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











Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(8): e1012409, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39146232

RESUMO

Flaviviridae is a family of positive-stranded RNA viruses, including human pathogens, such as Japanese encephalitis virus (JEV), dengue virus (DENV), Zika virus (ZIKV), and West Nile virus (WNV). Nuclear localization of the viral core protein is conserved among Flaviviridae, and this feature may be targeted for developing broad-ranging anti-flavivirus drugs. However, the mechanism of core protein translocation to the nucleus and the importance of nuclear translocation in the viral life cycle remain unknown. We aimed to identify the molecular mechanism underlying core protein nuclear translocation. We identified importin-7 (IPO7), an importin-ß family protein, as a nuclear carrier for Flaviviridae core proteins. Nuclear import assays revealed that core protein was transported into the nucleus via IPO7, whereas IPO7 deletion by CRISPR/Cas9 impaired their nuclear translocation. To understand the importance of core protein nuclear translocation, we evaluated the production of infectious virus or single-round-infectious-particles in wild-type or IPO7-deficient cells; both processes were significantly impaired in IPO7-deficient cells, whereas intracellular infectious virus levels were equivalent in wild-type and IPO7-deficient cells. These results suggest that IPO7-mediated nuclear translocation of core proteins is involved in the release of infectious virus particles of flaviviruses.


Assuntos
Transporte Ativo do Núcleo Celular , Núcleo Celular , Flavivirus , Humanos , Flavivirus/metabolismo , Flavivirus/fisiologia , Animais , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Replicação Viral/fisiologia , Proteínas do Core Viral/metabolismo , Proteínas do Core Viral/genética , Carioferinas/metabolismo , Carioferinas/genética , Infecções por Flavivirus/metabolismo , Infecções por Flavivirus/virologia , Chlorocebus aethiops , Células HEK293
2.
J Virol ; 98(9): e0063524, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39158346

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por AMP , Antivirais , Vírus da Encefalite Japonesa (Espécie) , Encefalite Japonesa , Metabolismo dos Lipídeos , Replicação Viral , Animais , Metabolismo dos Lipídeos/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Vírus da Encefalite Japonesa (Espécie)/efeitos dos fármacos , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Camundongos , Antivirais/farmacologia , Humanos , Encefalite Japonesa/tratamento farmacológico , Encefalite Japonesa/virologia , Proteínas Quinases Ativadas por AMP/metabolismo , Chalconas/farmacologia , Triterpenos/farmacologia , Proteínas não Estruturais Virais/metabolismo , Infecções por Flavivirus/tratamento farmacológico , Infecções por Flavivirus/virologia , Infecções por Flavivirus/metabolismo , Flavivirus/efeitos dos fármacos , Linhagem Celular
3.
Viruses ; 13(5)2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-34066055

RESUMO

Arthropod-borne flaviviruses, such as Zika virus (ZIKV), Usutu virus (USUV), and West Nile virus (WNV), are a growing cause of human illness and death around the world. Presently, no licensed antivirals to control them are available and, therefore, search for broad-spectrum antivirals, including host-directed compounds, is essential. The PI3K/Akt pathway controls essential cellular functions involved in cell metabolism and proliferation. Moreover, Akt has been found to participate in modulating replication in different viruses including the flaviviruses. In this work we studied the interaction of flavivirus NS5 polymerases with the cellular kinase Akt. In vitro NS5 phosphorylation experiments with Akt showed that flavivirus NS5 polymerases are phosphorylated and co-immunoprecipitate by Akt. Polymerase activity assays of Ala- and Glu-generated mutants for the Akt-phosphorylated residues also indicate that Glu mutants of ZIKV and USUV NS5s present a reduced primer-extension activity that was not observed in WNV mutants. Furthermore, treatment with Akt inhibitors (MK-2206, honokiol and ipatasertib) reduced USUV and ZIKV titers in cell culture but, except for honokiol, not WNV. All these findings suggest an important role for Akt in flavivirus replication although with specific differences among viruses and encourage further investigations to examine the PI3K/Akt/mTOR pathway as an antiviral potential target.


Assuntos
Infecções por Flavivirus/metabolismo , Infecções por Flavivirus/virologia , Flavivirus/fisiologia , Interações Hospedeiro-Patógeno , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Animais , Flavivirus/efeitos dos fármacos , Genoma Viral , Humanos , Mutação , Fases de Leitura Aberta , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Ligação Proteica , Proteoma , Proteômica/métodos , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Proteínas não Estruturais Virais/genética , Vírus do Nilo Ocidental/fisiologia , Zika virus/fisiologia , Infecção por Zika virus/metabolismo , Infecção por Zika virus/virologia
4.
J Virol ; 94(11)2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32161171

RESUMO

Flaviviruses encode one, two, or no N-linked glycosylation sites on their envelope proteins. Glycosylation can impact virus interactions with cell surface attachment factors and also may impact virion stability and virus replication. Envelope protein glycosylation has been identified as a virulence determinant for multiple flaviviruses, but the mechanisms by which glycosylation mediates pathogenesis remain unclear. In this Gem, we summarize current knowledge on flavivirus envelope protein glycosylation and its impact on viral infection and pathogenesis.


Assuntos
Infecções por Flavivirus/metabolismo , Flavivirus , Proteínas do Envelope Viral/metabolismo , Replicação Viral , Animais , Flavivirus/patogenicidade , Flavivirus/fisiologia , Infecções por Flavivirus/patologia , Glicosilação , Humanos
5.
J Interferon Cytokine Res ; 40(1): 1-18, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31436502

RESUMO

Flaviviruses cause significant human diseases putting more than 400 million people at risk annually worldwide. Because of migration and improved transportation, these viruses can be found on all continents (except Antarctica). Although a majority of the viruses are endemic in the tropics, a few [West Nile virus (WNV) and tick-borne encephalitis virus (TBEV)] have shown endemicity in Europe and North America. Currently, there are vaccines for the Yellow fever virus, Japanese encephalitis virus, and TBEV, but there is no effective vaccine and/or therapy against all other flaviviruses. Although there are intensive efforts to develop vaccines for Zika viruses, dengue viruses, and WNVs, there is the need for alternative or parallel antiviral therapeutic approaches. Suppressors of cytokine signaling (SOCS) and protein inhibitors of activated signal transducer and activator of transcription (STATs; PIAS), both regulatory proteins of the Janus kinase/STAT signaling pathway, have been explored as therapeutic targets in herpes simplex and vaccinia viruses, as well as in cancer therapy. In this review, we briefly describe the function of SOCS and PIAS and their therapeutic potential in flaviviral infections. [Figure: see text].


Assuntos
Infecções por Flavivirus/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Transativadores/metabolismo , Animais , Humanos , Transdução de Sinais
6.
J Virol ; 93(22)2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31462564

RESUMO

In mice, resistance to central nervous system (CNS) disease induced by members of the genus Flavivirus is conferred by an allele of the 2'-5' oligoadenylate synthetase 1b gene that encodes the inactive full-length protein (Oas1b-FL). The susceptibility allele encodes a C-terminally truncated protein (Oas1b-tr). We show that the efficiency of neuron infection in the brains of resistant and susceptible mice is similar after an intracranial inoculation of two flaviviruses, but amplification of viral proteins and double-stranded RNA (dsRNA) is inhibited in infected neurons in resistant mouse brains at later times. Active OAS proteins detect cytoplasmic dsRNA and synthesize short 2'-5'-linked oligoadenylates (2'-5'A) that interact with the latent endonuclease RNase L, causing it to dimerize and cleave single-stranded RNAs. To evaluate the contribution of RNase L to the resistance phenotype in vivo, we created a line of resistant RNase L-/- mice. Evidence of RNase L activation in infected RNase L+/+ mice was indicated by higher levels of viral RNA in the brains of infected RNase L-/- mice. Activation of type I interferon (IFN) signaling was detected in both resistant and susceptible brains, but Oas1a and Oas1b mRNA levels were lower in RNase L+/+ mice of both types, suggesting that activated RNase L also has a proflaviviral effect. Inhibition of virus replication was robust in resistant RNase L-/- mice, indicating that activated RNase L is not a critical factor in mediating this phenotype.IMPORTANCE The mouse genome encodes a family of Oas proteins that synthesize 2'-5'A in response to dsRNA. 2'-5'A activates the endonuclease RNase L to cleave single-stranded viral and cellular RNAs. The inactive, full-length Oas1b protein confers flavivirus-specific disease resistance. Although similar numbers of neurons were infected in resistant and susceptible brains after an intracranial virus infection, viral components amplified only in susceptible brains at later times. A line of resistant RNase L-/- mice was used to evaluate the contribution of RNase L to the resistance phenotype in vivo Activation of RNase L antiviral activity by flavivirus infection was indicated by increased viral RNA levels in the brains of RNase L-/- mice. Oas1a and Oas1b mRNA levels were higher in infected RNase L-/- mice, indicating that activated RNase L also have a proflaviviral affect. However, the resistance phenotype was equally robust in RNase L-/- and RNase L+/+ mice.


Assuntos
2',5'-Oligoadenilato Sintetase/metabolismo , Endorribonucleases/metabolismo , Infecções por Flavivirus/metabolismo , 2',5'-Oligoadenilato Sintetase/fisiologia , Nucleotídeos de Adenina/genética , Nucleotídeos de Adenina/metabolismo , Animais , Linhagem Celular , Endorribonucleases/genética , Endorribonucleases/fisiologia , Flavivirus/metabolismo , Infecções por Flavivirus/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligorribonucleotídeos/genética , Oligorribonucleotídeos/metabolismo , Fenótipo , RNA Viral/metabolismo , Ribonucleases/genética , Ribonucleases/metabolismo , Replicação Viral/efeitos dos fármacos
7.
J Virol ; 93(22)2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31462573

RESUMO

Avian Tembusu virus (TMUV) is a newly emerging avian pathogenic flavivirus in China and Southeast Asia with features of rapid spread, an expanding host range, and cross-species transmission. The mechanisms of its infection and pathogenesis remain largely unclear. Here, we investigated the tropism of this arbovirus in peripheral blood mononuclear cells of specific-pathogen-free (SPF) ducks and SPF chickens and identified monocytes/macrophages as the key targets of TMUV infection. In vivo studies in SPF ducks and SPF chickens with monocyte/macrophage clearance demonstrated that the infection of monocytes/macrophages was crucial for viral replication, transmission, and pathogenesis. Further genome-wide transcriptome analyses of TMUV-infected chicken macrophages revealed that host antiviral innate immune barriers were the major targets of TMUV in macrophages. Despite the activation of major pattern recognition receptor signaling, the inductions of alpha interferon (IFN-α) and IFN-ß were blocked by TMUV infection on transcription and translation levels, respectively. Meanwhile, TMUV inhibited host redox responses by repressing the transcription of genes encoding NADPH oxidase subunits and promoting Nrf2-mediated antioxidant responses. The recovery of either of the above-mentioned innate immune barriers was sufficient to suppress TMUV infection. Collectively, we identify an essential step of TMUV infection and reveal extensive subversion of host antiviral innate immune responses.IMPORTANCE Mosquito-borne flaviviruses include a group of pathogenic viruses that cause serious diseases in humans and animals, including dengue, West Nile, and Japanese encephalitis viruses. These flaviviruses are zoonotic and use animals, including birds, as amplifying and reservoir hosts. Avian Tembusu virus (TMUV) is an emerging mosquito-borne flavivirus that is pathogenic for many avian species and can infect cells derived from mammals and humans in vitro Although not currently pathogenic for primates, the infection of duck industry workers and the potential risk of TMUV infection in immunocompromised individuals have been highlighted. Thus, the prevention of TMUV in flocks is important for both avian and mammalian health. Our study reveals the escape of TMUV from the first line of the host defense system in the arthropod-borne transmission route of arboviruses, possibly helping to extend our understanding of flavivirus infection in birds and refine the design of anti-TMUV therapeutics.


Assuntos
Infecções por Flavivirus/imunologia , Flavivirus/imunologia , Animais , Galinhas/virologia , China , Patos/virologia , Feminino , Flavivirus/metabolismo , Infecções por Flavivirus/metabolismo , Especificidade de Hospedeiro/genética , Interações Hospedeiro-Patógeno/genética , Imunidade Inata/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Doenças das Aves Domésticas/virologia , Replicação Viral/efeitos dos fármacos
8.
J Virol ; 93(15)2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31118262

RESUMO

Schlafen 11 (Slfn11) is an interferon-stimulated gene that controls the synthesis of proteins by regulating tRNA abundance. Likely through this mechanism, Slfn11 has previously been shown to impair human immunodeficiency virus type 1 (HIV-1) infection and the expression of codon-biased open reading frames. Because replication of positive-sense single-stranded RNA [(+)ssRNA] viruses requires the immediate translation of the incoming viral genome, whereas negative-sense single-stranded RNA [(-)ssRNA] viruses carry at infection an RNA replicase that makes multiple translation-competent copies of the incoming viral genome, we reasoned that (+)ssRNA viruses will be more sensitive to the effect of Slfn11 on protein synthesis than (-)ssRNA viruses. To evaluate this hypothesis, we tested the effects of Slfn11 on the replication of a panel of ssRNA viruses in the human glioblastoma cell line A172, which naturally expresses Slfn11. Depletion of Slfn11 significantly increased the replication of (+)ssRNA viruses from the Flavivirus genus, including West Nile virus (WNV), dengue virus (DENV), and Zika virus (ZIKV), but had no significant effect on the replication of the (-)ssRNA viruses vesicular stomatitis virus (VSV) (Rhabdoviridae family) and Rift Valley fever virus (RVFV) (Phenuiviridae family). Quantification of the ratio of genome-containing viral particles to PFU indicated that Slfn11 impairs WNV infectivity. Intriguingly, Slfn11 prevented WNV-induced downregulation of a subset of tRNAs implicated in the translation of 11.8% of the viral polyprotein. Low-abundance tRNAs might promote optimal protein folding and enhance viral infectivity, as previously reported. In summary, this study demonstrates that Slfn11 restricts flavivirus replication by impairing viral infectivity.IMPORTANCE We provide evidence that the cellular protein Schlafen 11 (Slfn11) impairs replication of flaviviruses, including West Nile virus (WNV), dengue virus (DENV), and Zika virus (ZIKV). However, replication of single-stranded negative RNA viruses was not affected. Specifically, Slfn11 decreases the infectivity of WNV potentially by preventing virus-induced modifications of the host tRNA repertoire that could lead to enhanced viral protein folding. Furthermore, we demonstrate that Slfn11 is not the limiting factor of this novel broad antiviral pathway.


Assuntos
Infecções por Flavivirus/metabolismo , Flavivirus/fisiologia , Interações Hospedeiro-Patógeno/genética , Proteínas Nucleares/metabolismo , Replicação Viral , Linhagem Celular , Flavivirus/patogenicidade , Infecções por Flavivirus/virologia , Expressão Gênica , Técnicas de Silenciamento de Genes , Genoma Viral , Humanos , Interferon Tipo I/metabolismo , Cinética , Mutagênese Sítio-Dirigida , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Vírus de RNA/patogenicidade , Vírus de RNA/fisiologia , RNA de Transferência/genética , RNA de Transferência/metabolismo
9.
J Infect Dis ; 219(12): 2015-2025, 2019 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-30715407

RESUMO

Rocio virus (ROCV) is a highly neuropathogenic mosquito-transmitted flavivirus responsible for an unprecedented outbreak of human encephalitis during 1975-1976 in Sao Paulo State, Brazil. Previous studies have shown an increased number of inflammatory macrophages in the central nervous system (CNS) of ROCV-infected mice, implying a role for macrophages in the pathogenesis of ROCV. Here, we show that ROCV infection results in increased expression of CCL2 in the blood and in infiltration of macrophages into the brain. Moreover, we show, using CCR2 knockout mice, that CCR2 expression is essential for macrophage infiltration in the brain during ROCV infection and that the lack of CCR2 results in increased disease severity and mortality. Thus, our findings show the protective role of CCR2-mediated infiltration of macrophages in the brain during ROCV infection.


Assuntos
Encefalite/metabolismo , Infecções por Flavivirus/metabolismo , Flavivirus/patogenicidade , Macrófagos/metabolismo , Receptores CCR2/metabolismo , Animais , Encéfalo , Brasil , Encefalite/virologia , Feminino , Infecções por Flavivirus/virologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
10.
Cell Chem Biol ; 25(8): 1006-1016.e8, 2018 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-29937406

RESUMO

Viral envelope proteins are required for productive viral entry and initiation of infection. Although the humoral immune system provides ample evidence for targeting envelope proteins as an antiviral strategy, there are few pharmacological interventions that have this mode of action. In contrast to classical antiviral targets such as viral proteases and polymerases, viral envelope proteins as a class do not have a well-conserved active site that can be rationally targeted with small molecules. We previously identified compounds that inhibit dengue virus by binding to its envelope protein, E. Here, we show that these small molecules inhibit dengue virus fusion and map the binding site of these compounds to a specific pocket on E. We further demonstrate inhibition of Zika, West Nile, and Japanese encephalitis viruses by these compounds, providing pharmacological evidence for the pocket as a target for developing broad-spectrum antivirals against multiple, mosquito-borne flavivirus pathogens.


Assuntos
Antivirais/química , Antivirais/farmacologia , Infecções por Flavivirus/tratamento farmacológico , Flavivirus/efeitos dos fármacos , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Linhagem Celular , Sequência Conservada , Vírus da Dengue/química , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/fisiologia , Descoberta de Drogas , Flavivirus/química , Flavivirus/fisiologia , Infecções por Flavivirus/metabolismo , Infecções por Flavivirus/virologia , Humanos , Simulação de Acoplamento Molecular , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas do Envelope Viral/química , Replicação Viral/efeitos dos fármacos , Zika virus/química , Zika virus/efeitos dos fármacos , Zika virus/fisiologia
11.
Infect Genet Evol ; 63: 277-284, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29883772

RESUMO

The NS1protein, a nonstructural protein of Tembusu virus, plays a key role in the pathogenesis of TMUV. To research host proteins that interact with NS1 protein, the cDNA library of duck embryo fibroblasts (DEF) was successfully constructed. The recombinant plasmid, pGBKT7-NS1, was transformed into the yeast Y2H to be cloned and tested for autoactivation and toxicity. The autoactivation and toxicity test of bait showed that the yeast two hybrid test could be carried out normally. A total of 7 clones from the library were got by Yeast Two-Hybrid System, and 4 proteins, including RPS7, MORC3, GABARAPL1 and MTSS1, may be interacted with DTMUV NS1 after sequencing and blast. Then we chose the host protein of RPS7 for GST pull down assay and the recombinant plasmid of pGEX-6p-1-NS1and pEGFP-RPS7 were constructed. Then the proteins of GST-NS1 and GFP-RPS7 were successfully expressed in vitro for GST pull down assay. The results showed that there was a real interaction between the two proteins when the protein of GST-NS1-GFP-RPS7 was obtained eventually. The Real-time RT-PCR was used to detect the expression level of RPS7, MDM2 and P53 mRNA after the recombinant plasmid of pEGFP-NS1 was expressed in 293 T cells. It is showed that the expression of NS1 protein causes the low expression of RPS7 and MDM2 mRNA and eventually causes the high expression of P53 mRNA. This research lays the foundation for clarifying the pathogenic mechanism of Tembusu virus and the function of NS1 protein in virus propagation process.


Assuntos
Proteínas Aviárias/genética , Fibroblastos/virologia , Flavivirus/genética , Interações Hospedeiro-Patógeno/genética , Mapeamento de Interação de Proteínas/métodos , Proteínas não Estruturais Virais/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Proteínas Aviárias/metabolismo , Patos , Embrião não Mamífero , Fibroblastos/metabolismo , Flavivirus/metabolismo , Infecções por Flavivirus/genética , Infecções por Flavivirus/metabolismo , Infecções por Flavivirus/virologia , Expressão Gênica , Biblioteca Gênica , Humanos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Proteínas não Estruturais Virais/metabolismo
12.
J Immunol Res ; 2018: 6106582, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29888293

RESUMO

Flaviviridae family is a class of single-stranded RNA virus, which is fatal to human and animals and mainly prevalent in subtropic and tropic countries. Even though people and animals are barraged with flavivirus infection every year, we have not invented either vaccines or antiviral for most flavivirus infections yet. Innate immunity is the first line of defense in resisting pathogen invasion, serving an important role in a resisting virus. Toll-like receptors (TLRs) and retinoic acid-inducible gene I- (RIG-I-) like receptors (RLRs) are crucial pattern recognition receptors (PRRs) that play essential roles in recognizing and clearing pathogens, including resisting flavivirus. In the present review, we provide a significant reference for further research on the function of innate immunity in resisting flavivirus.


Assuntos
Proteína DEAD-box 58/metabolismo , Resistência à Doença/imunologia , Infecções por Flavivirus/imunologia , Infecções por Flavivirus/metabolismo , Flavivirus/imunologia , Interações Hospedeiro-Patógeno/imunologia , Receptores Toll-Like/metabolismo , Animais , Infecções por Flavivirus/virologia , Humanos , Transdução de Sinais
13.
J Chem Theory Comput ; 14(7): 3920-3932, 2018 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-29874075

RESUMO

During host cell infection by flaviviruses such as dengue and Zika, acidic pH within the endosome triggers a conformational change in the envelope protein on the outer surface of the virion. This results in exposure of the ∼15 residue fusion peptide (FP) region, freeing it to induce fusion between the viral and endosomal membranes. A better understanding of the conformational dynamics of the FP in the presence of membranes, and the basis for its selectivity for anionic lipid species present within the endosome, would facilitate its therapeutic targeting with antiviral drugs and antibodies. In this work, multiscale modeling, simulations, and free energy calculations (including a total of ∼75 µs of atomic-resolution sampling), combined with imaging total internal reflection fluorescence correlation spectroscopy experiments, were employed to investigate the mechanisms of interaction of FP variants with lipid bilayers. Wild-type FPs (in the presence or absence of a fluorescein isothiocyanate tag) were shown to possess a funneled conformational landscape governing their exit from solvent and penetration into the lipid phase and to exhibit an electrostatically favored >2-fold affinity for membranes containing anionic species over purely zwitterionic ones. Conversely, the landscape was abolished in a nonfunctional point mutant, leading to a 2-fold drop in host membrane affinity. Collectively, our data reveal how the highly conserved flavivirus FP has evolved to funnel its conformational space toward a maximally fusogenic state anchored within the endosomal membrane. Therapeutically targeting the accessible ensemble of FP conformations may represent a new, rational strategy for blocking viral infection.


Assuntos
Infecções por Flavivirus/metabolismo , Flavivirus/fisiologia , Lipídeos de Membrana/metabolismo , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Flavivirus/química , Infecções por Flavivirus/virologia , Interações Hospedeiro-Patógeno , Humanos , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Lipídeos de Membrana/química , Conformação Proteica , Termodinâmica , Proteínas Virais de Fusão/química
14.
Viruses ; 9(9)2017 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-28846635

RESUMO

Apoptosis is a type of programmed cell death that regulates cellular homeostasis by removing damaged or unnecessary cells. Its importance in host defenses is highlighted by the observation that many viruses evade, obstruct, or subvert apoptosis, thereby blunting the host immune response. Infection with Flaviviruses such as Japanese encephalitis virus (JEV), Dengue virus (DENV) and West Nile virus (WNV) has been shown to activate several signaling pathways such as endoplasmic reticulum (ER)-stress and AKT/PI3K pathway, resulting in activation or suppression of apoptosis in virus-infected cells. On the other hands, expression of some viral proteins induces or protects apoptosis. There is a discrepancy between induction and suppression of apoptosis during flavivirus infection because the experimental situation may be different, and strong links between apoptosis and other types of cell death such as necrosis may make it more difficult. In this paper, we review the effects of apoptosis on viral propagation and pathogenesis during infection with flaviviruses.


Assuntos
Apoptose/fisiologia , Infecções por Flavivirus/imunologia , Infecções por Flavivirus/metabolismo , Flavivirus/imunologia , Flavivirus/fisiologia , Animais , Morte Celular/imunologia , Morte Celular/fisiologia , Sobrevivência Celular , Vírus da Dengue/imunologia , Vírus da Dengue/fisiologia , Vírus da Encefalite Japonesa (Espécie)/imunologia , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Retículo Endoplasmático/virologia , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Estágios do Ciclo de Vida , Linfoma de Células B , Neoplasias/terapia , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Estresse Fisiológico , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/fisiologia
15.
J Virol ; 91(4)2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27974556

RESUMO

The Flavivirus genus contains several arthropod-borne viruses that pose global health threats, including dengue viruses (DENV), yellow fever virus (YFV), and Zika virus (ZIKV). In order to understand how these viruses replicate in human cells, we previously conducted genome-scale RNA interference screens to identify candidate host factors. In these screens, we identified ribosomal proteins RPLP1 and RPLP2 (RPLP1/2) to be among the most crucial putative host factors required for DENV and YFV infection. RPLP1/2 are phosphoproteins that bind the ribosome through interaction with another ribosomal protein, RPLP0, to form a structure termed the ribosomal stalk. RPLP1/2 were validated as essential host factors for DENV, YFV, and ZIKV infection in two human cell lines: A549 lung adenocarcinoma and HuH-7 hepatoma cells, and for productive DENV infection of Aedes aegypti mosquitoes. Depletion of RPLP1/2 caused moderate cell-line-specific effects on global protein synthesis, as determined by metabolic labeling. In A549 cells, global translation was increased, while in HuH-7 cells it was reduced, albeit both of these effects were modest. In contrast, RPLP1/2 knockdown strongly reduced early DENV protein accumulation, suggesting a requirement for RPLP1/2 in viral translation. Furthermore, knockdown of RPLP1/2 reduced levels of DENV structural proteins expressed from an exogenous transgene. We postulate that these ribosomal proteins are required for efficient translation elongation through the viral open reading frame. In summary, this work identifies RPLP1/2 as critical flaviviral host factors required for translation. IMPORTANCE: Flaviviruses cause important diseases in humans. Examples of mosquito-transmitted flaviviruses include dengue, yellow fever and Zika viruses. Viruses require a plethora of cellular factors to infect cells, and the ribosome plays an essential role in all viral infections. The ribosome is a complex macromolecular machine composed of RNA and proteins and it is responsible for protein synthesis. We identified two specific ribosomal proteins that are strictly required for flavivirus infection of human cells and mosquitoes: RPLP1 and RPLP2 (RPLP1/2). These proteins are part of a structure known as the ribosomal stalk and help orchestrate the elongation phase of translation. We show that flaviviruses are particularly dependent on the function of RPLP1/2. Our findings suggest that ribosome composition is an important factor for virus translation and may represent a regulatory layer for translation of specific cellular mRNAs.


Assuntos
Infecções por Flavivirus/metabolismo , Infecções por Flavivirus/virologia , Flavivirus/fisiologia , Interações Hospedeiro-Patógeno , Fosfoproteínas/metabolismo , Proteínas Ribossômicas/metabolismo , Proteínas Virais/metabolismo , Aedes/virologia , Animais , Linhagem Celular , Vírus da Dengue/fisiologia , Infecções por Flavivirus/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Fosfoproteínas/química , Fosfoproteínas/genética , Ligação Proteica , Multimerização Proteica , Proteínas Ribossômicas/química , Proteínas Ribossômicas/genética , Replicação Viral , Vírus da Febre Amarela/fisiologia
16.
Crit Rev Biochem Mol Biol ; 51(5): 379-394, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27685368

RESUMO

Viruses are obligate parasites that rely heavily on host cellular processes for replication. The small number of proteins typically encoded by a virus is faced with selection pressures that lead to the evolution of distinctive structural properties, allowing each protein to maintain its function under constraints such as small genome size, high mutation rate, and rapidly changing fitness conditions. One common strategy for this evolution is to utilize small building blocks to generate protein oligomers that assemble in multiple ways, thereby diversifying protein function and regulation. In this review, we discuss specific cases that illustrate how oligomerization is used to generate a single defined functional state, to modulate activity via different oligomeric states, or to generate multiple functional forms via different oligomeric states.


Assuntos
Multimerização Proteica , Proteínas Virais/química , Viroses/virologia , Vírus/química , Animais , Capsídeo/química , Capsídeo/imunologia , Capsídeo/metabolismo , Ebolavirus/química , Ebolavirus/imunologia , Ebolavirus/metabolismo , Flavivirus/química , Flavivirus/imunologia , Flavivirus/metabolismo , Infecções por Flavivirus/imunologia , Infecções por Flavivirus/metabolismo , Infecções por Flavivirus/virologia , HIV/química , HIV/imunologia , HIV/metabolismo , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/metabolismo , Doença pelo Vírus Ebola/virologia , Humanos , Modelos Moleculares , Conformação Proteica , Proteínas Virais/imunologia , Proteínas Virais/metabolismo , Viroses/imunologia , Viroses/metabolismo , Replicação Viral , Vírus/imunologia , Vírus/metabolismo
17.
Eksp Klin Farmakol ; 77(4): 33-41, 2014.
Artigo em Russo | MEDLINE | ID: mdl-25076758

RESUMO

In vitro immunohistochemical investigations on the human hepatoma cell line (Huh7) infected with hepatitis C virus (HCV) strain JFH-1 showed that AV0012 compound blocks the early stages of viral infection. AV0012 also blocked viral infection spread in tissue culture through the secreted virus and through tight cell-to-cell contact. AV0012 is a specific inhibitor of HCV but not of related pestivirus, flaviviruses and other RNA-containing viruses such as bovine diarrhea (BVDV), Venezuelan equine encephalitis (strain TC-83), dengue type 2 (New Guinea), yellow fever (strain 17D), west Nile fever, parainfluenza (type 3) virus, RSV (strain A2), and Rhinovirus (type 2 strain HGP). It is established that human serum does not significantly affect the antiviral activity of AV0012 in vitro. The drug combination studies with AV0012 and interferon alpha 2a in vitro showed that the two inhibitors act additively, which makes possible the use of this combination in clinical tests. AV0012 is highly soluble and stable in aqueous solutions and murine blood plasma, has limited metabolic stability, low binding to human plasma proteins, high permeability through biological membranes, and only interacts with isoenzymes 2D6 and 3A4 of human cytochrome P450. In animal pharmacokinetic studies, AV0012 was rapidly absorbed into the blood stream upon oral administration, showed sufficiently long half-elimination times, and had high oral bioavailability that reached 92% in monkeys. Further preclinical development of AV0012 is in progress.


Assuntos
Antivirais/farmacologia , Antivirais/farmacocinética , Hepacivirus/metabolismo , Hepatite C/tratamento farmacológico , Hepatite C/metabolismo , Animais , Antivirais/química , Bovinos , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Flavivirus/metabolismo , Infecções por Flavivirus/tratamento farmacológico , Infecções por Flavivirus/metabolismo , Haplorrinos , Humanos , Camundongos , Ratos
18.
Am J Trop Med Hyg ; 89(5): 1013-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24080631

RESUMO

Rocio virus (ROCV) caused an outbreak of human encephalitis during the 1970s in Brazil and its immunopathogenesis remains poorly understood. CC-chemokine receptor 5 (CCR5) is a chemokine receptor that binds to macrophage inflammatory protein (MIP-1 α). Both molecules are associated with inflammatory cells migration during infections. In this study, we demonstrated the importance of the CCR5 and MIP-1 α, in the outcome of viral encephalitis of ROCV-infected mice. CCR5 and MIP-1 α knockout mice survived longer than wild-type (WT) ROCV-infected animals. In addition, knockout mice had reduced inflammation in the brain. Assessment of brain viral load showed mice virus detection five days post-infection in wild-type and CCR5-/- mice, while MIP-1 α-/- mice had lower viral loads seven days post-infection. Knockout mice required a higher lethal dose than wild-type mice as well. The CCR5/MIP-1 α axis may contribute to migration of infected cells to the brain and consequently affect the pathogenesis during ROCV infection.


Assuntos
Encéfalo/patologia , Quimiocina CCL3/genética , Encefalite Viral/metabolismo , Infecções por Flavivirus/metabolismo , Flavivirus/fisiologia , Receptores CCR5/genética , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Movimento Celular , Quimiocina CCL3/deficiência , Encefalite Viral/mortalidade , Encefalite Viral/patologia , Encefalite Viral/virologia , Infecções por Flavivirus/mortalidade , Infecções por Flavivirus/patologia , Infecções por Flavivirus/virologia , Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Inflamação/metabolismo , Inflamação/mortalidade , Inflamação/patologia , Inflamação/virologia , Linfócitos/metabolismo , Linfócitos/patologia , Linfócitos/virologia , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica , Receptores CCR5/deficiência , Transdução de Sinais , Análise de Sobrevida , Carga Viral
20.
Novartis Found Symp ; 277: 23-39; discussion 40, 71-3, 251-3, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17319152

RESUMO

Flaviviruses are enveloped viruses with a single-stranded, 10.7kb positive-sense RNA genome. The genomic RNA, which has a 5' cap but no poly(A) tail, is translated as a single polyprotein that is then cleaved into three structural proteins and seven non-structural (NS) proteins by both viral and host proteases. The NS proteins include an RNA-dependent RNA polymerase (NS5), a helicase/protease (NS3), and other proteins that form part of the viral replication complex. Sequences and structures in the 5' and 3' untranslated regions (UTR) and capsid gene, including the cyclization sequences, the upstream AUG region, and the terminal 3' stem-loop, regulate translation, RNA synthesis and viral replication. We have also found that an RNA hairpin structure in the capsid coding region (cHP) influences start codon selection and viral replication of the flavivirus dengue virus (DENV). Peptide-conjugated phosphorodiamidate morpholino oligomers (P-PMOs) were used to further dissect the role of conserved regions of the 5' and 3' UTRs; several P-PMOs were shown to specifically inhibit DENV translation and/or RNA synthesis and, hence, are potentially useful as antiviral agents. Regarding the mechanism of DENV translation, we have shown that DENV undergoes canonical cap-dependent translation initiation as well as a non-canonical mechanism when cap-dependent translation is suppressed. Although much remains to be elucidated about the molecular biology of flavivirus infection, progress is being made towards defining the cis and trans factors that regulate flavivirus translation and replication.


Assuntos
Infecções por Flavivirus/genética , Flavivirus/genética , Genoma Viral , Iniciação Traducional da Cadeia Peptídica , RNA Viral/genética , Proteínas Virais/metabolismo , Replicação Viral , Animais , Infecções por Flavivirus/metabolismo , Humanos , RNA Viral/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA