Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 5179, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38898037

RESUMO

Viral genetic diversity presents significant challenges in developing antivirals with broad-spectrum activity and high barriers to resistance. Here we report development of proteolysis targeting chimeras (PROTACs) targeting the dengue virus envelope (E) protein through coupling of known E fusion inhibitors to ligands of the CRL4CRBN E3 ubiquitin ligase. The resulting small molecules block viral entry through inhibition of E-mediated membrane fusion and interfere with viral particle production by depleting intracellular E in infected Huh 7.5 cells. This activity is retained in the presence of point mutations previously shown to confer partial resistance to the parental inhibitors due to decreased inhibitor-binding. The E PROTACs also exhibit broadened spectrum of activity compared to the parental E inhibitors against a panel of mosquito-borne flaviviruses. These findings encourage further exploration of targeted protein degradation as a differentiated and potentially advantageous modality for development of broad-spectrum direct-acting antivirals.


Assuntos
Antivirais , Vírus da Dengue , Flavivirus , Proteólise , Internalização do Vírus , Humanos , Proteólise/efeitos dos fármacos , Animais , Antivirais/farmacologia , Flavivirus/efeitos dos fármacos , Flavivirus/genética , Flavivirus/metabolismo , Internalização do Vírus/efeitos dos fármacos , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/fisiologia , Vírus da Dengue/genética , Culicidae/virologia , Ubiquitina-Proteína Ligases/metabolismo , Proteínas do Envelope Viral/metabolismo , Linhagem Celular
2.
Antiviral Res ; 226: 105878, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38582134

RESUMO

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


Assuntos
Antivirais , Flavivirus , Serina Endopeptidases , Proteínas não Estruturais Virais , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Flavivirus/efeitos dos fármacos , Flavivirus/enzimologia , Serina Endopeptidases/metabolismo , Serina Endopeptidases/química , Antivirais/farmacologia , Antivirais/química , Humanos , RNA Helicases/metabolismo , RNA Helicases/química , RNA Helicases/genética , Inibidores de Proteases/farmacologia , Inibidores de Proteases/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/química , Proteases Virais , Nucleosídeo-Trifosfatase , RNA Helicases DEAD-box
3.
Antiviral Res ; 202: 105325, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35460703

RESUMO

Epidemics caused by flaviviruses occur globally; however, no antiviral drugs treating flaviviruses infections have yet been developed. Nafamostat (NM) is a protease inhibitor approved for pancreatitis and anti-coagulation. The anti-flavivirus potential of NM has yet to be determined. Here, utilizing in vitro and in vivo infection assays, we present that NM effectively inhibits Zika virus (ZIKV) and other flaviviruses in vitro. NM inhibited the production of ZIKV viral RNA and proteins originating from Asia and African lineage in human-, mouse- and monkey-derived cell lines and the in vivo anti-ZIKV efficacy of NM was verified. Mode-of-action analysis using time-of-drug-addition assay, infectivity inhibition assay, surface plasmon resonance assay, and molecular docking revealed that NM interacted with viral particles and blocked the early stage of infection by targeting the domain III of ZIKV envelope protein. Analysing the anti-flavivirus effects of NM-related compounds suggested that the antiviral effect depended on the unique structure of NM. These findings suggest the potential use of NM as an anti-flavivirus candidate, and a novel drug design approach targeting the flavivirus envelope protein.


Assuntos
Antivirais , Benzamidinas , Flavivirus , Guanidinas , Zika virus , Animais , Antivirais/química , Antivirais/farmacologia , Benzamidinas/química , Benzamidinas/farmacologia , Flavivirus/efeitos dos fármacos , Guanidinas/química , Guanidinas/farmacologia , Haplorrinos , Humanos , Camundongos , Simulação de Acoplamento Molecular , Proteínas do Envelope Viral/metabolismo , Zika virus/efeitos dos fármacos
4.
J Virol ; 96(2): e0177421, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-34757841

RESUMO

Alphaviruses and flaviviruses have class II fusion glycoproteins that are essential for virion assembly and infectivity. Importantly, the tip of domain II is structurally conserved between the alphavirus and flavivirus fusion proteins, yet whether these structural similarities between virus families translate to functional similarities is unclear. Using in vivo evolution of Zika virus (ZIKV), we identified several novel emerging variants, including an envelope glycoprotein variant in ß-strand c (V114M) of domain II. We have previously shown that the analogous ß-strand c and the ij loop, located in the tip of domain II of the alphavirus E1 glycoprotein, are important for infectivity. This led us to hypothesize that flavivirus E ß-strand c also contributes to flavivirus infection. We generated this ZIKV glycoprotein variant and found that while it had little impact on infection in mosquitoes, it reduced replication in human cells and mice and increased virus sensitivity to ammonium chloride, as seen for alphaviruses. In light of these results and given our alphavirus ij loop studies, we mutated a conserved alanine at the tip of the flavivirus ij loop to valine to test its effect on ZIKV infectivity. Interestingly, this mutation inhibited infectious virion production of ZIKV and yellow fever virus, but not West Nile virus. Together, these studies show that shared domains of the alphavirus and flavivirus class II fusion glycoproteins harbor structurally analogous residues that are functionally important and contribute to virus infection in vivo.IMPORTANCE Arboviruses are a significant global public health threat, yet there are no antivirals targeting these viruses. This problem is in part due to our lack of knowledge of the molecular mechanisms involved in the arbovirus life cycle. In particular, virus entry and assembly are essential processes in the virus life cycle and steps that can be targeted for the development of antiviral therapies. Therefore, understanding common, fundamental mechanisms used by different arboviruses for entry and assembly is essential. In this study, we show that flavivirus and alphavirus residues located in structurally conserved and analogous regions of the class II fusion proteins contribute to common mechanisms of entry, dissemination, and infectious-virion production. These studies highlight how class II fusion proteins function and provide novel targets for development of antivirals.


Assuntos
Alphavirus/fisiologia , Flavivirus/fisiologia , Proteínas Virais de Fusão/metabolismo , Vírion/metabolismo , Replicação Viral , Células A549 , Alphavirus/efeitos dos fármacos , Cloreto de Amônio/farmacologia , Animais , Culicidae/virologia , Flavivirus/efeitos dos fármacos , Humanos , Interferon Tipo I/deficiência , Camundongos , Camundongos Mutantes , Mutação , Domínios Proteicos , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Vírion/genética , Montagem de Vírus/genética , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/genética , Zika virus/efeitos dos fármacos , Zika virus/fisiologia , Infecção por Zika virus/virologia
5.
Viruses ; 13(10)2021 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-34696411

RESUMO

Viral proteases are indispensable for successful virion maturation, thus making them a prominent drug target. Their enzyme activity is tightly spatiotemporally regulated by expression in the precursor form with little or no activity, followed by activation via autoprocessing. These cleavage events are frequently triggered upon transportation to a specific compartment inside the host cell. Typically, precursor oligomerization or the presence of a co-factor is needed for activation. A detailed understanding of these mechanisms will allow ligands with non-canonical mechanisms of action to be designed, which would specifically modulate the initial irreversible steps of viral protease autoactivation. Binding sites exclusive to the precursor, including binding sites beyond the protease domain, can be exploited. Both inhibition and up-regulation of the proteolytic activity of viral proteases can be detrimental for the virus. All these possibilities are discussed using examples of medically relevant viruses including herpesviruses, adenoviruses, retroviruses, picornaviruses, caliciviruses, togaviruses, flaviviruses, and coronaviruses.


Assuntos
Antivirais/farmacologia , Inibidores de Protease Viral/farmacologia , Proteases Virais/metabolismo , Viroses/tratamento farmacológico , Adenovírus Humanos/efeitos dos fármacos , Adenovírus Humanos/metabolismo , Flavivirus/efeitos dos fármacos , Flavivirus/metabolismo , HIV-1/efeitos dos fármacos , Herpesviridae/efeitos dos fármacos , Herpesviridae/metabolismo , Humanos , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/metabolismo , Proteases Virais/biossíntese
6.
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
7.
Curr Opin Virol ; 49: 164-175, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34171540

RESUMO

The flavivirus are emerging and re-emerging arthropod-borne pathogens responsible for significant mortality and morbidity worldwide. The genus comprises more than 70 viruses, and despite genomic and structural similarities, infections by different flaviviruses result in different clinical presentations. In the absence of a safe and effective vaccine against these infections, the search for new strategies to inhibit viral infection is necessary. The life cycle of arboviruses begins with the entry process composed of multiple steps: attachment, internalization, endosomal escape and capsid uncoating. This mini-review describes factors and mechanisms involved in the viral entry as events required to take over the cellular machinery and host factors and cellular pathways commonly used by flaviviruses as possible approaches for developing broad-spectrum antiviral drugs.


Assuntos
Infecções por Flavivirus/virologia , Flavivirus/fisiologia , Internalização do Vírus , Animais , Antivirais/metabolismo , Antivirais/farmacologia , Endocitose , Flavivirus/efeitos dos fármacos , Flavivirus/patogenicidade , Infecções por Flavivirus/tratamento farmacológico , Interações Hospedeiro-Patógeno , Humanos , Receptores Virais/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus/efeitos dos fármacos , Replicação Viral
8.
Nat Commun ; 12(1): 3266, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34075032

RESUMO

The epidemic emergence of relatively rare and geographically isolated flaviviruses adds to the ongoing disease burden of viruses such as dengue. Structural analysis is key to understand and combat these pathogens. Here, we present a chimeric platform based on an insect-specific flavivirus for the safe and rapid structural analysis of pathogenic viruses. We use this approach to resolve the architecture of two neurotropic viruses and a structure of dengue virus at 2.5 Å, the highest resolution for an enveloped virion. These reconstructions allow improved modelling of the stem region of the envelope protein, revealing two lipid-like ligands within highly conserved pockets. We show that these sites are essential for viral growth and important for viral maturation. These findings define a hallmark of flavivirus virions and a potential target for broad-spectrum antivirals and vaccine design. We anticipate the chimeric platform to be widely applicable for investigating flavivirus biology.


Assuntos
Infecções por Flavivirus/terapia , Flavivirus/ultraestrutura , Proteínas do Envelope Viral/ultraestrutura , Vírion/ultraestrutura , Aedes/virologia , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Linhagem Celular , Chlorocebus aethiops , Microscopia Crioeletrônica , Dengue/terapia , Dengue/virologia , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/farmacologia , Desenho de Fármacos , Flavivirus/efeitos dos fármacos , Flavivirus/imunologia , Flavivirus/patogenicidade , Infecções por Flavivirus/virologia , Humanos , Mesocricetus , Modelos Moleculares , Conformação Molecular , Mutagênese Sítio-Dirigida , Mutação Puntual , Células Vero , Proteínas do Envelope Viral/metabolismo , Vacinas Virais/farmacologia , Vacinas Virais/uso terapêutico , Vírion/efeitos dos fármacos , Vírion/metabolismo
9.
Arch Virol ; 166(5): 1433-1438, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33683474

RESUMO

The recent introduction of Zika virus (ZIKV), the recurrence of dengue virus (DENV), and the lethality of yellow fever virus (YFV) have had a significant impact on Brazilian society and public health. Here, we targeted two cellular kinases implicated in cell proliferation and cancer that are also important for viral replication: mitogen-activated protein kinase kinase (MEK) and Src. We used two MEK inhibitors - trametinib and selumetinib - and two Src inhibitors - saracatinib and bosutinib - to inhibit ZIKV, DENV, and YFV replication in cell culture. The cytotoxicity of the four inhibitors was determined by the observation of abnormal morphology and quantification of adherent cells by crystal violet staining. The antiviral activity of these drugs was assessed based on the reduction of plaque-forming units in cell culture as evidence of the inhibition of the replication of the selected flaviviruses. All four inhibitors showed antiviral activity, but among them, trametinib was the safest and most efficacious against all of the viruses, inhibiting the replication of ZIKV and YFV by 1000-fold, and DENV2/3 by nearly 100-fold. This pan-antiviral effect shows that trametinib could be repurposed for the treatment of flaviviral infections.


Assuntos
Antivirais/farmacologia , Flavivirus/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Cricetinae , Flavivirus/classificação , Flavivirus/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Células Vero , Replicação Viral/efeitos dos fármacos , Quinases da Família src/antagonistas & inibidores
10.
Poult Sci ; 100(4): 100989, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33647721

RESUMO

The duck Tembusu virus (DTMUV) is a novel mosquito-borne Flavivirus which caused huge economic losses for poultry industries in Southeast Asia and China. Currently, no effective antiviral drugs against this virus have been reported. (-)-Epigallocatechin-3-gallate (EGCG), a polyphenol present in abundance in green tea, has recently been demonstrated to have an antiviral activity for many viruses; however, whether EGCG can inhibit DTMUV infection remains unknown. Here, we tried to explore the anti-DTMUV effects and mechanisms of EGCG both in vitro and in vivo. Several EGCG treatment regimens were used to study the comprehensive antiviral activity of EGCG in DTMUV-infected baby hamster kidney cell line (BHK-21). The DTMUV titers of mock- and EGCG-treated infected cell cultures were determined using the tissue culture infective dose assay and the DTMUV mRNA copy number as determined using quantitative Real Time PCR. Moreover, the therapeutic efficacy of EGCG against DTMUV was assessed in DTMUV-infected ducklings. Our results suggested that EGCG significantly reduced the viral infection in BHK-21 cells in a dose-dependent manner, as reflected by the reduction of virus titers, virus copy number, and the expressions of viral E protein. We also observed that EGCG exhibited direct virucidal abilities against DTMUV. Notably, a significant reduction in virus binding ability was also observed, indicating that EGCG possesses excellent inhibitory effects on the viral adsorption step. In addition, DTMUV replication was also suppressed in BHK-21 cells treated with EGCG after viral entry, likely because of upregulation of the levels of interferon alfa and interferon beta. Finally, we also proved that EGCG exhibited anti-DTMUV efficacy in a duckling infection model because the survival rate was significantly improved. This is the first study to demonstrate the protective effect of EGCG against DTMUV, suggesting its potential use as an antiviral drug for DTMUV infection.


Assuntos
Catequina/análogos & derivados , Infecções por Flavivirus , Flavivirus , Interferon Tipo I , Doenças das Aves Domésticas , Internalização do Vírus , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Catequina/farmacologia , Linhagem Celular , China , Cricetinae , Patos , Flavivirus/efeitos dos fármacos , Infecções por Flavivirus/tratamento farmacológico , Infecções por Flavivirus/veterinária , Interferon Tipo I/genética , Doenças das Aves Domésticas/tratamento farmacológico , Análise de Sobrevida , Resultado do Tratamento , Regulação para Cima/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos
11.
ChemMedChem ; 15(24): 2391-2419, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-32961008

RESUMO

Infections by flaviviruses, such as Dengue, West Nile, Yellow Fever and Zika viruses, represent a growing risk for global health. There are vaccines only for few flaviviruses while no effective treatments are available. Flaviviruses share epidemiological, structural, and ecologic features and often different viruses can co-infect the same host. Therefore, the identification of broad-spectrum inhibitors is highly desirable either for known flaviviruses or for viruses that likely will emerge in the future. Strategies targeting both virus and host factors have been pursued to identify broad-spectrum antiflaviviral agents. In this review, we describe the most promising and best characterized targets and their relative broad-spectrum inhibitors, identified by drug repurposing/libraries screenings and by focused medicinal chemistry campaigns. Finally, we discuss about future strategies to identify new broad-spectrum antiflavivirus agents.


Assuntos
Antivirais/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Infecções por Flavivirus/tratamento farmacológico , Flavivirus/efeitos dos fármacos , Animais , Antivirais/química , Antivirais/farmacocinética , Linhagem Celular Tumoral , Química Farmacêutica , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Flavivirus/química , Flavivirus/enzimologia , Humanos
12.
Antiviral Res ; 182: 104899, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32763313

RESUMO

Remdesivir was shown to inhibit RNA-dependent RNA-polymerases (RdRp) from distinct viral families such as from Filoviridae (Ebola) and Coronaviridae (SARS-CoV, SARS-CoV-2, MERS). In this study, we tested the ability of remdesivir to inhibit RdRps from the Flaviviridae family. Instead of remdesivir, we used the active species that is produced in cells from remdesivir, the appropriate triphosphate, which could be directly tested in vitro using recombinant flaviviral polymerases. Our results show that remdesivir can efficiently inhibit RdRps from viruses causing severe illnesses such as Yellow fever, West Nile fever, Japanese and Tick-borne encephalitis, Zika and Dengue. Taken together, this study demonstrates that remdesivir or its derivatives have the potential to become a broad-spectrum antiviral agent effective against many RNA viruses.


Assuntos
Trifosfato de Adenosina/análogos & derivados , Antivirais/farmacologia , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/virologia , Flavivirus/efeitos dos fármacos , Pneumonia Viral/tratamento farmacológico , Pneumonia Viral/virologia , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Trifosfato de Adenosina/química , Trifosfato de Adenosina/farmacologia , Antivirais/química , Betacoronavirus/efeitos dos fármacos , Betacoronavirus/enzimologia , COVID-19 , Flavivirus/enzimologia , Humanos , Concentração Inibidora 50 , Pandemias , Vírus de RNA/efeitos dos fármacos , Vírus de RNA/enzimologia , RNA Polimerase Dependente de RNA/metabolismo , SARS-CoV-2 , Tratamento Farmacológico da COVID-19
13.
Vet Microbiol ; 245: 108708, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32456819

RESUMO

The outbreak and spread of Tembusu virus (TMUV) has caused very large losses in the waterfowl-breeding industry since 2010. The viral envelope (E) protein, the principal surface protein of viral particles, plays a vital role in viral entry and fusion. In this study, two peptides derived from domain II (DII) and the stem of the TMUV envelope protein, TP1 and TP2, respectively, were tested for their antiviral activity. TP1 and TP2 inhibited TMUV infection in BHK-21 cells, and their 50% inhibitory concentrations (IC50) were 14.19 mg/L and 7.64 mg/L, respectively. Viral inhibition assays in different cell lines of avian origin showed that the inhibitory effects of TP1 and TP2 are not cell type dependent. Moreover, TP2 also exhibited inhibitory activity against Japanese encephalitis virus (JEV) infection. The two peptides inhibited antibody-mediated TMUV infection of duck peripheral blood lymphocytes. Co-immunoprecipitation assays and indirect enzyme-linked immunosorbent assays (ELISAs) indicated that both peptides interact with the surface of the TMUV virion. RNase digestion assays confirmed the release of viral RNA following incubation with TP1, while incubation with TP1 or TP2 interfered with the binding between TMUV and cells. Taken together, these results show that TP1 and TP2 may be developed into antiviral treatments against TMUV infection.


Assuntos
Antivirais/farmacologia , Flavivirus/efeitos dos fármacos , Peptídeos/farmacologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/farmacologia , Animais , Anticorpos Facilitadores , Antivirais/química , Linhagem Celular , Cricetinae , Patos , Concentração Inibidora 50 , Linfócitos/imunologia , Linfócitos/virologia , Peptídeos/química , RNA Viral/metabolismo , Organismos Livres de Patógenos Específicos
14.
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
15.
Rev Med Virol ; 30(4): e2100, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32101633

RESUMO

Flavivirus infections are a public health threat in the world that requires the development of safe and effective vaccines. Therefore, the understanding of the anti-flavivirus humoral immune response is fundamental to future studies on flavivirus pathogenesis and the design of anti-flavivirus therapeutics. This review aims to provide an overview of the current understanding of the function and involvement of flavivirus proteins in the humoral immune response as well as the ability of the anti-envelope (anti-E) antibodies to interfere (neutralizing antibodies) or not (non-neutralizing antibodies) with viral infection, and how they can, in some circumstances enhance dengue virus infection on Fc gamma receptor (FcγR) bearing cells through a mechanism known as antibody-dependent enhancement (ADE). Thus, the dual role of the antibodies against E protein poses a formidable challenge for vaccine development. Also, we discuss the roles of antibody binding stoichiometry (the concentration, affinity, or epitope recognition) in the neutralization of flaviviruses and the "breathing" of flavivirus virions in the humoral immune response. Finally, the relevance of some specific antibodies in the design and improvement of effective vaccines is addressed.


Assuntos
Suscetibilidade a Doenças/imunologia , Infecções por Flavivirus/imunologia , Infecções por Flavivirus/virologia , Flavivirus/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Humoral/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Flavivirus/efeitos dos fármacos , Infecções por Flavivirus/tratamento farmacológico , Humanos , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia
16.
Virology ; 541: 41-51, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31826845

RESUMO

The risk of infectious diseases caused by Flavivirus is increasing globally. Here, we developed a novel high-throughput screening (HTS) system to evaluate the inhibitory effects of compounds targeting the nuclear localization of the flavivirus core protein. We screened 4000 compounds based on their ability to inhibit the nuclear localization of the core protein, and identified over 20 compounds including inhibitors for cyclin dependent kinase and glycogen synthase kinase. The efficacy of the identified compounds to suppress viral growth was validated in a cell-based infection system. Remarkably, the nucleolus morphology was affected by the treatment with the compounds, suggesting that the nucleolus function is critical for viral propagation. The present HTS system provides a useful strategy for the identification of antivirals against flavivirus by targeting the nucleolar localization of the core protein.


Assuntos
Antivirais/farmacologia , Nucléolo Celular/efeitos dos fármacos , Flavivirus/efeitos dos fármacos , Proteínas do Core Viral/metabolismo , Transporte Ativo do Núcleo Celular , Nucléolo Celular/metabolismo , Nucléolo Celular/patologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Flavivirus/fisiologia , Células HEK293 , Ensaios de Triagem em Larga Escala , Humanos
17.
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
18.
Antiviral Res ; 172: 104636, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31654671

RESUMO

Dengue virus (DENV) is a global health problem that affects approximately 3.9 billion people worldwide. Since safety concerns were raised for the only licensed vaccine, Dengvaxia, and since the present treatment is only supportive care, the development of more effective therapeutic anti-DENV agents is urgently needed. In this report, we identified a potential small-molecule inhibitor, BP34610, via cell-based high-throughput screening (HTS) of 12,000 compounds using DENV-2 reporter viruses. BP34610 reduced the virus yields of type 2 DENV-infected cells with a 50% effective concentration (EC50) and selectivity index value of 0.48 ±â€¯0.06 µM and 197, respectively. Without detectable cytotoxicity, the compound inhibited not only all four serotypes of DENV but also Japanese encephalitis virus (JEV). Time-of-addition experiments suggested that BP34610 may act at an early stage of DENV virus infection. Sequencing analyses of several individual clones derived from BP34610-resistant viruses revealed a consensus amino acid substitution (S397P) in the N-terminal stem region of the E protein. Introduction of S397P into the DENV reporter viruses conferred an over 14.8-fold EC90 shift for BP34610. Importantly, the combination of BP34610 with a viral replication inhibitor, ribavirin, displayed synergistic enhancement of anti-DENV-2 activity. Our results identify an effective small-molecule inhibitor, BP34610, which likely targets the DENV E protein. BP34610 could be developed as an anti-flavivirus agent in the future.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Flavivirus/efeitos dos fármacos , Proteínas do Envelope Viral/efeitos dos fármacos , Animais , Antivirais/toxicidade , Linhagem Celular , Dengue/tratamento farmacológico , Sinergismo Farmacológico , Vírus da Encefalite Japonesa (Espécie)/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Humanos , Ribavirina/farmacologia , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
19.
Virus Res ; 273: 197739, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31493438

RESUMO

Duck Tembusu virus (DTMUV) is a new pathogen that produces an acute and potent disease in ducks which has caused serious economic losses in China. In this study, a virulent strain of DTMUV, designated as ZJSBL01, was attenuated by serial passages in BHK-21 cells supplied with 5-Fluorouracil (5-FU) for 50 passages to induce mutation and attenuation. Growth kinetics of different passages of ZJSBL01 strain in BHK-21 cells show that these viruses have similar replication characteristics. The virus was highly attenuated after 40 passages in BHK-21 cells supplied with 5-FU, based on mortality, morbidity, and viral load in inoculated Sheldrake ducklings. In addition, all of the ducklings immunized with ZJSBL01-P40, the virus obtained at passage 40 of ZJSBL01, showed seroconversion on day 14 post inoculation. Moreover, P40 did not cause clinical symptom for layding ducks. Immunization with ZJSBL01-P40 could provide effective protection against the virulent parental ZJSBL01 strain. Seventeen amino acid substitutions were observed in the polyprotein of ZJSBL01-P40 compared with parental ZJSBL01. These results indicate that ZJSBL01-P40 may be a live vaccine candidate for prevention of DTMUV-disease.


Assuntos
Patos/virologia , Flavivirus/efeitos dos fármacos , Flavivirus/genética , Fluoruracila/farmacologia , Inoculações Seriadas , Cultura de Vírus , Animais , Anticorpos Antivirais/sangue , Flavivirus/crescimento & desenvolvimento , Flavivirus/patogenicidade , Imunização , Cinética , Mutação , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Carga Viral/efeitos dos fármacos , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Replicação Viral/efeitos dos fármacos
20.
Bioorg Med Chem ; 27(18): 3963-3978, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31351847

RESUMO

Currently, more than 70 flaviviruses were identified and reported in the literature, whose Dengue (DENV), Zika (ZIKV), and West Nile (WNV) viruses have been responsible for millions of cases of infections worldwide, mainly in developing countries. These viruses are transmitted by the bite of mosquitoes from genus Aedes, or Culex and, in some cases, Stegomyia. Despite numerous efforts to identify a selective, safe, and effective antiviral agent, there is no currently approved drug for the treatment of flaviviral infections. Then, current pharmacological therapy has the objective to treat the clinical symptoms. Various peptidomimetics and peptide-derivatives have been synthesized and evaluated against several biological targets from flaviviruses with different applications, such as diagnosis, E protein inhibitors, entry inhibitors, virucidal inhibitors, and also viral replication inhibitors. Flaviviral replication depends on the NS3pro that is completely activated when it is complexed to its cofactor, NS2B; forming a viral enzymatic complex. The development of NS2B-NS3pro inhibitors is considered a challenging work due to its active site is shallow and open-pocket. In this work, we report all advances involving peptidomimetics, peptide-derived, and peptide-hybrids found in the literature. In sense, we discuss the influence of different functional groups in the activity and selectivity. Moreover, the first inhibitors reported in the literature as covalent ligands, comprising two basic residues followed by an electrophilic moiety that binds to the catalytic serine (Ser135-O-) are also discussed in details, such as trifluoromethyl ketones, aldehydes, and boronic acids. Furthermore, it is presented the influence of introducing transition metals, providing metallopeptide inhibitors; and cyclization of linear peptides, generating cyclic and macrocyclic peptide inhibitors. Finally, we provide the most accurate state of the art found in the literature, which can be utilized to design new and effective antiviral agents.


Assuntos
Dengue/tratamento farmacológico , Flavivirus/efeitos dos fármacos , Peptídeos/uso terapêutico , Inibidores de Proteases/uso terapêutico , Vírus do Nilo Ocidental/efeitos dos fármacos , Infecção por Zika virus/tratamento farmacológico , Humanos , Peptídeos/farmacologia , Inibidores de Proteases/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA