Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Biochem Biophys Res Commun ; 668: 35-41, 2023 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-37235917

RESUMO

The recent outbreak of COVID-19 has created a serious health crisis with fatFal infectious viral diseases, such as Severe Acute Respiratory Syndrome (SARS). The nsp13, a helicase of coronaviruses is an essential element for viral replication that unwinds secondary structures of DNA and RNA, and is thus considered a major therapeutic target for treatment. The replication of coronaviruses and other retroviruses occurs in the cytoplasm of infected cells, in association with viral replication organelles, called virus-induced cytosolic double-membrane vesicles (DMVs). In addition, an increase in cytosolic Ca2+ concentration accelerates viral replication. However, the molecular mechanism of nsp13 in the presence of Ca2+ is not well understood. In this study, we applied biochemical methods and single-molecule techniques to demonstrate how nsp13 achieves its unwinding activity while performing ATP hydrolysis in the presence of Ca2+. Our study found that nsp13 could efficiently unwind double stranded (ds) DNA under physiological concentration of Ca2+ of cytosolic DMVs. These findings provide new insights into the properties of nsp13 in the range of calcium in cytosolic DMVs.


Assuntos
Cálcio , DNA , Conformação de Ácido Nucleico , RNA Helicases , Imagem Individual de Molécula , Proteínas não Estruturais Virais , Cálcio/metabolismo , Cálcio/farmacologia , DNA/química , DNA/efeitos dos fármacos , DNA/metabolismo , Magnésio/metabolismo , Magnésio/farmacologia , Conformação de Ácido Nucleico/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Replicação Viral , Citosol/metabolismo , Hidrólise/efeitos dos fármacos , RNA Helicases/efeitos dos fármacos , RNA Helicases/metabolismo , Proteínas não Estruturais Virais/efeitos dos fármacos , Proteínas não Estruturais Virais/metabolismo , Transferência Ressonante de Energia de Fluorescência , Eletroforese em Gel de Poliacrilamida , Relação Dose-Resposta a Droga , Transcrição Gênica
2.
Molecules ; 26(22)2021 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-34833913

RESUMO

Dengue fever is a dangerous infectious endemic disease that affects over 100 nations worldwide, from Africa to the Western Pacific, and is caused by the dengue virus, which is transmitted to humans by an insect bite of Aedes aegypti. Millions of citizens have died as a result of dengue fever and dengue hemorrhagic fever across the globe. Envelope (E), serine protease (NS3), RNA-directed RNA polymerase (NS5), and non-structural protein 1 (NS1) are mostly required for cell proliferation and survival. Some of the diterpenoids and their derivatives produced by nature possess anti-dengue viral properties. The goal of the computational study was to scrutinize the effectiveness of diterpenoids and their derivatives against dengue viral proteins through in silico study. Methods: molecular docking was performed to analyze the binding affinity of compounds against four viral proteins: the envelope (E) protein, the NS1 protein, the NS3 protein, and the NS5 protein. Results: among the selected drug candidates, triptolide, stevioside, alepterolic acid, sphaeropsidin A, methyl dodovisate A, andrographolide, caesalacetal, and pyrimethamine have demonstrated moderate to good binding affinities (-8.0 to -9.4 kcal/mol) toward the selected proteins: E protein, NS3, NS5, and NS1 whereas pyrimethamine exerts -7.5, -6.3, -7.8, and -6.6 kcal/mol with viral proteins, respectively. Interestingly, the binding affinities of these lead compounds were better than those of an FDA-approved anti-viral medication (pyrimethamine), which is underused in dengue fever. Conclusion: we can conclude that diterpenoids can be considered as a possible anti-dengue medication option. However, in vivo investigation is recommended to back up the conclusions of this study.


Assuntos
Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Diterpenos/farmacologia , Antivirais/química , Antivirais/farmacocinética , Sítios de Ligação , Simulação por Computador , Dengue/tratamento farmacológico , Dengue/virologia , Diterpenos/química , Diterpenos/farmacocinética , Desenho de Fármacos , Humanos , Simulação de Acoplamento Molecular , Compostos Fitoquímicos/química , Compostos Fitoquímicos/farmacocinética , Compostos Fitoquímicos/farmacologia , Ligação Proteica , RNA Helicases/química , RNA Helicases/efeitos dos fármacos , RNA Helicases/metabolismo , Serina Endopeptidases/química , Serina Endopeptidases/efeitos dos fármacos , Serina Endopeptidases/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/efeitos dos fármacos , Proteínas não Estruturais Virais/metabolismo
3.
Mol Biol Cell ; 32(21): ar18, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34432484

RESUMO

Stress granules (SGs) are ribonucleoprotein functional condensates that form under stress conditions in all eukaryotic cells. Although their stress-survival function is far from clear, SGs have been implicated in the regulation of many vital cellular pathways. Consequently, SG dysfunction is thought to be a mechanistic point of origin for many neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS). Additionally, SGs are thought to play a role in pathogenic pathways as diverse as viral infection and chemotherapy resistance. There is a growing consensus on the hypothesis that understanding the mechanistic regulation of SG physical properties is essential to understanding their function. Although the internal dynamics and condensation mechanisms of SGs have been broadly investigated, there have been fewer investigations into the timing of SG formation and clearance in live cells. Because the lifetime of SG persistence can be a key factor in their function and tendency toward pathological dysregulation, SG clearance mechanisms deserve particular attention. Here we show that resveratrol and its analogues piceatannol, pterostilbene, and 3,4,5,4'-tetramethoxystilbene induce G3BP-dependent SG formation with atypically rapid clearance kinetics. Resveratrol binds to G3BP, thereby reducing its protein-protein association valency. We suggest that altering G3BP valency is a pathway for the formation of uniquely transient SGs.


Assuntos
DNA Helicases/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , RNA Helicases/metabolismo , Proteínas com Motivo de Reconhecimento de RNA/metabolismo , Resveratrol/farmacologia , Grânulos de Estresse/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Grânulos Citoplasmáticos/efeitos dos fármacos , Grânulos Citoplasmáticos/metabolismo , DNA Helicases/efeitos dos fármacos , Células HEK293 , Células HeLa , Humanos , Cinética , Proteínas de Ligação a Poli-ADP-Ribose/efeitos dos fármacos , RNA Helicases/efeitos dos fármacos , Proteínas com Motivo de Reconhecimento de RNA/efeitos dos fármacos , Ribonucleoproteínas/metabolismo , Grânulos de Estresse/efeitos dos fármacos
4.
Biomed Res Int ; 2020: 6237160, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33102585

RESUMO

Coronaviruses have been reported previously due to their association with the severe acute respiratory syndrome (SARS). After SARS, these viruses were known to be causing Middle East respiratory syndrome (MERS) and caused 35% evanescence amid victims pursuing remedial care. Nowadays, beta coronaviruses, members of Coronaviridae, family order Nidovirales, have become subjects of great importance due to their latest pandemic originating from Wuhan, China. The virus named as human-SARS-like coronavirus-2 contains four structural as well as sixteen nonstructural proteins encoded by single-stranded ribonucleic acid of positive polarity. As there is no vaccine available to treat the infection caused by these viruses, there is a dire need for taking necessary steps against this virus. Herein, we have targeted two nonstructural proteins of SARS-CoV-2, namely, methyltransferase (nsp16) and helicase (nsp13), respectively, due to their substantial activity in viral pathogenesis. A total of 2035 compounds were analyzed for their pharmacokinetics and pharmacological properties. The screened 108 compounds were docked against both targeted proteins and were compared with previously reported known compounds. Compounds with high binding affinity were analyzed for their reactivity through DFT analysis, and binding was analyzed using molecular dynamics simulations. Through the analyses performed in this study, it is concluded that EryvarinM, Silydianin, Osajin, and Raddeanine can be considered potential inhibitors for MTase, while TomentodiplaconeB, Osajin, Sesquiterpene Glycoside, Rhamnetin, and Silydianin for helicase after these compounds are validated thoroughly using in vitro and in vivo protocols.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Compostos Fitoquímicos/química , Compostos Fitoquímicos/farmacologia , SARS-CoV-2/efeitos dos fármacos , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/química , Monofosfato de Adenosina/farmacologia , Alanina/análogos & derivados , Alanina/química , Alanina/farmacologia , Antimetabólitos/química , Antimetabólitos/farmacologia , Antivirais/química , COVID-19/epidemiologia , COVID-19/virologia , China/epidemiologia , Dioxolanos/química , Dioxolanos/farmacologia , Fluoroquinolonas/química , Fluoroquinolonas/farmacologia , Humanos , Metiltransferases/efeitos dos fármacos , Simulação de Acoplamento Molecular , Nelfinavir/química , Nelfinavir/farmacologia , Piperazinas/química , Piperazinas/farmacologia , Conformação Proteica , RNA Helicases/efeitos dos fármacos , SARS-CoV-2/química , SARS-CoV-2/isolamento & purificação , SARS-CoV-2/metabolismo , Inibidores da Topoisomerase II/química , Inibidores da Topoisomerase II/farmacologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo
5.
Virol Sin ; 35(3): 321-329, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32500504

RESUMO

The ongoing outbreak of Coronavirus Disease 2019 (COVID-19) has become a global public health emergency. SARS-coronavirus-2 (SARS-CoV-2), the causative pathogen of COVID-19, is a positive-sense single-stranded RNA virus belonging to the family Coronaviridae. For RNA viruses, virus-encoded RNA helicases have long been recognized to play pivotal roles during viral life cycles by facilitating the correct folding and replication of viral RNAs. Here, our studies show that SARS-CoV-2-encoded nonstructural protein 13 (nsp13) possesses the nucleoside triphosphate hydrolase (NTPase) and RNA helicase activities that can hydrolyze all types of NTPs and unwind RNA helices dependently of the presence of NTP, and further characterize the biochemical characteristics of these two enzymatic activities associated with SARS-CoV-2 nsp13. Moreover, we found that some bismuth salts could effectively inhibit both the NTPase and RNA helicase activities of SARS-CoV-2 nsp13 in a dose-dependent manner. Thus, our findings demonstrate the NTPase and helicase activities of SARS-CoV-2 nsp13, which may play an important role in SARS-CoV-2 replication and serve as a target for antivirals.


Assuntos
Betacoronavirus/metabolismo , Bismuto/farmacologia , Metiltransferases/metabolismo , Nucleosídeo-Trifosfatase/efeitos dos fármacos , RNA Helicases/efeitos dos fármacos , Sais/farmacologia , Proteínas não Estruturais Virais/metabolismo , Adenosina Trifosfatases/efeitos dos fármacos , Adenosina Trifosfatases/metabolismo , Betacoronavirus/enzimologia , Betacoronavirus/genética , COVID-19 , Infecções por Coronavirus/virologia , Humanos , Metiltransferases/genética , Nucleosídeo-Trifosfatase/genética , Nucleosídeo-Trifosfatase/metabolismo , Pandemias , Pneumonia Viral/virologia , RNA Helicases/genética , RNA Helicases/metabolismo , Proteínas Recombinantes , SARS-CoV-2 , Síndrome Respiratória Aguda Grave , Proteínas não Estruturais Virais/genética , Replicação Viral
6.
J Virol ; 92(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29237842

RESUMO

RNA-remodeling proteins, including RNA helicases and chaperones, act to remodel RNA structures and/or protein-RNA interactions and are required for all processes involving RNAs. Although many viruses encode RNA helicases and chaperones, their in vitro activities and their roles in infected cells largely remain elusive. Noroviruses are a diverse group of positive-strand RNA viruses in the family Caliciviridae and constitute a significant and potentially fatal threat to human health. Here, we report that the protein NS3 encoded by human norovirus has both ATP-dependent RNA helicase activity that unwinds RNA helices and ATP-independent RNA-chaperoning activity that can remodel structured RNAs and facilitate strand annealing. Moreover, NS3 can facilitate viral RNA synthesis in vitro by norovirus polymerase. NS3 may therefore play an important role in norovirus RNA replication. Lastly, we demonstrate that the RNA-remodeling activity of NS3 is inhibited by guanidine hydrochloride, an FDA-approved compound, and, more importantly, that it reduces the replication of the norovirus replicon in cultured human cells. Altogether, these findings are the first to demonstrate the presence of RNA-remodeling activities encoded by Caliciviridae and highlight the functional significance of NS3 in the noroviral life cycle.IMPORTANCE Noroviruses are a diverse group of positive-strand RNA viruses, which annually cause hundreds of millions of human infections and over 200,000 deaths worldwide. For RNA viruses, cellular or virus-encoded RNA helicases and/or chaperones have long been considered to play pivotal roles in viral life cycles. However, neither RNA helicase nor chaperoning activity has been demonstrated to be associated with any norovirus-encoded proteins, and it is also unknown whether norovirus replication requires the participation of any viral or cellular RNA helicases/chaperones. We found that a norovirus protein, NS3, not only has ATP-dependent helicase activity, but also acts as an ATP-independent RNA chaperone. Also, NS3 can facilitate in vitro viral RNA synthesis, suggesting the important role of NS3 in norovirus replication. Moreover, NS3 activities can be inhibited by an FDA-approved compound, which also suppresses norovirus replicon replication in human cells, raising the possibility that NS3 could be a target for antinoroviral drug development.


Assuntos
Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Norovirus/enzimologia , Norovirus/genética , RNA Helicases/genética , RNA Helicases/metabolismo , Proteínas não Estruturais Virais/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Guanidina/antagonistas & inibidores , Humanos , Estágios do Ciclo de Vida , Chaperonas Moleculares/efeitos dos fármacos , Norovirus/efeitos dos fármacos , Norovirus/crescimento & desenvolvimento , Nucleosídeo-Trifosfatase/genética , Nucleosídeo-Trifosfatase/metabolismo , Ligação Proteica , Dobramento de Proteína , RNA Helicases/efeitos dos fármacos , RNA Viral/química , RNA Viral/efeitos dos fármacos , RNA Viral/genética , RNA Viral/metabolismo , Replicon/efeitos dos fármacos , Alinhamento de Sequência , Análise de Sequência , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/efeitos dos fármacos , Proteínas não Estruturais Virais/genética , Replicação Viral/efeitos dos fármacos , Replicação Viral/fisiologia
7.
Antiviral Res ; 143: 218-229, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28461069

RESUMO

The recent re-emergence of Zika virus (ZIKV)1, a member of the Flaviviridae family, has become a global emergency. Currently, there are no effective methods of preventing or treating ZIKV infection, which causes severe neuroimmunopathology and is particularly harmful to the developing fetuses of infected pregnant women. However, the pathology induced by ZIKV is unique among flaviviruses, and knowledge of the biology of other family members cannot easily be extrapolated to ZIKV. Thus, structure-function studies of ZIKV proteins are urgently needed to facilitate the development of effective preventative and therapeutic agents. Like other flaviviruses, ZIKV expresses an NS2B-NS3 protease, which consists of the NS2B cofactor and the NS3 protease domain and is essential for cleavage of the ZIKV polyprotein precursor and generation of fully functional viral proteins. Here, we report the enzymatic characterization of ZIKV protease, and we identify structural scaffolds for allosteric small-molecule inhibitors of this protease. Molecular modeling of the protease-inhibitor complexes suggests that these compounds bind to the druggable cavity in the NS2B-NS3 protease interface and affect productive interactions of the protease domain with its cofactor. The most potent compound demonstrated efficient inhibition of ZIKV propagation in vitro in human fetal neural progenitor cells and in vivo in SJL mice. The inhibitory scaffolds could be further developed into valuable research reagents and, ultimately, provide a roadmap for the selection of efficient inhibitors of ZIKV infection.


Assuntos
Sítio Alostérico , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Proteínas não Estruturais Virais/química , Zika virus/enzimologia , Sequência de Aminoácidos , Animais , Antivirais/antagonistas & inibidores , Antivirais/química , Sequência de Bases , Ativação Enzimática , Feminino , Flavivirus/química , Expressão Gênica , Humanos , Concentração Inibidora 50 , Camundongos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , RNA Helicases/química , RNA Helicases/efeitos dos fármacos , Fatores de Transcrição SOXB1/genética , Alinhamento de Sequência , Serina Endopeptidases/química , Serina Endopeptidases/efeitos dos fármacos , Células-Tronco , Proteínas não Estruturais Virais/efeitos dos fármacos , Proteínas Virais/química , Proteínas Virais/genética , Zika virus/química , Zika virus/genética , Zika virus/crescimento & desenvolvimento , Infecção por Zika virus/virologia
8.
Antiviral Res ; 143: 186-194, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28457855

RESUMO

Zika virus (ZIKV) is an arthropod-borne flavivirus that causes newborn microcephaly and Guillian-Barré syndrome in adults. No therapeutics are available to treat ZIKV infection or other flaviviruses. In this study, we explored the inhibitory effect of glycosaminoglycans and analogues against ZIKV infection. Highly sulfated heparin, dextran sulfate and suramin significantly inhibited ZIKV infection in Vero cells. De-sulfated heparin analogues lose inhibitory effect, implying that sulfonate groups are critical for viral inhibition. Suramin, an FDA-approved anti-parasitic drug, inhibits ZIKV infection with 3-5 log10 PFU viral reduction with IC50 value of ∼2.5-5 µg/ml (1.93 µM-3.85 µM). A time-of-drug-addition study revealed that suramin remains potent even when administrated at 1-24 hpi. Suramin inhibits ZIKV infection by preventing viral adsorption, entry and replication. Molecular dynamics simulation revealed stronger interaction of suramin with ZIKV NS3 helicase than with the envelope protein. Suramin warrants further investigation as a potential antiviral candidate for ZIKV infection. Heparan sulfate (HS) is a cellular attachment receptor for multiple flaviviruses. However, no direct ZIKV-heparin interaction was observed in heparin-binding analysis, and downregulate or removal of cellular HS with sodium chlorate or heparinase I/III did not inhibit ZIKV infection. This indicates that cell surface HS is not utilized by ZIKV as an attachment receptor.


Assuntos
Suramina/antagonistas & inibidores , Infecção por Zika virus/prevenção & controle , Zika virus/efeitos dos fármacos , Animais , Anticorpos Antivirais , Cloratos/farmacologia , Chlorocebus aethiops , DNA Helicases/metabolismo , Sulfato de Dextrana/antagonistas & inibidores , Flavivirus/efeitos dos fármacos , Glicosaminoglicanos/farmacologia , Heparina/análogos & derivados , Heparina/química , Heparina/farmacologia , Heparitina Sulfato/farmacologia , Concentração Inibidora 50 , Camundongos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , RNA Helicases/química , RNA Helicases/efeitos dos fármacos , Serina Endopeptidases/química , Serina Endopeptidases/efeitos dos fármacos , Suramina/administração & dosagem , Células Vero , Proteínas do Envelope Viral/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Zika virus/fisiologia , Infecção por Zika virus/virologia
9.
Antiviral Res ; 142: 141-147, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28336347

RESUMO

The Zika virus presents a serious risk for global health. Crystal structures of different constructs of the Zika virus NS2B-NS3 protease (NS2B-NS3pro) have been determined with the aim to provide a basis for rational drug discovery. In these structures, the C-terminal ß-hairpin of NS2B, NS2Bc, was observed to be either disordered (open conformation) or bound to NS3pro complementing the substrate binding site (closed conformation). Enzymatically active constructs of flaviviral NS2B-NS3 proteases commonly used for inhibitor testing contain a covalent peptide linker between NS2B and NS3pro. Using a linked construct of Zika virus NS2B-NS3pro, we studied the location of NS2Bc relative to NS3pro in solution by pseudocontact shifts generated by a paramagnetic lanthanide tag attached to NS3pro. Both closed and open conformations were observed with different inhibitors. As the NS2B co-factor is involved in substrate binding of flaviviral NS2B-NS3 proteases, the destabilization of the closed conformation in the linked construct makes it an attractive tool to search for inhibitors that interfere with the formation of the enzymatically active, closed conformation.


Assuntos
Proteínas não Estruturais Virais/química , Zika virus/enzimologia , Sítios de Ligação , Ácidos Borônicos/antagonistas & inibidores , Dipeptídeos/antagonistas & inibidores , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Inibidores de Proteases/química , Conformação Proteica , RNA Helicases/química , RNA Helicases/efeitos dos fármacos , Serina Endopeptidases/química , Serina Endopeptidases/efeitos dos fármacos , Proteínas não Estruturais Virais/efeitos dos fármacos , Proteínas não Estruturais Virais/genética
10.
EMBO J ; 33(4): 371-84, 2014 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-24488111

RESUMO

In Drosophila, Dicer-1 produces microRNAs (miRNAs) from pre-miRNAs, whereas Dicer-2 generates small interfering RNAs from long double-stranded RNA (dsRNA), a process that requires ATP hydrolysis. We previously showed that inorganic phosphate inhibits Dicer-2 cleavage of pre-miRNAs, but not long dsRNAs. Here, we report that phosphate-dependent substrate discrimination by Dicer-2 reflects dsRNA substrate length. Efficient processing by Dicer-2 of short dsRNA requires a 5' terminal phosphate and a two-nucleotide, 3' overhang, but does not require ATP. Phosphate inhibits cleavage of such short substrates. In contrast, cleavage of longer dsRNA requires ATP but no specific end structure: phosphate does not inhibit cleavage of these substrates. Mutation of a pair of conserved arginine residues in the Dicer-2 PAZ domain blocked cleavage of short, but not long, dsRNA. We propose that inorganic phosphate occupies a PAZ domain pocket required to bind the 5' terminal phosphate of short substrates, blocking their use and restricting pre-miRNA processing in flies to Dicer-1. Our study helps explain how a small molecule can alter the substrate specificity of a nucleic acid processing enzyme.


Assuntos
Proteínas de Drosophila/efeitos dos fármacos , Drosophila melanogaster/metabolismo , MicroRNAs/metabolismo , Fosfatos/farmacologia , RNA Helicases/efeitos dos fármacos , Ribonuclease III/efeitos dos fármacos , Substituição de Aminoácidos , Animais , Arginina , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Mutagênese Sítio-Dirigida , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , RNA Helicases/química , RNA Helicases/genética , RNA Helicases/metabolismo , RNA de Cadeia Dupla/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Ribonuclease III/química , Ribonuclease III/genética , Ribonuclease III/metabolismo , Especificidade por Substrato
11.
Protein Expr Purif ; 92(2): 156-62, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24084007

RESUMO

Dengue virus (DENV), a member of the flavivirus genus, affects 50-100 million people in tropical and sub-tropical regions. The DENV protease domain is located at the N-terminus of the NS3 protease and requires for its enzymatic activity a hydrophilic segment of the NS2B that acts as a cofactor. The protease is an important antiviral drug target because it plays a crucial role in virus replication by cleaving the genome-coded polypeptide into mature functional proteins. Currently, there are no drugs to inhibit DENV protease activity. Most structural and functional studies have been conducted using protein constructs containing the NS3 protease domain connected to a soluble segment of the NS2B membrane protein via a nine-residue linker. For in vitro structural and functional studies, it would be useful to produce a natural form of the DENV protease containing the NS3 protease domain and the full-length NS2B protein. Herein, we describe the expression and purification of a natural form of DENV protease (NS2BFL-NS3pro) containing the full-length NS2B protein and the protease domain of NS3 (NS3pro). The protease was expressed and purified in detergent micelles necessary for its folding. Our results show that this purified protein was active in detergent micelles such as lyso-myristoyl phosphatidylcholine (LMPC). These findings should facilitate further structural and functional studies of the protease and will facilitate drug discovery targeting DENV.


Assuntos
Lisofosfatidilcolinas/farmacologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/efeitos dos fármacos , Sequência de Aminoácidos , Detergentes/química , Estabilidade Enzimática , Lisofosfatidilcolinas/química , Micelas , Dados de Sequência Molecular , Estrutura Terciária de Proteína , RNA Helicases/química , RNA Helicases/efeitos dos fármacos , RNA Helicases/genética , RNA Helicases/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Serina Endopeptidases/química , Serina Endopeptidases/efeitos dos fármacos , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
12.
Eur J Med Chem ; 62: 199-205, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23353753

RESUMO

This communication describes the synthesis and inhibitory activities of thirty-seven novel C-terminal agmatine dipeptides used as screening compounds to study the structure-activity relationship between active-site peptidomimetics and the West Nile virus (WNV) NS2B/NS3 serine protease. Our efforts lead to the discovery of a novel agmatine dipeptide inhibitor (compound 33, IC50 2.6 ± 0.3 µM) with improved inhibitory activity in comparison to the most potent inhibitor described in our recent report [IC50 4.7 ± 1.2 µM; Lim et al., Eur. J. Med. Chem. 46 (2011) 3130-3134]. In addition, our study cleared the contention surrounding the previous X-ray co-crystallization study and an enzyme inhibition report on the binding conformation adopted by active-site peptide aldehydes. Our data should provide valuable insights into the design of future peptidomimetic antivirals against WNV infections.


Assuntos
Agmatina/farmacologia , Dipeptídeos/farmacologia , Inibidores de Proteases/farmacologia , Proteínas não Estruturais Virais/efeitos dos fármacos , Vírus do Nilo Ocidental/enzimologia , Agmatina/síntese química , Agmatina/química , Cristalografia por Raios X , Dipeptídeos/síntese química , Dipeptídeos/química , Relação Dose-Resposta a Droga , Modelos Moleculares , Estrutura Molecular , Inibidores de Proteases/síntese química , Inibidores de Proteases/química , RNA Helicases/efeitos dos fármacos , Serina Endopeptidases/efeitos dos fármacos , Software , Relação Estrutura-Atividade , Vírus do Nilo Ocidental/efeitos dos fármacos
13.
J Nat Prod ; 75(4): 650-4, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22394195

RESUMO

The hepatitis C virus (HCV) causes one of the most prevalent chronic infectious diseases in the world, hepatitis C, which ultimately develops into liver cancer through cirrhosis. The NS3 protein of HCV possesses nucleoside triphosphatase (NTPase) and RNA helicase activities. As both activities are essential for viral replication, NS3 is proposed as an ideal target for antiviral drug development. In this study, we identified manoalide (1) from marine sponge extracts as an RNA helicase inhibitor using a high-throughput screening photoinduced electron transfer (PET) system that we previously developed. Compound 1 inhibits the RNA helicase and ATPase activities of NS3 in a dose-dependent manner, with IC(50) values of 15 and 70 µM, respectively. Biochemical kinetic analysis demonstrated that 1 does not affect the apparent K(m) value (0.31 mM) of NS3 ATPase activity, suggesting that 1 acts as a noncompetitive inhibitor. The binding of NS3 to single-stranded RNA was inhibited by 1. Manoalide (1) also has the ability to inhibit the ATPase activity of human DHX36/RHAU, a putative RNA helicase. Taken together, we conclude that 1 inhibits the ATPase, RNA binding, and helicase activities of NS3 by targeting the helicase core domain conserved in both HCV NS3 and DHX36/RHAU.


Assuntos
Hepacivirus/metabolismo , Terpenos/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Sequência de Bases , Humanos , Estrutura Molecular , Nucleosídeo-Trifosfatase/efeitos dos fármacos , Nucleosídeo-Trifosfatase/metabolismo , RNA Helicases/efeitos dos fármacos , RNA Helicases/metabolismo
14.
Bioorg Khim ; 37(6): 723-44, 2011.
Artigo em Russo | MEDLINE | ID: mdl-22497074
15.
Curr Opin Pharmacol ; 9(6): 680-7, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19828376

RESUMO

The pathogen recognition receptors (PRRs) initiate immediate responses against infection and tissue damage to protect the host from microbial invasion. In response to mucosal damage, intestinal PRR signaling initiates damage repair processes. Recent advances appear to link PRR abnormalities and inflammatory as well as neoplastic intestinal disorders. Emerging evidence suggests a dual role of PRRs, in which they may simultaneously induce tumorigenesis and antitumor immunity. PRR may induce tumor cell proliferation by activating cell survival signaling mainly via NF-kappaB, but this signal can activate dendritic cells to promote antitumor immunity. TLR signaling within the tumor cells may result in evasion of immune surveillance, propagation of metastatic growth, or rather, induction of tumor cell apoptosis depending on ligands. Epithelial cells induce endogenous PRR ligands when damaged or during neoplastic transformation. Targeted manipulation of PRR signaling may provide emerging opportunities for the development of new therapeutic strategies for many gastrointestinal diseases.


Assuntos
Neoplasias Gastrointestinais/metabolismo , Trato Gastrointestinal/metabolismo , Receptores de Reconhecimento de Padrão/fisiologia , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Enterocolite , Neoplasias Gastrointestinais/tratamento farmacológico , Neoplasias Gastrointestinais/etiologia , Neoplasias Gastrointestinais/imunologia , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/fisiologia , Proteínas Adaptadoras de Sinalização NOD/efeitos dos fármacos , Proteínas Adaptadoras de Sinalização NOD/metabolismo , RNA Helicases/efeitos dos fármacos , RNA Helicases/metabolismo , Receptores de Reconhecimento de Padrão/imunologia , Receptores Toll-Like/efeitos dos fármacos , Receptores Toll-Like/metabolismo
16.
Antimicrob Agents Chemother ; 52(6): 1901-11, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18391043

RESUMO

The development of effective therapies for hepatitis C virus (HCV) must take into account genetic variation among HCV strains. Response rates to interferon-based treatments, including the current preferred treatment of pegylated alpha interferon administered with ribavirin, are genotype specific. Of the numerous HCV inhibitors currently in development as antiviral drugs, nucleoside analogs that target the conserved NS5B active site seem to be quite effective against diverse HCV strains. To test this hypothesis, we examined the effects of a panel of nucleotide analogs, including ribavirin triphosphate (RTP) and several chain-terminating nucleoside triphosphates, on the activities of purified HCV NS5B polymerases derived from genotype 1a, 1b, and 2a strains. Unlike the genotype-specific effects on NS5B activity reported previously for nonnucleoside inhibitors (F. Pauwels, W. Mostmans, L. M. Quirynen, L. van der Helm, C. W. Boutton, A. S. Rueff, E. Cleiren, P. Raboisson, D. Surleraux, O. Nyanguile, and K. A. Simmen, J. Virol. 81:6909-6919, 2007), only minor differences in inhibition were observed among the various genotypes; thus, nucleoside analogs that are current drug candidates may be more promising for treatment of a broader variety of HCV strains. We also examined the effects of RTP on the HCV NS3 helicase/ATPase. As with the polymerase, only minor differences were observed among 1a-, 1b-, and 2a-derived enzymes. RTP did not inhibit the rate of NS3 helicase-catalyzed DNA unwinding but served instead as a substrate to fuel unwinding. NS3 added to RNA synthesis reactions relieved inhibition of the polymerase by RTP, presumably due to RTP hydrolysis. These results suggest that NS3 can limit the incorporation of ribavirin into viral RNA, thus reducing its inhibitory or mutagenic effects.


Assuntos
Hepacivirus/classificação , Hepacivirus/efeitos dos fármacos , Nucleotídeos/farmacologia , Proteínas não Estruturais Virais/efeitos dos fármacos , Adenosina Trifosfatases/efeitos dos fármacos , Adenosina Trifosfatases/metabolismo , Antivirais/farmacologia , Genótipo , Hepacivirus/enzimologia , Hepacivirus/genética , Humanos , Nucleotídeos/química , RNA Helicases/efeitos dos fármacos , RNA Helicases/metabolismo , RNA Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo
17.
J Gen Virol ; 88(Pt 8): 2223-2227, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17622626

RESUMO

A recombinant form of yellow fever virus (YFV) NS3 protease, linked via a nonapeptide to the minimal NS2B co-factor sequence (CF40-gly-NS3pro190), was expressed in Escherichia coli and shown to be catalytically active. It efficiently cleaved the fluorogenic tetrapeptide substrate Bz-norleucine-lysine-arginine-arginine-AMC, which was previously optimized for dengue virus NS2B/3 protease. A series of small peptidic inhibitors based on this substrate sequence readily inhibited its enzymic activity. To understand the structure-activity relationship of the inhibitors, they were docked into a homology model of the YFV NS2B/NS3 protease structure. The results revealed that the P1 and P2 positions are most important for inhibitor binding, whilst the P3 and P4 positions have much less effect. These findings indicate that the characteristics of YFV protease are very similar to those reported for dengue and West Nile virus proteases, and suggest that pan-flavivirus NS3 protease drugs may be developed for flaviviral diseases.


Assuntos
Inibidores Enzimáticos/farmacologia , Oligopeptídeos/metabolismo , Proteínas não Estruturais Virais/efeitos dos fármacos , Proteínas não Estruturais Virais/metabolismo , Vírus da Febre Amarela/enzimologia , Sequência de Aminoácidos , Antivirais/farmacologia , Sítios de Ligação/fisiologia , Cinética , Dados de Sequência Molecular , Oligopeptídeos/química , RNA Helicases/química , RNA Helicases/efeitos dos fármacos , RNA Helicases/genética , RNA Helicases/metabolismo , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Serina Endopeptidases/química , Serina Endopeptidases/efeitos dos fármacos , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Especificidade por Substrato , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética
18.
PLoS One ; 2(3): e303, 2007 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-17375189

RESUMO

The core of the exon-junction complex consists of Y14, Magoh, MLN51 and eIF4AIII, a DEAD-box RNA helicase. MLN51 stimulates the ATPase activity of eIF4AIII, whilst the Y14-Magoh complex inhibits it. We show that the MLN51-dependent stimulation increases both the affinity of eIF4AIII for ATP and the rate of enzyme turnover; the K(M) is decreased by an order of magnitude and k(cat) increases 30 fold. Y14-Magoh do inhibit the MLN51-stimulated ATPase activity, but not back to background levels. The ATP-bound form of the eIF4AIII-MLN51 complex has a 100-fold higher affinity for RNA than the unbound form and ATP hydrolysis reduces this affinity. MLN51 stimulates the RNA-helicase activity of eIF4AIII, suggesting that this activity may be functionally important.


Assuntos
Fator de Iniciação 4A em Eucariotos/genética , Éxons/genética , RNA Helicases/metabolismo , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Bases , Fator de Iniciação 4A em Eucariotos/isolamento & purificação , Fator de Iniciação 4A em Eucariotos/metabolismo , Humanos , Cinética , Oligodesoxirribonucleotídeos , Reação em Cadeia da Polimerase/métodos , Biossíntese de Proteínas , RNA/genética , RNA Helicases/efeitos dos fármacos , RNA Helicases/genética , RNA Mensageiro/genética , Especificidade por Substrato , Transcrição Gênica
19.
Hepatology ; 43(6): 1392-5, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16729317

RESUMO

Helicases are a ubiquitous class of enzymes involved in nearly all aspects of DNA and RNA metabolism. Despite recent progress in understanding their mechanism of action, limited resolution has left inaccessible the detailed mechanisms by which these enzymes couple the rearrangement of nucleic acid structures to the binding and hydrolysis of ATP. Observing individual mechanistic cycles of these motor proteins is central to understanding their cellular functions. Here we follow in real time, at a resolution of two base pairs and 20 ms, the RNA translocation and unwinding cycles of a hepatitis C virus helicase (NS3) monomer. NS3 is a representative superfamily-2 helicase essential for viral replication, and therefore a potentially important drug target. We show that the cyclic movement of NS3 is coordinated by ATP in discrete steps of 11 +/- 3 base pairs, and that actual unwinding occurs in rapid smaller substeps of 3.6 +/- 1.3 base pairs, also triggered by ATP binding, indicating that NS3 might move like an inchworm. This ATP-coupling mechanism is likely to be applicable to other non-hexameric helicases involved in many essential cellular functions. The assay developed here should be useful in investigating a broad range of nucleic acid translocation motors.


Assuntos
Hepacivirus/genética , Hepatite C/enzimologia , Hepatite C/genética , RNA Helicases/metabolismo , Antivirais/uso terapêutico , Hepacivirus/efeitos dos fármacos , Hepacivirus/enzimologia , Hepatite C/tratamento farmacológico , Humanos , Conformação de Ácido Nucleico , RNA Helicases/efeitos dos fármacos , RNA Helicases/genética , Fatores de Risco , Sensibilidade e Especificidade , Índice de Gravidade de Doença
20.
Nat Rev Drug Discov ; 4(10): 845-53, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16184083

RESUMO

Although there has been considerable progress in the development of antiviral agents in recent years, there is still a pressing need for new drugs both to improve on the properties of existing agents and to combat the problem of viral resistance. Helicases, both viral and human, have recently emerged as novel targets for the treatment of viral infections. Here, we discuss the role of these enzymes, factors affecting their potential as drug targets and progress in the development of agents that inhibit their activity using the hepatitis C virus-encoded helicase NS3 and the cellular helicase DDX3 adopted for use by HIV-1 as examples.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , RNA Helicases/efeitos dos fármacos , RNA Viral/efeitos dos fármacos , Humanos , RNA Helicases/genética , RNA Helicases/uso terapêutico , RNA Viral/metabolismo , RNA Viral/uso terapêutico , Tecnologia Farmacêutica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA