RESUMEN
Flavivirus infection capitalizes on cellular lipid metabolism to remodel the cellular intima, creating a specialized lipid environment conducive to viral replication, assembly, and release. The Japanese encephalitis virus (JEV), a member of the Flavivirus genus, is responsible for significant morbidity and mortality in both humans and animals. Currently, there are no effective antiviral drugs available to combat JEV infection. In this study, we embarked on a quest to identify anti-JEV compounds within a lipid compound library. Our research led to the discovery of two novel compounds, isobavachalcone (IBC) and corosolic acid (CA), which exhibit dose-dependent inhibition of JEV proliferation. Time-of-addition assays indicated that IBC and CA predominantly target the late stage of the viral replication cycle. Mechanistically, JEV nonstructural proteins 1 and 2A (NS1 and NS2A) impede 5'-adenosine monophosphate (AMP)-activated protein kinase (AMPK) activation by obstructing the liver kinase B1 (LKB1)-AMPK interaction, resulting in decreased p-AMPK expression and a consequent upsurge in lipid synthesis. In contrast, IBC and CA may stimulate AMPK by binding to its active allosteric site, thereby inhibiting lipid synthesis essential for JEV replication and ultimately curtailing viral infection. Most importantly, in vivo experiments demonstrated that IBC and CA protected mice from JEV-induced mortality, significantly reducing viral loads in the brain and mitigating histopathological alterations. Overall, IBC and CA demonstrate significant potential as effective anti-JEV agents by precisely targeting AMPK-associated signaling pathways. These findings open new therapeutic avenues for addressing infections caused by Flaviviruses. IMPORTANCE: This study is the inaugural utilization of a lipid compound library in antiviral drug screening. Two lipid compounds, isobavachalcone (IBC) and corosolic acid (CA), emerged from the screening, exhibiting substantial inhibitory effects on the Japanese encephalitis virus (JEV) proliferation in vitro. In vivo experiments underscored their efficacy, with IBC and CA reducing viral loads in the brain and mitigating JEV-induced histopathological changes, effectively shielding mice from fatal JEV infection. Intriguingly, IBC and CA may activate 5'-adenosine monophosphate (AMP)-activated protein kinase (AMPK) by binding to its active site, curtailing the synthesis of lipid substances, and thus suppressing JEV proliferation. This indicates AMPK as a potential antiviral target. Remarkably, IBC and CA demonstrated suppression of multiple viruses, including Flaviviruses (JEV and Zika virus), porcine herpesvirus (pseudorabies virus), and coronaviruses (porcine deltacoronavirus and porcine epidemic diarrhea virus), suggesting their potential as broad-spectrum antiviral agents. These findings shed new light on the potential applications of these compounds in antiviral research.
Asunto(s)
Proteínas Quinasas Activadas por AMP , Antivirales , Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa , Metabolismo de los Lípidos , Replicación Viral , Animales , Metabolismo de los Lípidos/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/fisiología , Ratones , Antivirales/farmacología , Humanos , Encefalitis Japonesa/tratamiento farmacológico , Encefalitis Japonesa/virología , Proteínas Quinasas Activadas por AMP/metabolismo , Chalconas/farmacología , Triterpenos/farmacología , Proteínas no Estructurales Virales/metabolismo , Infecciones por Flavivirus/tratamiento farmacológico , Infecciones por Flavivirus/virología , Infecciones por Flavivirus/metabolismo , Flavivirus/efectos de los fármacos , Línea CelularRESUMEN
The envelope (E) protein of the Japanese encephalitis virus (JEV) is a key protein for virus infection and adsorption of host cells, which determines the virulence of the virus and regulates the intensity of inflammatory response. The mutation of multiple aa residues in the E protein plays a critical role in the attenuated strain of JEV. This study demonstrated that the Asp to Gly, Ser, and His mutation of the E389 site, respectively, the replication ability of the viruses in cells was significantly reduced, and the viral neuroinvasiveness was attenuated to different degrees. Among them, the mutation at E389 site enhanced the E protein flexibility contributed to the attenuation of neuroinvasiveness. In contrast, less flexibility of E protein enhanced the neuroinvasiveness of the strain. Our results indicate that the mechanism of attenuation of E389 aa mutation attenuates neuroinvasiveness is related to increased flexibility of the E protein. In addition, the increased flexibility of E protein enhanced the viral sensitivity to heparin inhibition in vitro, which may lead to a decrease in the viral load entering brain. These results suggest that E389 residue is a potential site affecting JEV virulence, and the flexibility of the E protein of aa at this site plays an important role in the determination of neuroinvasiveness.
Asunto(s)
Virus de la Encefalitis Japonesa (Especie) , Proteínas del Envoltorio Viral , Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/fisiología , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/química , Animales , Línea Celular , Virulencia , Replicación Viral , Encefalitis Japonesa/virología , Humanos , Heparina/farmacología , Sustitución de Aminoácidos , Mutación Missense , Ratones , Mutación , Factores de Virulencia/genética , Glicoproteínas de MembranaRESUMEN
Japanese encephalitis virus (JEV) genotype I (GI) replicates more efficiently than genotype III (GIII) in birds, and this difference is considered to be one of the reasons for the JEV genotype shift. In this study, we utilized duck embryo fibroblasts and domestic ducklings as in vitro and in vivo models of a JEV amplifying avian host to identify the viral determinants of the differing replication efficiency between the GI and GIII strains in birds. GI strains induced significantly lower levels of interferon (IFN)-α and ß production than GIII strains, an effect orrelated with the enhanced replication efficiency of GI strains over GIII strains. By using a series of chimeric viruses with exchange of viral structural and non-structural (NS) proteins, we identified NS5 as the viral determinant of the differences in IFN-α and ß induction and replication efficiency between the GI and III strains. NS5 inhibited IFN-α and ß production induced by poly(I:C) stimulation and harbored 11 amino acid variations, of which the NS5-V372A and NS5-H386Y variations were identified to co-contribute to the differences in IFN-α and ß induction and replication efficiency between the strains. The NS5-V372A and NS5-H386Y variations resulted in alterations in the number of hydrogen bonds formed with neighboring residues, which were associated with the different ability of the GI and GIII strains to inhibit IFN-α and ß production. Our findings indicated that the NS5-V372A and NS5-H386Y variations enabled GI strains to inhibit IFN-α and ß production more efficiently than GIII strains for antagonism of the IFN-I mediated antiviral response, thereby leading to the replication and host adaption advantages of GI strains over GIII strains in birds. These findings provide new insight into the molecular basis of the JEV genotype shift.
Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/inmunología , Interferón-alfa/farmacología , Interferón beta/farmacología , Mutación , Proteínas no Estructurales Virales/genética , Replicación Viral/genética , Animales , Antivirales/farmacología , Patos , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/genética , Encefalitis Japonesa/tratamiento farmacológico , Encefalitis Japonesa/virología , Interacciones Huésped-Patógeno , Ratones , Unión Proteica , Porcinos , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacosRESUMEN
INTRODUCTION: Invasion of viruses into the brain causes viral encephalitis, which can be fatal and causes permanent brain damage. The blood-brain barrier (BBB) protects the brain by excluding harmful substances and microbes. Brain microvascular endothelial cells are important components of the BBB; however, the mechanisms of antiviral reactions in these cells have not been fully elucidated. Zinc-finger antiviral protein (ZAP) is a molecule that restricts the infection of various viruses, and there are 2 major isoforms: ZAPL and ZAPS. Toll-like receptor 3 (TLR3), a pattern-recognition receptor against viral double-stranded RNA, is implicated in antiviral innate immune reactions. The aim of this study was to investigate the expression of ZAP in cultured hCMEC/D3 human brain microvascular endothelial cells treated with an authentic TLR3 agonist polyinosinic-polycytidylic acid (poly IC). METHODS: hCMEC/D3 cells were cultured and treated with poly IC. Expression of ZAPL and ZAPS mRNA was investigated using quantitative reverse transcription-polymerase chain reaction, and protein expression of these molecules was examined using western blotting. The role of nuclear factor-κB (NF-κB) was examined using the NF-κB inhibitor, SN50. The roles of interferon (IFN)-ß, IFN regulatory factor 3 (IRF3), tripartite motif protein 25 (TRIM25), and retinoic acid-inducible gene-I (RIG-I) in poly IC-induced ZAPS expression were examined using RNA interference. Propagation of Japanese encephalitis virus (JEV) was examined using a focus-forming assay. RESULTS: ZAPS mRNA and protein expression was upregulated by poly IC, whereas the change of ZAPL mRNA and protein levels was minimal. Knockdown of IRF3 or TRIM25 decreased the poly IC-induced upregulation of ZAPS, whereas knockdown of IFN-ß or RIG-I did not affect ZAPS upregulation. SN50 did not affect ZAPS expression. Knockdown of ZAP enhanced JEV propagation. CONCLUSION: ZAPL and ZAPS were expressed in hCMEC/D3 cells, and ZAPS expression was upregulated by poly IC. IRF3 and TRIM25 are involved in poly IC-induced upregulation of ZAPS. ZAP may contribute to antiviral reactions in brain microvascular endothelial cells and protect the brain from invading viruses such as JEV.
Asunto(s)
Antivirales , Cerebro , Virus de la Encefalitis Japonesa (Especie) , Células Endoteliales , Microvasos , Receptor Toll-Like 3 , Humanos , Antivirales/inmunología , Antivirales/farmacología , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , FN-kappa B/metabolismo , Poli I-C/farmacología , ARN Mensajero/metabolismo , Receptor Toll-Like 3/inmunología , Zinc , Microvasos/efectos de los fármacos , Microvasos/inmunología , Cerebro/irrigación sanguínea , Cerebro/inmunología , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/inmunologíaRESUMEN
Microglia being the resident macrophage of brain provides neuroprotection following diverse microbial infections. Japanese encephalitis virus (JEV) invades the CNS, resulting in neuroinflammation, which turns the neuroprotective role of microglia detrimental as characterized by increased microglial activation and neuronal death. Several host factors, including microRNAs, play vital roles in regulating virus-induced inflammation. In the current study, we demonstrate that the expression of miR-301a is increased in JEV-infected microglial cells and human brain. Overexpression of miR-301a augments the JEV-induced inflammatory response, whereas inhibition of miR-301a completely reverses the effects. Mechanistically, NF-κB-repressing factor (NKRF) functioning as inhibitor of NF-κB activation is identified as a potential target of miR-301a in JEV infection. Consequently, miR-301a-mediated inhibition of NKRF enhances nuclear translocation of NF-κB, which, in turn, resulted in amplified inflammatory response. Conversely, NKRF overexpression in miR-301a-inhibited condition restores nuclear accumulation of NF-κB to a basal level. We also observed that JEV infection induces classical activation (M1) of microglia that drives the production of proinflammatory cytokines while suppressing alternative activation (M2) that could serve to dampen the inflammatory response. Furthermore, in vivo neutralization of miR-301a in mouse brain restores NKRF expression, thereby reducing inflammatory response, microglial activation, and neuronal apoptosis. Thus, our study suggests that the JEV-induced expression of miR-301a positively regulates inflammatory response by suppressing NKRF production, which might be targeted to manage viral-induced neuroinflammation.
Asunto(s)
Antivirales/farmacología , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/tratamiento farmacológico , Encefalitis Japonesa/inmunología , Interferon beta-1b/farmacología , MicroARNs/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Animales , Células Cultivadas , Virus de la Encefalitis Japonesa (Especie)/metabolismo , Encefalitis Japonesa/metabolismo , Femenino , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Proteínas Represoras/metabolismoRESUMEN
Kaempferol, a plant-derived flavonoid, has been reported to have activity against Japanese encephalitis virus (JEV) in BHK-21 cells. To determine the broader utility of this compound, we initially evaluated the activity of kaempferol against JEV and dengue virus (DENV) in HEK293T/17 cells. Results showed no significant antiviral activity against either virus. We subsequently investigated the activity of kaempferol against both JEV and DENV in BHK-21 cells. Results showed a significant inhibition of JEV infection but, surprisingly, a significant enhancement of DENV infection. The effect of kaempferol on both host protein expression and transcription was investigated and both transcriptional and translational inhibitory effects were observed, although a more marked effect was observed on host cell protein expression. Markedly, while GRP78 was increased in DENV infected cells treated with kaempferol, it was not increased in JEV infected cells treated with kaempferol. These results show that cellular alteration induced by one compound can have opposite effects on viruses from the same family, suggesting the presence of distinct replication strategies for these two viruses.
Asunto(s)
Virus del Dengue/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Quempferoles/farmacología , Animales , Línea Celular , Cricetinae , Dengue/tratamiento farmacológico , Dengue/genética , Encefalitis Japonesa/tratamiento farmacológico , Encefalitis Japonesa/genética , Chaperón BiP del Retículo Endoplásmico , Células HEK293 , Proteínas de Choque Térmico/genética , Humanos , Biosíntesis de Proteínas/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Replicación Viral/efectos de los fármacosRESUMEN
Flaviviruses are emerging arthropod-borne viruses posing a great threat to human beings worldwide. The E dimer configuration of the flavivirus was prominent during viral assembly, maturation and entry. Neutralization antibodies targeting E dimer played the important role in controlling the flavivirus infection. Previously, the ideal drug target of small molecular inhibitors of JEV was viral proteases and polymerases. The crystal structure of JEV E protein showed a conserved pocket in it is important at membrane fusion step. Recently, a set of anti-virus drugs has been found by virtual screening. Here, we show that the fusion-loop pocket of JEV E protein was a conservative region and an ideal drug target. ChemDiv-3 from virtual screening as the lead compound was found to show a relatively modest inhibition effect for JEV in vitro and in vivo test and could interfere with the formation of JEV sE dimer. ChemDiv-3 interacts with the amino acid residues ASN 313, PRO 314, ALA 315, and VAL 323 in E protein via hydrogen bonds for occupation of the fusion-loop pocket. The key binding sites LYS 312, ALA 513 and THR 317 forming the fusion-loop pocket are the same and other auxiliary sites are similar among the flavivirus. Taken together, the fusion-loop pocket of the flavivirus could be one promising target for drug discovery.
Asunto(s)
Antivirales/química , Antivirales/farmacología , Virus de la Encefalitis Japonesa (Especie)/química , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Bases de Datos Farmacéuticas , Modelos Animales de Enfermedad , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Virus de la Encefalitis Japonesa (Especie)/genética , Encefalitis Japonesa/tratamiento farmacológico , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Multimerización de Proteína/efectos de los fármacos , Estructura Cuaternaria de Proteína/efectos de los fármacos , Relación Estructura-Actividad , Interfaz Usuario-Computador , Proteínas del Envoltorio Viral/genéticaRESUMEN
Neurotropic viruses, such as the rabies virus (RABV) and Japanese encephalitis virus (JEV), induce neuronal dysfunction and complication, causing neuronal damage. Currently, there are still no effective clinical treatments for neuronal injury caused by neurotropic viruses. Memantine, a drug capable of passing through the blood-brain barrier, noncompetitively and reversibly binds to n-methyl- d-aspartic acid (NMDA) receptors. Memantine is used to treat Alzheimer's disease by blocking the activation of extra axonal ion channels, thus preventing neuronal degeneration by inhibiting the abnormal cytosolic Ca 2+ increase. To explore whether memantine can alleviate neurological disturbances caused by RABV and JEV, the following experiments were carried out: (1) for primary neurons cultured in vitro infected with RABV, the addition of memantine showed neuroprotection. (2) In the RABV challenge experiments, memantine had limited therapeutic effect, mildly extending the survival time of mice. In contrast, memantine significantly prolonged the survival time of mice infected with JEV, by reducing the intravascular cuff and inflammatory cell infiltration in mice. Furthermore, memantine decreases the amount of JEV virus in mice brain.
Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Memantina/farmacología , Neuronas/efectos de los fármacos , Neuronas/virología , Fármacos Neuroprotectores/farmacología , Virus de la Rabia/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/virología , Células Cultivadas , Femenino , Ratones , Ratones Endogámicos C57BL , Neuronas/patologíaRESUMEN
Japanese encephalitis virus (JEV), a mosquito-borne flavivirus, occasionally causes severe central nervous system disorders in the risk zone where more than 3 billion people reside. Our prior studies demonstrated antiviral potential of 4,5-dihydrofuran-3-carboxylate compound CW-33 (ethyl 2-(3',5'-dimethylanilino)-4-oxo-4,5-dihydrofuran-3-carboxylate) and its derivative CW-33A ((ethyl 2-(2-fluoroanilino)-4-oxo-4,5-dihydrofuran-3-carboxylate) against JEV infection ((Int. J. Mol. Sci. 2016, 17: E1386; Sci. Rep. 2018, 8: 16595). This study synthesized six new CW-33 derivatives containing chloro, or bromo groups at the C-2, C-3, or C-4 of anilino ring of CW-33, and assessed the antiviral activity and mechanisms of these chloro- and bromo-anilino substitutedderivatives. CW-33K, CW-33L and CW-33M had the bromo-substituents at the C-2, C-3, or C-4 of anilino ring of CW-33, respectively, showing the higher anti-JEV activity than CW-33 and other derivatives. CW-33K (ethyl 2-(2-bromoanilino)-4-oxo-4,5-dihydrofuran-3-carboxylate) exhibited the highest antiviral efficacy and therapeutic index. The IC50 value of CW-33K was less than 5⯵M for reducing JEV-induced cytopathic effect, virus infectivity and virus yield. CW-33K significantly inhibited the JEV replication at the early and late stages, suppressing viral RNA synthesis and intracellular JEV particle production. The study demonstrated that the CW-33 derivative with a bromosubstitutionat the C-2 anilino ring improved the antiviral activity JEV, providing the structure-antiviral activity relationship for the development of anti-JEV agents.
Asunto(s)
Antivirales/uso terapéutico , Efecto Citopatogénico Viral/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Antivirales/farmacología , HumanosRESUMEN
Diphenyleneiodonium (DPI) and N-acetyl-l-cysteine (NAC), two widely used anti-oxidants, were employed to evaluate the role of oxidative stress in Japanese encephalitis virus (JEV) induced autophagy, stress responses and replication. DPI and NAC exerted opposite effects on ROS levels in JEV infected mouse neuronal cells (Neuro2a), mouse embryonic fibroblasts (MEFs) and human epithelial cells (HeLa). While NAC effectively quenched ROS, DPI enhanced ROS levels, suggesting that DPI induces oxidative stress in JEV infected cells. DPI treatment of JEV infected Neuro2a cells further blocked autophagy induction and activation of all three arms of the ER stress pathway, and, inhibited virus particle release. Autophagy induction in JEV infection has been previously shown to be linked to the activation of XBP1 and ATF6 ER stress sensors. Our data suggests that DPI mediated block of autophagy is a result of inhibition of ER stress responses and is not associated with an anti-oxidative effect. Since DPI has a wide inhibitory potential for all Flavin dependent enzymes, it is likely that the signalling pathways for ER stress and autophagy during JEV infection are modulated by DPI sensitive enzymes.
Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Compuestos Onio/farmacología , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Autofagia/efectos de los fármacos , Células Cultivadas , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Virus de la Encefalitis Japonesa (Especie)/fisiología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células HeLa , Humanos , Ratones , Estrés Oxidativo/efectos de los fármacos , Porcinos , Replicación Viral/efectos de los fármacosRESUMEN
Japanese encephalitis virus (JEV), an arthropod-borne flavivirus, is a major cause of acute viral encephalitis in humans. No approved drug is available for the specific treatment of JEV infections, and the available vaccines are not effective against all clinical JEV isolates. In the study described here, a high-throughput screening of an FDA-approved drug library for inhibitors of JEV was performed. Five hit drugs that inhibited JEV infection with a selective index of >10 were identified. The antiviral activities of these five hit drugs against other flavivirus, including Zika virus, were also validated. As three of the five hit drugs were calcium inhibitors, additional types of calcium inhibitors that confirmed that calcium is essential for JEV infection, most likely during viral replication, were utilized. Adaptive mutant analysis uncovered that replacement of Q130, located in transmembrane domain 3 of the nonstructural NS4B protein, which is relatively conserved in flaviviruses, with R or K conferred JEV resistance to manidipine, a voltage-gated Ca2+ channel (VGCC) inhibitor, without an apparent loss of the viral growth profile. Furthermore, manidipine was indicated to protect mice against JEV-induced lethality by decreasing the viral load in the brain, while it abrogated the histopathological changes associated with JEV infection. This study provides five antiflavivirus candidates and identifies cytoplasmic calcium to be a novel antiviral target for the treatment of JEV infection. The findings reported here provide therapeutic possibilities for combating infections caused by flaviviruses.IMPORTANCE No approved therapy for the treatment of Japanese encephalitis virus infection is currently available. Repurposing of approved drugs would accelerate the development of a therapeutic stratagem. In this study, we screened a library of FDA-approved drugs and identified five hit drugs, especially calcium inhibitors, exerting antiflavivirus activity that blocked viral replication. The in vivo efficacy and toxicity of manidipine were investigated with a mouse model of JEV infection, and the viral target was identified by generating an adaptive mutant.
Asunto(s)
Antivirales/farmacología , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Encefalitis Japonesa/tratamiento farmacológico , Ensayos Analíticos de Alto Rendimiento/métodos , Replicación Viral/efectos de los fármacos , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Aprobación de Drogas , Encefalitis Japonesa/virología , Femenino , Interacciones Huésped-Patógeno , Humanos , Ratones , Ratones Endogámicos BALB C , Estados Unidos , United States Food and Drug Administration , Carga ViralRESUMEN
Japanese encephalitis (JE) is a viral encephalitis disease caused by infection with the Japanese encephalitis virus (JEV). The virus can cross the blood-brain barrier and cause death or long-term sequela in infected humans or animals. In this study, we first investigated the distribution of JEV infection in brain and further analyzed the dynamic change in inflammation related genes, chemokines, as well as pathological characteristics. Results demonstrated that CCR2 and CCR5 antagonist could significantly inhibit the inflammation. The mice treated with CCR2 and CCR5 antagonists had a higher survival rate between 60% and 70%, respectively. In summary, our study thoroughly illustrated the characteristics of the dynamic change in inflammation related genes and chemokines induced by JEV infection. We further indicated that CCR5 and CCR2 are potential targets for treatment of JE.
Asunto(s)
Antagonistas de los Receptores CCR5/farmacología , Quimiocinas/metabolismo , Encefalitis Japonesa/tratamiento farmacológico , Encefalitis Japonesa/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Receptores de Quimiocina/antagonistas & inhibidores , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Chlorocebus aethiops , Citocinas/metabolismo , Modelos Animales de Enfermedad , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos C57BL , Receptores CCR2/antagonistas & inhibidores , Receptores CCR5 , Células VeroRESUMEN
Japanese encephalitis virus (JEV), known to affect children, is a major cause of severe encephalopathy. Its prevalence has been percolated over wider regions of Southern Asia. JEV is associated with neurodegeneration, severe inflammation, increased oxidative stress and elevated levels of stress linked proteins. Four groups of 15 mice each (4-5 weeks old BALB/c mice of either sex) was used for the study. Mice were intravenously infected with lethal dose of 3 × 105 pfu of JEV, followed by mortality after 8 days. On the next day and onwards, the animals were administered intraperitonially with (-)-tetrahydropalmatine (LTHP) solution (0.1 mg/mL in PBS) for the next 7 days. Animals exhibited protection against JEV infection, after being administered with LTHP. Reduction in levels of, viral population, caspase-2 expression, reactive oxygen and nitrogen species, microglial cells and proinflammatory mediators, stress linked protein molecules and neuronal apoptosis was exhibited in JEV infected animals treated with LTHP. The effects produced by the administration of LTHP indicated its possible use to treat JEV in mouse model. Potential to reduce viral count in brain and subsequent neuronal apoptosis, reduction in mediators of inflammation and oxidative stress, strictly advocate the use of LTHP for treatment of JEV. Thus, the present investigation indorses LTHP as a potentially strong drug candidate for the treatment of JEV infection due to its neuroprotective, anti-inflammatory, antiviral and anti-oxidative effect.
Asunto(s)
Antivirales/administración & dosificación , Alcaloides de Berberina/administración & dosificación , Virus de la Encefalitis Japonesa (Especie)/fisiología , Encefalitis Japonesa/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Animales , Caspasa 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Encefalitis Japonesa/genética , Encefalitis Japonesa/metabolismo , Encefalitis Japonesa/virología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/virología , Especies Reactivas de Oxígeno/metabolismoRESUMEN
Dengue virus (DENV) infection is a major public health problem worldwide; however, specific antiviral drugs against it are not available. Hence, identifying effective antiviral agents for the prevention of DENV infection is important. In this study, we showed that the reportedly highly biologically active green-tea component epigallocatechin gallate (EGCG) inhibited dengue virus infection regardless of infecting serotype, but no or minimal inhibition was observed with other flaviviruses, including Japanese encephalitis virus, yellow fever virus, and Zika virus. EGCG exerted its antiviral effect mainly at the early stage of infection, probably by interacting directly with virions to prevent virus infection. Our results suggest that EGCG specifically targets DENV and might be used as a lead structure to develop an antiviral drug for use against the virus.
Asunto(s)
Antivirales/farmacología , Catequina/análogos & derivados , Virus del Dengue/efectos de los fármacos , Té/química , Virión/efectos de los fármacos , Antivirales/aislamiento & purificación , Catequina/aislamiento & purificación , Catequina/farmacología , Virus del Dengue/fisiología , Relación Dosis-Respuesta a Droga , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/fisiología , Especificidad de la Especie , Virión/fisiología , Internalización del Virus/efectos de los fármacos , Virus de la Fiebre Amarilla/efectos de los fármacos , Virus de la Fiebre Amarilla/fisiología , Virus Zika/efectos de los fármacos , Virus Zika/fisiologíaRESUMEN
Japanese encephalitis (JE) is a vector-borne disease caused by the Japanese encephalitis virus (JEV). JEV is transmitted by mosquitoes to a wide range of vertebrate hosts, including birds and mammals. Domestic animals, especially pigs, are generally implicated as reservoirs of the virus, while humans are not part of the natural transmission cycle and cannot pass the virus to other hosts. Although JEV infection is very common in endemic areas (many countries in Asia), less than 1% of people affected develop clinical disease, and severe disease affects about 1 case per 250 JEV infections. Although rare, severe disease can be devastating; among the 30,000-50,000 global cases per year, approximately 20-30% of patients die and 30-50% of survivors develop significant neurological sequelae. JE is a significant public health problem for residents in endemic areas and may constitute a substantial risk for travelers to these areas. The epidemiology of JE and its risk to travelers have changed, and continue to evolve. The rapid economic growth of Asian countries has led to a surge in both inbound and outbound travel, making Asia the second most-visited region in the world after Europe, with 279 million international travelers in 2015. The top destination is China, followed by Thailand, Hong Kong, Malaysia and Japan, and the number of travelers is forecast to reach 535 million by 2030 (+ 4.9% per year). Because of the lack of treatment and the infeasibility of eliminating the vector, vaccination is recognized as the most efficacious means of preventing JE. The IC51 vaccine (IXIARO®) is a purified, inactivated, whole virus vaccine against JE. It is safe, well tolerated, efficacious and can be administered to children, adults and the elderly. The vaccination schedule involves administering 2 doses four weeks apart. For adults, a rapid schedule (0-7 days) is available, which could greatly enhance the feasibility of its use. Healthcare workers should inform both short- and long-term travelers of the risk of JE in each period of the year and recommend vaccination. Indeed, it has been shown that short-term travelers are also at risk, not only in rural environments, but also in cities and coastal towns, especially in tourist localities where excursions to country areas are organized.
Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Encefalitis Japonesa/prevención & control , Vacunas contra la Encefalitis Japonesa/administración & dosificación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Asia , Ensayos Clínicos Fase III como Asunto , Descubrimiento de Drogas , Humanos , Vacunas contra la Encefalitis Japonesa/efectos adversos , Persona de Mediana Edad , Ensayos Clínicos Controlados Aleatorios como Asunto , Medicina del Viajero , Adulto JovenRESUMEN
Japanese encephalitis virus (JEV) is the most common etiological agent of epidemic viral encephalitis. JEV encodes a single methyltransferase (MTase) domain located at the N-terminal region of the viral nonstructural protein NS5. JEV MTase is essential for viral replication and specifically catalyzes methylation of the viral RNA cap, which occurs exclusively in the cytoplasm. Therefore, JEV MTase is a potential target for antiviral therapy. Here, we identified specific and avid RNA aptamer (Kd â¼ 12 nM) with modified 2'-O-methyl pyrimidines against JEV MTase. The RNA aptamer efficiently inhibited viral cap methylation activity of MTase and interfered with JEV production in cells. Moreover, we generated a 24-mer truncated aptamer that could specifically bind to JEV MTase with high affinity (Kd â¼16 nM). The 24-mer aptamer efficiently inhibited JEV production and replication in cells. Therefore, MTase-specific RNA aptamer might be useful as an anti-JEV agent.
Asunto(s)
Antivirales/farmacología , Aptámeros de Nucleótidos/farmacología , Metiltransferasas/antagonistas & inhibidores , Caperuzas de ARN/metabolismo , ARN Viral/metabolismo , Proteínas no Estructurales Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Animales , Antivirales/química , Aptámeros de Nucleótidos/química , Línea Celular , Cricetinae , Virus de la Encefalitis Japonesa (Especie)/química , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/fisiología , Metilación/efectos de los fármacos , Metiltransferasas/genética , Conformación de Ácido Nucleico , Caperuzas de ARN/genética , ARN Viral/genética , Técnica SELEX de Producción de Aptámeros , Transfección , Proteínas no Estructurales Virales/genéticaRESUMEN
Mosquitoes are natural vectors for many etiologic agents of human viral diseases. Mosquito-borne flaviviruses can persistently infect the mosquito central nervous system without causing dramatic pathology or influencing the mosquito behavior and lifespan. The mechanism by which the mosquito nervous system resists flaviviral infection is still largely unknown. Here we report that an Aedes aegypti homologue of the neural factor Hikaru genki (AaHig) efficiently restricts flavivirus infection of the central nervous system. AaHig was predominantly expressed in the mosquito nervous system and localized to the plasma membrane of neural cells. Functional blockade of AaHig enhanced Dengue virus (DENV) and Japanese encephalitis virus (JEV), but not Sindbis virus (SINV), replication in mosquito heads and consequently caused neural apoptosis and a dramatic reduction in the mosquito lifespan. Consistently, delivery of recombinant AaHig to mosquitoes reduced viral infection. Furthermore, the membrane-localized AaHig directly interfaced with a highly conserved motif in the surface envelope proteins of DENV and JEV, and consequently interrupted endocytic viral entry into mosquito cells. Loss of either plasma membrane targeting or virion-binding ability rendered AaHig nonfunctional. Interestingly, Culex pipien pallens Hig also demonstrated a prominent anti-flavivirus activity, suggesting a functionally conserved function for Hig. Our results demonstrate that an evolutionarily conserved antiviral mechanism prevents lethal flaviviral infection of the central nervous system in mosquitoes, and thus may facilitate flaviviral transmission in nature.
Asunto(s)
Aedes/inmunología , Encéfalo/inmunología , Virus del Dengue/inmunología , Virus de la Encefalitis Japonesa (Especie)/inmunología , Interacciones Huésped-Patógeno , Proteínas del Tejido Nervioso/metabolismo , Neuronas/inmunología , Aedes/efectos de los fármacos , Aedes/virología , Animales , Antivirales/metabolismo , Antivirales/farmacología , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/virología , Línea Celular , Membrana Celular/efectos de los fármacos , Virus del Dengue/efectos de los fármacos , Virus del Dengue/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/fisiología , Femenino , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Proteínas de Insectos/antagonistas & inhibidores , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Proteínas de Insectos/farmacología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/farmacología , Neuronas/efectos de los fármacos , Neuronas/virología , Filogenia , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacosRESUMEN
Japanese encephalitis virus (JEV), a mosquito-borne flavivirus, has five genotypes (I, II, III, IV, and V). JEV genotype I circulates widely in some Asian countries. However, current JEV vaccines based on genotype III strains show low neutralizing capacities against genotype I variants. In addition, JE has no specific treatment, except a few supportive treatments. Compound CW-33, an intermediate synthesized derivative of furoquinolines, was investigated for its antiviral activities against JEV in this study. CW-33 exhibited the less cytotoxicity to Syrian baby hamster kidney (BHK-21) and human medulloblastoma (TE761) cells. CW-33 dose-dependently reduced the cytopathic effect and apoptosis of JEV-infected cells. Supernatant virus yield assay pinpointed CW-33 as having potential anti-JEV activity with IC50 values ranging from 12.7 to 38.5 µM. Time-of-addition assay with CW-33 indicated that simultaneous and post-treatment had no plaque reduction activity, but continuous and simultaneous treatments proved to have highly effective antiviral activity, with IC50 values of 32.7 and 48.5 µM, respectively. CW-33 significantly moderated JEV-triggered Ca(2+) overload, which correlated with the recovery of mitochondria membrane potential as well as the activation of Akt/mTOR and Jak/STAT1 signals in treated infected cells. Phosphopeptide profiling by LC-MS/MS revealed that CW-33 upregulated proteins from the enzyme modulator category, such as protein phosphatase inhibitor 2 (I-2), Rho GTPase-activating protein 35, ARF GTPase-activating protein GIT2, and putative 3-phosphoinositide-dependent protein kinase 2. These enzyme modulators identified were associated with the activation of Akt/mTOR and Jak/STAT1 signals. Meanwhile, I-2 treatment substantially inhibited the apoptosis of JEV-infected cells. The results demonstrated that CW-33 exhibited a significant potential in the development of anti-JEV agents.
Asunto(s)
Antivirales/farmacología , Calcio/metabolismo , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/metabolismo , Animales , Antivirales/química , Apoptosis/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Cricetinae , Proteínas Activadoras de GTPasa , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mesocricetus , Quinolinas/química , Quinolinas/farmacología , Factor de Transcripción STAT1/metabolismo , Espectrometría de Masas en Tándem , Replicación Viral/efectos de los fármacosRESUMEN
UNLABELLED: Nanomaterials have the characteristics associated with high surface-to-volume ratios and have been explored for their antiviral activity. Despite some success, cytotoxicity has been an issue in nanomaterial-based antiviral strategies. We previously developed a novel method to fully exfoliate montmorillonite clay to generate the most fundamental units of nanoscale silicate platelet (NSP). We further modified NSP by capping with various surfactants and found that the surfactant-modified NSP (NSQ) was less cytotoxic. In this study, we tested the antiviral potentials of a series of natural-clay-derived nanomaterials. Among the derivatives, NSP modified with anionic sodium dodecyl sulfate (NSQc), but not the pristine clay, unmodified NSP, a silver nanoparticle-NSP hybrid, NSP modified with cationic n-octadecanylamine hydrochloride salt, or NSP modified with nonionic Triton X-100, significantly suppressed the plaque-forming ability of Japanese encephalitis virus (JEV) at noncytotoxic concentrations. NSQc also blocked infection with dengue virus (DEN) and influenza A virus. Regarding the antiviral mechanism, NSQc interfered with viral binding through electrostatic interaction, since its antiviral activity can be neutralized by Polybrene, a cationic polymer. Furthermore, NSQc reduced the lethality of JEV and DEN infection in mouse challenge models. Thus, the surfactant-modified exfoliated nanoclay NSQc may be a novel nanomaterial with broad and potent antiviral activity. IMPORTANCE: Nanomaterials have being investigated as antimicrobial agents, yet their antiviral potential is overshadowed by their cytotoxicity. By using a novel method, we fully exfoliated montmorillonite clay to generate the most fundamental units of nanoscale silicate platelet (NSP). Here, we show that the surfactant-modified NSP (NSQ) is less cytotoxic and that NSQc (NSP modified with sodium dodecyl sulfate) could potently block infection by dengue virus (DEN), Japanese encephalitis virus (JEV), and influenza A virus at noncytotoxic concentrations. For the antiviral mechanism, we find that the electrostatic interaction between the negatively charged NSQc and the positively charged virus particles blocks viral binding. Furthermore, we used mouse challenge models of JEV and DEN to demonstrate the in vivo antiviral potential of NSQc. Thus, NSQc may function as a potent and safe antiviral nanohybrid against several viruses, and our success in synthesizing surfactant-modified NSP with antiviral activity may shed some light on future antiviral development.
Asunto(s)
Antivirales/farmacología , Bentonita/farmacología , Virus del Dengue/efectos de los fármacos , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus de la Influenza A/efectos de los fármacos , Nanoestructuras/uso terapéutico , Tensoactivos/química , Animales , Antivirales/química , Bentonita/química , Virus del Dengue/fisiología , Virus de la Encefalitis Japonesa (Especie)/fisiología , Encefalitis Japonesa , Humanos , Virus de la Influenza A/fisiología , Ratones , Ratones Endogámicos C57BL , Nanoestructuras/química , Octoxinol , Virosis/tratamiento farmacológico , Virosis/virologíaRESUMEN
The flaviviruses dengue, West Nile, and Japanese encephalitis represent three major mosquito-borne viruses worldwide. These pathogens impact the lives of millions of individuals and potentially could affect non-endemic areas already colonized by mosquito vectors. Unintentional transport of infected vectors (Aedes and Culex spp.), traveling within endemic areas, rapid adaptation of the insects into new geographic locations, climate change, and lack of medical surveillance have greatly contributed to the increase in flaviviral infections worldwide. The mechanisms by which flaviviruses alter the immune and the central nervous system have only recently been examined despite the alarming number of infections, related deaths, and increasing global distribution. In this review, we will discuss the expansion of the geographic areas affected by flaviviruses, the potential threats to previously unaffected countries, the mechanisms of pathogenesis, and the potential therapeutic interventions to limit the devastating consequences of these viruses.