Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Med Virol ; 95(1): e28400, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36511115

RESUMEN

Enteroviral 2A proteinase (2Apro ), a well-established and important viral functional protein, plays a key role in shutting down cellular cap-dependent translation, mainly via its proteolytic activity, and creating optimal conditions for Enterovirus survival. Accumulated data show that viruses take advantage of various signaling cascades for their life cycle; studies performed by us and others have demonstrated that the extracellular signal-regulated kinase (ERK) pathway is essential for enterovirus A71 (EV-A71) and other viruses replication. We recently showed that ERK1/2 is required for the proteolytic activity of viral 2Apro ; however, the mechanism underlying the regulation of 2Apro remains unknown. Here, we demonstrated that the 125th residue Ser125 of EV-A71 2Apro or Thr125 of coxsackievirus B3 2Apro , which is highly conserved in the Enterovirus, was phosphorylated by ERK1/2. Importantly, 2Apro with phosphor-Ser/Thr125 had much stronger proteolytic activity toward eukaryotic initiation factor 4GI and rendered the virus more efficient for multiplication and pathogenesis in hSCARB2 knock-in mice than that in nonphospho-Ser/Thr125A (S/T125A) mutants. Notably, phosphorylation-mimic mutations caused deleterious changes in 2Apro catalytic function (S/T125D/E) and in viral propagation (S125D). Crystal structure simulation analysis showed that Ser125 phosphorylation in EV-A71 2Apro enabled catalytic Cys to adopt an optimal conformation in the catalytic triad His-Asp-Cys, which enhances 2Apro proteolysis. Therefore, we are the first to report Ser/Thr125 phosphorylation of 2Apro increases enteroviral adaptation to the host to ensure enteroviral multiplication, causing pathogenicity. Additionally, weakened viruses containing a S/T125A mutation could be a general strategy to develop attenuated Enterovirus vaccines.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Proteínas Virales , Animales , Ratones , Antígenos Virales/metabolismo , Enterovirus Humano A/genética , Enterovirus Humano A/metabolismo , Infecciones por Enterovirus/virología , Fosforilación , Proteolisis , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/fisiología
2.
J Immunol ; 205(11): 3167-3178, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33127822

RESUMEN

Deubiquitinating enzymes (DUBs) are cysteine proteases that reverse the ubiquitination by removing ubiquitins from the target protein. The human genome encodes ∼100 potential DUBs, which can be classified into six families, influencing multiple cellular processes, such as antiviral responses, inflammatory responses, apoptosis, etc. To systematically explore the role of DUBs involved in antiviral immunity, we performed an RNA interference-based screening that contains 97 human DUBs. We identified that ubiquitin-specific protease (USP) 39 expression modulates the antiviral activity, which is, to our knowledge, a previously unknown function of this enzyme. Small interfering RNA knockdown of USP39 significantly enhanced viral replication, whereas overexpression of USP39 had an opposite effect. Mechanistically, USP39 does not affect the production of type I IFN but significantly promotes JAK/STAT downstream of type I signaling by enhancing IFN-stimulated response elements promoter activity and expression of IFN-stimulated genes. Interestingly, USP39, previously considered not to have the deubiquitinase activity, in this study is proved to interact with STAT1 and sustain its protein level by deubiqutination. Furthermore, we found that through novel mechanism USP39 can significantly decrease K6-linked but not K48-linked ubiquitination of STAT1 for degradation. Taken together, these findings uncover that USP39 is, to our knowledge, a new deubiquitinase that positively regulates IFN-induced antiviral efficacy.


Asunto(s)
Antivirales/metabolismo , Interferón Tipo I/metabolismo , Factor de Transcripción STAT1/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Apoptosis/fisiología , Línea Celular , Femenino , Células HEK293 , Humanos , Ratones , Interferencia de ARN/fisiología , Transducción de Señal/fisiología , Ubiquitinación/fisiología , Ubiquitinas/metabolismo
3.
Bioorg Med Chem ; 24(16): 3472-82, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27288186

RESUMEN

Enterovirus 71 (EV71) is a kind of RNA virus and one of the two causes of Hand, foot and mouth disease (HFMD). Inhibitors that target key components of Ras/Raf/MEK/ERK pathway in host cells could impair replication of EV71. A series of 3-benzyl-1,3-benzoxazine-2,4-diones were designed from a specific MEK inhibitor G8935, by replacing the double bond between C3 and C4 within the coumarin scaffold with amide bond. One compound (9f) showed submicromolar inhibitory activity among the 12 derivatives. Further optimization on 9f led to two active compounds (9k and 9m) with nanomolar bioactivities (55nM and 60nM). The results of enzymatic assays also demonstrated that this series of compounds were allosteric inhibitors of unphosphorylated MEK1. The binding mode of compound 9k was predicted by molecular dynamic simulation and the key interactions were same as published MEK1/2 allosteric inhibitors. In the cell-based assays, compounds 9k and 9m could effectively suppress the ERK1/2 pathway, expression of EV71 VP1, and EV71 induced cytopathic effect (CPE) in rhabdomyosarcoma (RD) cells.


Asunto(s)
Antivirales/farmacología , Enterovirus Humano A/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Regulación Alostérica , Antivirales/química , Línea Celular , Descubrimiento de Drogas , Humanos , Inhibidores de Proteínas Quinasas/química
4.
Org Biomol Chem ; 13(44): 10825-33, 2015 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-26313718

RESUMEN

It has been demonstrated that passenger strand cleavage is important for the activation of RNA-induced silencing complex (RISC), which is a crucial step for siRNA-mediated gene silencing. Herein, we report that isonucleotide (isoNA) modification around the cleavage site of the passenger strand would affect the in vitro potency of modified siRNAs by altering the motion pattern of the Ago2-PAZ domain. According to western blotting, q-PCR and antiviral test results, we proved that D-isonucleotide (isoNA) modification at the position 8 of the passenger strand (siMek1-S08D), which is adjacent to the cleavage site, markedly improved the in vitro potency of the modified siRNA, whereas siRNAs with D-isoNA incorporation at position 9 (siMek1-S09D) or L-isoNA incorporation at positions 8 and 9 (siMek1-S08L, siMek1-S09L) displayed lower activity compared to native siRNA. Kinetics evaluation of passenger strand cleavage induced by T. thermophilus Ago (Tt-Ago) showed that D-isoNA modification at position 8 of the passenger strand had no significant influence on the cleavage rate, but L-isoNA modification at position 8 slowed the cleavage rate markedly. Moreover, the results of molecular dynamics simulations showed that D-isoNA modification at position 8 affected the open-close motion of the PAZ domain in the Ago/siRNA complex, which may promote the loading of RISC and release of a passenger strand cleavage product, and consequently accelerate the activation of RISC and enhance silencing activity. However, D-isoNA modification at position 9 or L-isoNA modification at position 8 or 9 exerted opposite influences on the motion of the Ago-PAZ domain.


Asunto(s)
Proteínas Argonautas/metabolismo , Proteínas Bacterianas/metabolismo , ARN Interferente Pequeño/metabolismo , Thermus thermophilus/metabolismo , Proteínas Argonautas/química , Proteínas Bacterianas/química , Secuencia de Bases , Células HEK293 , Humanos , Modelos Moleculares , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , Thermus thermophilus/química , Transfección
5.
Beijing Da Xue Xue Bao Yi Xue Ban ; 47(6): 914-9, 2015 Dec 18.
Artículo en Zh | MEDLINE | ID: mdl-26679650

RESUMEN

OBJECTIVE: To determine the prevalence of Salmonella paratyphi C phage (SPC-P1) in different Salmonella serovars and to identify the integration sites in host genome. METHODS: Based on the complete genome of SPC-P1 in S. paratyphi C RKS4594, 6 pairs of primers were designed and used to amplify the fragments of SPC-P1 in 11 S. typhi, 11 S. paratyphi A, 12 S. paratyphi B and 23 S. paratyphi C strains. At the same time, 100 complete genomes of Salmonella including 20 serovars available in National Center for Biotechnology Information (NCBI) database were downloaded and aligned by Mauve 2.3.1 to determine the prevalence of SPC-P1 in these serovars. Primers were designed according to the integration sites of SPC-P1 in the genome of RKS4594, and used to amplify ten strains having SPC-P1 in the genome. The PCR products were sequenced to investigate the integration sites of SPC-P1. RESULTS: SPC-P1 was widely distributed in S.paratyphi C genome. In the study, 14 strains had all 6 fragments and 2 strains had 3-5 fragments. All the amplified fragments showed expected sizes. In contrast, in the genomes of S. typhi, S. paratyphi A and S. paratyphi B, no or only 1-2 fragments could be amplified, and the sizes were smaller than expected. The results from Mauve showed that only in the genome of S.choleraesuis, which was a close relative of S. paratyphi C, there existed an almost complete genome of SPC-P1. The insertion site of SPC-P1 in all the ten S. paratyphi C strains tested was between pgtE and yfdC genes. CONCLUSION: SPC-P1 is a unique virulence factor of S. paratyphi C. It may play roles in the host range and pathogenicity of S.paratyphi C.


Asunto(s)
Fagos de Salmonella/fisiología , Salmonella paratyphi C/virología , Integración Viral , Cartilla de ADN , Reacción en Cadena de la Polimerasa , Serogrupo
6.
Molecules ; 18(5): 6057-91, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23698055

RESUMEN

In order to find novel antiviral agents, a series of allosteric MEK1 inhibitors were designed and synthesized. Based on docking results, multiple optimizations were made on the coumarin scaffold. Some of the derivatives showed excellent MEK1 binding affinity in the appropriate enzymatic assays and displayed obvious inhibitory effects on the ERK pathway in a cellular assay. These compounds also significantly inhibited virus (EV71) replication in HEK293 and RD cells. Several compounds showed potential as agents for the treatment of viral infective diseases, with the most potent compound 18 showing an IC50 value of 54.57 nM in the MEK1 binding assay.


Asunto(s)
Antivirales , Enterovirus Humano A/fisiología , Infecciones por Enterovirus/tratamiento farmacológico , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas , Replicación Viral/efectos de los fármacos , Antivirales/síntesis química , Antivirales/química , Antivirales/farmacología , Línea Celular Tumoral , Cumarinas , Infecciones por Enterovirus/enzimología , Células HEK293 , Humanos , MAP Quinasa Quinasa 1/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología
7.
J Med Chem ; 66(14): 9363-9375, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37424079

RESUMEN

The farnesoid X receptor (FXR) is a ligand-activated nuclear receptor. Activation of FXR significantly impacts the expressions of the pivotal genes involved in bile acid metabolism, inflammation, fibrosis, and homeostasis of lipid and glucose, leading to considerable interests in developing FXR agonists for the treatment of nonalcoholic steatohepatitis (NASH) or other FXR-relevant diseases. Herein, we describe the design, optimization, and characterization of a series of N-methylene-piperazinyl derivatives as the nonbile acid FXR agonists. Particularly, compound 23 (HPG1860), a potent full FXR agonist, shows high selectivity, favorable ADME and pharmacokinetics profile, along with favorable in vivo activities demonstrated in both rodent PD model and HFD-CCl4 model and is currently in clinical development in patients with NASH in phase II.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Receptores Citoplasmáticos y Nucleares , Ácidos y Sales Biliares , Inflamación , Relación Estructura-Actividad
8.
Mater Today Bio ; 21: 100721, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37502829

RESUMEN

Development of lysosomes and mitochondria dual-targeting photosensitizer with the virtues of near-infrared (NIR) emission, highly efficient reactive oxygen generation, good phototoxicity and biocompatibility is highly desirable in the field of imaging-guided photodynamic therapy (PDT) for cancer. Herein, a new positively charged amphiphilic organic compound (2-(2-(5-(7-(4-(diphenylamino)phenyl)benzo[c][1,2,5]thiadiazol-4-yl)thiophen-2-yl)vinyl)-3-methylbenzo[d]thiazol-3-ium iodide) (ADB) based on a D-A-π-A structure is designed and comprehensively investigated. ADB demonstrates special lysosomes and mitochondria dual-organelles targeting, bright NIR aggregation-induced emission (AIE) at 736 â€‹nm, high singlet oxygen (1O2) quantum yield (0.442), as well as good biocompatibility and photostability. In addition, ADB can act as a two-photon imaging agent for the elaborate observation of living cells and blood vessel networks of tissues. Upon light irradiation, obvious decrease of mitochondrial membrane potential (MMP), abnormal mitochondria morphology, as well as phagocytotic vesicles and lysosomal disruption in cells are observed, which further induce cell apoptosis and resulting in enhanced antitumor activity for cancer treatment. In vivo experiments reveal that ADB can inhibit tumor growth efficiently upon light exposure. These findings demonstrate that this dual-organelles targeted ADB has great potential for clinical imaging-guided photodynamic therapy, and this work provides a new avenue for the development of multi-organelles targeted photosensitizers for highly efficient cancer treatment.

9.
Viruses ; 14(8)2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-36016362

RESUMEN

Intensive efforts have been made to develop models of hRSV infection or disease using various animals. However, the limitations such as semi-permissiveness and short duration of infection have impeded their applications in both the pathogenesis of hRSV and therapeutics development. Here, we present a mouse model based on a Rag2 gene knockout using CRISPR/Cas9 technology. Rag2-/- mice sustained high viral loads upon intranasal inoculation with hRSV. The average peak titer rapidly reached 1 × 109.8 copies/g and 1c106 TCID50 in nasal cavity, as well as 1 × 108 copies/g and 1 × 105 TCID50 in the lungs up to 5 weeks. Mild interstitial pneumonia, severe bronchopneumonia, elevated cytokines and NK cells were seen in Rag2-/- mice. A humanized monoclonal antibody showed strong antiviral activity in this animal model, implying that Rag2-/- mice that support long-term stable infection are a useful tool for studying the transmission and pathogenesis of human RSV, as well as evaluating therapeutics.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Animales , Modelos Animales de Enfermedad , Humanos , Pulmón , Ratones , Virus Sincitial Respiratorio Humano/genética , Carga Viral
10.
Virol Sin ; 35(1): 64-72, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31512106

RESUMEN

Enterovirus A71 (EV-A71) is the major pathogen responsible for the severe hand, foot and mouth disease worldwide, for which few effective antiviral drugs are presently available. Interferon-α (IFN-α) has been used in antiviral therapy for decades; it has been reported that EV-A71 antagonizes the antiviral activity of IFN-α based on viral 2Apro-mediated reduction of the interferon-alpha receptor 1 (IFNAR1); however, the mechanism remains unknown. Here, we showed a significant increase in IFNAR1 protein induced by IFN-α in RD cells, whereas EV-A71 infection caused obvious down-regulation of the IFNAR1 protein and blockage of IFN-α signaling. Subsequently, we observed that EV-A71 2Apro inhibited IFNAR1 translation by cleavage of the eukaryotic initiation factor 4GI (eIF4GI), without affecting IFNAR1 mRNA levels induced by IFN-α. The inhibition of IFNAR1 translation also occurred in puromycin-induced apoptotic cells when caspase-3 cleaved eIF4GI. Importantly, we verified that 2Apro could activate cellular caspase-3, which was subsequently involved in eIF4GI cleavage mediated by 2Apro. Furthermore, inhibition of caspase-3 activation resulted in the partial restoration of IFNAR1 in cells transfected with 2A or infected with EV-A71, suggesting the pivotal role of both viral 2Apro and caspase-3 activation in the disturbance of IFN-α signaling. Collectively, we elucidate a novel mechanism by which cellular caspase-3 contributes to viral 2Apro-mediated down-regulation of IFNAR1 at the translation level during EV-A71 infection, indicating that caspase-3 inhibition could be a potential complementary strategy to improve clinical anti-EV-A71 therapy with IFN-α.


Asunto(s)
Caspasa 3/genética , Regulación hacia Abajo , Enterovirus Humano A/inmunología , Interacciones Huésped-Patógeno , Biosíntesis de Proteínas , Receptor de Interferón alfa y beta/genética , Caspasa 3/inmunología , Línea Celular Tumoral , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Receptor de Interferón alfa y beta/inmunología , Rabdomiosarcoma , Transducción de Señal
11.
Eur J Med Chem ; 196: 112271, 2020 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-32305784

RESUMEN

A series of hybrids of MEK inhibitor and nitric oxide donor have been designed and synthesized. Compound 18h [4-(3-((3-(2-fluoro-3-((N-methylsulfamoyl)amino)benzyl)-4-methyl-2-oxo-2H-chromen-7-yl)oxy) propoxy)-3-(phenylsulfonyl)-1,2,5-oxadiazole 2-oxide] was proven to be more potent than the clinical compound RO5126766 in MDA-MB-231 cells. Compound 18h can significantly reduce the levels of pMEK and pERK, induce cell apoptosis in MDA-MB-231 cells, and release NO in cells efficiently, suggesting that these hybrids, while displaying the properties of both MEK inhibitors and NO donors have a mechanism of action different from that of MEK inhibitors and NO donors. Thus, we are able to report a series of multitarget hybrids with better antitumor potency than a known MEK inhibitor and NO donor.


Asunto(s)
Antineoplásicos/farmacología , Cumarinas/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cumarinas/síntesis química , Cumarinas/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HCT116 , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Simulación del Acoplamiento Molecular , Estructura Molecular , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
12.
Environ Sci Pollut Res Int ; 25(29): 29335-29344, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30121767

RESUMEN

The effects of combined exposure to perfluoroalkyl acids (PFAAs) and heavy metals (HMs) including cadmium (Cd), copper (Cu), zinc (Zn), nickel (Ni), and lead (Pb) on earthworms (Eisenia fetida) were investigated. The results have demonstrated that the concentrations of labile acid exchangeable Cd, Zn, Ni, Pb, and Cu in soil were enhanced in addition of PFAAs. With PFAAs, the uptake of Cd, Zn, Ni, Pb, and Cu in earthworms was increased compared to those without PFAAs with the order of Cd > Zn > Pb > Ni > Cu. In the presence of HMs, the average biota-to-soil accumulation factors (BSAFs) of PFAAs in earthworms were decreased by 0.498-0.729 times for perfluorooctane sulfonate (PFOS) and 0.606-0.978 times for perfluorooctanoic acid (PFOA), indicating decrease rates of PFOS were higher than those of PFOA. And different levels of HMs led to insignificant different responses on the inhibiting effects of PFAAs uptake in earthworms. The increase of Cd in fraction C (associated with cytosol) and decrease of PFAAs in fraction C and fraction P (associated with tissue fragments, cell membranes, and intact cells) especially for fraction C were revealed when they were combined, suggesting cytosolic PFAAs and Cd were susceptibly mutual effected. This study indicated that PFAAs and metals mutually affected their bioaccumulation and subcellular distribution in earthworms, which will help to understand the fate and risks of PFAAs and metals in co-contaminated soil.


Asunto(s)
Fluorocarburos/toxicidad , Metales Pesados/toxicidad , Oligoquetos/efectos de los fármacos , Contaminantes del Suelo/toxicidad , Ácidos Alcanesulfónicos/farmacocinética , Ácidos Alcanesulfónicos/toxicidad , Animales , Disponibilidad Biológica , Caprilatos/farmacocinética , Caprilatos/toxicidad , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , China , Ecotoxicología/métodos , Fluorocarburos/farmacocinética , Metales Pesados/análisis , Metales Pesados/farmacocinética , Oligoquetos/metabolismo , Suelo/química , Contaminantes del Suelo/análisis , Contaminantes del Suelo/farmacocinética
13.
Antiviral Res ; 160: 10-16, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30315876

RESUMEN

Activation of the ERK signaling cascade in host cells has been demonstrated to be essential for enterovirus A71 (EV-A71) replication. Our previous study showed that MEK kinase, which specially activated downstream ERK kinase, is an important and potential target against EV-A71. Furthermore, we reported that a series of substituted 3-benzylcoumarins designed and synthesized as well as verified for inhibiting the MEK-ERK cascade were found to be effective on anti-EV-A71. In this study, we further demonstrated that two substituted 3-benzylcoumarins designated as 13 and 14 were more effective anti-MEK/ERK activity, less cytotoxicity and stronger antiviral effect represented by inhibition of viral-induced CPE, the expression of viral proteins and the replication of the viral genome, as well as the production of progeny virions, compared to those of U0126, an available MEK inhibitor, and sorafenib, a multiple-targeted kinase inhibitor in clinical use. Moreover, we explored that the likely mechanism of action of these two test compounds were to block EV-A71 2A dependent IRES-driven activity essential for successful viral replication. Hence, our results suggest that two substituted 3-benzylcoumarins 13 and 14 could be candidates as potential anti-EV-A71 agents.


Asunto(s)
Antivirales/farmacología , Cumarinas/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Enterovirus Humano A/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Antivirales/química , Cumarinas/química , Inhibidores de Cisteína Proteinasa/química , Enterovirus Humano A/crecimiento & desarrollo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología
14.
J Virol Methods ; 248: 87-91, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28629711

RESUMEN

A versatile single-step method is described for constructing a picornavirus replicon RNA with precise ends to facilitate improved understanding of viral genome function and mimic native virus replication in host cells as far as possible. The key innovation in this new approach is the use of a bridge primer to both introduce a ribozyme sequence for cis-cleavage of RNA to generate precise 5' ends of EV71 RNA and also mediate overlapping assembly of two fragments. Using an EV71 replicon as a test case, precise ends for the viral replicon were shown to be important for efficient virus replication. Thus, our work provides a novel efficient way to generating higher efficient viral replicon with precise ends and this novel method can be applied to other picornaviruses' research.


Asunto(s)
Biología Molecular/métodos , Picornaviridae/genética , ARN Viral/genética , Replicón , Línea Celular , Cartilla de ADN , Genoma Viral , ARN Catalítico , Replicación Viral
15.
Antiviral Res ; 143: 13-21, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28351508

RESUMEN

In a previous study the ERK1/2 pathway was found to be crucially involved in positive regulation of the enterovirus A 71(EV-A71) IRES (vIRES), thereby contributing to the efficient replication of an important human enterovirus causing death in young children (<5yrs) worldwide. This study focuses on unraveling more about the detailed mechanism of ERK's involvement in this regulation of vIRES. Through the use of siRNAs and specifically pharmacological inhibitor U0126, the ERK cascade was shown to positively regulate EV-A71-mediated cleavage of eIF4GI that established the cellular conditions which favour vIRES-dependent translation. Site-directed mutagenesis of the viral 2A protease (2Apro) was undertaken to show that the positive regulation of virus replication by the ERK cascade was mediated through effects on both the cis-cleavage of the viral polyprotein by 2Apro and its trans-cleavage of cellular eIF4GI. This ERK-2Apro linked network coordinating vIRES efficiency was also found in other important human enteroviruses. This identification of the ERK cascade as having a key role in maintaining the 2Apro proteolytic activity required to maximize enterovirus IRES activity, expands current understanding of the diverse functions of the ERK signaling cascade in the regulation of viral translation, therefore providing a potentially comprehensive drug target for anti-enterovirus infection.


Asunto(s)
Antivirales/farmacología , Enterovirus Humano A/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Sitios Internos de Entrada al Ribosoma/fisiología , Transducción de Señal/fisiología , Proteínas Virales/metabolismo , Butadienos/antagonistas & inhibidores , Línea Celular Tumoral , Enterovirus Humano A/enzimología , Infecciones por Enterovirus/tratamiento farmacológico , Infecciones por Enterovirus/virología , Células HEK293 , Humanos , Sitios Internos de Entrada al Ribosoma/genética , Mutagénesis Sitio-Dirigida , Nitrilos/antagonistas & inhibidores , Poliproteínas , ARN Interferente Pequeño , Rabdomiosarcoma , Transducción de Señal/efectos de los fármacos , Proteínas Virales/genética , Replicación Viral/efectos de los fármacos
16.
Infect Genet Evol ; 45: 83-89, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27566335

RESUMEN

Currently, it is still controversial that if the pathogenicity of EV-A71 causing severe or mild hand, foot, and mouth disease (HFMD) is associated with viral nucleotide or amino acid sequence(s). In this study, 19 clinical strains were detected in samples from diagnosed patients of EV-A71-caused HFMD with mild or severe symptoms. Then, VP1-2A fragment sequences of 19 EV-A71 isolates were determined, the phylogenetic analysis, based on VP1 sequences of 19 EV-A71 stains in this study and which of 62 EV-A71 strains with different clinical phenotypes reported before, were carried out. Our results showed that no difference in the genotype and evolution distribution was observed among the EV-A71 strains mentioned above. Furthermore, two EV-A71 isolates, which with much close evolutionary relationship but different clinical manifestations, were purified by plaque assay, the complete genome sequencing was done, and deduced amino acid sequence analysis of 11 proteins coded by EV-A71 was carried out. Eight variable amino acid sites were found and further verified with those of 62 strains reported before. Our study provides further evidence that the potential pathogenicity of EV-A71 causing severe or mild HFMD seems not to be associated with viral genotype and even the amino acid substitution.


Asunto(s)
Enterovirus/genética , Enfermedad de Boca, Mano y Pie/virología , Aminoácidos , Proteínas de la Cápside/genética , Estudios de Cohortes , Enterovirus/clasificación , Genoma Viral/genética , Enfermedad de Boca, Mano y Pie/diagnóstico , Enfermedad de Boca, Mano y Pie/fisiopatología , Humanos , Fenotipo , Filogenia , ARN Viral/análisis , ARN Viral/genética , Análisis de Secuencia de ARN
17.
Viruses ; 7(3): 1344-56, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25803100

RESUMEN

It has been demonstrated that MEK1, one of the two MEK isoforms in Raf-MEK-ERK1/2 pathway, is essential for successful EV71 propagation. However, the distinct function of ERK1 and ERK2 isoforms, the downstream kinases of MEKs, remains unclear in EV71 replication. In this study, specific ERK siRNAs and selective inhibitor U0126 were applied. Silencing specific ERK did not significantly impact on the EV71-caused biphasic activation of the other ERK isoform, suggesting the EV71-induced activations of ERK1 and ERK2 were non-discriminative and independent to one another. Knockdown of either ERK1 or ERK2 markedly impaired progeny EV71 propagation (both by more than 90%), progeny viral RNA amplification (either by about 30% to 40%) and protein synthesis (both by around 70%), indicating both ERK1 and ERK2 were critical and not interchangeable to EV71 propagation. Moreover, suppression of EV71 replication by inhibiting both early and late phases of ERK1/2 activation showed no significant difference from that of only blocking the late phase, supporting the late phase activation was more importantly responsible for EV71 life cycle. Taken together, this study for the first time identified both ERK1 and ERK2 were required for EV71 efficient replication and further verified the important role of MEK1-ERK1/2 in EV71 replication.


Asunto(s)
Enterovirus Humano A/fisiología , Interacciones Huésped-Patógeno , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Replicación Viral , Línea Celular , Inhibidores Enzimáticos/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética
18.
Antiviral Res ; 106: 80-5, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24680956

RESUMEN

The activation of ERK and p38 signal cascade in host cells has been demonstrated to be essential for picornavirus enterovirus 71 (EV71) replication and up-regulation of virus-induced cyclooxygenase-2 (COX-2)/prostaglandins E2 (PGE2) expression. The aim of this study was to examine the effects of sorafenib, a clinically approved anti-cancer multi-targeted kinase inhibitor, on the propagation and pathogenesis of EV71, with a view to its possible mechanism and potential use in the design of therapy regimes for Hand foot and mouth disease (HFMD) patients with life threatening neurological complications. In this study, non-toxic concentrations of sorafenib were shown to inhibit the yield of infectious progeny EV71 (clinical BC08 strain) by about 90% in three different cell types. A similar inhibitory effect of sorafenib was observed on the synthesis of both viral genomic RNA and the VP1 protein. Interestingly, sorafenib exerted obvious inhibition of the EV71 internal ribosomal entry site (IRES)-mediated translation, the first step in picornavirus replication, by linking it to a firefly luciferase reporter gene. Sorafenib was also able to prevent both EV71-induced CPE and the activation of ERK and p38, which contributes to up-regulation COX-2/PGE2 expression induced by the virus. Overall, this study shows that sorafenib strongly inhibits EV71 replication at least in part by regulating viral IRES-dependent translation of viral proteins, indicating a novel potential strategy for the treatment of HFMD patients with severe neurological complications. To our knowledge, this is the first report that investigates the mechanism by which sorafenib inhibits EV71 replication.


Asunto(s)
Antivirales/farmacología , Enterovirus Humano A/efectos de los fármacos , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Virales/biosíntesis , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Efecto Citopatogénico Viral/efectos de los fármacos , Enterovirus Humano A/crecimiento & desarrollo , Enterovirus Humano A/fisiología , Humanos , Niacinamida/farmacología , Sorafenib , Proteínas Virales/antagonistas & inhibidores
19.
Antiviral Res ; 101: 82-92, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24263113

RESUMEN

Enterovirus 71 (EV71), a member of Picornaviridae, is one of the major pathogens of human hand, foot and mouth disease. EV71 mainly infects children and causes severe neurological complications and even death. The pathogenesis of EV71 infection is largely unknown, and no clinically approved vaccine or effective treatment is available to date. Here we described a novel bioluminescence imaging approach for EV71 detection. In this approach, a plasmid-based reporter was constructed to express the fusion protein AmN(Q/G)BC, a split firefly luciferase mutant, which can be specifically cleaved by EV71 protease 3C(pro). Upon cleavage, the splitting fusion protein restores luciferase activity. Our test confirmed that AmN(Q/G)BC was specifically cleaved by 3C(pro) and EV71 and restored the luciferase activity to a degree that corresponds to the 3C(pro) and virus doses in cells and mice. The anti-EV71 effect of GW5074 and U0126, two mitogen-activated protein kinase (MAPK) inhibitors, was evaluated using this approach to validate its application of screening anti-EV71 agents. We found that the AmN(Q/G)BC reporter efficiently monitored the inhibitory effect of GW5074 and U0126 on EV71 infection under in vitro and in vivo conditions. The data from AmN(Q/G)BC reporter were consistent with Western blotting and histopathology examination. Taken together, this real-time imaging approach can quantitatively monitor the efficacy of anti-EV71 agents and is valuable for anti-EV71 drug screening and evaluation, especially, under in vivo conditions.


Asunto(s)
Antivirales/farmacología , Cisteína Endopeptidasas/metabolismo , Enterovirus Humano A/efectos de los fármacos , Mediciones Luminiscentes/métodos , Imagen Óptica/métodos , Proteínas Virales/metabolismo , Proteasas Virales 3C , Animales , Antivirales/aislamiento & purificación , Línea Celular , Cisteína Endopeptidasas/genética , Evaluación Preclínica de Medicamentos/métodos , Luciferasas de Luciérnaga/genética , Luciferasas de Luciérnaga/metabolismo , Ratones , Ratones Endogámicos ICR , Proteínas Virales/genética
20.
Antiviral Res ; 93(1): 110-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22101247

RESUMEN

The role of the MEK1-ERK signaling cascade in the replication cycle of Enterovirus 71 (EV71), the primary cause of hand, foot and mouth disease (HFMD), has been analyzed. In vitro infection with EV71 induced a biphasic activation of ERK. The two phases of activation appeared to be triggered by different mechanisms, with the first phase being activated by the binding of viral particles to the membrane receptor of host cells and the second probably being in response to the production of new virus particles. Inhibition of ERK activation by U0126 was found to severely impair virus production. A similar reduction in EV71 replication was also observed when MEK1 expression was subject to knockdown using specific siRNAs. By contrast knockdown of MEK2 expression showed that it was dispensable for virus replication cycle, despite both MEK isoforms being activated and translocated to the nucleus equally well in response to virus infection. Overall, this study suggests distinct functions of the two isoforms of MEK in EV71 replication cycle, with an essential role for MEK1 in stimulating the ERK signaling cascade to promote virus replication. Taken together with our previous work on herpes simplex virus type 2 (HSV2) this study highlights MEK1 as a potential broad antiviral molecular target.


Asunto(s)
Enterovirus Humano A/fisiología , MAP Quinasa Quinasa 1/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Replicación Viral , Butadienos/farmacología , Efecto Citopatogénico Viral/efectos de los fármacos , Enterovirus Humano A/efectos de los fármacos , Enterovirus Humano A/efectos de la radiación , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Inhibidores Enzimáticos/farmacología , Células HEK293 , Humanos , Isoenzimas/genética , Nitrilos/farmacología , Transporte de Proteínas , ARN Interferente Pequeño , Transfección , Rayos Ultravioleta , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA