Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 97(11): e0149723, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37877719

RESUMEN

IMPORTANCE: Duck Tembusu virus (DTMUV) is an emerging pathogenic flavivirus that replicates well in mosquito, bird, and mammalian cells. An in vivo study revealed that BALB/c mice and Kunming mice were susceptible to DTMUV after intracerebral inoculation. Moreover, there are no reports about DTMUV-related human disease, but antibodies against DTMUV and viral RNA were detected in the serum samples of duck industry workers. This information implies that DTMUV has expanded its host range and poses a threat to mammalian health. Thus, understanding the pathogenic mechanism of DTMUV is crucial for identifying potential antiviral targets. In this study, we discovered that NS3 can induce the mitochondria-mediated apoptotic pathway through the PERK/PKR pathway; it can also interact with voltage-dependent anion channel 2 to induce apoptosis. Our findings provide a theoretical basis for understanding the pathogenic mechanism of DTMUV infection and identifying potential antiviral targets and may also serve as a reference for exploring the pathogenesis of other flaviviruses.


Asunto(s)
Apoptosis , Patos , Infecciones por Flavivirus , Flavivirus , Especificidad del Huésped , Animales , Humanos , Antivirales/farmacología , Patos/virología , eIF-2 Quinasa/metabolismo , Flavivirus/enzimología , Flavivirus/patogenicidad , Infecciones por Flavivirus/diagnóstico , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/transmisión , Infecciones por Flavivirus/virología , Mitocondrias/metabolismo , Terapia Molecular Dirigida/tendencias , Zoonosis Virales/diagnóstico , Zoonosis Virales/inmunología , Zoonosis Virales/transmisión , Zoonosis Virales/virología , Canal Aniónico 2 Dependiente del Voltaje/metabolismo
2.
J Cell Biochem ; 124(1): 127-145, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36502494

RESUMEN

Numerous pathogens affecting human is present in the flavivirus family namely west nile, dengue, yellow fever, and zika which involves in development of global burden and distressing the environment economically. Till date, no approved drugs are available for targeting these viruses. The threat which urged the identification of small molecules for the inhibition of these viruses is the spreading of serious viral diseases. The recent outbreak of zika and dengue infections postured a solemn risk to worldwide public well-being. RNA-dependent RNA polymerase (RdRp) is the supreme adaptable enzymes of all the RNA viruses which is responsible for the replication and transcription of genome among the structural and nonstructural proteins of flaviviruses. It is understood that the RdRp of the flaviviruses are similar stating that the japanese encephalitis and west nile shares 70% identity with zika whereas the dengue serotype 2 and 3 shares the identity of 76% and 81%, respectively. In this study, we investigated the binding site of four flaviviral RdRp and provided insights into various interaction of the molecules using the computational approach. Our study helps in recognizing the potent compounds that could inhibit the viral protein as a common inhibitor. Additionally, with the conformational stability analysis, we proposed the possible mechanism of inhibition of the identified common small molecule toward RdRp of flavivirus. Finally, this study could be an initiative for the identification of common inhibitors and can be explored further for understanding the mechanism of action through in vitro studies for the study on efficacy.


Asunto(s)
Reposicionamiento de Medicamentos , Flavivirus , ARN Polimerasa Dependiente del ARN , Humanos , Dengue/tratamiento farmacológico , Flavivirus/efectos de los fármacos , Flavivirus/enzimología , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Virales/metabolismo , Virus Zika/efectos de los fármacos , Virus Zika/enzimología , Infección por el Virus Zika/tratamiento farmacológico
3.
J Virol ; 96(14): e0041822, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35758665

RESUMEN

The positive-sense flavivirus RNA genome bears a cap 1 structure essential for RNA stability and viral protein translation, and the formation of cap 1 requires the virally encoded nonstructural protein NS5 harboring guanylyltransferase (GTase), cap guanine N7 methyltransferase (N7 MTase), and 5'-nucleotide ribose 2'-O MTase activities in its single-domain MTase module. Despite numerous MTase-containing structures reported, the structural evidence for a critical GMP-enzyme intermediate formation and RNA repositioning when transitioning among different reactions is missing. Here, we report 10 high-resolution MTase crystal structures of Omsk hemorrhagic fever virus (OHFV), a representative high-consequence tick-borne flavivirus, capturing previously unidentified GMP-arginine adduct structures and a rarely observed capped RNA conformation. These structures help us thread capping events in the canonical model with a structure-based hypothesis involving the flipping of the 5' nucleotide, while the observation of an m7GMP-arginine adduct is compatible with an alternate capping model that decouples the N7 and 2'-O methylation steps. IMPORTANCE The methyltransferase (MTase) domain of flavivirus NS5 is unique in harboring guanylyltransferase (GTase), N7 MTase, and 2'-O MTase activities, playing a central role in viral RNA capping. However, the detailed mechanisms of the multistep capping process remain elusive. Here, we report 10 crystal structures of a flavivirus MTase to help understand the guanylyl transfer from GTP to the GTase itself and the transition between guanylyl transfer and methylation steps. In particular, a previously unobserved GMP-arginine covalent intermediate was captured multiple times in MTase crystal soaking trials with GTP present in the soaking solution, supporting its role in bridging the guanylyl transfer from GTP to the GTase and subsequent transfer to the 5'-diphosphate RNA.


Asunto(s)
Flavivirus , Modelos Moleculares , Proteínas no Estructurales Virales , Arginina , Virus de la Encefalitis Transmitidos por Garrapatas/enzimología , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Flavivirus/enzimología , Flavivirus/genética , Guanosina Trifosfato/metabolismo , Metiltransferasas/metabolismo , Nucleotidiltransferasas/química , Caperuzas de ARN/metabolismo , ARN Viral/metabolismo , Proteínas no Estructurales Virales/química
4.
J Immunol ; 203(12): 3374-3385, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31704883

RESUMEN

Duck Tembusu virus (DTMUV) is a newly emerged causative agent of avian disease. The protease-dependent immune evasion of flaviviruses has been reported; however, the molecular details of this process are unclear. In this study, we found that DTMUV nonstructural protein 2B-3, a NS2B3 protease, can inhibit IFN-ß production. DTMUV NS2B3 inhibited RIG-I-, MDA5-, MAVS-, and STING-directed IFN-ß transcription, but not TBK1- and IRF7-mediated induction of IFN-ß. Further analysis showed that DTMUV NS2B3 could cleave duck STING (duSTING); the cleavage was dependent on the protease activity of NS2B3. Moreover, the STING cleavage event occurred in a not-strictly-species-specific manner. The scissile bond of duSTING cleaved by NS2B3 was mapped between the R84 and G85 residues. The ability of NS2B3 to reduce duSTING cleavage-resistant mutant-mediated IFN-ß, and ISG production was significantly reduced, demonstrating that duSTING cleavage is essential for NS2B3-induced suppression of type I IFN responses. Remarkably, the binding of NS2B3 to duSTING, which is a prerequisite for cleavage, was found to depend on NS2B, but not NS3, the cofactor of the enzyme. Unexpectedly, we found that the region between aa residues 221-225 of duSTING, distal from the site of the scissile bond, was essential for the binding of NS2B3 to duSTING and/or the cleavage of duSTING by NS2B3. Thus, we identified the molecular mechanism by which DTMUV subverts the host innate immunity using its protease. More importantly, our study provides insight into NS2B3-mediated STING cleavage events in general.


Asunto(s)
Endopeptidasas/metabolismo , Infecciones por Flavivirus/veterinaria , Flavivirus/enzimología , Interferón beta/biosíntesis , Proteínas de la Membrana/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Línea Celular , Técnica del Anticuerpo Fluorescente , Genes Reporteros , Interacciones Huésped-Patógeno , Humanos , Unión Proteica , Proteolisis
5.
Int J Mol Sci ; 21(7)2020 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-32260545

RESUMEN

Nuclear magnetic resonance (NMR) spectroscopy plays important roles in structural biology and drug discovery, as it is a powerful tool to understand protein structures, dynamics, and ligand binding under physiological conditions. The protease of flaviviruses is an attractive target for developing antivirals because it is essential for the maturation of viral proteins. High-resolution structures of the proteases in the absence and presence of ligands/inhibitors were determined using X-ray crystallography, providing structural information for rational drug design. Structural studies suggest that proteases from Dengue virus (DENV), West Nile virus (WNV), and Zika virus (ZIKV) exist in open and closed conformations. Solution NMR studies showed that the closed conformation is predominant in solution and should be utilized in structure-based drug design. Here, we reviewed solution NMR studies of the proteases from these viruses. The accumulated studies demonstrated that NMR spectroscopy provides additional information to understand conformational changes of these proteases in the absence and presence of substrates/inhibitors. In addition, NMR spectroscopy can be used for identifying fragment hits that can be further developed into potent protease inhibitors.


Asunto(s)
Cisteína Endopeptidasas/química , Flavivirus/enzimología , Proteínas Virales/química , Antivirales/química , Antivirales/farmacología , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/farmacología , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/metabolismo
6.
Molecules ; 25(23)2020 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-33287144

RESUMEN

The RNA-dependent RNA polymerase (RdRp) is an essential enzyme for the viral replication process, catalyzing the viral RNA synthesis using a metal ion-dependent mechanism. In recent years, RdRp has emerged as an optimal target for the development of antiviral drugs, as demonstrated by recent approvals of sofosbuvir and remdesivir against Hepatitis C virus (HCV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), respectively. In this work, we overview the main sequence and structural features of the RdRp of emerging RNA viruses such as Coronaviruses, Flaviviruses, and HCV, as well as inhibition strategies implemented so far. While analyzing the structural information available on the RdRp of emerging RNA viruses, we provide examples of success stories such as for HCV and SARS-CoV-2. In contrast, Flaviviruses' story has raised attention about how the lack of structural details on catalytically-competent or ligand-bound RdRp strongly hampers the application of structure-based drug design, either in repurposing and conventional approaches.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Virus ARN/enzimología , ARN Polimerasa Dependiente del ARN/química , Amidas/química , Amidas/farmacología , Coronavirus/efectos de los fármacos , Coronavirus/enzimología , Coronavirus/genética , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Flavivirus/efectos de los fármacos , Flavivirus/enzimología , Flavivirus/genética , Hepacivirus/efectos de los fármacos , Hepacivirus/enzimología , Hepacivirus/genética , Humanos , Pirazinas/química , Pirazinas/farmacología , Infecciones por Virus ARN/epidemiología , Virus ARN/efectos de los fármacos , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
7.
PLoS Pathog ; 13(5): e1006411, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28542603

RESUMEN

The flavivirus genome encodes a single polyprotein precursor requiring multiple cleavages by host and viral proteases in order to produce the individual proteins that constitute an infectious virion. Previous studies have revealed that the NS2B cofactor of the viral NS2B-NS3 heterocomplex protease displays a conformational dynamic between active and inactive states. Here, we developed a conformational switch assay based on split luciferase complementation (SLC) to monitor the conformational change of NS2B and to characterize candidate allosteric inhibitors. Binding of an active-site inhibitor to the protease resulted in a conformational change of NS2B and led to significant SLC enhancement. Mutagenesis of key residues at an allosteric site abolished this induced conformational change and SLC enhancement. We also performed a virtual screen of NCI library compounds to identify allosteric inhibitors, followed by in vitro biochemical screening of the resultant candidates. Only three of these compounds, NSC135618, 260594, and 146771, significantly inhibited the protease of Dengue virus 2 (DENV2) in vitro, with IC50 values of 1.8 µM, 11.4 µM, and 4.8 µM, respectively. Among the three compounds, only NSC135618 significantly suppressed the SLC enhancement triggered by binding of active-site inhibitor in a dose-dependent manner, indicating that it inhibits the conformational change of NS2B. Results from virus titer reduction assays revealed that NSC135618 is a broad spectrum flavivirus protease inhibitor, and can significantly reduce titers of DENV2, Zika virus (ZIKV), West Nile virus (WNV), and Yellow fever virus (YFV) on A549 cells in vivo, with EC50 values in low micromolar range. In contrast, the cytotoxicity of NSC135618 is only moderate with CC50 of 48.8 µM on A549 cells. Moreover, NSC135618 inhibited ZIKV in human placental and neural progenitor cells relevant to ZIKV pathogenesis. Results from binding, kinetics, Western blot, mass spectrometry and mutagenesis experiments unambiguously demonstrated an allosteric mechanism for inhibition of the viral protease by NSC135618.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Flavivirus/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas no Estructurales Virales/química , Regulación Alostérica , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Flavivirus/química , Flavivirus/enzimología , Flavivirus/genética , Cinética , Conformación Proteica , ARN Helicasas/antagonistas & inhibidores , ARN Helicasas/química , ARN Helicasas/genética , ARN Helicasas/metabolismo , Serina Endopeptidasas/química , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
8.
Mol Phylogenet Evol ; 118: 58-63, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28951254

RESUMEN

Viruses belonging to the Flaviviridae family have been an important health concern for humans, animals and birds alike. No specific treatment is available yet for many of the viral infections caused by the members of this family. Lack of specific drugs against these viruses is mainly due to lack of protein structure information. It has been known that protein backbone fluctuation pattern is highly conserved in protein pairs with similar folds, in spite of the lack of sequence similarity. We hypothesized that this concept should also hold true for proteins (especially enzymes) of viruses included in different genera of the Flaviviridae family, as we know that the sequence similarity between them is low. Using available NS3 protease crystal structures of the Flaviviridae family, our preliminary results have shown that the Cα (i.e. backbone) fluctuation patterns are highly similar between Flaviviruses and a Hepacivirus (i.e. hepatitis C virus, HCV). This has to be validated further experimentally.


Asunto(s)
Evolución Molecular , Flavivirus/enzimología , Hepacivirus/enzimología , Proteínas no Estructurales Virales/clasificación , Secuencia de Aminoácidos , Animales , Humanos , Funciones de Verosimilitud , Filogenia , Estructura Terciaria de Proteína , ARN Helicasas/química , ARN Helicasas/clasificación , ARN Helicasas/genética , Alineación de Secuencia , Serina Endopeptidasas/química , Serina Endopeptidasas/clasificación , Serina Endopeptidasas/genética , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética
9.
Adv Exp Med Biol ; 1062: 175-186, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29845533

RESUMEN

Infections with flaviviruses are a continuing public health threat. In addition to vaccine development and vector control, the search for antiviral agents that alleviate symptoms in patients are of considerable interest. Among others, the flaviviral protease NS2B-NS3 is a promising drug target to inhibit viral replication. Flaviviral proteases share a high degree of structural similarity and substrate-recognition profile, which may facilitate a strategy towards development of pan-flaviviral protease inhibitors. However, the success of various drug discovery attempts during the last decade has been limited by the nature of the viral enzyme as well as a lack of robust structural templates. Small-molecular, structurally diverse protease inhibitors have been reported to reach affinities in the lower micromolar range. Peptide-based, substrate-derived compounds are often nanomolar inhibitors, however, with highly compromised drug-likeness. With some exceptions, the antiviral cellular activity of most of the reported compounds have been patchy and insufficient for further development. Recent progress has been made in the elucidation of inhibitor binding using different structural methods. This will hopefully lead to more rational attempts for the identification of various lead compounds that may be successful in cellular assays, animal models and ultimately translated to patients.


Asunto(s)
Antivirales/farmacología , Infecciones por Flavivirus/virología , Flavivirus/efectos de los fármacos , Flavivirus/enzimología , Inhibidores de Proteasas/farmacología , Animales , Descubrimiento de Drogas , Flavivirus/genética , Flavivirus/fisiología , Humanos , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
10.
J Gen Virol ; 95(Pt 4): 763-778, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24486628

RESUMEN

The 5' end of eukaryotic mRNA contains the type-1 (m7GpppNm) or type-2 (m7GpppNmNm) cap structure. Many viruses have evolved various mechanisms to develop their own capping enzymes (e.g. flavivirus and coronavirus) or to 'steal' caps from host mRNAs (e.g. influenza virus). Other viruses have developed 'cap-mimicking' mechanisms by attaching a peptide to the 5' end of viral RNA (e.g. picornavirus and calicivirus) or by having a complex 5' RNA structure (internal ribosome entry site) for translation initiation (e.g. picornavirus, pestivirus and hepacivirus). Here we review the diverse viral RNA capping mechanisms. Using flavivirus as a model, we summarize how a single methyltransferase catalyses two distinct N-7 and 2'-O methylations of viral RNA cap in a sequential manner. For antiviral development, a structural feature unique to the flavivirus methyltransferase was successfully used to design selective inhibitors that block viral methyltransferase without affecting host methyltransferases. Functionally, capping is essential for prevention of triphosphate-triggered innate immune activation; N-7 methylation is critical for enhancement of viral translation; and 2'-O methylation is important for subversion of innate immune response during viral infection. Flaviviruses defective in 2'-O methyltransferase are replicative, but their viral RNAs lack 2'-O methylation and are recognized and eliminated by the host immune response. Such mutant viruses could be rationally designed as live attenuated vaccines. This concept has recently been proved with Japanese encephalitis virus and dengue virus. The findings obtained with flavivirus should be applicable to other RNA viruses.


Asunto(s)
Flavivirus/enzimología , Flavivirus/metabolismo , Análogos de Caperuza de ARN , Procesamiento Postranscripcional del ARN , ARN Viral/metabolismo , ARNt Metiltransferasas/metabolismo , Evasión Inmune , Metilación , Biosíntesis de Proteínas
11.
Structure ; 32(8): 1099-1109.e3, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38781970

RESUMEN

Flaviviruses are single-stranded positive-sense RNA (+RNA) viruses that are responsible for several (re)emerging diseases such as yellow, dengue, or West Nile fevers. The Zika epidemic highlighted their dangerousness when a relatively benign virus known since the 1950s turned into a deadly pathogen. The central protein for their replication is NS5 (non-structural protein 5), which is composed of the N-terminal methyltransferase (MTase) domain and the C-terminal RNA-dependent RNA-polymerase (RdRp) domain. It is responsible for both RNA replication and installation of the 5' RNA cap. We structurally and biochemically analyzed the Ntaya virus MTase and RdRp domains and we compared their properties to other flaviviral NS5s. The enzymatic centers are well conserved across Flaviviridae, suggesting that the development of drugs targeting all flaviviruses is feasible. However, the enzymatic activities of the isolated proteins were significantly different for the MTase domains.


Asunto(s)
Metiltransferasas , Modelos Moleculares , ARN Polimerasa Dependiente del ARN , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , ARN Polimerasa Dependiente del ARN/metabolismo , ARN Polimerasa Dependiente del ARN/química , Metiltransferasas/metabolismo , Metiltransferasas/química , Cristalografía por Rayos X , Flavivirus/enzimología , Flavivirus/metabolismo , Unión Proteica , Secuencia de Aminoácidos , Dominios Proteicos , Virus de la Encefalitis Transmitidos por Garrapatas/metabolismo
12.
Antiviral Res ; 226: 105878, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38582134

RESUMEN

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


Asunto(s)
Antivirales , Flavivirus , Serina Endopeptidasas , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Flavivirus/efectos de los fármacos , Flavivirus/enzimología , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/química , Antivirales/farmacología , Antivirales/química , Humanos , ARN Helicasas/metabolismo , ARN Helicasas/química , ARN Helicasas/genética , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/química , Proteasas Virales , Nucleósido-Trifosfatasa , ARN Helicasas DEAD-box
13.
J Virol ; 86(16): 8730-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22674988

RESUMEN

Arthropod-borne flavivirus infection causes serious morbidity and mortality worldwide, but there are currently no effective antiflaviviral chemotherapeutics available for human use. Therefore, it is critical that new therapeutics against virus-specific targets be developed. To identify new compounds that may be used as broadly active flavivirus therapeutics, we have performed a high-throughput screening of 235,456 commercially available compounds for small-molecule inhibitors of the dengue virus NS5 RNA capping enzyme. We identified a family of compounds, the 2-thioxothiazolidin-4-ones, that show potent biochemical inhibition of capping enzyme GTP binding and guanylyltransferase function. During the course of structure-activity relationship analysis, a molecule within this family, (E)-{3-[5-(4-tert-butylbenzylidene)-4-oxo-2-thioxo-1,3-thiazolidin-3-yl]propanoic acid} (BG-323), was found to possess significant antiviral activity in a dengue virus subgenomic replicon assay. Further testing of BG-323 demonstrated that this molecule is able to reduce the replication of infectious West Nile virus and yellow fever virus in cell culture with low toxicity. The results of this study describe the first inhibitor that targets the GTP-binding/guanylyltransferase activity of the flavivirus RNA capping enzyme.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Flavivirus/efectos de los fármacos , Flavivirus/enzimología , Nucleotidiltransferasas/antagonistas & inhibidores , Proteínas no Estructurales Virales/antagonistas & inhibidores , Antivirales/química , Antivirales/aislamiento & purificación , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/aislamiento & purificación , Guanosina Trifosfato/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Pruebas de Sensibilidad Microbiana , Relación Estructura-Actividad , Tiazoles/química , Tiazoles/aislamiento & purificación , Tiazoles/farmacología
14.
Antiviral Res ; 210: 105516, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36586467

RESUMEN

Flaviviruses are important human pathogens and include dengue (DENV), West Nile (WNV), Yellow fever virus (YFV), Japanese encephalitis (JEV) and Zika virus (ZIKV). DENV, transmitted by mosquitoes, causes diseases ranging in severity from mild dengue fever with non-specific flu-like symptoms to fatal dengue hemorrhagic fever and dengue shock syndrome. DENV infections are caused by four serotypes, DENV1-4, which interact differently with antibodies in blood serum. The incidence of DENV infection has increased dramatically in recent decades and the CDC estimates 400 million dengue infections occur each year, resulting in ∼25,000 deaths mostly among children and elderly people. Similarly, ZIKV infections are caused by infected mosquito bites to humans, can be transmitted sexually and through blood transfusions. If a pregnant woman is infected, the virus can cross the placental barrier and can spread to her fetus, causing severe brain malformations in the child including microcephaly and other birth defects. It is noteworthy that the neurological manifestations of ZIKV were also observed in DENV endemic regions, suggesting that pre-existing antibody response to DENV could augment ZIKV infection. WNV, previously unknown in the US (and known to cause only mild disease in Middle East), first arrived in New York city in 1999 (NY99) and spread throughout the US and Canada by Culex mosquitoes and birds. WNV is now endemic in North America. Thus, emerging and re-emerging flaviviruses are significant threat to human health. However, vaccines are available for only a limited number of flaviviruses, and antiviral therapies are not available for any flavivirus. Hence, there is an urgent need to develop therapeutics that interfere with essential enzymatic steps, such as protease in the flavivirus lifecycle as these viruses possess significant threat to future pandemics. In this review, we focus on our E. coli expression of NS2B hydrophilic domain (NS2BH) covalently linked to NS3 protease domain (NS3Pro) in their natural context which is processed by the combined action of both subunits of the NS2B-NS3Pro precursor. Biochemical activities of the viral protease such as solubility and autoproteolysis of NS2BH-NS3Pro linkage depended on the C-terminal portion of NS2BH linked to the NS3Pro domain. Since 2008, we also focus on the use of the recombinant protease in high throughput screens and characterization of small molecular compounds identified in these screens.


Asunto(s)
Infecciones por Flavivirus , Flavivirus , Péptido Hidrolasas , Animales , Femenino , Humanos , Embarazo , Dengue/prevención & control , Virus del Dengue , Flavivirus/enzimología , Pandemias , Placenta , Virus Zika , Infección por el Virus Zika/prevención & control , Infecciones por Flavivirus/prevención & control
15.
J Biol Chem ; 286(16): 14362-72, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21349834

RESUMEN

Flavivirus NS5 protein encodes methyltransferase and RNA-dependent RNA polymerase (RdRp) activities. Structural analysis of flavivirus RdRp domains uncovered two conserved cavities (A and B). Both cavities are located in the thumb subdomains and represent potential targets for development of allosteric inhibitors. In this study, we used dengue virus as a model to analyze the function of the two RdRp cavities. Amino acids from both cavities were subjected to mutagenesis analysis in the context of genome-length RNA and recombinant NS5 protein; residues critical for viral replication were subjected to revertant analysis. For cavity A, we found that only one (Lys-756) of the seven selected amino acids is critical for viral replication. Alanine substitution of Lys-756 did not affect the RdRp activity, suggesting that this residue functions through a nonenzymatic mechanism. For cavity B, all four selected amino acids (Leu-328, Lys-330, Trp-859, and Ile-863) are critical for viral replication. Biochemical and revertant analyses showed that three of the four mutated residues (Leu-328, Trp-859, and Ile-863) function at the step of initiation of RNA synthesis, whereas the fourth residue (Lys-330) functions by interacting with the viral NS3 helicase domain. Collectively, our results have provided direct evidence for the hypothesis that cavity B, but not cavity A, from dengue virus NS5 polymerase could be a target for rational drug design.


Asunto(s)
Flavivirus/enzimología , Proteínas no Estructurales Virales/química , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Cricetinae , ARN Polimerasas Dirigidas por ADN/química , Diseño de Fármacos , Enzimas/química , Lisina/química , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , ARN Polimerasa Dependiente del ARN/química , Homología de Secuencia de Aminoácido , Triptófano/química , Células Vero , Replicación Viral
16.
J Biol Chem ; 285(42): 32586-95, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20685660

RESUMEN

The flavivirus methyltransferase (MTase) sequentially methylates the N7 and 2'-O positions of the viral RNA cap (GpppA-RNA → m(7)GpppA-RNA → m(7)GpppAm-RNA), using S-adenosyl-l-methionine (AdoMet) as a methyl donor. We report here that sinefungin (SIN), an AdoMet analog, inhibits several flaviviruses through suppression of viral MTase. The crystal structure of West Nile virus MTase in complex with SIN inhibitor at 2.0-Šresolution revealed a flavivirus-conserved hydrophobic pocket located next to the AdoMet-binding site. The pocket is functionally critical in the viral replication and cap methylations. In addition, the N7 methylation efficiency was found to correlate with the viral replication ability. Thus, SIN analogs with modifications that interact with the hydrophobic pocket are potential specific inhibitors of flavivirus MTase.


Asunto(s)
Flavivirus/enzimología , Metiltransferasas/química , Metiltransferasas/metabolismo , Estructura Terciaria de Proteína , Adenosina/análogos & derivados , Adenosina/química , Secuencia de Aminoácidos , Animales , Antifúngicos/química , Sitios de Unión , Cristalografía por Rayos X , Humanos , Metiltransferasas/genética , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Caperuzas de ARN/química , ARN Viral/química , ARN Viral/metabolismo , Virus del Nilo Occidental/enzimología
17.
RNA ; 15(12): 2340-50, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19850911

RESUMEN

The 5'-end of the flavivirus genome harbors a methylated (m7)GpppA(2'OMe) cap structure, which is generated by the virus-encoded RNA triphosphatase, RNA (guanine-N7) methyltransferase, nucleoside 2'-O-methyltransferase, and RNA guanylyltransferase. The presence of the flavivirus guanylyltransferase activity in NS5 has been suggested by several groups but has not been empirically proven. Here we provide evidence that the N-terminus of the flavivirus NS5 protein is a true RNA guanylyltransferase. We demonstrate that GTP can be used as a substrate by the enzyme to form a covalent GMP-enzyme intermediate via a phosphoamide bond. Mutational studies also confirm the importance of a specific lysine residue in the GTP binding site for the enzymatic activity. We show that the GMP moiety can be transferred to the diphosphate end of an RNA transcript harboring an adenosine as the initiating residue. We also demonstrate that the flavivirus RNA triphosphatase (NS3 protein) stimulates the RNA guanylyltransferase activity of the NS5 protein. Finally, we show that both enzymes are sufficient and necessary to catalyze the de novo formation of a methylated RNA cap structure in vitro using a triphosphorylated RNA transcript. Our study provides biochemical evidence that flaviviruses encode a complete RNA capping machinery.


Asunto(s)
Biocatálisis , Flavivirus/enzimología , Nucleotidiltransferasas/metabolismo , Caperuzas de ARN/metabolismo , Proteínas no Estructurales Virales/metabolismo , Guanosina Monofosfato/metabolismo , Nucleotidiltransferasas/genética , Caperuzas de ARN/química , Especificidad por Sustrato , Transcripción Genética , Proteínas no Estructurales Virales/genética
18.
Enzymes ; 49: 265-303, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34696835

RESUMEN

Flaviviruses such as dengue, Japanese encephalitis, West Nile, Yellow Fever and Zika virus, cause viral hemorrhagic fever and encephalitis in humans. However, antiviral therapeutics to treat or prevent flavivirus infections are not yet available. Thus, there is pressing need to develop therapeutics and vaccines that target flavivirus infections. All flaviviruses carry a positive-sense single-stranded RNA genome, which encodes ten proteins; three structural proteins form the virus shell, and seven nonstructural (NS) proteins are involved in replication of the viral genome. While all NS proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5) are part of a functional membrane-bound replication complex, enzymatic activities required for flaviviral replication reside in only two NS proteins, NS3 and NS5. NS3 functions as a protease, helicase, and triphosphatase, and NS5 as a capping enzyme, methyltransferase, and RNA-dependent RNA polymerase. In this chapter, we provide an overview of viral replication focusing on the structure and function of NS3 and NS5 replicases. We further describe strategies and examples of current efforts to identify potential flavivirus inhibitors against NS3 and NS5 enzymatic activities that can be developed as therapeutic agents to combat flavivirus infections.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Flavivirus , Proteínas no Estructurales Virales , Flavivirus/enzimología , ARN Polimerasa Dependiente del ARN , Proteínas no Estructurales Virales/genética , Replicación Viral
19.
Viruses ; 14(1)2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-35062249

RESUMEN

Flaviviruses are known to cause a variety of diseases in humans in different parts of the world. There are very limited numbers of antivirals to combat flavivirus infection, and therefore new drug targets must be explored. The flavivirus NS2B-NS3 proteases are responsible for the cleavage of the flavivirus polyprotein, which is necessary for productive viral infection and for causing clinical infections; therefore, they are a promising drug target for devising novel drugs against different flaviviruses. This review highlights the structural details of the NS2B-NS3 proteases of different flaviviruses, and also describes potential antiviral drugs that can interfere with the viral protease activity, as determined by various studies. Moreover, optimized in vitro reaction conditions for studying the NS2B-NS3 proteases of different flaviviruses may vary and have been incorporated in this review. The increasing availability of the in silico and crystallographic/structural details of flavivirus NS2B-NS3 proteases in free and drug-bound states can pave the path for the development of promising antiflavivirus drugs to be used in clinics. However, there is a paucity of information available on using animal cells and models for studying flavivirus NS2B-NS3 proteases, as well as on the testing of the antiviral drug efficacy against NS2B-NS3 proteases. Therefore, on the basis of recent studies, an effort has also been made to propose potential cellular and animal models for the study of flavivirus NS2B-NS3 proteases for the purposes of exploring flavivirus pathogenesis and for testing the efficacy of possible drugs targets, in vitro and in vivo.


Asunto(s)
Antivirales/farmacología , Descubrimiento de Drogas , Infecciones por Flavivirus/virología , Flavivirus/enzimología , Péptido Hidrolasas/metabolismo , ARN Helicasas/metabolismo , Serina Endopeptidasas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Virus del Dengue , Reducción Gradual de Medicamentos , Virus de la Encefalitis Japonesa (Especie) , Flavivirus/genética , Humanos , Péptido Hidrolasas/genética , Poliproteínas , ARN Helicasas/genética , Serina Endopeptidasas/genética , Proteínas no Estructurales Virales/genética , Proteinas del Complejo de Replicasa Viral , Virus del Nilo Occidental , Virus de la Fiebre Amarilla , Virus Zika
20.
Biochim Biophys Acta ; 1794(1): 50-60, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18930844

RESUMEN

West Nile virus (WNV) is a member of the Flaviviridae family which includes a number of important human pathogens. The WNV NS5 protein harbors an RNA-dependent RNA polymerase activity which is required both for replication and transcription of the viral genome. To extend our studies on the role of metal ions in the activity of flaviviral polymerases, we have used fluorescence spectroscopy, circular dichroism, and a combination of chemical and thermal denaturation assays to monitor the consequences of metal ion binding to the enzyme. We demonstrate that the binding of magnesium is not critical for the structural stabilization of the enzyme. Moreover, structural studies indicate that the protein does not undergo conformational change upon the binding of magnesium ions. Additional binding assays also indicate that the interaction of magnesium ions with the enzyme does not significantly stimulate the interaction with the RNA or NTP substrates. The inability of cobalt hexamine, an exchange-inert metal complex structurally analogous to magnesium hexahydrate, to support the catalytic activity also allowed us to demonstrate a direct role of magnesium ions in the catalytic activity of the enzyme. Finally, a three-dimensional structural model of the active center of the enzyme was generated which highlighted the importance of two aspartate residues involved in the coordination of two metal ions. Mutational analyses confirmed the importance of these two amino acids for the binding of magnesium ions. Our data provide further insight into the precise role of magnesium ions for the RNA polymerase activity of the protein, and more importantly, highlight key differences between the RNA polymerases of the Flaviviridae family.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/metabolismo , Flavivirus/enzimología , Metales/metabolismo , Secuencia de Bases , Sitios de Unión , Cloruros/química , Cloruros/metabolismo , Dicroismo Circular , Cobalto/química , Cobalto/metabolismo , Iones/metabolismo , Magnesio/química , Magnesio/metabolismo , Mutación , Desnaturalización Proteica , Espectrometría de Fluorescencia , Termodinámica , Urea/química , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Virus del Nilo Occidental/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA