Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Antiviral Res ; 210: 105516, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36586467

RESUMO

Flaviviruses are important human pathogens and include dengue (DENV), West Nile (WNV), Yellow fever virus (YFV), Japanese encephalitis (JEV) and Zika virus (ZIKV). DENV, transmitted by mosquitoes, causes diseases ranging in severity from mild dengue fever with non-specific flu-like symptoms to fatal dengue hemorrhagic fever and dengue shock syndrome. DENV infections are caused by four serotypes, DENV1-4, which interact differently with antibodies in blood serum. The incidence of DENV infection has increased dramatically in recent decades and the CDC estimates 400 million dengue infections occur each year, resulting in ∼25,000 deaths mostly among children and elderly people. Similarly, ZIKV infections are caused by infected mosquito bites to humans, can be transmitted sexually and through blood transfusions. If a pregnant woman is infected, the virus can cross the placental barrier and can spread to her fetus, causing severe brain malformations in the child including microcephaly and other birth defects. It is noteworthy that the neurological manifestations of ZIKV were also observed in DENV endemic regions, suggesting that pre-existing antibody response to DENV could augment ZIKV infection. WNV, previously unknown in the US (and known to cause only mild disease in Middle East), first arrived in New York city in 1999 (NY99) and spread throughout the US and Canada by Culex mosquitoes and birds. WNV is now endemic in North America. Thus, emerging and re-emerging flaviviruses are significant threat to human health. However, vaccines are available for only a limited number of flaviviruses, and antiviral therapies are not available for any flavivirus. Hence, there is an urgent need to develop therapeutics that interfere with essential enzymatic steps, such as protease in the flavivirus lifecycle as these viruses possess significant threat to future pandemics. In this review, we focus on our E. coli expression of NS2B hydrophilic domain (NS2BH) covalently linked to NS3 protease domain (NS3Pro) in their natural context which is processed by the combined action of both subunits of the NS2B-NS3Pro precursor. Biochemical activities of the viral protease such as solubility and autoproteolysis of NS2BH-NS3Pro linkage depended on the C-terminal portion of NS2BH linked to the NS3Pro domain. Since 2008, we also focus on the use of the recombinant protease in high throughput screens and characterization of small molecular compounds identified in these screens.


Assuntos
Infecções por Flavivirus , Flavivirus , Peptídeo Hidrolases , Animais , Feminino , Humanos , Gravidez , Dengue/prevenção & controle , Vírus da Dengue , Flavivirus/enzimologia , Pandemias , Placenta , Zika virus , Infecção por Zika virus/prevenção & controle , Infecções por Flavivirus/prevenção & controle
2.
Nat Commun ; 12(1): 2530, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33953197

RESUMO

Flaviviruses use a ~70 nucleotide stem-loop structure called stem-loop A (SLA) at the 5' end of the RNA genome as a promoter for RNA synthesis. Flaviviral polymerase NS5 specifically recognizes SLA to initiate RNA synthesis and methylate the 5' guanosine cap. We report the crystal structures of dengue (DENV) and Zika virus (ZIKV) SLAs. DENV and ZIKV SLAs differ in the relative orientations of their top stem-loop helices to bottom stems, but both form an intermolecular three-way junction with a neighboring SLA molecule. To understand how NS5 engages SLA, we determined the SLA-binding site on NS5 and modeled the NS5-SLA complex of DENV and ZIKV. Our results show that the gross conformational differences seen in DENV and ZIKV SLAs can be compensated by the differences in the domain arrangements in DENV and ZIKV NS5s. We describe two binding modes of SLA and NS5 and propose an SLA-mediated RNA synthesis mechanism.


Assuntos
Flavivirus/genética , Regiões Promotoras Genéticas , RNA Viral/química , RNA Viral/genética , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Sítios de Ligação , Cristalografia por Raios X , Vírus da Dengue/genética , Ligação Proteica , RNA Viral/metabolismo , Replicação Viral/fisiologia , Zika virus/genética
3.
Virology ; 552: 32-42, 2021 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-33059318

RESUMO

The HEK-293 cell line was created in 1977 by transformation of primary human embryonic kidney cells with sheared adenovirus type 5 DNA. A previous study determined that the HEK-293 cells have neuronal markers rather than kidney markers. In this study, we tested the hypothesis whether Zika virus (ZIKV), a neurotropic virus, is able to infect and replicate in the HEK-293 cells. We show that the HEK-293 cells infected with ZIKV support viral replication as shown by indirect immunofluorescence (IFA) and quantitative reverse transcriptase-PCR (qRT-PCR). We performed RNA-seq analysis on the ZIKV-infected and the control uninfected HEK-293 cells and find 659 genes that are differentially transcribed in ZIKV-infected HEK-293 cells as compared to uninfected cells. The results show that the top 10 differentially transcribed and upregulated genes are involved in antiviral and inflammatory responses. Seven upregulated genes, IFNL1, DDX58, CXCL10, ISG15, KCNJ15, IFNIH1, and IFIT2, were validated by qRT-PCR. Altogether, our findings show that ZIKV infection alters host gene expression by affecting their antiviral and inflammatory responses.


Assuntos
Regulação da Expressão Gênica , Inflamação/virologia , Infecção por Zika virus/metabolismo , Infecção por Zika virus/virologia , Zika virus/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Quimiocina CXCL10/metabolismo , Citocinas/metabolismo , Proteína DEAD-box 58/metabolismo , Técnica Indireta de Fluorescência para Anticorpo/métodos , Células HEK293 , Interações entre Hospedeiro e Microrganismos , Humanos , Helicase IFIH1 Induzida por Interferon/metabolismo , Interferons/metabolismo , Interleucinas/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Proteínas de Ligação a RNA/metabolismo , RNA-Seq , Receptores Imunológicos/metabolismo , Ubiquitinas/metabolismo , Zika virus/imunologia , Infecção por Zika virus/imunologia
4.
Proc Natl Acad Sci U S A ; 117(49): 31365-31375, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33229545

RESUMO

When Zika virus emerged as a public health emergency there were no drugs or vaccines approved for its prevention or treatment. We used a high-throughput screen for Zika virus protease inhibitors to identify several inhibitors of Zika virus infection. We expressed the NS2B-NS3 Zika virus protease and conducted a biochemical screen for small-molecule inhibitors. A quantitative structure-activity relationship model was employed to virtually screen ∼138,000 compounds, which increased the identification of active compounds, while decreasing screening time and resources. Candidate inhibitors were validated in several viral infection assays. Small molecules with favorable clinical profiles, especially the five-lipoxygenase-activating protein inhibitor, MK-591, inhibited the Zika virus protease and infection in neural stem cells. Members of the tetracycline family of antibiotics were more potent inhibitors of Zika virus infection than the protease, suggesting they may have multiple mechanisms of action. The most potent tetracycline, methacycline, reduced the amount of Zika virus present in the brain and the severity of Zika virus-induced motor deficits in an immunocompetent mouse model. As Food and Drug Administration-approved drugs, the tetracyclines could be quickly translated to the clinic. The compounds identified through our screening paradigm have the potential to be used as prophylactics for patients traveling to endemic regions or for the treatment of the neurological complications of Zika virus infection.


Assuntos
Antivirais/análise , Antivirais/farmacologia , Avaliação Pré-Clínica de Medicamentos , Ensaios de Triagem em Larga Escala , Inibidores de Proteases/análise , Inibidores de Proteases/farmacologia , Zika virus/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Inteligência Artificial , Chlorocebus aethiops , Modelos Animais de Doenças , Imunocompetência , Concentração Inibidora 50 , Metaciclina/farmacologia , Camundongos Endogâmicos C57BL , Inibidores de Proteases/uso terapêutico , Relação Quantitativa Estrutura-Atividade , Bibliotecas de Moléculas Pequenas , Células Vero , Infecção por Zika virus/tratamento farmacológico , Infecção por Zika virus/virologia
5.
J Virol ; 93(15)2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31092581

RESUMO

Dengue virus (DENV) causes frequent epidemics infecting ∼390 million people annually in over 100 countries. There are no approved vaccines or antiviral drugs for treatment of infected patients. However, there is a novel approach to control DENV transmission by the mosquito vectors, Aedes aegypti and Aedes albopictus, using the Wolbachia symbiont. The wMelPop strain of Wolbachia suppresses DENV transmission and shortens the mosquito life span. However, the underlying mechanism is poorly understood. To clarify this mechanism, either naive A. albopictus (C6/36) or wMelPop-C6/36 cells were infected with DENV serotype 2 (DENV2). Analysis of host transcript profiles by transcriptome sequencing (RNAseq) revealed that the presence of wMelPop dramatically altered the mosquito host cell transcription in response to DENV2 infection. The viral RNA evolved from wMelPop-C6/36 cells contained low-frequency mutations (∼25%) within the coding region of transmembrane domain 1 (TMD1) of E protein. Mutations with >97% frequencies were distributed within other regions of E, the NS5 RNA-dependent RNA polymerase (NS5POL) domain, and the TMDs of NS2A, NS2B, and NS4B. Moreover, while DENV2-infected naive C6/36 cells showed syncytium formation, DENV2-infected wMelPop-C6/36 cells did not. The Wolbachia-induced mutant DENV2 can readily infect and replicate in naive C6/36 cells, whereas in mutant DENV2-infected BHK-21 or Vero cells, virus replication was delayed. In LLC-MK2 cells, the mutant failed to produce plaques. Additionally, in BHK-21 cells, many mutations in the viral genome reverted to the wild type (WT) and compensatory mutations in NS3 gene appeared. Our results indicate that wMelPop impacts significantly the interactions of DENV2 with mosquito and mammalian host cells.IMPORTANCE Mosquito-borne diseases are of global significance causing considerable morbidity and mortality throughout the world. Dengue virus (DENV; serotypes 1 to 4), a member of the Flavivirus genus of the Flaviviridae family, causes millions of infections annually. Development of a safe vaccine is hampered due to absence of cross-protection and increased risk in secondary infections due to antibody-mediated immune enhancement. Infection of vector mosquitoes with Wolbachia bacteria offers a novel countermeasure to suppress DENV transmission, but the mechanisms are poorly understood. In this study, the host transcription profiles and viral RNA sequences were analyzed in naive A. albopictus (C6/36) and wMelPop-C6/36 cells by RNAseq. Our results showed that the wMelPop symbiont caused profound changes in host transcription profiles and morphology of DENV2-infected C6/36 cells. Accumulation of several mutations throughout DENV2 RNA resulted in loss of infectivity of progeny virions. Our findings offer new insights into the mechanism of Wolbachia-mediated suppression of DENV transmission.


Assuntos
Aedes , Vírus da Dengue/crescimento & desenvolvimento , Regulação da Expressão Gênica , Genoma Viral , Interações Microbianas , Mutação , Wolbachia/crescimento & desenvolvimento , Animais , Linhagem Celular , Cercopithecus , Cricetinae , Vírus da Dengue/genética , Perfilação da Expressão Gênica , Interações entre Hospedeiro e Microrganismos , Análise de Sequência de RNA , Replicação Viral
6.
Antiviral Res ; 162: 71-78, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30529358

RESUMO

The dengue virus is considered to be a globally important human pathogen prevalent in tropical and subtropical regions of the world. According to a recent estimate, the disease burden due to DENV infections is ∼390 million infections per year globally in ∼100 countries including the southern US, Puerto Rico and Hawaii, resulting in nearly ∼25,000 deaths mostly among children. Despite the significant morbidity and mortality that results from DENV infections, there is currently no effective chemotherapeutic treatment for DENV infections. We identified curcumin as an inhibitor of DENV2 NS2B/NS3protease in a previous high-throughput screening (HTS) campaign. We synthesized four analogues of curcumin (curcuminoids) and tested the in vitro protease inhibition activity and inhibition of replication by cell-based assays. The results revealed that curcumin is a weak inhibitor of the viral protease. However, the analogues exhibited more potent inhibition of DENV infectivity in plaque assays suggesting that the cellular pathway(s) required for viral replication and/or assembly are targeted by these compounds. Further analysis shows that inhibition of genes involved in lipid biosynthesis, and of actin polymerization by curcuminoids, are likely to be involved as their mode of action in DENV2-infected cells. Three of the curcumin derivatives possess good selectivity indices (SI) (>10) when compared to the parent curcumin.


Assuntos
Acetil-CoA Carboxilase/antagonistas & inibidores , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Diarileptanoides/farmacologia , Ácido Graxo Sintases/antagonistas & inibidores , Animais , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Vírus da Dengue/fisiologia , Diarileptanoides/análogos & derivados , Humanos , Macaca mulatta , Replicação Viral/efeitos dos fármacos
7.
J Biol Chem ; 292(23): 9465-9479, 2017 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-28396347

RESUMO

Four serotypes of mosquito-borne dengue virus (DENV), evolved from a common ancestor, are human pathogens of global significance for which there is no vaccine or antiviral drug available. The N-terminal domain of DENV NS5 has guanylyltransferase and methyltransferase (MTase), and the C-terminal region has the polymerase (POL), all of which are important for 5'-capping and RNA replication. The crystal structure of NS5 shows it as a dimer, but the functional evidence for NS5 dimer is lacking. Our studies showed that the substitution of DENV2 NS5 MTase or POL for DENV4 NS5 within DENV2 RNA resulted in a severe attenuation of replication in the transfected BHK-21 cells. A replication-competent species was evolved with the acquired mutations in the DENV2 and DENV4 NS5 MTase or POL domain or in the DENV2 NS3 helicase domain in the DENV2 chimera RNAs by repeated passaging of infected BHK-21 or mosquito cells. The linker region of seven residues in NS5, rich in serotype-specific residues, is important for the recovery of replication fitness in the chimera RNA. Our results, taken together, provide genetic evidence for a serotype-specific interaction between NS3 and NS5 as well as specific interdomain interaction within NS5 required for RNA replication. Genome-wide RNAseq analysis revealed the distribution of adaptive mutations in RNA quasispecies. Those within NS3 and NS5 are located at the surface and/or within the NS5 dimer interface, providing a functional significance to the crystal structure NS5 dimer.


Assuntos
Vírus da Dengue/fisiologia , RNA Viral , Sorogrupo , Proteínas não Estruturais Virais , Replicação Viral/fisiologia , Animais , Linhagem Celular , Cricetinae , Culicidae , Humanos , Domínios Proteicos , RNA Helicases/química , RNA Helicases/genética , RNA Helicases/imunologia , RNA Helicases/metabolismo , RNA Viral/biossíntese , RNA Viral/química , RNA Viral/genética , RNA Viral/imunologia , Serina Endopeptidases/química , Serina Endopeptidases/genética , Serina Endopeptidases/imunologia , Serina Endopeptidases/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/metabolismo
8.
Antiviral Res ; 137: 141-150, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27889529

RESUMO

In a previous study, twelve antimalarial compounds, amodiaquine (AQ) and derivatives, were shown to have potent anti-dengue viral (DENV) activity by using the stable DENV2 Renilla luciferase reporter replicon expressing BHK-21 cells, infectivity (plaque), and the qRT-PCR assays. In this study, we performed molecular modeling on these compounds to determine their stereo-electronic properties required for optimal antiviral activity. Based on the similarity of calculated stereo-electronic profiles, specifically the electrostatic potential profiles of the compounds, and in silico screening of related compounds from literature, we identified three additional compounds, Quinacrine (QC), Mefloquine (MQ), and GSK369796. Analysis of their antiviral activities indicated that all three compounds have high anti-DENV activity in the DENV2 replicon expressing cells with EC50 values of 5.30 ± 1.31 µM (QC), 3.22 ± 0.37 µM (MQ), and 5.06 ± 0.86 µM (GSK369796). The infectivity assays revealed the EC50 values of 7.09 ± 1.67 µM (QC), 4.36 ± 0.31 µM (MQ) and 3.03 ± 0.35 µM (GSK369796). The mode of action of these compounds is through inhibition of autophagy, thereby affecting DENV2 replication. Moreover, these compounds also showed antiviral activity against the rapidly emerging Zika virus (ZIKV) with EC50 values of 2.27 ± 0.14 µM (QC), 3.95 ± 0.21 µM (MQ), and 2.57 ± 0.09 µM (GSK369796).


Assuntos
Antimaláricos/farmacologia , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Zika virus/efeitos dos fármacos , Amodiaquina/análogos & derivados , Amodiaquina/química , Amodiaquina/farmacologia , Antimaláricos/química , Autofagia/efeitos dos fármacos , Simulação por Computador , Vírus da Dengue/fisiologia , Descoberta de Drogas , Humanos , Mefloquina/química , Mefloquina/farmacologia , Quinacrina/química , Quinacrina/farmacologia , Replicon/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Zika virus/fisiologia
9.
Antiviral Res ; 134: 6-16, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27539384

RESUMO

The mosquito-borne dengue virus serotypes 1-4 (DENV1-4) and West Nile virus (WNV) cause serious illnesses worldwide associated with considerable morbidity and mortality. According to the World Health Organization (WHO) estimates, there are about 390 million infections every year leading to ∼500,000 dengue haemorrhagic fever (DHF) cases and ∼25,000 deaths, mostly among children. Antiviral therapies could reduce the morbidity and mortality associated with flaviviral infections, but currently there are no drugs available for treatment. In this study, a high-throughput screening assay for the Dengue protease was employed to screen ∼120,000 small molecule compounds for identification of inhibitors. Eight of these inhibitors have been extensively analyzed for inhibition of the viral protease in vitro and cell-based viral replication using Renilla luciferase reporter replicon, infectivity (plaque) and cytotoxicity assays. Three of these compounds were identified as potent inhibitors of DENV and WNV proteases, and viral replication of DENV2 replicon and infectious RNA. Fluorescence quenching, kinetic analysis and molecular modeling of these inhibitors into the structure of NS2B-NS3 protease suggest a mode of inhibition for three compounds that they bind to the substrate binding pocket.


Assuntos
Flavivirus/efeitos dos fármacos , Peptídeo Hidrolases/efeitos dos fármacos , Inibidores de Proteases/isolamento & purificação , Inibidores de Proteases/farmacologia , Bibliotecas de Moléculas Pequenas/química , Antivirais/química , Antivirais/isolamento & purificação , Antivirais/farmacologia , Sítios de Ligação , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/enzimologia , Descoberta de Drogas/métodos , Flavivirus/enzimologia , Fluorescência , Ensaios de Triagem em Larga Escala/métodos , Cinética , Luciferases de Renilla/genética , Modelos Moleculares , Inibidores de Proteases/química , Replicon/efeitos dos fármacos , Proteínas não Estruturais Virais/química , Ensaio de Placa Viral , Replicação Viral/efeitos dos fármacos , Vírus do Nilo Ocidental/efeitos dos fármacos , Vírus do Nilo Ocidental/enzimologia
10.
PLoS Pathog ; 12(2): e1005451, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26895240

RESUMO

Flavivirus nonstructural protein 5 (NS5) consists of methyltransferase (MTase) and RNA-dependent RNA polymerase (RdRp) domains, which catalyze 5'-RNA capping/methylation and RNA synthesis, respectively, during viral genome replication. Although the crystal structure of flavivirus NS5 is known, no data about the quaternary organization of the functional enzyme are available. We report the crystal structure of dengue virus full-length NS5, where eight molecules of NS5 are arranged as four independent dimers in the crystallographic asymmetric unit. The relative orientation of each monomer within the dimer, as well as the orientations of the MTase and RdRp domains within each monomer, is conserved, suggesting that these structural arrangements represent the biologically relevant conformation and assembly of this multi-functional enzyme. Essential interactions between MTase and RdRp domains are maintained in the NS5 dimer via inter-molecular interactions, providing evidence that flavivirus NS5 can adopt multiple conformations while preserving necessary interactions between the MTase and RdRp domains. Furthermore, many NS5 residues that reduce viral replication are located at either the inter-domain interface within a monomer or at the inter-molecular interface within the dimer. Hence the X-ray structure of NS5 presented here suggests that MTase and RdRp activities could be coordinated as a dimer during viral genome replication.


Assuntos
Vírus da Dengue/metabolismo , Metiltransferases/genética , Multimerização Proteica , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Estrutura Terciária de Proteína , RNA Polimerase Dependente de RNA/metabolismo , Proteínas não Estruturais Virais/química
11.
Methods ; 91: 20-34, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26272247

RESUMO

Establishment of in vitro systems to study mechanisms of RNA synthesis for positive strand RNA viruses have been very useful in the past and have shed light on the composition of protein and RNA components, optimum conditions, the nature of the products formed, cis-acting RNA elements and trans-acting protein factors required for efficient synthesis. In this review, we summarize our current understanding regarding the requirements for flavivirus RNA synthesis in vitro. We describe details of reaction conditions, the specificity of template used by either the multi-component membrane-bound viral replicase complex or by purified, recombinant RNA-dependent RNA polymerase. We also discuss future perspectives to extend the boundaries of our knowledge.


Assuntos
Flavivirus/metabolismo , Técnicas de Amplificação de Ácido Nucleico/métodos , RNA Viral/biossíntese , RNA Polimerase Dependente de RNA/metabolismo , Flavivirus/genética
12.
Viruses ; 7(8): 4640-56, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26287232

RESUMO

Genome replication in flavivirus requires (-) strand RNA synthesis, (+) strand RNA synthesis, and 51-RNA capping and methylation. To carry out viral genome replication, flavivirus assembles a replication complex, consisting of both viral and host proteins, on the cytoplasmic side of the endoplasmic reticulum (ER) membrane. Two major components of the replication complex are the viral non-structural (NS) proteins NS3 and NS5. Together they possess all the enzymatic activities required for genome replication, yet how these activities are coordinated during genome replication is not clear. We provide an overview of the flaviviral genome replication process, the membrane-bound replication complex, and recent crystal structures of full-length NS5. We propose a model of how NS3 and NS5 coordinate their activities in the individual steps of (-) RNA synthesis, (+) RNA synthesis, and 51-RNA capping and methylation.


Assuntos
Flavivirus/fisiologia , Capuzes de RNA/biossíntese , RNA Viral/biossíntese , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Flavivirus/genética , Regulação Viral da Expressão Gênica , Humanos , RNA Helicases/metabolismo , Serina Endopeptidases/metabolismo
13.
Nucleic Acids Res ; 43(2): 1069-80, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25564528

RESUMO

RNA helicases impact RNA structure and metabolism from transcription through translation, in part through protein interactions with transcription factors. However, there is limited knowledge on the role of transcription factor influence upon helicase activity. RNA helicase A (RHA) is a DExH-box RNA helicase that plays multiple roles in cellular biology, some functions requiring its activity as a helicase while others as a protein scaffold. The oncogenic transcription factor EWS-FLI1 requires RHA to enable Ewing sarcoma (ES) oncogenesis and growth; a small molecule, YK-4-279 disrupts this complex in cells. Our current study investigates the effect of EWS-FLI1 upon RHA helicase activity. We found that EWS-FLI1 reduces RHA helicase activity in a dose-dependent manner without affecting intrinsic ATPase activity; however, the RHA kinetics indicated a complex model. Using separated enantiomers, only (S)-YK-4-279 reverses the EWS-FLI1 inhibition of RHA helicase activity. We report a novel RNA binding property of EWS-FLI1 leading us to discover that YK-4-279 inhibition of RHA binding to EWS-FLI1 altered the RNA binding profile of both proteins. We conclude that EWS-FLI1 modulates RHA helicase activity causing changes in overall transcriptome processing. These findings could lead to both enhanced understanding of oncogenesis and provide targets for therapy.


Assuntos
RNA Helicases DEAD-box/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Proteína Proto-Oncogênica c-fli-1/metabolismo , Proteína EWS de Ligação a RNA/metabolismo , RNA Helicases DEAD-box/antagonistas & inibidores , RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/genética , Indóis/farmacologia , Modelos Moleculares , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , RNA/metabolismo , Proteínas Recombinantes/metabolismo
14.
J Biol Chem ; 289(32): 22385-400, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-24904061

RESUMO

Flavivirus NS3 and NS5 are required in viral replication and 5'-capping. NS3 has NS2B-dependent protease, RNA helicase, and 5'-RNA triphosphatase activities. NS5 has 5'-RNA methyltransferase (MT)/guanylyltransferase (GT) activities within the N-terminal 270 amino acids and the RNA-dependent RNA polymerase (POL) activity within amino acids 271-900. A chimeric NS5 containing the D4MT/D4GT and the D2POL domains in the context of wild-type (WT) D2 RNA was constructed. RNAs synthesized in vitro were transfected into baby hamster kidney cells. The viral replication was analyzed by an indirect immunofluorescence assay to monitor NS1 expression and by quantitative real-time PCR. WT D2 RNA-transfected cells were NS1- positive by day 5, whereas the chimeric RNA-transfected cells became NS1-positive ∼30 days post-transfection in three independent experiments. Sequence analysis covering the entire genome revealed the appearance of a single K74I mutation within the D4MT domain ∼16 days post-transfection in two experiments. In the third, D290N mutation in the conserved NS3 Walker B motif appeared ≥16 days post-transfection. A time course study of serial passages revealed that the 30-day supernatant had gradually evolved to gain replication fitness. Trans-complementation by co-expression of WT D2 NS5 accelerated viral replication of chimeric RNA without changing the K74I mutation. However, the MT and POL activities of NS5 WT D2 and the chimeric NS5 proteins with or without the K74I mutation are similar. Taken together, our results suggest that evolution of the functional interactions involving the chimeric NS5 protein encoded by the viral genome species is essential for gain of viral replication fitness.


Assuntos
Vírus da Dengue/genética , Vírus da Dengue/fisiologia , RNA Viral/genética , Proteínas não Estruturais Virais/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Cricetinae , Vírus da Dengue/classificação , Aptidão Genética , Genoma Viral , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Conformação Proteica , Estrutura Terciária de Proteína , RNA/genética , Homologia de Sequência de Aminoácidos , Sorotipagem , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/fisiologia , Virulência/genética , Virulência/fisiologia , Replicação Viral/genética
15.
Methods Mol Biol ; 1138: 131-50, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24696335

RESUMO

Dengue virus serotypes 1-4 are members of mosquito-borne flavivirus genus of Flaviviridae family that encode one long open reading frame (ORF) that is translated to a polyprotein. Both host and virally encoded proteases function in the processing of the polyprotein by co-translational and posttranslational mechanisms to yield 10 mature proteins prior to viral RNA replication. To study cis- and trans-acting factors involved in viral RNA replication, many groups [1-8] have constructed cDNAs encoding West Nile virus (WNV), DENV, or yellow fever virus reporter replicon RNAs. The replicon plasmids constructed in our laboratory for WNV [9] and the DENV4 replicon described here are arranged in the order of 5'-untranslated region (UTR), the N-terminal coding sequence of capsid (C), Renilla luciferase (Rluc) reporter gene with a translation termination codon, and an internal ribosome entry site (IRES) element from encephalomyocarditis virus (EMCV) for cap-independent translation of the downstream ORF that codes for a polyprotein precursor, CterE-NS1-NS2A-NS2B-NS3-NS4A-NS4B-NS5, followed by the 3'-UTR. In the second DENV4 replicon, the Rluc gene is fused sequentially downstream to the 20 amino acid (aa) FMDV 2A protease coding sequence, neomycin resistance gene (Neo(r)), a termination codon, and the EMCV leader followed by the same polyprotein coding sequence and 3'-UTR as in the first replicon. The first replicon is useful to study by transient transfection experiments the cis-acting elements and trans-acting factors involved in viral RNA replication. The second DENV4 replicon is used to establish a stable monkey kidney (Vero) cell line by transfection of replicon RNA and selection in the presence of the G418, an analog of neomycin. This replicon is useful for screening and identifying antiviral compounds that are potential inhibitors of viral replication.


Assuntos
Vírus da Dengue/genética , Vírus da Dengue/fisiologia , Genoma Viral/genética , Biologia Molecular/métodos , Replicon/genética , Replicação Viral/fisiologia , Animais , Linhagem Celular , Clonagem Molecular , Eletroporação , Genes Reporter , Vetores Genéticos/metabolismo , Recombinação Homóloga/genética , Humanos , Luciferases de Renilla/metabolismo , Plasmídeos/metabolismo , Reação em Cadeia da Polimerase , Saccharomyces cerevisiae/metabolismo , Transcrição Gênica , Transformação Genética
16.
Methods Mol Biol ; 1138: 151-60, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24696336

RESUMO

The use of cDNA infectious clones or subgenomic replicons is indispensable in studying flavivirus biology. Mutating nucleotides or amino acid residues gives important clues to their function in the viral life cycle. However, a major challenge to the establishment of a reverse genetics system for flaviviruses is the instability of their nucleotide sequences in Escherichia coli. Thus, direct cloning using conventional restriction enzyme-based procedures usually leads to unwanted rearrangements of the construct. In this chapter, we discuss a cloning strategy that bypasses traditional cloning procedures. We take advantage of the observations from previous studies that (1) unstable sequences in bacteria can be cloned in eukaryotic systems and (2) Saccharomyces cerevisiae has a well-studied genetics system to introduce sequences using homologous recombination. We describe a protocol to perform targeted mutagenesis in a subgenomic dengue virus 2 replicon. Our method makes use of homologous recombination in yeast using a linearized replicon and a PCR product containing the desired mutation. Constructs derived from this method can be propagated in E. coli with improved stability. Thus, yeast in vivo recombination provides an excellent strategy to genetically engineer flavivirus infectious clones or replicons because this system is compatible with inherently unstable sequences of flaviviruses and is not restricted by the limitations of traditional cloning procedures.


Assuntos
Vírus da Dengue/genética , Biologia Molecular/métodos , Mutagênese/genética , RNA Viral/genética , Recombinação Genética , Replicon/genética , Saccharomyces cerevisiae/genética , Regiões 3' não Traduzidas/genética , Reparo do DNA/genética , Plasmídeos/metabolismo , Reação em Cadeia da Polimerase , Transformação Genética
17.
Methods Mol Biol ; 1138: 331-44, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24696346

RESUMO

Dengue virus (DENV), a member of mosquito-borne flavivirus genus in the Flaviviridae family, is an important human pathogen of global significance. DENV infections are the most common arbovirus infections in the world, causing more than ~300 million cases annually. Although majority of infections result in simple self-limiting disease known as dengue fever which resolve in 7-10 days, ~500,000 cases lead to more severe complications known as dengue hemorrhagic fever/dengue shock syndrome, more frequently observed in secondary infections due to an antibody-dependent enhancement mechanism, resulting in ~25,000 deaths. Currently, there are no vaccines or antiviral drug available for the treatment of DENV infections. Several viral and host proteins have been identified as potential targets for drug development. Some of the viral targets have enzyme activities that play essential roles in viral RNA replication for which in vitro high-throughput screening (HTS) assays have been developed. In this chapter, we describe an in vitro assay for the viral serine protease that has been successfully adapted to HTS format and has been used to screen several thousand compounds to identify inhibitors of the viral protease.


Assuntos
Vírus da Dengue/enzimologia , Descoberta de Drogas , Ensaios de Triagem em Larga Escala/métodos , Inibidores de Proteases/análise , Serina Endopeptidases/metabolismo , Bibliotecas de Moléculas Pequenas/análise , Vírus da Dengue/efeitos dos fármacos , Humanos , Inibidores de Proteases/farmacologia , Proteínas Recombinantes/metabolismo , Reprodutibilidade dos Testes , Serina Endopeptidases/isolamento & purificação , Bibliotecas de Moléculas Pequenas/farmacologia , Estatística como Assunto , Fatores de Tempo , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/isolamento & purificação
18.
Methods Mol Biol ; 1138: 345-60, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24696347

RESUMO

Dengue virus serotypes 1-4 (DENV1-4) are mosquito-borne human pathogens of global significance causing ~390 million cases annually worldwide. The virus infections cause in general a self-limiting disease, known as dengue fever, but occasionally also more severe forms, especially during secondary infections, dengue hemorrhagic fever and dengue shock syndrome causing ~25,000 deaths annually. The DENV genome contains a single-strand positive sense RNA, approximately 11 kb in length. The 5'-end has a type I cap structure. The 3'-end has no poly(A) tail. The viral RNA has a single long open reading frame that is translated by the host translational machinery to yield a polyprotein precursor. Processing of the polyprotein precursor occurs co-translationally by cellular proteases and posttranslationally by the viral serine protease in the endoplasmic reticulum (ER) to yield three structural proteins (capsid (C), precursor membrane (prM), and envelope (E) and seven nonstructural (NS) proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5). The active viral protease consists of both NS2B, an integral membrane protein in the ER, and the N-terminal part of NS3 (180 amino acid residues) that contains the trypsin-like serine protease domain having a catalytic triad of H51, D75, and S135. The C-terminal part of NS3, ~170-618 amino acid residues, encodes an NTPase/RNA helicase and 5'-RNA triphosphatase activities; the latter enzyme is required for the first step in 5'-capping. The cleavage sites of the polyprotein by the viral protease consist of two basic amino acid residues such as KR, RR, or QR, followed by short chain amino acid residues, G, S, or T. Since the cleavage of the polyprotein by the viral protease is absolutely required for assembly of the viral replicase, blockage of NS2B/NS3pro activity provides an effective means for designing dengue virus (DENV) small-molecule therapeutics. Here we describe the screening of small-molecule inhibitors against DENV2 protease.


Assuntos
Antivirais/análise , Vírus da Dengue/enzimologia , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios Enzimáticos/métodos , Plasmídeos/metabolismo , Inibidores de Proteases/análise , Proteínas não Estruturais Virais/metabolismo , Antivirais/farmacologia , Humanos , Concentração Inibidora 50 , Cinética , Inibidores de Proteases/farmacologia , RNA Helicases/antagonistas & inibidores , RNA Helicases/metabolismo , Serina Endopeptidases/metabolismo , Bibliotecas de Moléculas Pequenas/análise , Bibliotecas de Moléculas Pequenas/farmacologia , Especificidade por Substrato/efeitos dos fármacos , Temperatura , Proteínas não Estruturais Virais/antagonistas & inibidores
19.
Methods Mol Biol ; 1138: 361-73, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24696348

RESUMO

Dengue virus (DENV), a member of mosquito-borne flavivirus, causes self-limiting dengue fever as well as life-threatening dengue hemorrhagic fever and dengue shock syndrome. Its positive sense RNA genome has a cap at the 5'-end and no poly(A) tail at the 3'-end. The viral RNA encodes a single polyprotein, C-prM-E-NS1-NS2A-NS2B-NS3-NS4A-NS4B-NS5. The polyprotein is processed into 3 structural proteins (C, prM, and E) and 7 nonstructural (NS) proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, NS5). NS3 and NS5 are multifunctional enzymes performing various tasks in viral life cycle. The N-terminal domain of NS5 has distinct GTP and S-adenosylmethionine (SAM) binding sites. The role of GTP binding site is implicated in guanylyltransferase (GTase) activity of NS5. The SAM binding site is involved in both N-7 and 2'-O-methyltransferase (MTase) activities involved in formation of type I cap. The C-terminal domain of NS5 catalyzes RNA-dependent RNA polymerase (RdRp) activity involved in RNA synthesis. We describe the construction of the MTase domain of NS5 in an E. coli expression vector, purification of the enzyme, and conditions for enzymatic assays of N7- and 2'O-methyltransferase activities that yield the final type I 5'-capped RNA ((7Me)GpppA2'OMe-RNA).


Assuntos
Vírus da Dengue/enzimologia , Ensaios Enzimáticos/métodos , Escherichia coli/metabolismo , Plasmídeos/metabolismo , Proteínas não Estruturais Virais/isolamento & purificação , Cromatografia em Camada Fina , Análogos de Capuz de RNA/metabolismo , Capuzes de RNA/metabolismo , RNA Viral/metabolismo , Especificidade por Substrato
20.
Antiviral Res ; 106: 125-34, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24680954

RESUMO

Dengue virus serotypes 1-4 (DENV1-4) are transmitted by mosquitoes which cause most frequent arboviral infections in the world resulting in ∼390 million cases with ∼25,000 deaths annually. There is no vaccine or antiviral drug currently available for human use. Compounds containing quinoline scaffold were shown to inhibit flavivirus NS2B-NS3 protease (NS2B-NS3pro) with good potencies. In this study, we screened quinoline derivatives, which are known antimalarial drugs for inhibition of DENV2 and West Nile virus (WNV) replication using the corresponding replicon expressing cell-based assays. Amodiaquine (AQ), one of the 4-aminoquinoline drugs, inhibited DENV2 infectivity measured by plaque assays, with EC50 and EC90 values of 1.08±0.09µM and 2.69±0.47 µM, respectively, and DENV2 RNA replication measured by Renilla luciferase reporter assay, with EC50 value of 7.41±1.09µM in the replicon expressing cells. Cytotoxic concentration (CC50) in BHK-21 cells was 52.09±4.25µM. The replication inhibition was confirmed by plaque assay of the extracellular virions as well as by qRT-PCR of the intracellular and extracellular viral RNA levels. AQ was stable for at least 96h and had minor inhibitory effect on entry, translation, and post-replication stages in the viral life cycle. DENV protease, 5'-methyltransferase, and RNA-dependent RNA polymerase do not seem to be targets of AQ. Both p-hydroxyanilino and diethylaminomethyl moieties are important for AQ to inhibit DENV2 replication and infectivity. Our results support AQ as a promising candidate for anti-flaviviral therapy.


Assuntos
Amodiaquina/farmacologia , Antimaláricos/farmacologia , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/fisiologia , Replicação Viral/efeitos dos fármacos , Amodiaquina/toxicidade , Animais , Antimaláricos/toxicidade , Antivirais/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Testes de Sensibilidade Microbiana , Ensaio de Placa Viral , Vírus do Nilo Ocidental/efeitos dos fármacos , Vírus do Nilo Ocidental/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...