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.
Artículo en Inglés | MEDLINE | ID: mdl-37235917

RESUMEN

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.


Asunto(s)
Calcio , ADN , Conformación de Ácido Nucleico , ARN Helicasas , Imagen Individual de Molécula , Proteínas no Estructurales Virales , Calcio/metabolismo , Calcio/farmacología , ADN/química , ADN/efectos de los fármacos , ADN/metabolismo , Magnesio/metabolismo , Magnesio/farmacología , Conformación de Ácido Nucleico/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Replicación Viral , Citosol/metabolismo , Hidrólisis/efectos de los fármacos , ARN Helicasas/efectos de los fármacos , ARN Helicasas/metabolismo , Proteínas no Estructurales Virales/efectos de los fármacos , Proteínas no Estructurales Virales/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Electroforesis en Gel de Poliacrilamida , Relación Dosis-Respuesta a Droga , Transcripción Genética
2.
Molecules ; 26(22)2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34833913

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Diterpenos/farmacología , Antivirales/química , Antivirales/farmacocinética , Sitios de Unión , Simulación por Computador , Dengue/tratamiento farmacológico , Dengue/virología , Diterpenos/química , Diterpenos/farmacocinética , Diseño de Fármacos , Humanos , Simulación del Acoplamiento Molecular , Fitoquímicos/química , Fitoquímicos/farmacocinética , Fitoquímicos/farmacología , Unión Proteica , ARN Helicasas/química , ARN Helicasas/efectos de los fármacos , ARN Helicasas/metabolismo , Serina Endopeptidasas/química , Serina Endopeptidasas/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/efectos de los fármacos , Proteínas del Envoltorio Viral/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/efectos de los fármacos , Proteínas no Estructurales Virales/metabolismo
3.
Mol Biol Cell ; 32(21): ar18, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34432484

RESUMEN

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.


Asunto(s)
ADN Helicasas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Resveratrol/farmacología , Gránulos de Estrés/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Gránulos Citoplasmáticos/efectos de los fármacos , Gránulos Citoplasmáticos/metabolismo , ADN Helicasas/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Cinética , Proteínas de Unión a Poli-ADP-Ribosa/efectos de los fármacos , ARN Helicasas/efectos de los fármacos , Proteínas con Motivos de Reconocimiento de ARN/efectos de los fármacos , Ribonucleoproteínas/metabolismo , Gránulos de Estrés/efectos de los fármacos
4.
Biomed Res Int ; 2020: 6237160, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33102585

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Fitoquímicos/química , Fitoquímicos/farmacología , SARS-CoV-2/efectos de los fármacos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/química , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/química , Alanina/farmacología , Antimetabolitos/química , Antimetabolitos/farmacología , Antivirales/química , COVID-19/epidemiología , COVID-19/virología , China/epidemiología , Dioxolanos/química , Dioxolanos/farmacología , Fluoroquinolonas/química , Fluoroquinolonas/farmacología , Humanos , Metiltransferasas/efectos de los fármacos , Simulación del Acoplamiento Molecular , Nelfinavir/química , Nelfinavir/farmacología , Piperazinas/química , Piperazinas/farmacología , Conformación Proteica , ARN Helicasas/efectos de los fármacos , SARS-CoV-2/química , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/metabolismo , Inhibidores de Topoisomerasa II/química , Inhibidores de Topoisomerasa II/farmacología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo
5.
Virol Sin ; 35(3): 321-329, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32500504

RESUMEN

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.


Asunto(s)
Betacoronavirus/metabolismo , Bismuto/farmacología , Metiltransferasas/metabolismo , Nucleósido-Trifosfatasa/efectos de los fármacos , ARN Helicasas/efectos de los fármacos , Sales (Química)/farmacología , Proteínas no Estructurales Virales/metabolismo , Adenosina Trifosfatasas/efectos de los fármacos , Adenosina Trifosfatasas/metabolismo , Betacoronavirus/enzimología , Betacoronavirus/genética , COVID-19 , Infecciones por Coronavirus/virología , Humanos , Metiltransferasas/genética , Nucleósido-Trifosfatasa/genética , Nucleósido-Trifosfatasa/metabolismo , Pandemias , Neumonía Viral/virología , ARN Helicasas/genética , ARN Helicasas/metabolismo , Proteínas Recombinantes , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave , Proteínas no Estructurales Virales/genética , Replicación Viral
6.
J Virol ; 92(5)2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29237842

RESUMEN

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.


Asunto(s)
Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Norovirus/enzimología , Norovirus/genética , ARN Helicasas/genética , ARN Helicasas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Secuencia de Aminoácidos , Línea Celular , Guanidina/antagonistas & inhibidores , Humanos , Estadios del Ciclo de Vida , Chaperonas Moleculares/efectos de los fármacos , Norovirus/efectos de los fármacos , Norovirus/crecimiento & desarrollo , Nucleósido-Trifosfatasa/genética , Nucleósido-Trifosfatasa/metabolismo , Unión Proteica , Pliegue de Proteína , ARN Helicasas/efectos de los fármacos , ARN Viral/química , ARN Viral/efectos de los fármacos , ARN Viral/genética , ARN Viral/metabolismo , Replicón/efectos de los fármacos , Alineación de Secuencia , Análisis de Secuencia , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/efectos de los fármacos , Proteínas no Estructurales Virales/genética , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología
7.
Antiviral Res ; 143: 218-229, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28461069

RESUMEN

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.


Asunto(s)
Sitio Alostérico , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Proteínas no Estructurales Virales/química , Virus Zika/enzimología , Secuencia de Aminoácidos , Animales , Antivirales/antagonistas & inhibidores , Antivirales/química , Secuencia de Bases , Activación Enzimática , Femenino , Flavivirus/química , Expresión Génica , Humanos , Concentración 50 Inhibidora , Ratones , Modelos Moleculares , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , ARN Helicasas/química , ARN Helicasas/efectos de los fármacos , Factores de Transcripción SOXB1/genética , Alineación de Secuencia , Serina Endopeptidasas/química , Serina Endopeptidasas/efectos de los fármacos , Células Madre , Proteínas no Estructurales Virales/efectos de los fármacos , Proteínas Virales/química , Proteínas Virales/genética , Virus Zika/química , Virus Zika/genética , Virus Zika/crecimiento & desarrollo , Infección por el Virus Zika/virología
8.
Antiviral Res ; 143: 186-194, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28457855

RESUMEN

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.


Asunto(s)
Suramina/antagonistas & inhibidores , Infección por el Virus Zika/prevención & control , Virus Zika/efectos de los fármacos , Animales , Anticuerpos Antivirales , Cloratos/farmacología , Chlorocebus aethiops , ADN Helicasas/metabolismo , Sulfato de Dextran/antagonistas & inhibidores , Flavivirus/efectos de los fármacos , Glicosaminoglicanos/farmacología , Heparina/análogos & derivados , Heparina/química , Heparina/farmacología , Heparitina Sulfato/farmacología , Concentración 50 Inhibidora , Ratones , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , ARN Helicasas/química , ARN Helicasas/efectos de los fármacos , Serina Endopeptidasas/química , Serina Endopeptidasas/efectos de los fármacos , Suramina/administración & dosificación , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Virus Zika/fisiología , Infección por el Virus Zika/virología
9.
Antiviral Res ; 142: 141-147, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28336347

RESUMEN

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.


Asunto(s)
Proteínas no Estructurales Virales/química , Virus Zika/enzimología , Sitios de Unión , Ácidos Borónicos/antagonistas & inhibidores , Dipéptidos/antagonistas & inhibidores , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Inhibidores de Proteasas/química , Conformación Proteica , ARN Helicasas/química , ARN Helicasas/efectos de los fármacos , Serina Endopeptidasas/química , Serina Endopeptidasas/efectos de los fármacos , Proteínas no Estructurales Virales/efectos de los fármacos , Proteínas no Estructurales Virales/genética
10.
EMBO J ; 33(4): 371-84, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24488111

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila/efectos de los fármacos , Drosophila melanogaster/metabolismo , MicroARNs/metabolismo , Fosfatos/farmacología , ARN Helicasas/efectos de los fármacos , Ribonucleasa III/efectos de los fármacos , Sustitución de Aminoácidos , Animales , Arginina , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Mutagénesis Sitio-Dirigida , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , ARN Helicasas/química , ARN Helicasas/genética , ARN Helicasas/metabolismo , ARN Bicatenario/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Ribonucleasa III/química , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Especificidad por Sustrato
11.
Protein Expr Purif ; 92(2): 156-62, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24084007

RESUMEN

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.


Asunto(s)
Lisofosfatidilcolinas/farmacología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/efectos de los fármacos , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/efectos de los fármacos , Secuencia de Aminoácidos , Detergentes/química , Estabilidad de Enzimas , Lisofosfatidilcolinas/química , Micelas , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , ARN Helicasas/química , ARN Helicasas/efectos de los fármacos , ARN Helicasas/genética , ARN Helicasas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Serina Endopeptidasas/química , Serina Endopeptidasas/efectos de los fármacos , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
12.
Eur J Med Chem ; 62: 199-205, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23353753

RESUMEN

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.


Asunto(s)
Agmatina/farmacología , Dipéptidos/farmacología , Inhibidores de Proteasas/farmacología , Proteínas no Estructurales Virales/efectos de los fármacos , Virus del Nilo Occidental/enzimología , Agmatina/síntesis química , Agmatina/química , Cristalografía por Rayos X , Dipéptidos/síntesis química , Dipéptidos/química , Relación Dosis-Respuesta a Droga , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química , ARN Helicasas/efectos de los fármacos , Serina Endopeptidasas/efectos de los fármacos , Programas Informáticos , Relación Estructura-Actividad , Virus del Nilo Occidental/efectos de los fármacos
13.
J Nat Prod ; 75(4): 650-4, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22394195

RESUMEN

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.


Asunto(s)
Hepacivirus/metabolismo , Terpenos/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Secuencia de Bases , Humanos , Estructura Molecular , Nucleósido-Trifosfatasa/efectos de los fármacos , Nucleósido-Trifosfatasa/metabolismo , ARN Helicasas/efectos de los fármacos , ARN Helicasas/metabolismo
14.
Bioorg Khim ; 37(6): 723-44, 2011.
Artículo en Ruso | MEDLINE | ID: mdl-22497074
15.
Curr Opin Pharmacol ; 9(6): 680-7, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19828376

RESUMEN

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.


Asunto(s)
Neoplasias Gastrointestinales/metabolismo , Tracto Gastrointestinal/metabolismo , Receptores de Reconocimiento de Patrones/fisiología , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Enterocolitis , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/etiología , Neoplasias Gastrointestinales/inmunología , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/fisiología , Proteínas Adaptadoras de Señalización NOD/efectos de los fármacos , Proteínas Adaptadoras de Señalización NOD/metabolismo , ARN Helicasas/efectos de los fármacos , ARN Helicasas/metabolismo , Receptores de Reconocimiento de Patrones/inmunología , Receptores Toll-Like/efectos de los fármacos , Receptores Toll-Like/metabolismo
16.
Antimicrob Agents Chemother ; 52(6): 1901-11, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18391043

RESUMEN

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.


Asunto(s)
Hepacivirus/clasificación , Hepacivirus/efectos de los fármacos , Nucleótidos/farmacología , Proteínas no Estructurales Virales/efectos de los fármacos , Adenosina Trifosfatasas/efectos de los fármacos , Adenosina Trifosfatasas/metabolismo , Antivirales/farmacología , Genotipo , Hepacivirus/enzimología , Hepacivirus/genética , Humanos , Nucleótidos/química , ARN Helicasas/efectos de los fármacos , ARN Helicasas/metabolismo , ARN Viral/metabolismo , Proteínas no Estructurales Virales/metabolismo
17.
J Gen Virol ; 88(Pt 8): 2223-2227, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17622626

RESUMEN

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.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Oligopéptidos/metabolismo , Proteínas no Estructurales Virales/efectos de los fármacos , Proteínas no Estructurales Virales/metabolismo , Virus de la Fiebre Amarilla/enzimología , Secuencia de Aminoácidos , Antivirales/farmacología , Sitios de Unión/fisiología , Cinética , Datos de Secuencia Molecular , Oligopéptidos/química , ARN Helicasas/química , ARN Helicasas/efectos de los fármacos , ARN Helicasas/genética , ARN Helicasas/metabolismo , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Serina Endopeptidasas/química , Serina Endopeptidasas/efectos de los fármacos , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Especificidad por Sustrato , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética
18.
PLoS One ; 2(3): e303, 2007 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-17375189

RESUMEN

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.


Asunto(s)
Factor 4A Eucariótico de Iniciación/genética , Exones/genética , ARN Helicasas/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Bases , Factor 4A Eucariótico de Iniciación/aislamiento & purificación , Factor 4A Eucariótico de Iniciación/metabolismo , Humanos , Cinética , Oligodesoxirribonucleótidos , Reacción en Cadena de la Polimerasa/métodos , Biosíntesis de Proteínas , ARN/genética , ARN Helicasas/efectos de los fármacos , ARN Helicasas/genética , ARN Mensajero/genética , Especificidad por Sustrato , Transcripción Genética
19.
Hepatology ; 43(6): 1392-5, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16729317

RESUMEN

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.


Asunto(s)
Hepacivirus/genética , Hepatitis C/enzimología , Hepatitis C/genética , ARN Helicasas/metabolismo , Antivirales/uso terapéutico , Hepacivirus/efectos de los fármacos , Hepacivirus/enzimología , Hepatitis C/tratamiento farmacológico , Humanos , Conformación de Ácido Nucleico , ARN Helicasas/efectos de los fármacos , ARN Helicasas/genética , Factores de Riesgo , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad
20.
Nat Rev Drug Discov ; 4(10): 845-53, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16184083

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Antivirales/uso terapéutico , ARN Helicasas/efectos de los fármacos , ARN Viral/efectos de los fármacos , Humanos , ARN Helicasas/genética , ARN Helicasas/uso terapéutico , ARN Viral/metabolismo , ARN Viral/uso terapéutico , Tecnología Farmacéutica/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA