Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Int J Mol Sci ; 21(24)2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33333737

RESUMO

Protein kinases (PKs) are enzymes that catalyze the transfer of the terminal phosphate group from ATP to a protein acceptor, mainly to serine, threonine, and tyrosine residues. PK catalyzed phosphorylation is critical to the regulation of cellular signaling pathways that affect crucial cell processes, such as growth, differentiation, and metabolism. PKs represent attractive targets for drugs against a wide spectrum of diseases, including viral infections. Two different approaches are being applied in the search for antivirals: compounds directed against viral targets (direct-acting antivirals, DAAs), or against cellular components essential for the viral life cycle (host-directed antivirals, HDAs). One of the main drawbacks of DAAs is the rapid emergence of drug-resistant viruses. In contrast, HDAs present a higher barrier to resistance development. This work reviews the use of chemicals that target cellular PKs as HDAs against virus of the Flaviviridae family (Flavivirus and Hepacivirus), thus being potentially valuable therapeutic targets in the control of these pathogens.


Assuntos
Infecções por Flaviviridae/tratamento farmacológico , Hepacivirus/efeitos dos fármacos , Hepatite C Crônica/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Antivirais/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Caseína Quinase I/antagonistas & inibidores , Caseína Quinase I/metabolismo , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Flaviviridae/efeitos dos fármacos , Flaviviridae/enzimologia , Infecções por Flaviviridae/enzimologia , Hepacivirus/enzimologia , Hepacivirus/metabolismo , Hepatite C Crônica/metabolismo , Humanos , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo
2.
Antiviral Res ; 184: 104881, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32768411

RESUMO

N-linked glycosylation is the most common form of protein glycosylation and is required for the proper folding, trafficking, and/or receptor binding of some host and viral proteins. As viruses lack their own glycosylation machinery, they are dependent on the host's machinery for these processes. Certain iminosugars are known to interfere with the N-linked glycosylation pathway by targeting and inhibiting α-glucosidases I and II in the endoplasmic reticulum (ER). Perturbing ER α-glucosidase function can prevent these enzymes from removing terminal glucose residues on N-linked glycans, interrupting the interaction between viral glycoproteins and host chaperone proteins that is necessary for proper folding of the viral protein. Iminosugars have demonstrated broad-spectrum antiviral activity in vitro and in vivo against multiple viruses. This review discusses the broad activity of iminosugars against Flaviviridae. Iminosugars have shown favorable activity against multiple members of the Flaviviridae family in vitro and in murine models of disease, although the activity and mechanism of inhibition can be virus-specfic. While iminosugars are not currently approved for the treatment of viral infections, their potential use as future host-targeted antiviral (HTAV) therapies continues to be investigated.


Assuntos
Infecções por Flaviviridae/tratamento farmacológico , Flaviviridae/efeitos dos fármacos , Inibidores de Glicosídeo Hidrolases , Glicosilação/efeitos dos fármacos , Imino Açúcares/farmacologia , Proteínas Virais/metabolismo , Animais , Antivirais/farmacologia , Flaviviridae/genética , Interações entre Hospedeiro e Microrganismos , Humanos , Imino Açúcares/química , Camundongos , alfa-Glucosidases
3.
J Comput Aided Mol Des ; 33(11): 997-1008, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31773464

RESUMO

Imbalanced datasets, comprising of more inactive compounds relative to the active ones, are a common challenge in ligand-based model building workflows for drug discovery. This is particularly true for neglected tropical diseases since efforts to identify therapeutics for these diseases are often limited. In this report, we analyze the performance of several undersampling strategies in modeling the Dengue Virus 2 (DENV2) inhibitory activity, as well as the anti-flaviviral activities for the West Nile (WNV) and Zika (ZIKV) viruses. To this end, we build datasets comprising of 1218 (159 actives and 1059 inactives), 1044 (132 actives and 912 inactives) and 302 (75 actives and 227 inactives) molecules with known DENV2, WNV and ZIKV inhibitory activity profiles, respectively. We develop ensemble classifiers for these endpoints and compare the performance of the different undersampling algorithms on external sets. It is observed that data pruning algorithms yield superior performance relative to data selection algorithms. The best overall performance is provided by the one-sided selection algorithm with test set balanced accuracy (BACC) values of 0.84, 0.74 and 0.77 for the DENV2, WNV and ZIKV inhibitory activities, respectively. For the model building, we use the recently proposed GT-STAF information indices, and compare the predictivity of 3 molecular fragmentation approaches: connected subgraphs, substructure and alogp atom types, which are observed to show comparable performance. On the other hand, a combination of indices based on these fragmentation strategies enhances the predictivity of the built ensembles. The built models could be useful for screening new molecules with possible DENV, WNV and ZIKV inhibitory activities. ADMET modelers are encouraged to adopt undersampling algorithms in their workflows when dealing with imbalanced datasets.


Assuntos
Antivirais/farmacologia , Descoberta de Drogas/métodos , Flaviviridae/efeitos dos fármacos , Máquina de Vetores de Suporte , Antivirais/química , Vírus da Dengue/efeitos dos fármacos , Infecções por Flaviviridae/tratamento farmacológico , Humanos , Vírus do Nilo Ocidental/efeitos dos fármacos , Zika virus/efeitos dos fármacos
4.
J Virol ; 93(22)2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31462558

RESUMO

Enteric viruses exploit bacterial components, including lipopolysaccharides (LPS) and peptidoglycan (PG), to facilitate infection in humans. Because of their origin in the bat enteric system, we wondered if severe acute respiratory syndrome coronavirus (SARS-CoV) or Middle East respiratory syndrome CoV (MERS-CoV) also use bacterial components to modulate infectivity. To test this question, we incubated CoVs with LPS and PG and evaluated infectivity, finding no change following LPS treatment. However, PG from Bacillus subtilis reduced infection >10,000-fold, while PG from other bacterial species failed to recapitulate this. Treatment with an alcohol solvent transferred inhibitory activity to the wash, and mass spectrometry revealed surfactin, a cyclic lipopeptide antibiotic, as the inhibitory compound. This antibiotic had robust dose- and temperature-dependent inhibition of CoV infectivity. Mechanistic studies indicated that surfactin disrupts CoV virion integrity, and surfactin treatment of the virus inoculum ablated infection in vivo Finally, similar cyclic lipopeptides had no effect on CoV infectivity, and the inhibitory effect of surfactin extended broadly to enveloped viruses, including influenza, Ebola, Zika, Nipah, chikungunya, Una, Mayaro, Dugbe, and Crimean-Congo hemorrhagic fever viruses. Overall, our results indicate that peptidoglycan-associated surfactin has broad viricidal activity and suggest that bacteria by-products may negatively modulate virus infection.IMPORTANCE In this article, we consider a role for bacteria in shaping coronavirus infection. Taking cues from studies of enteric viruses, we initially investigated how bacterial surface components might improve CoV infection. Instead, we found that peptidoglycan-associated surfactin is a potent viricidal compound that disrupts virion integrity with broad activity against enveloped viruses. Our results indicate that interactions with commensal bacterial may improve or disrupt viral infections, highlighting the importance of understanding these microbial interactions and their implications for viral pathogenesis and treatment.


Assuntos
Lipopeptídeos/farmacologia , Peptídeos Cíclicos/farmacologia , Peptidoglicano/metabolismo , Vírus de RNA/efeitos dos fármacos , Animais , Linhagem Celular , Chlorocebus aethiops , Infecções por Coronavirus/virologia , Flaviviridae/efeitos dos fármacos , Lipopeptídeos/imunologia , Lipopeptídeos/metabolismo , Coronavírus da Síndrome Respiratória do Oriente Médio/metabolismo , Peptídeos Cíclicos/imunologia , Peptídeos Cíclicos/metabolismo , Peptidoglicano/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Síndrome Respiratória Aguda Grave/virologia , Células Vero , Viroses/metabolismo
6.
Antiviral Res ; 167: 6-12, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30849420

RESUMO

We report the design, synthesis, and biological evaluation of a class of 1H-pyrido[2,1-b][1,3]benzothiazol-1-ones originated from compound 1, previously identified as anti-flavivirus agent. Some of the new compounds showed activity in low µM range with reasonable selectivity against Dengue 2, Yellow fever (Bolivia strain), and West Nile viruses. One of the most interesting molecules, compound 16, showed broad antiviral activity against additional flaviviruses such as Dengue 1, 3 and 4, Zika, Japanese encephalitis, several strains of Yellow fever, and tick-borne encephalitis viruses. Compound 16 did not exert any effect on alphaviruses and phleboviruses and its activity was maintained in YFV infected cells from different species. The activity of 16 appears specific for flavivirus with respect to other virus families, suggesting, but not proving, that it might be targeting a viral factor. We demonstrated that the antiviral effect of 16 is not related to reduced viral RNA synthesis or virion release. On the contrary, viral particles grown in the presence of 16 showed reduced infectivity, being unable to perform a second round of infection. The chemical class herein presented thus emerges as suitable to provide pan-flavivirus inhibitors.


Assuntos
Antivirais , Flaviviridae/efeitos dos fármacos , Oxazocinas , Piridinas , Animais , Antivirais/síntese química , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Vírus da Encefalite Transmitidos por Carrapatos/efeitos dos fármacos , Humanos , Oxazocinas/síntese química , Oxazocinas/farmacologia , Piridinas/síntese química , Piridinas/farmacologia , RNA Viral/efeitos dos fármacos , Vírion/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Vírus do Nilo Ocidental/efeitos dos fármacos , Vírus da Febre Amarela/efeitos dos fármacos , Zika virus/efeitos dos fármacos
7.
Artigo em Inglês | MEDLINE | ID: mdl-30181371

RESUMO

The virus family Flaviviridae encompasses several viruses, including (re)emerging viruses which cause widespread morbidity and mortality throughout the world. Members of this virus family are positive-strand RNA viruses and replicate their genome in close association with reorganized intracellular host cell membrane compartments. This evolutionarily conserved strategy facilitates efficient viral genome replication and contributes to evasion from host cell cytosolic defense mechanisms. We have previously described the identification of a small-compound inhibitor, K22, which exerts a potent antiviral activity against a broad range of coronaviruses by targeting membrane-bound viral RNA replication. To analyze the antiviral spectrum of this inhibitor, we assessed the inhibitory potential of K22 against several members of the Flaviviridae family, including the reemerging Zika virus (ZIKV). We show that ZIKV is strongly affected by K22. Time-of-addition experiments revealed that K22 acts during a postentry phase of the ZIKV life cycle, and combination regimens of K22 together with ribavirin (RBV) or interferon alpha (IFN-α) further increased the extent of viral inhibition. Ultrastructural electron microscopy studies revealed severe alterations of ZIKV-induced intracellular replication compartments upon infection of K22-treated cells. Importantly, the antiviral activity of K22 was demonstrated against several other members of the Flaviviridae family. It is tempting to speculate that K22 exerts its broad antiviral activity against several positive-strand RNA viruses via a similar mechanism and thereby represents an attractive candidate for development as a panviral inhibitor.


Assuntos
Antivirais/farmacologia , Membrana Celular/efeitos dos fármacos , Infecções por Flaviviridae/tratamento farmacológico , Flaviviridae/efeitos dos fármacos , Aedes , Animais , Linhagem Celular , Membrana Celular/virologia , Chlorocebus aethiops , Infecções por Flaviviridae/virologia , Humanos , Interferon-alfa/farmacologia , RNA Viral/genética , Ribavirina/farmacologia , Células Vero , Replicação Viral/efeitos dos fármacos
8.
Nat Rev Microbiol ; 16(3): 125-142, 2018 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-29430005

RESUMO

Members of the Flaviviridae virus family comprise a large group of enveloped viruses with a single-strand RNA genome of positive polarity. Several genera belong to this family, including the Hepacivirus genus, of which hepatitis C virus (HCV) is the prototype member, and the Flavivirus genus, which contains both dengue virus and Zika virus. Viruses of these genera differ in many respects, such as the mode of transmission or the course of infection, which is either predominantly persistent in the case of HCV or acutely self-limiting in the case of flaviviruses. Although the fundamental replication strategy of Flaviviridae members is similar, during the past few years, important differences have been discovered, including the way in which these viruses exploit cellular resources to facilitate viral propagation. These differences might be responsible, at least in part, for the various biological properties of these viruses, thus offering the possibility to learn from comparisons. In this Review, we discuss the current understanding of how Flaviviridae viruses manipulate and usurp cellular pathways in infected cells. Specifically, we focus on comparing strategies employed by flaviviruses with those employed by hepaciviruses, and we discuss the importance of these interactions in the context of viral replication and antiviral therapies.


Assuntos
Flaviviridae/fisiologia , Replicação Viral/fisiologia , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Flaviviridae/efeitos dos fármacos , Regulação Viral da Expressão Gênica/fisiologia , Humanos , RNA Viral , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos
9.
Appl Biochem Biotechnol ; 185(3): 799-814, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29349531

RESUMO

A precipitous increase in the number of flaviviral infections has been noted over the last 5 years. Despite these outbreaks, treatment protocols for infected individuals remain ambiguous. Numerous studies have identified NITD008 as a potent flavivirus inhibitor; however, clinical testing was dismissed due to undesirable toxic effects. The binding landscape of NITD008 in complex with five detrimental flaviviruses at the RNA-dependent RNA polymerase active sites was explored. An "all-in-one" pharmacophore model was created for the design of small molecules that may inhibit a broad spectrum of flaviviruses. This pharmacophore model approach serves as a robust cornerstone, thus assisting medicinal experts in the composition of multifunctional inhibitors that will eliminate cross-resistance and toxicity and enhance patient adherence.


Assuntos
Adenosina/análogos & derivados , Antivirais/farmacologia , Descoberta de Drogas/métodos , Flaviviridae/efeitos dos fármacos , Modelos Químicos , RNA Polimerase Dependente de RNA/efeitos dos fármacos , Adenosina/química , Adenosina/metabolismo , Adenosina/farmacologia , Domínio Catalítico , Flaviviridae/enzimologia , Flaviviridae/fisiologia , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , RNA Polimerase Dependente de RNA/metabolismo , Termodinâmica , Replicação Viral/efeitos dos fármacos
10.
J Virol ; 92(2)2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29093094

RESUMO

The family Flaviviridae consists of four genera, Flavivirus, Pestivirus, Pegivirus, and Hepacivirus, and comprises important pathogens of human and animals. Although the construction of recombinant viruses carrying reporter genes encoding fluorescent and bioluminescent proteins has been reported, the stable insertion of foreign genes into viral genomes retaining infectivity remains difficult. Here, we applied the 11-amino-acid subunit derived from NanoLuc luciferase to the engineering of the Flaviviridae viruses and then examined the biological characteristics of the viruses. We successfully generated recombinant viruses carrying the split-luciferase gene, including dengue virus, Japanese encephalitis virus, hepatitis C virus (HCV), and bovine viral diarrhea virus. The stability of the viruses was confirmed by five rounds of serial passages in the respective susceptible cell lines. The propagation of the recombinant luciferase viruses in each cell line was comparable to that of the parental viruses. By using a purified counterpart luciferase protein, this split-luciferase assay can be applicable in various cell lines, even when it is difficult to transduce the counterpart gene. The efficacy of antiviral reagents against the recombinant viruses could be monitored by the reduction of luciferase expression, which was correlated with that of viral RNA, and the recombinant HCV was also useful to examine viral dynamics in vivo Taken together, our findings indicate that the recombinant Flaviviridae viruses possessing the split NanoLuc luciferase gene generated here provide powerful tools to understand viral life cycle and pathogenesis and a robust platform to develop novel antivirals against Flaviviridae viruses.IMPORTANCE The construction of reporter viruses possessing a stable transgene capable of expressing specific signals is crucial to investigations of viral life cycle and pathogenesis and the development of antivirals. However, it is difficult to maintain the stability of a large foreign gene, such as those for fluorescence and bioluminescence, after insertion into a viral genome. Here, we successfully generated recombinant Flaviviridae viruses carrying the 11-amino-acid subunit derived from NanoLuc luciferase and demonstrated that these viruses are applicable to in vitro and in vivo experiments, suggesting that these recombinant Flaviviridae viruses are powerful tools for increasing our understanding of viral life cycle and pathogenesis and that these recombinant viruses will provide a robust platform to develop antivirals against Flaviviridae viruses.


Assuntos
Flaviviridae/genética , Expressão Gênica , Genes Reporter , Recombinação Genética , Animais , Antivirais/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos , Flaviviridae/efeitos dos fármacos , Genoma Viral , Hepacivirus/genética , Humanos , Camundongos , Mutagênese Insercional
11.
Viruses ; 9(10)2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28972551

RESUMO

Flaviviruses including Zika, Dengue and Hepatitis C virus cause debilitating diseases in humans, and the former are emerging as global health concerns with no antiviral treatments. We investigated Sophora Flavecens, used in Chinese medicine, as a source for antiviral compounds. We isolated Sophoraflavenone G and found that it inhibited Hepatitis C replication, but not Sendai or Vesicular Stomatitis Virus. Pre- and post-infection treatments demonstrated anti-flaviviral activity against Dengue and Zika virus, via viral RNA polymerase inhibition. These data suggest that Sophoraflavenone G represents a promising candidate regarding anti-Flaviviridae research.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Flavanonas/farmacologia , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Sophora/química , Zika virus/efeitos dos fármacos , Antivirais/química , Antivirais/isolamento & purificação , Linhagem Celular , Dengue/tratamento farmacológico , Vírus da Dengue/enzimologia , Vírus da Dengue/fisiologia , Descoberta de Drogas , Flavanonas/química , Flavanonas/isolamento & purificação , Flaviviridae/efeitos dos fármacos , Hepacivirus/efeitos dos fármacos , Humanos , Raízes de Plantas/química , Replicação Viral/efeitos dos fármacos , Zika virus/enzimologia , Zika virus/fisiologia , Infecção por Zika virus/tratamento farmacológico
12.
Antiviral Res ; 146: 161-163, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28912011

RESUMO

The recent epidemic of Zika virus (ZIKV) in the Americas and its association with fetal and neurological complications has shown the need to develop a treatment. Repurposing of drugs that are already FDA approved or in clinical development may shorten drug development timelines in case of emerging viral diseases like ZIKV. Initial studies have shown conflicting results when testing sofosbuvir developed for treatment of infections with another Flaviviridae virus, hepatitis C virus. We hypothesized that the conflicting results could be explained by differences in intracellular processing of the compound. We assessed the antiviral activity of sofosbuvir and mericitabine against ZIKV using Vero, A549, and Huh7 cells and measured the level of the active sofosbuvir metabolite by mass spectrometry. Mericitabine did not show activity, while sofosbuvir inhibited ZIKV with an IC50 of ∼4 µM, but only in Huh7 cells. This correlated with differences in intracellular concentration of the active triphosphate metabolite of sofosbuvir, GS-461203 or 007-TP, which was 11-342 times higher in Huh7 cells compared to Vero and A549 cells. These results show that a careful selection of cell system for repurposing trials of prodrugs is needed for evaluation of antiviral activity. Furthermore, the intracellular levels of 007-TP in tissues and cell types that support ZIKV replication in vivo should be determined to further investigate the potential of sofosbuvir as anti-ZIKV compound.


Assuntos
Antivirais/farmacologia , Sofosbuvir/farmacologia , Replicação Viral/efeitos dos fármacos , Zika virus/efeitos dos fármacos , Células A549 , Animais , Linhagem Celular , Chlorocebus aethiops , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Reposicionamento de Medicamentos , Flaviviridae/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Células Vero , Zika virus/fisiologia
13.
Virol J ; 14(1): 95, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28506240

RESUMO

Flaviviruses are small viruses with single-stranded RNA, which include the yellow fever virus, dengue virus, West Nile virus, Japanese encephalitis virus, tick-borne encephalitis virus, and Zika virus; and are causal agents of the most important emerging diseases that have no available treatment to date. In recent years, the strategy has focused on the development of replication inhibitors of these viruses designed to act mainly by affecting the activity of enzyme proteins, such as NS3 and NS5, which perform important functions in the viral replication process. This article describes the importance of flaviviruses and the development of molecules used as inhibitors of viral replication in this genus.


Assuntos
Antivirais/isolamento & purificação , Antivirais/farmacologia , Descoberta de Drogas/tendências , Infecções por Flaviviridae/epidemiologia , Flaviviridae/efeitos dos fármacos , Flaviviridae/fisiologia , Replicação Viral/efeitos dos fármacos , Infecções por Flaviviridae/virologia , Humanos
14.
J Gen Virol ; 97(10): 2552-2565, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27489039

RESUMO

Natural 2'-modified nucleosides are the most widely used antiviral therapy. In their triphosphorylated form, also known as nucleotide analogues, they target the active site of viral polymerases. Viral polymerases have an overall right-handed structure that includes the palm, fingers and thumb domains. These domains are further subdivided into structurally conserved motifs A-G, common to all viral polymerases. The structural motifs encapsulate the allosteric/initiation (N1) and orthosteric/catalytic (N2) nucleotide-binding sites. The current study investigated how nucleotide analogues explore the N2 site of viral polymerases from three genera of the family Flaviviridae using a stochastic, biophysical, Metropolis Monte Carlo-based software. The biophysical simulations showed a statistical distinction in nucleotide-binding energy and exploration between phylogenetically related viral polymerases. This distinction is clearly demonstrated by the respective analogue contacts made with conserved viral polymerase residues, the heterogeneous dynamics of structural motifs, and the orientation of the nucleotide analogues within the N2 site. Being able to simulate what occurs within viral-polymerase-binding sites can prove useful in rational drug designs against viruses.


Assuntos
Antivirais/farmacologia , RNA Polimerases Dirigidas por DNA/antagonistas & inibidores , RNA Polimerases Dirigidas por DNA/química , Flaviviridae/efeitos dos fármacos , Flaviviridae/enzimologia , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/química , Antivirais/química , Sítios de Ligação , Domínio Catalítico , RNA Polimerases Dirigidas por DNA/metabolismo , Desenho de Fármacos , Flaviviridae/química , Flaviviridae/genética , Humanos , Nucleosídeos/química , Nucleosídeos/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
15.
Mol Biosyst ; 12(7): 2080-93, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-26864387

RESUMO

The rate of Flaviviridae family virus infections worldwide has increased dramatically in the last few years. In addition, infections caused by arthropod vector viruses including Hepatitis C, West Nile, Dengue fever, Yellow fever and Japanese encephalitis are emerging throughout the world. Based on a recent taxon update, the Flaviviridae family comprises four main genera; Flavivirus, Hepacivirus, Pestivirus and a recent genus Pegivirus. Although the new scientific classification plays a key role in providing useful information about the relationships between viruses, many new documented viruses remain unclassified. Furthermore, based on the different results of several studies the classification is unclear. In an effort to provide more insights into the classification of viruses, a holistic evolutionary study of the two viral enzymes NS3 helicase and NS5 RNA-dependent RNA polymerase (RdRp) has been conducted in this study. These two viral enzymes are very crucial for the inhibition of viruses due to the fact that they are involved in the survival, proliferation and transmission of viruses. The main goal of this study is the presentation of two novel updated phylogenetic trees of the enzymes NS3 helicase and NS5 RdRp as a reliable phylogeny "map" to correlate the information of the closely related viruses and identify new possible targets for the Flaviviridae family virus inhibition. Despite the earliest trials for drugs against Flaviviridae related viruses, no antiviral drug vaccine has been available to date. Therefore there is an urgent need for research towards the development of efficient antiviral agents.


Assuntos
Flaviviridae/genética , Domínios e Motivos de Interação entre Proteínas , Proteínas não Estruturais Virais/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Antivirais/farmacologia , Sequência Conservada , Bases de Dados Genéticas , Flaviviridae/efeitos dos fármacos , Flaviviridae/enzimologia , Modelos Moleculares , Filogenia , Conformação Proteica , RNA Helicases/antagonistas & inibidores , RNA Helicases/química , RNA Helicases/genética , Análise de Sequência de DNA , Serina Endopeptidases/química , Serina Endopeptidases/genética , Relação Estrutura-Atividade , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/química
16.
Viruses ; 6(7): 2531-50, 2014 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-24967693

RESUMO

RNA viruses are capable of rapid spread and severe or potentially lethal disease in both animals and humans. The development of reverse genetics systems for manipulation and study of RNA virus genomes has provided platforms for designing and optimizing viral mutants for vaccine development. Here, we review the impact of RNA virus reverse genetics systems on past and current efforts to design effective and safe viral therapeutics and vaccines.


Assuntos
Genoma Viral , Infecções por Vírus de RNA/prevenção & controle , Genética Reversa/métodos , Vacinas Virais/genética , Animais , Coronaviridae/efeitos dos fármacos , Coronaviridae/genética , Coronaviridae/imunologia , Flaviviridae/efeitos dos fármacos , Flaviviridae/genética , Flaviviridae/imunologia , Engenharia Genética , Humanos , Orthomyxoviridae/efeitos dos fármacos , Orthomyxoviridae/genética , Orthomyxoviridae/imunologia , Paramyxoviridae/efeitos dos fármacos , Paramyxoviridae/genética , Paramyxoviridae/imunologia , Infecções por Vírus de RNA/imunologia , Infecções por Vírus de RNA/virologia , Vacinas Virais/administração & dosagem , Vacinas Virais/biossíntese
17.
Curr Pharm Des ; 20(21): 3428-44, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24001228

RESUMO

Viruses belonging to the Flaviviridae family primarily spread through arthropod vectors, and are the major causes of illness and death around the globe. The Flaviviridae family consists of 3 genera which include the Flavivirus genus (type species, yellow fever virus) as the largest genus, the Hepacivirus (type species, hepatitis C virus) and the Pestivirus (type species, bovine virus diarrhea). The flaviviruses (Flavivirus genus) are small RNA viruses transmitted by mosquitoes and ticks that take over host cell machinery in order to propagate. However, hepaciviruses and pestiviruses are not antropod-borne. Despite the extensive research and public health concern associated with flavivirus diseases, to date, there is no specific treatment available for any flavivirus infections, though commercially available vaccines for yellow fever, Japanese encephalitis and tick-born encephalitis exist. Due to the global threat of viral pandemics, there is an urgent need for new drugs. In many countries, patients with severe cases of flavivirus infections are treated only by supportive care, which includes intravenous fluids, hospitalization, respiratory support, and prevention of secondary infections. This review discusses the strategies used towards the discovery of antiviral drugs, focusing on rational drug design against Dengue virus (DENV), West Nile virus (WNV), Japanese encephalitis virus (JEV), Yellow Fever virus (YFV) and Hepatitis C virus (HCV). Only modified peptidic, nonpeptidic, natural compounds and fragment-based inhibitors (typically of mass less than 300 Da) against structural and non-structural proteins are discussed.


Assuntos
Antivirais/farmacologia , Descoberta de Drogas/métodos , Flaviviridae/efeitos dos fármacos , Antivirais/uso terapêutico , Desenho de Fármacos , Flaviviridae/enzimologia , Infecções por Flaviviridae/tratamento farmacológico , Humanos , Terapia de Alvo Molecular/métodos , Proteínas Virais/antagonistas & inibidores
18.
Expert Opin Drug Discov ; 7(5): 417-28, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22439769

RESUMO

INTRODUCTION: The members of the family Flaviviridae, including West Nile virus, yellow fever virus and dengue virus, are important human pathogens that are expanding their impact around the globe. The four serotypes of dengue infect 50-100 million people each year, yet the only clinical treatment is supportive care to reduce symptoms. Drugs that employ novel inhibition mechanisms and targets are urgently needed to combat the growing incidence of dengue worldwide. AREAS COVERED: The authors discuss recently discovered flavivirus inhibitors with a focus on antivirals targeting non-enzymatic proteins of the dengue virus lifecycle. Specifically, the authors discuss the flaviviruses, the need for novel inhibitors and the criteria for successful antiviral drug development. Current literature describing new advances in antiviral therapy at each stage of the flavivirus lifecycle (entry, endosomal escape, viral RNA processing and replication, assembly and immune evasion) are evaluated and summarized. EXPERT OPINION: Overall, the prognosis of flavivirus antiviral drug development is positive: new effective compounds have been discovered and studied. However, repurposing existing compounds and a greater translation to the clinical setting are recommended in order to combat the growing threat of flaviviruses.


Assuntos
Antivirais/química , Descoberta de Drogas/métodos , Flaviviridae/efeitos dos fármacos , Infecções por Flavivirus/tratamento farmacológico , RNA Viral/antagonistas & inibidores , Proteínas do Envelope Viral/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Alcaloides de Amaryllidaceae/farmacologia , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Vírus da Dengue/efeitos dos fármacos , Infecções por Flavivirus/virologia , Humanos , Camundongos , Nanotubos/química , Peptídeos/farmacologia , Fenantridinas/farmacologia , RNA Interferente Pequeno/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Vírus do Nilo Ocidental/efeitos dos fármacos , Vírus da Febre Amarela/efeitos dos fármacos
19.
Bioorg Med Chem ; 19(23): 7070-84, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22047799

RESUMO

In this study three new classes of linear N-tricyclic compounds, derived by condensation of the quinoline nucleus with 1,2,3-triazole, imidazole or pyrazine, were synthesized, obtaining triazolo[4,5-g]quinolines, imidazo[4,5-g]quinolines and pyrido[2,3-g]quinoxalines, respectively. Title compounds were tested in cell-based assays for cytotoxicity and antiviral activity against RNA viruses representative of the three genera of the Flaviviridae family, that is BVDV (Pestivirus), YFV (Flavivirus) and HCV (Hepacivirus). Quinoline derivatives were also tested against representatives of other RNA virus families containing single-stranded, either positive-sense (ssRNA(+)) or negative-sense (RNA(-)), and double-stranded genomes (dsRNA), as well as against representatives of two DNA virus families. Some quinolines showed moderate, although selective activity against CVB-5, Reo-1 and RSV. However, derivatives belonging to all classes showed activity against BVDV. Among the most potent were the bis-triazoloquinoline 1m, the imidazoquinolines 2e and 2h, and the pyridoquinoxalines 4h, 4j and 5n (EC(50) range 1-5 µM). When tested in a replicon assay, compound 2h was the sole derivative to also display anti-HCV activity (EC(50)=3.1 µM). In enzyme assays, 1m, 2h, 5m and 5n proved to be potent inhibitors of the BVDV RNA-dependent RNA polymerase (RdRp), while only 2h also inhibited the recombinant HCV enzyme.


Assuntos
Antivirais/química , Antivirais/farmacologia , Quinolinas/síntese química , Quinolinas/farmacologia , RNA Polimerase Dependente de RNA/antagonistas & inibidores , Antivirais/síntese química , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Flaviviridae/efeitos dos fármacos , Flaviviridae/enzimologia , Humanos , Estrutura Molecular , Relação Estrutura-Atividade
20.
Chem Biol Drug Des ; 77(6): 441-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21414145

RESUMO

With the aim of identifying novel lead compounds active against emergent human infectious diseases, a series of 2,3-dihydro-4H-pyridinone derivatives has been prepared and evaluated for antiviral activity. Compounds were evaluated in vitro in cell-based assays for cytotoxicity and against a wide spectrum of viruses. In the antiviral screening, several compounds showed to be fairly active against viruses belonging to the Flaviviridae family. The Pestiviruses (bovine viral diarrhoea virus) were inhibited by 4a cis (CC(50) > 100 µm; EC(50) = 14 µm), compounds 4c cis and 6a showed a significant activity against Flaviviruses (Yellow Fever Virus) (CC(50) > 100 µm; EC(50) = 18µm, CC(50) > 100 µm; EC(50) = 10 µm). Among these, compound 6a displayed great inhibitory activity against Hepaciviruses (hepatitis C virus) in replicon assay [CC(50) > 100 µm; EC(50) (1b) = 4 µm]. In vitro inhibitory activity against the HCV RNA-dependent RNA polymerase (NS5B) of title compounds is discussed. The antiviral screening of viral strains indicated that compound 6a can be selected as promising tool in novel anti-flaviviruses development.


Assuntos
Antivirais/química , Antivirais/farmacologia , Infecções por Flaviviridae/tratamento farmacológico , Flaviviridae/efeitos dos fármacos , Piridonas/química , Piridonas/farmacologia , Animais , Linhagem Celular , Humanos , Testes de Sensibilidade Microbiana , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...