Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Cell Rep ; 42(5): 112489, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37167063

RESUMO

Upon recognizing danger signals produced by virally infected neurons, macrophages in the central nervous system (CNS) secrete multiple inflammatory cytokines to accelerate neuron apoptosis. The understanding is limited about which key effectors regulate macrophage-neuron crosstalk upon infection. We have used neurotropic-virus-infected murine models to identify that vascular endothelial growth factor receptor 3 (VEGFR-3) is upregulated in the CNS macrophages and that virally infected neurons secrete the ligand VEGF-C. When cultured with VEGF-C-containing supernatants from virally infected neurons, VEGFR-3+ macrophages suppress tumor necrosis factor α (TNF-α) secretion to reduce neuron apoptosis. Vegfr-3ΔLBD/ΔLBD (deletion of ligand-binding domain in myeloid cells) mice or mice treated with the VEGFR-3 kinase inhibitor exacerbate the severity of encephalitis, TNF-α production, and neuron apoptosis post Japanese encephalitis virus (JEV) infection. Activating VEGFR-3 or blocking TNF-α can reduce encephalitis and neuronal damage upon JEV infection. Altogether, we show that the inducible VEGF-C/VEGFR-3 module generates protective crosstalk between neurons and macrophages to alleviate CNS viral infection.


Assuntos
Vírus da Encefalite Japonesa (Espécie) , Encefalite Japonesa , Camundongos , Animais , Fator de Necrose Tumoral alfa/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Ligantes , Fator A de Crescimento do Endotélio Vascular/metabolismo , Encefalite Japonesa/metabolismo , Encefalite Japonesa/patologia , Vírus da Encefalite Japonesa (Espécie)/metabolismo , Neurônios/metabolismo , Macrófagos/metabolismo
2.
Elife ; 112022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35972780

RESUMO

Zika virus (ZIKV) can be transmitted from mother to fetus during pregnancy, causing adverse fetal outcomes. Several studies have indicated that ZIKV can damage the fetal brain directly; however, whether the ZIKV-induced maternal placental injury contributes to adverse fetal outcomes is sparsely defined. Here, we demonstrated that ZIKV causes the pyroptosis of placental cells by activating the executor gasdermin E (GSDME) in vitro and in vivo. Mechanistically, TNF-α release is induced upon the recognition of viral genomic RNA by RIG-I, followed by activation of caspase-8 and caspase-3 to ultimately escalate the GSDME cleavage. Further analyses revealed that the ablation of GSDME or treatment with TNF-α receptor antagonist in ZIKV-infected pregnant mice attenuates placental pyroptosis, which consequently confers protection against adverse fetal outcomes. In conclusion, our study unveils a novel mechanism of ZIKV-induced adverse fetal outcomes via causing placental cell pyroptosis, which provides new clues for developing therapies for ZIKV-associated diseases.


Assuntos
Placenta , Complicações Infecciosas na Gravidez , Piroptose , Infecção por Zika virus , Animais , Feminino , Feto , Humanos , Camundongos , Placenta/patologia , Placenta/virologia , Proteínas Citotóxicas Formadoras de Poros , Gravidez , Complicações Infecciosas na Gravidez/virologia , RNA Viral , Fator de Necrose Tumoral alfa , Zika virus/patogenicidade , Infecção por Zika virus/complicações
3.
Microbiol Spectr ; 10(3): e0141722, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35695552

RESUMO

In flavivirus, the furin-mediated cleavage of prM is mandatory to produce infectious particles, and the immature particles containing uncleaved prM cannot undergo membrane fusion and release to the extracellular environment. However, the detailed relationship between viral replication or pathogenicity and furin in Japanese encephalitis virus (JEV) hasn't been clarified. Here, JEV with the mutations in furin cleavage sites and its nearby were constructed. Compared with WT virus, the mutant virus showed enhanced cleavage efficiency of prM protein and increased replication ability. Furthermore, we found that the mutations mainly promote genomic replication and assembly of JEV. However, the mutant formed smaller plaques than WT virus in plaque forming assay, indicating the lower cytopathogenicity of mutant virus. To assess the virulence of JEV mutant, an in vivo assay was performed using a mouse model. A higher survival rate and attenuated neuroinflammation were observed in JEV mutant-infected mice than those of WT-infected mice, suggesting the cleavage of prM by furin was closely related to viral virulence. These findings will provide new understanding on JEV pathogenesis and contribute to the development of novel JEV vaccines. IMPORTANCE Japanese encephalitis virus (JEV) is the leading cause of viral encephalitis epidemics in Southeast Asia, affecting mostly children, with high morbidity and mortality. During the viral maturation process, prM is cleaved into M by the cellular endoprotease furin in the acidic secretory system. After cleavage of the prM protein, mature virions are exocytosed. Here, the mutant in furin cleavage sites and its nearby was constructed, and the results showed that the mutant virus with enhanced replication mainly occurred in the process of genomic replication and assembly. Meanwhile, the mutant showed an attenuated virulence than WT virus in vivo. Our study contributes to understanding the function of prM and M proteins and provides new clues for live vaccine designation for JEV.


Assuntos
Vírus da Encefalite Japonesa (Espécie) , Linhagem Celular , Vírus da Encefalite Japonesa (Espécie)/genética , Vírus da Encefalite Japonesa (Espécie)/metabolismo , Furina/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Virulência , Replicação Viral
4.
Arch Virol ; 167(3): 849-859, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35119507

RESUMO

Japanese encephalitis (JE) is a zoonotic epidemic disease caused by Japanese encephalitis virus (JEV), and currently, no medicines are available to treat this disease. Autophagy modulators play an important role in the treatment of tumors, heart disease, and some viral diseases. The aim of this study was to investigate the effects of autophagy modulators on JEV infection and the host response in mice. The experimental mice were grouped as follows: DMEM (control), JEV, JEV+rapamycin (JEV+Rapa), JEV+wortmannin (JEV+Wort), JEV+chloroquine (JEV+CQ), Rapa, Wort, and CQ. The control group was treated with DMEM. The mice in other groups were infected with 105 PFU of JEV, and Rapa, Wort, and CQ were administered 2 h prior to JEV challenge and then administered daily for 10 consecutive days. All mice were monitored for neurological signs and survival. The damage of subcellular structures in the mouse brain was evaluated by transmission electron microscopy. The distribution of virus in the mouse brain was determined by RNAScope staining and immunohistochemical staining. The neuroinflammatory responses in the brain were examined via quantitative real-time PCR, and the signal pathways involved in neuroinflammation were identified by Western blot. The mice in the JEV+Wort and JEV+CQ groups showed milder neurological symptoms, less damage to the mitochondria in the brain tissue, and a higher survival rate than those in the JEV+Rapa and JEV groups. Compared with the JEV+Rapa and JEV groups, the distribution of JEV in the brain of mice in the JEV+Wort and JEV+CQ groups was lower, and the inflammatory response was weaker. No significant difference was observed in the expression of the PI3K/AKT/NF-κB pathway in mouse brain among the different groups. Our study suggests that the autophagy inhibitors Wort and CQ reduce JEV infection and weaken the inflammatory response, which does not depend on the PI3K/AKT/NF-κB pathway in mouse brain.


Assuntos
Vírus da Encefalite Japonesa (Espécie) , Encefalite Japonesa , Animais , Autofagia , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Encefalite Japonesa/tratamento farmacológico , Inflamação/tratamento farmacológico , Camundongos , Fosfatidilinositol 3-Quinases
5.
Microbiol Spectr ; 9(3): e0166121, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34756071

RESUMO

Type I interferon (IFN-I) is a key component of the host innate immune system. To establish efficient replication, viruses have developed several strategies to escape from the host IFN response. Japanese encephalitis virus (JEV) NS1', a larger NS1-related protein, is known to inhibit the mitochondrial antiviral signaling (MAVS)-mediated IFN-ß induction by increasing the binding of transcription factors (CREB and c-Rel) to the microRNA 22 (miRNA-22) promoter. However, the mechanism by which NS1' induces the recruitment of CREB and c-Rel onto the miRNA-22 promoter is unknown. Here, we found that JEV NS1' protein interacts with the host cyclin-dependent kinase 1 (CDK1) protein. Mechanistically, NS1' interrupts the CDC25C phosphatase-mediated dephosphorylation of CDK1, which prolongs the phosphorylation status of CDK1 and leads to the inhibition of MAVS-mediated IFN-ß induction. Furthermore, the CREB phosphorylation and c-Rel activation through the IκBα phosphorylation were observed to be enhanced upon the augmentation of CDK1 phosphorylation by NS1'. The abrogation of CDK1 activity by a small-molecule inhibitor significantly suppressed the JEV replication in vitro and in vivo. Moreover, the administration of CDK1 inhibitor protected the wild-type mice from JEV-induced lethality but showed no effect on the MAVS-/- mice challenged with JEV. In conclusion, our study provides new insight into the mechanism of JEV immune evasion, which may lead to the development of novel therapeutic options to treat JEV infection. IMPORTANCE Japanese encephalitis virus (JEV) is the main cause of acute human encephalitis in Asia. The unavailability of specific treatment for Japanese encephalitis demands a better understanding of the basic cellular mechanisms that contribute to the onset of disease. The present study identifies a novel interaction between the JEV NS1' protein and the cellular CDK1 protein, which facilitates the JEV replication by dampening the cellular antiviral response. This study sheds light on a novel mechanism of JEV replication, and thus our findings could be employed for developing new therapies against JEV infection.


Assuntos
Proteína Quinase CDC2/metabolismo , Vírus da Encefalite Japonesa (Espécie)/imunologia , Evasão da Resposta Imune/imunologia , Interferon beta/imunologia , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Animais , Proteína Quinase CDC2/antagonistas & inibidores , Proteína de Ligação a CREB/metabolismo , Linhagem Celular Tumoral , Cricetinae , Encefalite Japonesa/imunologia , Células HeLa , Humanos , Imunidade Inata/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Inibidor de NF-kappaB alfa/metabolismo , Fosforilação/genética , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-rel/metabolismo , Fosfatases cdc25/metabolismo
6.
Monoclon Antib Immunodiagn Immunother ; 40(2): 60-64, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33900823

RESUMO

The DNA fragment encoding predicted main antigenic region, aa 14-245 on N protein of Rift Valley virus (RVFV) was cloned into the vector pET-28a (+) and p3xFLAG-CMV-10. The recombinant pET-28a-N1 protein was expressed in Escherichia coli BL21 (DE3) with 1 mM isopropyl-b-thio-galactopyranoside at 37°C for 5 hours, and purified by protein purifier. Three monoclonal antibodies (mAbs) named 3A5, 3A6, and 3A7 against N protein were obtained by fusing mouse myeloma cell line SP2/0 with spleen lymphocytes from pET-28a-N1 protein-immunized mice. Finally, the mAbs were characterized by enzyme-linked immunosorbent assays, indirect immunofluorescent assays, and Western blot. The results show that all the mAbs possess high specificity and react with both prokaryotic and eukaryotic N protein, which could provide important materials for the research on the function of N protein and the diagnostic methods of RVFV.


Assuntos
Anticorpos Monoclonais/imunologia , Proteínas do Nucleocapsídeo/antagonistas & inibidores , Febre do Vale de Rift/terapia , Vírus da Febre do Vale do Rift/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral , Humanos , Camundongos , Proteínas do Nucleocapsídeo/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Febre do Vale de Rift/imunologia , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/efeitos dos fármacos , Vírus da Febre do Vale do Rift/patogenicidade
7.
Virulence ; 12(1): 968-980, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33724154

RESUMO

Thousands of human deaths occur annually due to Japanese encephalitis (JE), caused by Japanese encephalitis virus. During the virus infection of the central nervous system, reactive gliosis, uncontrolled inflammatory response, and neuronal cell death are considered as the characteristic features of JE. To date, no specific treatment has been approved to overcome JE, indicating a need for the development of novel therapies. In this article, we focused on basic biological mechanisms in glial (microglia and astrocytes) and neuronal cells that contribute to the onset of neuroinflammation and neuronal cell damage during Japanese encephalitis virus infection. We also provided comprehensive knowledge about anti-JE therapies tested in clinical or pre-clinical settings, and discussed recent therapeutic strategies that could be employed for JE treatment. The improved understanding of JE pathogenesis might lay a foundation for the development of novel therapies to halt JE.Abbreviations AKT: a serine/threonine-specific protein kinase; AP1: activator protein 1; ASC: apoptosis-associated speck-like protein containing a CARD; ASK1: apoptosis signal-regulated kinase 1; ATF3/4/6: activating transcription factor 3/4/6; ATG5/7: autophagy-related 5/7; BBB: blood-brain barrier; Bcl-3/6: B-cell lymphoma 3/6 protein; CCL: C-C motif chemokine ligand; CCR2: C-C motif chemokine receptor 2; CHOP: C/EBP homologous protein; circRNA: circular RNA; CNS: central nervous system; CXCL: C-X-C motif chemokine ligand; dsRNA: double-stranded RNA; EDEM1: endoplasmic reticulum degradation enhancer mannosidase alpha-like 1; eIF2-ɑ: eukaryotic initiation factor 2 alpha; ER: endoplasmic reticulum; ERK: extracellular signal-regulated kinase; GRP78: 78-kDa glucose-regulated protein; ICAM: intercellular adhesion molecule; IFN: interferon; IL: interleukin; iNOS: inducible nitric oxide synthase; IRAK1/2: interleukin-1 receptor-associated kinase 1/2; IRE-1: inositol-requiring enzyme 1; IRF: interferon regulatory factor; ISG15: interferon-stimulated gene 15; JE: Japanese encephalitis; JEV: Japanese encephalitis virus; JNK: c-Jun N-terminal kinase; LAMP2: lysosome-associated membrane protein type 2; LC3-I/II: microtubule-associated protein 1 light chain 3-I/II; lncRNA: long non-coding RNA; MAPK: mitogen-activated protein kinase; miR/miRNA: microRNA; MK2: mitogen-activated protein kinase-activated protein kinase 2; MKK4: mitogen-activated protein kinase kinase 4; MLKL: mixed-linage kinase domain-like protein; MMP: matrix metalloproteinase; MyD88: myeloid differentiation factor 88; Nedd4: neural precursor cell-expressed developmentally downregulated 4; NF-κB: nuclear factor kappa B; NKRF: nuclear factor kappa B repressing factor; NLRP3: NLR family pyrin domain containing 3; NMDAR: N-methyl-D-aspartate receptor; NO: nitric oxide; NS2B/3/4: JEV non-structural protein 2B/3/4; P: phosphorylation. p38: mitogen-activated protein kinase p38; PKA: protein kinase A; PAK4: p21-activated kinase 4; PDFGR: platelet-derived growth factor receptor; PERK: protein kinase R-like endoplasmic reticulum kinase; PI3K: phosphoinositide 3-kinase; PTEN: phosphatase and tensin homolog; Rab7: Ras-related GTPase 7; Raf: proto-oncogene tyrosine-protein kinase Raf; Ras: a GTPase; RIDD: regulated IRE-1-dependent decay; RIG-I: retinoic acid-inducible gene I; RIPK1/3: receptor-interacting protein kinase 1/3; RNF11/125: RING finger protein 11/125; ROS: reactive oxygen species; SHIP1: SH2-containing inositol 5' phosphatase 1; SOCS5: suppressor of cytokine signaling 5; Src: proto-oncogene tyrosine-protein kinase Src; ssRNA = single-stranded RNA; STAT: signal transducer and activator of transcription; TLR: toll-like receptor; TNFAIP3: tumor necrosis factor alpha-induced protein 3; TNFAR: tumor necrosis factor alpha receptor; TNF-α: tumor necrosis factor-alpha; TRAF6: tumor necrosis factor receptor-associated factor 6; TRIF: TIR-domain-containing adapter-inducing interferon-ß; TRIM25: tripartite motif-containing 25; VCAM: vascular cell adhesion molecule; ZO-1: zonula occludens-1.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/patogenicidade , Encefalite Japonesa/complicações , Inflamação/virologia , Doenças do Sistema Nervoso/virologia , Neurônios/patologia , Animais , Apoptose , Morte Celular , Encefalite Japonesa/virologia , Chaperona BiP do Retículo Endoplasmático , Humanos , Camundongos , Neurônios/virologia , Proto-Oncogene Mas , Transdução de Sinais , Virulência
8.
Antiviral Res ; 182: 104930, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32898585

RESUMO

Japanese encephalitis virus (JEV) is one of the most important culex transmitted-flaviviruses, which can cause encephalitis in humans. Although non-structural protein 1 (NS1) of JEV does not stimulate neutralizing antibodies, this protein can provide high immunoprotection in vivo. The protective epitopes and the protective mechanism of NS1 still remain unclear. In this study, we generated five different monoclonal antibodies (mAbs) targeting the NS1 protein of JEV. In vitro experiments revealed that none of these five antibodies neutralized the JEV infection. In mouse protection studies, one of these mAbs, designated 2B8, provided a therapeutic effect against JEV lethal challenge (70% survival rate). Using peptide mapping analysis, we found that mAb 2B8 reacted with the epitope 225PETHTLWGD233 in the NS1 protein, in which any mutations among amino acid residues T228, H229, L231 or W232 could cause binding failure of 2B8 to the NS1 protein. Furthermore, mice immunized with KLH-polypeptide (225PETHTLWGD233) showed reduced mortality following JEV challenge. Collectively, we found a new protective epitope in the JEV NS1 protein. These results may facilitate the development of therapeutic agent and subunit-based vaccines based on the NS1 protein.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Vírus da Encefalite Japonesa (Espécie)/imunologia , Encefalite Japonesa/prevenção & controle , Mapeamento de Epitopos , Epitopos/imunologia , Proteínas não Estruturais Virais/imunologia , Animais , Sítios de Ligação de Anticorpos , Linhagem Celular , Cricetinae , Encefalite Japonesa/imunologia , Imunização , Rim/citologia , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/administração & dosagem , Peptídeos/imunologia , Células Sf9 , Spodoptera
9.
BMC Genomics ; 21(1): 409, 2020 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-32552669

RESUMO

BACKGROUND: Japanese encephalitis virus (JEV) is one of the common causes of acute encephalitis in humans. Japanese encephalitis is characterized by the uncontrolled release of inflammatory cytokines, which ultimately results in neuronal cell damage. In recent years, with the advancement of high-throughput sequencing technology, studies have shown that circRNAs, by competing with endogenous miRNAs, play a vital role in the pathology of CNS diseases. However, it is unknown whether circRNAs participate in JEV-induced neuroinflammation. RESULTS: By employing Illumina RNA-sequencing, we identified 180 circRNAs and 58 miRNAs that showed significant differential expression in JEV-infected mice brain tissues. The functional enrichment analyses revealed that these differentially regulated circRNAs were predominantly related to neurotransmission, histone modifications, transcription misregulation, and inflammation-associated calcium signaling pathway. Our established competing endogenous RNA (ceRNA) interaction network suggested the correlation of several circRNAs, miRNAs, and mRNAs in regulating the inflammatory response during JEV infection. Among the predicted interactions, the correlation between circ_0000220, miR-326-3p, and BCL3/MK2/TRIM25 mRNAs was experimentally validated by knockdown or overexpression of the non-coding RNA entities in cultured mouse microglia. The knockdown of circ_0000220 or overexpression of miR-326-3p caused a lower production of JEV-induced inflammatory cytokines. CONCLUSIONS: Conclusively, our study provides new insights into the host response to JEV infection and proposes the circRNA-targeting therapeutic interventions to rein in Japanese encephalitis.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/patogenicidade , Encefalite Japonesa/genética , Sequenciamento do Exoma/métodos , MicroRNAs/genética , RNA Circular/genética , Animais , Proteína 3 do Linfoma de Células B/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Microglia/química , Microglia/citologia , Proteínas Serina-Treonina Quinases/genética , Análise de Sequência de RNA/métodos , Fatores de Transcrição/genética
10.
Antiviral Res ; 179: 104810, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32360948

RESUMO

Flaviviruses are considered to be major emerging human pathogens globally. Currently available anti-flavivirus approaches are ineffective, thus there is a desperate need for broad-spectrum drugs that can be active against existing and emerging flaviviruses. Artemisinin has been found to cause an antiviral effect against several viruses; however, its antiviral effect against flaviviruses remains unexplored. Here the antiviral activity of artemisinin against flaviviruses such as JEV, DENV, and ZIKV was evaluated by measuring the hallmark features of virus replication both in vitro and in vivo. Mechanistically, the artemisinin-induced antiviral effect was associated with enhanced host type I interferon response. The blocking of interferon signaling inhibited the artemisinin-induced interferon-stimulated genes expression and rescued the artemisinin-suppressed virus replication. This study demonstrated for the first time the antiviral activity of artemisinin against flaviviruses with a novel antiviral mechanism. The therapeutic application of artemisinin may constitute a broad-spectrum approach to cure infections caused by flaviviruses.


Assuntos
Antivirais/farmacologia , Artemisininas/farmacologia , Flavivirus/efeitos dos fármacos , Interferon Tipo I/imunologia , Replicação Viral/efeitos dos fármacos , Células A549 , Animais , Descoberta de Drogas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Flavivirus/classificação , Humanos , Camundongos Endogâmicos C57BL , Alvéolos Pulmonares/citologia
11.
J Virol ; 94(15)2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32461319

RESUMO

Japanese encephalitis virus (JEV) infection alters microRNA (miRNA) expression in the central nervous system (CNS). However, the mechanism contributing to miRNA regulation in the CNS is not known. We discovered global degradation of mature miRNA in mouse brains and neuroblastoma (NA) cells after JEV infection. Integrative analysis of miRNAs and mRNAs suggested that several significantly downregulated miRNAs and their targeted mRNAs were clustered into an inflammation pathway. Transfection with miRNA 466d-3p (miR-466d-3p) decreased interleukin-1ß (IL-1ß) expression and inhibited JEV replication in NA cells. However, miR-466d-3p expression increased after JEV infection in the presence of cycloheximide, indicating that viral protein expression reduced miR-466d-3p expression. We generated all the JEV coding proteins and demonstrated NS3 helicase protein to be a potent miRNA suppressor. The NS3 proteins of Zika virus, West Nile virus, and dengue virus serotype 1 (DENV-1) and DENV-2 also decreased miR-466d-3p expression. Results from helicase-blocking assays and in vitro unwinding assays demonstrated that NS3 could unwind pre-miR-466d and induce miRNA dysfunction. Computational models and an RNA immunoprecipitation assay revealed arginine-rich domains of NS3 to be crucial for pre-miRNA binding and degradation of host miRNAs. Importantly, site-directed mutagenesis of conserved residues in NS3 revealed that R226G and R202W reduced the binding affinity and degradation of pre-miR-466d. These results expand the function of flavivirus helicases beyond unwinding duplex RNA to degrade pre-miRNAs. Hence, we revealed a new mechanism for NS3 in regulating miRNA pathways and promoting neuroinflammation.IMPORTANCE Host miRNAs have been reported to regulate JEV-induced inflammation in the CNS. We found that JEV infection could reduce expression of host miRNA. The helicase region of the NS3 protein bound specifically to miRNA precursors and could lead to incorrect unwinding of miRNA precursors, thereby reducing the expression of mature miRNAs. This observation led to two major findings. First, our results suggested that JEV NS3 protein induced miR-466d-3p degradation, which promoted IL-1ß expression and JEV replication. Second, arginine molecules on NS3 were the main miRNA-binding sites, because we demonstrated that miRNA degradation was abolished if arginines at R226 and R202 were mutated. Our study provides new insights into the molecular mechanism of JEV and reveals several amino acid sites that could be mutated for a JEV vaccine.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/fisiologia , Regulação da Expressão Gênica , Interleucina-1beta/biossíntese , MicroRNAs/metabolismo , Estabilidade de RNA , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Animais , Linhagem Celular Tumoral , Cricetinae , Mesocricetus , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , RNA Helicases/genética , RNA Helicases/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Proteínas não Estruturais Virais/genética
12.
Monoclon Antib Immunodiagn Immunother ; 39(1): 12-16, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32004438

RESUMO

The DNA fragment encoding predicted main antigenic region, aa 1-300 on Gp protein of Ebola virus (EBOV) was cloned into the vector pGEX-KG. The recombinant GST-tagged Gp-300 was expressed in Escherichia coli BL21 (DE3) by induction with 1 mM isopropyl-1-thio-b-d-galactoside and purified by dialysis. Four monoclonal antibodies (mAbs) named 1C4, 2A3, 2G7, and 2H9 against Gp protein were generated by fusing mouse myeloma cell line SP2/0 with spleen lymphocytes from Gp-300 protein-immunized mice. The activity of the mAbs was then characterized by enzyme-linked immunosorbent assay, indirect immunofluorescent assays (IFA), and western blot analysis. The results demonstrated that all the mAbs showed high specificity and sensitivity in IFA and in western blot analysis, which indicated that these mAbs against Gp protein of EBOV may be used as valuable tools for analysis of the protein functions and pathogenesis of EBOV.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Ebolavirus/imunologia , Proteínas do Envelope Viral/antagonistas & inibidores , Animais , Ensaio de Imunoadsorção Enzimática , Técnica Indireta de Fluorescência para Anticorpo , Camundongos , Camundongos Endogâmicos BALB C
13.
Viruses ; 11(11)2019 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-31683742

RESUMO

Flaviviruses, such as Zika virus (ZIKV), Japanese encephalitis virus (JEV), Dengue virus (DENV), and West Nile virus (WNV), are important arthropod-borne pathogens that present an immense global health problem. Their unpredictable disease severity, unusual clinical features, and severe neurological manifestations underscore an urgent need for antiviral interventions. Furin, a host proprotein convertase, is a key contender in processing flavivirus prM protein to M protein, turning the inert virus to an infectious particle. For this reason, the current study was planned to evaluate the antiviral activity of decanoyl-Arg-Val-Lys-Arg-chloromethylketone, a specific furin inhibitor, against flaviviruses, including ZIKV and JEV. Analysis of viral proteins revealed a significant increase in the prM/E index of ZIKV or JEV in dec-RVKR-cmk-treated Vero cells compared to DMSO-treated control cells, indicating dec-RVKR-cmk inhibits prM cleavage. Plaque assay, qRT-PCR, and immunofluorescence assay revealed a strong antiviral activity of dec-RVKR-cmk against ZIKV and JEV in terms of the reduction in virus progeny titer and in viral RNA and protein production in both mammalian cells and mosquito cells. Time-of-drug addition assay revealed that the maximum reduction of virus titer was observed in post-infection treatment. Furthermore, our results showed that dec-RVKR-cmk exerts its inhibitory action on the virus release and next round infectivity but not on viral RNA replication. Taken together, our study highlights an interesting antiviral activity of dec-RVKR-cmk against flaviviruses.


Assuntos
Clorometilcetonas de Aminoácidos/farmacologia , Flavivirus/efeitos dos fármacos , Furina/antagonistas & inibidores , Proteínas do Envelope Viral , Animais , Antivirais/farmacologia , Linhagem Celular , Chlorocebus aethiops , Vírus da Encefalite Japonesa (Espécie)/efeitos dos fármacos , Furina/metabolismo , Células Vero , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/metabolismo , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Zika virus/efeitos dos fármacos
14.
Vet Microbiol ; 238: 108430, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31648727

RESUMO

Japanese Encephalitis Virus (JEV) is an important zoonotic flavivirus transmitted by mosquitos. JEV infection in sows primarily manifests as a reproductive disease such as abortion and transient infertility while in infected boars, it can cause orchitis. Previous studies mainly focused on the pathogenesis of human encephalitis caused by JEV infection, while few concentrations have been made to unveil the potential mechanism of reproductive dysfunction in JEV-infected pigs. In this study, histopathological analysis and immunohistochemistry staining was performed on testis of JEV-infected boars, indicating that JEV could infect testicular cells and cause inflammatory changes in testis. In vitro assays reveal that primary swine testicular cells and swine testis (ST) cells are highly permissive to JEV and significant inflammatory response was shown during JEV infection. Mechanically, we found that JEV infection increases the expression of retinoic acid-inducible gene I (RIG-I) and activates transcription factor NF-κB. Production of pro-inflammatory cytokines was greatly reduced in JEV infected testicular cells after knockout of RIG-I or treatment with the NF-κB specific inhibitor. In addition, activation of NF-κB was also significantly suppressed upon RIG-I knockout. Taken together, our results reveal that JEV could infect boar testicles, and RIG-I-NF-κB signaling pathway is involved in JEV-induced inflammation in swine testicular cells.


Assuntos
Proteína DEAD-box 58/metabolismo , Encefalite Japonesa/veterinária , NF-kappa B/metabolismo , Orquite/veterinária , Sus scrofa , Doenças dos Suínos/fisiopatologia , Animais , Células Cultivadas , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Encefalite Japonesa/complicações , Encefalite Japonesa/fisiopatologia , Técnicas In Vitro , Inflamação , Masculino , Orquite/etiologia , Transdução de Sinais/imunologia , Suínos , Doenças dos Suínos/virologia
15.
J Colloid Interface Sci ; 542: 198-206, 2019 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-30739009

RESUMO

Multiple viruses can cause infection and death of millions annually. Of these, flaviviruses are found to be highly prevalent in recent years with no distinctive antiviral therapies. Therefore, there is a desperate need for broad-spectrum antiviral drugs that can be active against a large number of existing and emerging viruses. Herein, we prepared a kind of benzoxazine monomer derived carbon dots (BZM-CDs) and demonstrated their infection-blocking ability against life-threatening flaviviruses (Japanese encephalitis, Zika, and dengue viruses) and non-enveloped viruses (porcine parvovirus and adenovirus-associated virus). It was found that BZM-CDs could directly bind to the surface of the virion, and eventually the first step of virus-cell interaction was impeded. The developed nanoparticles are active against both flaviviruses and non-enveloped viruses in vitro. Thus, the application of BZM-CDs may constitute an intriguing broad-spectrum approach to rein in viral infections.


Assuntos
Antivirais/farmacologia , Benzoxazinas/farmacologia , Carbono/química , Nanopartículas/química , Pontos Quânticos , Animais , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Cricetinae , Flavivirus/efeitos dos fármacos , Células HEK293 , Humanos , Tamanho da Partícula , Propriedades de Superfície , Células Vero , Vírion/efeitos dos fármacos
16.
Virol Sin ; 33(6): 515-523, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30542978

RESUMO

Japanese encephalitis virus (JEV) is a mosquito-borne virus and the major cause of viral encephalitis in Asia. NS1', a 52-amino acid C-terminal extension of NS1, is generated with a -1 programmed ribosomal frameshift and is only present in members of the Japanese encephalitis serogroup of flaviviruses. Previous studies demonstrated that NS1' plays a vital role in virulence, but the mechanism is unclear. In this study, an NS1' defected (rG66A) virus was generated. We found that rG66A virus was less virulent than its parent virus (pSA14) in wild-type mice. However, similar mortality caused by the two viruses was observed in an IFNAR knockout mouse model. Moreover, we found that rG66A virus induced a greater type I interferon (IFN) response than that by pSA14, and JEV NS1' significantly inhibited the production of IFN-ß and IFN-stimulated genes. Taken together, our results reveal that NS1' plays a vital role in blocking type I IFN production to help JEV evade antiviral immunity and benefit viral replication.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/patogenicidade , Encefalite Japonesa/imunologia , Interferon beta/antagonistas & inibidores , Proteínas não Estruturais Virais/imunologia , Células A549 , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Feminino , Células HeLa , Humanos , Evasão da Resposta Imune , Interferon beta/imunologia , Camundongos , Camundongos Knockout , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Células Vero , Virulência , Replicação Viral
17.
Front Immunol ; 9: 1148, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29910805

RESUMO

Japanese encephalitis is a neuropathological disorder caused by Japanese encephalitis virus (JEV), which is characterized by severe pathological neuroinflammation and damage to the blood-brain barrier (BBB). Inflammatory cytokines/chemokines can regulate the expression of tight junction (TJ) proteins and are believed to be a leading cause of BBB disruption, but the specific mechanisms remain unclear. IP-10 is the most abundant chemokine produced in the early stage of JEV infection, but its role in BBB disruption is unknown. The administration of IP-10-neutralizing antibody ameliorated the decrease in TJ proteins and restored BBB integrity in JEV-infected mice. In vitro study showed IP-10 and JEV treatment did not directly alter the permeability of the monolayers of endothelial cells. However, IP-10 treatment promoted tumor necrosis factor alpha (TNF-α) production and IP-10-neutralizing antibody significantly reduced the production of TNF-α. Thus, TNF-α could be a downstream cytokine of IP-10, which decreased TJ proteins and damaged BBB integrity. Further study indicated that JEV infection can stimulate upregulation of the IP-10 receptor CXCR3 on astrocytes, resulting in TNF-α production through the JNK-c-Jun signaling pathway. Consequently, TNF-α affected the expression and cellular distribution of TJs in brain microvascular endothelial cells and led to BBB damage during JEV infection. Regarding regulation of the BBB, the IP-10/TNF-α cytokine axis could be considered a potential target for the development of novel therapeutics in BBB-related neurological diseases.


Assuntos
Barreira Hematoencefálica/metabolismo , Quimiocina CXCL10/metabolismo , Encefalite Japonesa/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Animais , Astrócitos/metabolismo , Astrócitos/virologia , Linhagem Celular , Modelos Animais de Doenças , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Encefalite Japonesa/etiologia , Endotélio/metabolismo , Feminino , Expressão Gênica , Sistema de Sinalização das MAP Quinases , Camundongos , Permeabilidade , Fator de Necrose Tumoral alfa/genética
18.
Artigo em Inglês | MEDLINE | ID: mdl-28680855

RESUMO

Japanese encephalitis virus (JEV) targets central nervous system, resulting in neuroinflammation with typical features of neuronal death along with hyper activation of glial cells. Exploring the mechanisms responsible for the JEV-caused inflammatory response remains a pivotal area of research. In the present study, we have explored the function of p21-activated kinase 4 (PAK4) in JEV-mediated inflammatory response in human astrocytes. The results showed that JEV infection enhances the phosphorylation of PAK4 in U251 cells and mouse brain. Knockdown of PAK4 resulted in decreased expression of inflammatory cytokines that include tumor necrosis factor alpha, interleukin-6, interleukin-1ß, and chemokine (C-C motif) ligand 5 and interferon ß upon JEV infection, suggesting that PAK4 signaling promotes JEV-mediated inflammation. In addition, we found that knockdown of PAK4 led to the inhibition of MAPK signaling including ERK, p38 MAPK and JNK, and also resulted in the reduced nuclear translocation of NF-κB and phosphorylation of AP-1. These results demonstrate that PAK4 signaling actively promotes JEV-mediated inflammation in human astrocytes via MAPK-NF-κB/AP-1 pathway, which will provide a new insight into the molecular mechanism of the JEV-induced inflammatory response.


Assuntos
Astrócitos/imunologia , Vírus da Encefalite Japonesa (Espécie)/imunologia , Encefalite Japonesa/imunologia , Inflamação/imunologia , Transdução de Sinais , Quinases Ativadas por p21/imunologia , Animais , Astrócitos/virologia , Encéfalo/imunologia , Encéfalo/virologia , Linhagem Celular , Citocinas/metabolismo , Proteína DEAD-box 58/metabolismo , Encefalite Japonesa/virologia , Técnicas de Silenciamento de Genes , Glioma/imunologia , Humanos , Inflamação/metabolismo , Interferon beta/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Camundongos , NF-kappa B/metabolismo , Fosforilação , Receptor 3 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Quinases Ativadas por p21/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Biosci Rep ; 37(1)2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-27986864

RESUMO

Inflammation is the body's normal self-protection mechanism to eliminate pathogens and resist pathogen invasion. The excessive inflammatory response may lead to inflammatory lesions. The mechanisms accounting for inflammation remain hazy. miRNAs have been proposed to have crucial effects on inflammation. In the present study, we reported that lipopolysaccharide (LPS)-stimulation increased the expression levels of inflammatory cytokines and the cell-cycle progression was suppressed in RAW264.7 cells. Meanwhile, the expression of miR-322 was significantly down-regulated after LPS treatment. Bioinformatics predictions revealed a potential binding site of miR-322 in 3'-UTR of NF-κB1 (p50) and it was further confirmed by luciferase assay. Moreover, both the mRNA and protein levels of NF-κB1 (p50) were down-regulated by miR-322 in RAW264.7 cells. Subsequently, we demonstrated that miR-322 mimics decrease in the expression levels of inflammatory cytokines and cell-cycle repression can be rescued following LPS treatment in RAW264.7 cells. The anti-inflammatory cytokines expression including IL-4 and IL-10 were significantly up-regulated. Furthermore, miR-322 could also promote RAW264.7 cells proliferation. These results demonstrate that miR-322 is a negative regulator of inflammatory response by targeting NF-κB1 (p50).


Assuntos
Proliferação de Células , Inflamação/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Macrófagos/metabolismo , MicroRNAs/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Regiões 3' não Traduzidas , Animais , Ciclo Celular , Regulação para Baixo , Lipopolissacarídeos , Macrófagos/citologia , Camundongos , MicroRNAs/genética , Células RAW 264.7 , RNA Mensageiro/metabolismo
20.
Oncotarget ; 7(47): 76667-76683, 2016 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-27705941

RESUMO

MicroRNAs (miRNAs) are small non-coding RNAs that play important roles in regulating the host immune response. Here we found that miR-22 is induced in glial cells upon stimulation with poly(I:C). Overexpression of miR-22 in the cultured cells resulted in decreased activity of interferon regulatory factor-3 and nuclear factor-kappa B, which in turn led to reduced expression of interferon-ß and inflammatory cytokines, including tumor necrosis factor-α, interleukin-1ß, interleukin-6, and chemokine (C-C motif) ligand 5, upon stimulation with poly(I:C), whereas knockdown of miR-22 had the opposite effect. We used a combination of bioinformatics and experimental techniques to demonstrate that mitochondrial antiviral signaling protein (MAVS), which positively regulates type I interferon production, is a novel target of miR-22. Overexpression of miR-22 decreased the activity of a luciferase reporter containing the MAVS 3'-untranslated region and led to decreased MAVS mRNA and protein levels. In contrast, ectopic expression of miR-22 inhibitor led to elevated MAVS expression. Collectively, our results demonstrate that miR-22 negatively regulates poly(I:C)-induced production of type I interferon and inflammatory cytokines via targeting MAVS.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Citocinas/biossíntese , Mediadores da Inflamação/metabolismo , Interferon Tipo I/biossíntese , MicroRNAs/genética , Poli I-C/farmacologia , Linhagem Celular Tumoral , Células Cultivadas , Vírus da Encefalite Japonesa (Espécie) , Expressão Gênica , Regulação da Expressão Gênica , Genes Reporter , Interações Hospedeiro-Patógeno/genética , Humanos , Fator Regulador 3 de Interferon/metabolismo , NF-kappa B/metabolismo , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA