


Asunto(s)
Clonación Molecular/métodos , ADN Complementario/genética , ADN Viral/genética , ARN Viral/genética , Virus Zika/genética , Adenosina/análogos & derivados , Adenosina/farmacología , Aedes , Animales , Antivirales/farmacología , Línea Celular , Chlorocebus aethiops , Cricetinae , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Genoma Viral/genética , Células Vero , Virus Zika/efectos de los fármacos , Virus Zika/aislamiento & purificación
15.
J Virol ; 90(12): 5735-5749, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27053551

RESUMEN

UNLABELLED: Flavivirus nonstructural protein 2B (NS2B) is a transmembrane protein that functions as a cofactor for viral NS3 protease. The cytoplasmic region (amino acids 51 to 95) alone of NS2B is sufficient for NS3 protease activity, whereas the role of transmembrane domains (TMDs) remains obscure. Here, we demonstrate for the first time that flavivirus NS2B plays a critical role in virion assembly. Using Japanese encephalitis virus (JEV) as a model, we performed a systematic mutagenesis at the flavivirus conserved residues within the TMDs of NS2B. As expected, some mutations severely attenuated (L38A and R101A) or completely destroyed (G12L) viral RNA synthesis. Interestingly, two mutations (G37L and P112A) reduced viral RNA synthesis and blocked virion assembly. None of the mutations affected NS2B-NS3 protease activity. Because mutations G37L and P112A affected virion assembly, we selected revertant viruses for these two mutants. For mutant G37L, replacement with G37F, G37H, G37T, or G37S restored virion assembly. For mutant P112A, insertion of K at position K127 (leading to K127KK) of NS2B rescued virion assembly. A biomolecular fluorescent complementation (BiFC) analysis demonstrated that (i) mutation P112A selectively weakened NS2B-NS2A interaction and (ii) the adaptive mutation K127KK restored NS2B-NS2A interaction. Collectively, our results demonstrate that, in addition to being a cofactor for NS3 protease, flavivirus NS2B also functions in viral RNA replication, as well as virion assembly. IMPORTANCE: Many flaviviruses are important human pathogens. Understanding the molecular mechanisms of the viral infection cycle is essential for vaccine and antiviral development. In this study, we demonstrate that the TMDs of JEV NS2B participate in both viral RNA replication and virion assembly. A viral genetic study and a BiFC assay demonstrated that interaction between NS2B and NS2A may participate in modulating viral assembly in the flavivirus life cycle. Compensatory-mutation analysis confirmed that there was a correlation between viral assembly and NS2B-NS2A interaction. TMDs of NS2B may serve as novel antiviral targets to prevent flavivirus infection, and the structure determination of NS2B will help us to understand the functional mechanism of NS2B in viral RNA replication and assembly. The results have uncovered a new function of flavivirus NS2B in virion assembly, possibly through interaction with the NS2A protein.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/fisiología , ARN Viral/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Ensamble de Virus , Replicación Viral , Análisis Mutacional de ADN , Virus de la Encefalitis Japonesa (Especie)/química , Virus de la Encefalitis Japonesa (Especie)/genética , Humanos , Mutagénesis , Dominios Proteicos
16.
Arch Virol ; 162(11): 3417-3423, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28779235

RESUMEN

Japanese encephalitis virus (JEV), an important pathogen in Eastern and Southern Asia and the Pacific, has spread to Australia and other territories in recent years. Although the vaccine for JEV has been used in some countries, development of efficient antiviral drugs is still an urgent requirement. Replicon systems have been widely used in the research of viral replication and antiviral screening for West Nile virus (WNV), yellow fever virus (YFV) and dengue virus (DENV). Here, a novel JEV replicon harboring the Rluc and Pac gene (JEV-Pac-Rluc-Rep) was constructed. Furthermore, we established a BHK-21 cell line harboring JEV-Pac-Rluc-Rep (BHK-21/PAC/Rluc cell line) through continuous puromycin selection. Characterization of cell line stability showed that the replicon RNA could persistently replicate in this cell line for at least up to 10 rounds of passage. Using a known flavivirus inhibitor, the JEV replicon cell line was validated for antiviral screening. The JEV replicon cell line will be a valuable tool for both compound screening and viral replication studies.


Asunto(s)
Antivirales/uso terapéutico , Virus de la Encefalitis Japonesa (Especie)/fisiología , Animales , Línea Celular , Cricetinae , Puromicina , Replicón/genética , Replicón/fisiología , Replicación Viral
17.
J Gen Virol ; 96(Pt 6): 1264-1275, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25575708

RESUMEN

Flavivirus NS4A and NS4B are important membrane proteins for viral replication that are assumed to serve as the scaffold for the formation of replication complexes. We previously demonstrated that a single Lys-to-Arg mutation at position 79 in NS4A (NS4A-K79R) significantly impaired Japanese encephalitis virus (JEV) replication. In this study, the mutant virus was subject to genetic selection to search for the potential interaction between NS4A and other viral components. Sequencing of the recovered viruses revealed that, in addition to an A97E change in NS4A itself, a Y3N compensatory mutation located in NS4B had emerged from independent selections. Mutagenesis analysis, using a genome-length RNA and a replicon of JEV, demonstrated that both adaptive mutations greatly restored the replication defect caused by NS4A-K79R. Our results, for the first time to our knowledge, clearly showed the genetic interaction between NS4A and NS4B, although the mechanism underlying their interaction is unknown.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/fisiología , Mapeo de Interacción de Proteínas , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Sustitución de Aminoácidos , Animales , Análisis Mutacional de ADN , Virus de la Encefalitis Japonesa (Especie)/genética , Mutación Missense , Selección Genética , Supresión Genética , Proteínas no Estructurales Virales/genética
18.
J Virol ; 88(20): 11915-23, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25100827

RESUMEN

Enterovirus 71 (EV71) is a major viral pathogen in China and Southeast Asia. There is no clinically approved vaccine or antiviral therapy for EV71 infection. NITD008, an adenosine analog, is an inhibitor of flavivirus that blocks viral RNA synthesis. Here we report that NITD008 has potent antiviral activity against EV71. In cell culture, the compound inhibits EV71 at a 50% effective concentration of 0.67 µM and a 50% cytotoxic concentration of 119.97 µM. When administered at 5 mg/kg in an EV71 mouse model, the compound reduced viral loads in various organs and completely prevented clinical symptoms and death. To study the antiviral mechanism and drug resistance, we selected escape mutant viruses by culturing EV71 with increasing concentrations of NITD008. Resistance mutations were reproducibly mapped to the viral 3A and 3D polymerase regions. Resistance analysis with recombinant viruses demonstrated that either a 3A or a 3D mutation alone could lead to resistance to NITD008. A combination of both 3A and 3D mutations conferred higher resistance, suggesting a collaborative interplay between the 3A and 3D proteins during viral replication. The resistance results underline the importance of combination therapy required for EV71 treatment. Importance: Human enterovirus 71 (EV71) has emerged as a major cause of viral encephalitis in children worldwide, especially in the Asia-Pacific region. Vaccines and antivirals are urgently needed to prevent and treat EV71 infections. In this study, we report the in vitro and in vivo efficacy of NITD008 (an adenosine analog) as an inhibitor of EV71. The efficacy results validated the potential of nucleoside analogs as antiviral drugs for EV71 infections. Mechanistically, we showed that mutations in the viral 3A and 3D polymerases alone or in combination could confer resistance to NITD008. The resistance results suggest an intrinsic interaction between viral proteins 3A and 3D during replication, as well as the importance of combination therapy for the treatment of EV71 infections.


Asunto(s)
Adenosina/análogos & derivados , Adenosina/farmacología , Antivirales/farmacología , Enterovirus Humano A/efectos de los fármacos , Animales , Antivirales/uso terapéutico , Chlorocebus aethiops , Farmacorresistencia Viral/genética , Enterovirus Humano A/crecimiento & desarrollo , Enterovirus Humano A/fisiología , Infecciones por Enterovirus/tratamiento farmacológico , Infecciones por Enterovirus/fisiopatología , Infecciones por Enterovirus/virología , Ratones , Mutación , Células Vero , Carga Viral , Ensayo de Placa Viral , Replicación Viral
19.
J Gen Virol ; 95(Pt 4): 806-815, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24362961

RESUMEN

A full-length genome infectious clone is a powerful tool for functional assays in virology. In this study, using a chemical synthesized complete genome of Japanese encephalitis virus (JEV) strain SA14 (GenBank accession no. U14163), we constructed a full-length genomic cDNA clone of JEV. The recovered virus from the cDNA clone replicated poorly in baby hamster kidney (BHK-21) cells and in suckling mice brain. Following serial passage in BHK-21 cells, adaptive mutations within the NS2B and NS4A proteins were recovered in the passaged viruses leading to viruses with a large-plaque phenotype. Mutagenesis analysis, using a genome-length RNA and a replicon of JEV, demonstrated that the adaptive mutations restored replication to different degrees, and the restoration efficiencies were in the order: NS2B-T102M

Asunto(s)
Adaptación Biológica , Virus de la Encefalitis Japonesa (Especie)/fisiología , Mutación Missense , Pase Seriado , Proteínas no Estructurales Virales/genética , Replicación Viral , Animales , Encéfalo/virología , Línea Celular , Cricetinae , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Virus de la Encefalitis Japonesa (Especie)/genética , Encefalitis Japonesa/patología , Encefalitis Japonesa/virología , Femenino , Ratones , Ratones Endogámicos BALB C , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Genética Inversa , Proteínas no Estructurales Virales/metabolismo , Virulencia
20.
Virol Sin ; 39(2): 301-308, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38452856

RESUMEN

Hand, foot, and mouth disease (HFMD) is a common pediatric illness mainly caused by enteroviruses, which are important human pathogens. Currently, there are no available antiviral agents for the therapy of enterovirus infection. In this study, an excellent high-content antiviral screening system utilizing the EV-A71-eGFP reporter virus was developed. Using this screening system, we screened a drug library containing 1042 natural compounds to identify potential EV-A71 inhibitors. Fangchinoline (FAN), a bis-benzylisoquinoline alkaloid, exhibits potential inhibitory effects against various enteroviruses that cause HFMD, such as EV-A71, CV-A10, CV-B3 and CV-A16. Further investigations revealed that FAN targets the early stage of the enterovirus life cycle. Through the selection of FAN-resistant EV-A71 viruses, we demonstrated that the VP1 protein could be a potential target of FAN, as two mutations in VP1 (E145G and V258I) resulted in viral resistance to FAN. Our research suggests that FAN is an efficient inhibitor of EV-A71 and has the potential to be a broad-spectrum antiviral drug against human enteroviruses.


Asunto(s)
Antivirales , Bencilisoquinolinas , Farmacorresistencia Viral , Antivirales/farmacología , Humanos , Bencilisoquinolinas/farmacología , Farmacorresistencia Viral/genética , Replicación Viral/efectos de los fármacos , Enterovirus Humano A/efectos de los fármacos , Enterovirus Humano A/genética , Evaluación Preclínica de Medicamentos , Genes Reporteros , Ensayos Analíticos de Alto Rendimiento , Proteínas de la Cápside/genética , Proteínas de la Cápside/antagonistas & inhibidores , Enterovirus/efectos de los fármacos , Enterovirus/genética , Línea Celular , Proteínas Fluorescentes Verdes/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA