Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Microbiol Spectr ; 10(1): e0138821, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34985336

RESUMO

Enterovirus 71 (EV71) is the major pathogen of hand, foot, and mouth disease. In severe cases, it can cause life-threatening neurological complications, such as aseptic meningitis and polio-like paralysis. There are no specific antiviral treatments for EV71 infections. In a previous study, the host protein growth arrest and DNA damage-inducible protein 34 (GADD34) expression was upregulated during EV71 infection determined by ribosome profiling and RNA-sequencing. Here, we investigated the interactions of host protein GADD34 and EV71 during infections. Rhabdomyosarcoma (RD) cells were infected with EV71 resulting in a significant increase in expression of GADD34 mRNA and protein. Through screening of EV71 protein we determined that the non-structural precursor protein 3CD is responsible for upregulating GADD34. EV71 3CD increased the RNA and protein levels of GADD34, while the 3CD mutant Y441S could not. 3CD upregulated GADD34 translation via the upstream open reading frame (uORF) of GADD34 5'untranslated regions (UTR). EV71 replication was attenuated by the knockdown of GADD34. The function of GADD34 to dephosphorylate eIF2α was unrelated to the upregulation of EV71 replication, but the PEST 1, 2, and 3 regions of GADD34 were required. GADD34 promoted the EV71 internal ribosome entry site (IRES) activity through the PEST repeats and affected several other viruses. Finally, GADD34 amino acids 563 to 565 interacted with 3CD, assisting GADD34 to target the EV71 IRES. Our research reveals a new mechanism by which GADD34 promotes viral IRES and how the EV71 non-structural precursor protein 3CD regulates host protein expression to support viral replication. IMPORTANCE Identification of host factors involved in viral replication is an important approach in discovering viral pathogenic mechanisms and identifying potential therapeutic targets. Previously, we screened host proteins that were upregulated by EV71 infection. Here, we report the interaction between the upregulated host protein GADD34 and EV71. EV71 non-structural precursor protein 3CD activates the RNA and protein expression of GADD34. Our study reveals that 3CD regulates the uORF of the 5'-UTR to increase GADD34 translation, providing a new explanation for how viral proteins regulate host protein expression. GADD34 is important for EV71 replication, and the key functional domains of GADD34 that promote EV71 are PEST 1, 2, and 3 regions. We report that GADD34 promotes viral IRES for the first time and this process is independent of its eIF2α phosphatase activity.


Assuntos
Enterovirus Humano A/genética , Enterovirus Humano A/metabolismo , Doença de Mão, Pé e Boca/metabolismo , Biossíntese de Proteínas , Proteína Fosfatase 1/metabolismo , Proteínas não Estruturais Virais/metabolismo , Regiões 5' não Traduzidas , Motivos de Aminoácidos , Linhagem Celular , Enterovirus Humano A/química , Doença de Mão, Pé e Boca/genética , Doença de Mão, Pé e Boca/virologia , Interações Hospedeiro-Patógeno , Humanos , Sítios Internos de Entrada Ribossomal , Fases de Leitura Aberta , Ligação Proteica , Proteína Fosfatase 1/química , Proteína Fosfatase 1/genética , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Replicação Viral
2.
Vitam Horm ; 117: 17-46, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34420580

RESUMO

The emergence and re-emergence of viral pathogens capable of causing epidemics or pandemics pose a serious healthcare burden. Small molecule antivirals used in conventional therapy have given rise to the severe problem of viral resistance against them. Peptides are generally considered safe, effective and are less likely to induce viral resistance. Antiviral peptides can be identified from screening of phage display of combinational peptide libraries, peptide array libraries or designed against viral targets. Limitations of peptides such as bioavailability can be improved with chemical modifications. Nanotechnology can further improve the stability of peptides in systemic circulation and enhance the antiviral activity of peptides, making them an appealing therapeutic option.


Assuntos
Antivirais , Enterovirus Humano A , Antivirais/química , Antivirais/farmacologia , Antivirais/uso terapêutico , Enterovirus Humano A/química , Humanos , Peptídeos/química , Peptídeos/farmacologia , Peptídeos/uso terapêutico
3.
Int J Mol Sci ; 22(16)2021 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-34445463

RESUMO

Enterovirus A71 (EV-A71) is a major neurovirulent agent capable of causing severe hand, foot and mouth disease (HFMD) associated with neurological complications and death. Currently, no FDA-approved antiviral is available for the treatment of EV-A71 infections. The flavonoid silymarin was shown to exert virucidal effects, but the binding site on the capsid was unknown. In this study, the ligand interacting site of silymarin was determined in silico and validated in vitro. Moreover, the potential of EV-A71 to develop resistance against silymarin was further evaluated. Molecular docking of silymarin with the capsid of EV-A71 indicated that silymarin binds to viral protein 1 (VP1) of EV-A71, specifically at the GH loop of VP1. The in vitro binding of silymarin with VP1 of EV-A71 was validated using recombinant VP1 through ELISA competitive binding assay. Continuous passaging of EV-A71 in the presence of silymarin resulted in the emergence of a mutant carrying a substitution of isoleucine by threonine (I97T) at position 97 of the BC loop of EV-A71. The mutation was speculated to overcome the inhibitory effects of silymarin. This study provides functional insights into the underlying mechanism of EV-A71 inhibition by silymarin, but warrants further in vivo evaluation before being developed as a potential therapeutic agent.


Assuntos
Antivirais/química , Proteínas do Capsídeo/química , Capsídeo/química , Enterovirus Humano A/química , Simulação de Acoplamento Molecular , Silimarina/química , Proteínas do Capsídeo/genética , Linhagem Celular Tumoral , Farmacorresistência Viral Múltipla/genética , Enterovirus Humano A/genética , Humanos , Mutação , Estrutura Secundária de Proteína
4.
J Virol ; 95(12)2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-33789997

RESUMO

Enteroviruses belong to the genus Enterovirus of the family Picornaviridae and include four human enterovirus groups (EV-A to -D): the epidemic of enteroviruses such as human enterovirus A71 (EV-A71) and coxsackievirus A16 (CVA16) is a threat to global public health. Enteroviral protein 2C is the most conserved nonstructural protein among all enteroviruses and possesses RNA helicase activity that plays pivotal roles during enteroviral life cycles, which makes 2C an attractive target for developing antienterovirus drugs. In this study, we designed a peptide, named 2CL, based on the structure of EV-A71 2C. This peptide effectively impaired the oligomerization of EV-A71 2C protein and inhibited the RNA helicase activities of 2C proteins encoded by EV-A71 and CVA16, both of which belong to EV-A, and showed potent antiviral efficacy against EV-A71 and CVA16 in cells. Moreover, the 2CL treatment elicited a strong in vivo protective efficacy against lethal EV-A71 challenge. In addition, the antiviral strategy of targeting the 2C helicase activity can be applied to inhibit the replication of EV-B. Either 2CL or B-2CL, the peptide redesigned based on the 2CL-corresponding sequence of EV-Bs, could exert effective antiviral activity against two important EV-Bs, coxsackievirus B3 and echovirus 11. Together, our findings demonstrated that targeting the helicase activity of 2C with a rationally designed peptide is an efficient antiviral strategy against enteroviruses, and 2CL and B-2CL show promising clinical potential to be further developed as broad-spectrum antienterovirus drugs.IMPORTANCE Enteroviruses are a large group of positive-sense single-stranded RNA viruses and include numerous human pathogens, such as enterovirus A71 (EV-A71), coxsackieviruses, and echoviruses. However, no approved EV antiviral drugs are available. Enteroviral 2C is the most conserved nonstructural protein among all enteroviruses and contains the RNA helicase activity critical for the viral life cycle. Herein, according to the structure of EV-A71 2C, we designed a peptide that effectively inhibited the RNA helicase activities of EV-A71- and coxsackievirus A16 (CVA16)-encoded 2C proteins. Moreover, this peptide exerted potent antiviral effects against EV-A71 and CVA16 in cells and elicited therapeutic efficacy against lethal EV-A71 challenge in vivo Furthermore, we demonstrate that the strategy of targeting the 2C helicase activity can be used for other relevant enteroviruses, including coxsackievirus B3 and echovirus 11. In summary, our findings provide compelling evidence that the designed peptides targeting the helicase activity of 2C could be broad-spectrum antivirals for enteroviruses.


Assuntos
Antivirais/farmacologia , Proteínas de Transporte/antagonistas & inibidores , Enterovirus Humano A/efeitos dos fármacos , Infecções por Enterovirus/tratamento farmacológico , Peptídeos/farmacologia , RNA Helicases/antagonistas & inibidores , Proteínas não Estruturais Virais/antagonistas & inibidores , Animais , Antivirais/química , Antivirais/uso terapêutico , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Linhagem Celular , Desenho de Fármacos , Enterovirus Humano A/química , Enterovirus Humano A/fisiologia , Enterovirus Humano B/efeitos dos fármacos , Enterovirus Humano B/fisiologia , Infecções por Enterovirus/virologia , Humanos , Camundongos , Camundongos Endogâmicos ICR , Peptídeos/química , Peptídeos/uso terapêutico , RNA Helicases/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/efeitos dos fármacos
5.
Emerg Microbes Infect ; 7(1): 162, 2018 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-30254193

RESUMO

Coxsackievirus A16 (CVA16) is an etiologic agent of hand, foot, and mouth disease (HFMD) that affects young children, and although typically self-limited, severe complications, and fatal cases have been reported. Due to the lack of specific medication and vaccines against CVA16, there is currently a need to develop effective antivirals to better control CVA16 infections in epidemic areas. In this study, we identified the tannins chebulagic acid (CHLA) and punicalagin (PUG) as small molecules that can efficiently disrupt the CVA16 infection of human rhabdomyosarcoma cells. Both compounds significantly reduced CVA16 infectivity at micromolar concentrations without apparent cytotoxicity. A mechanistic analysis revealed that the tannins particularly targeted the CVA16 entry phase by inactivating cell-free viral particles and inhibiting viral binding. Further examination by molecular docking analysis pinpointed the targets of the tannins in the fivefold axis canyon region of the CVA16 capsid near the pocket entrance that functions in cell surface receptor binding. We suggest that CHLA and PUG are efficient antagonists of CVA16 entry and could be of value as antiviral candidates or as starting points for developing molecules to treat CVA16 infections.


Assuntos
Antivirais/farmacologia , Proteínas do Capsídeo/metabolismo , Enterovirus Humano A/efeitos dos fármacos , Infecções por Enterovirus/virologia , Bibliotecas de Moléculas Pequenas/farmacologia , Ligação Viral/efeitos dos fármacos , Antivirais/química , Benzopiranos/química , Benzopiranos/farmacologia , Proteínas do Capsídeo/antagonistas & inibidores , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Enterovirus Humano A/química , Enterovirus Humano A/genética , Enterovirus Humano A/fisiologia , Glucosídeos/química , Glucosídeos/farmacologia , Humanos , Taninos Hidrolisáveis/química , Taninos Hidrolisáveis/farmacologia , Simulação de Acoplamento Molecular , Bibliotecas de Moléculas Pequenas/química , Taninos/química , Taninos/farmacologia
6.
Acta Virol ; 62(1): 50-57, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29521103

RESUMO

There is still no effective clinical antiviral drug against human enterovirus 71 (EV71) infection, which causes hand, foot and mouth disease (HFMD) in children. Scavenger receptor class B member 2 (SCARB2) is an important receptor of EV71 as it plays a vital role in the early steps of viral infection. In this study, recombinant SCARB2 protein was expressed and purified in a prokaryotic expression system, and was identified by western blot with a monoclonal antibody and mass spectrometry analysis. Detection of the sera from mice immunized with the recombinant SCARB2 protein using ELISA and western blot showed good immunogenicity of the recombinant protein. Furthermore, in the neutralization test cytopathic effect was significantly decreased when EV71 was incubated with the immune sera before infection. In summary, the SCARB2 protein was expressed successfully, and the immune sera showed obvious antiviral effect against EV71. This study provides useful information about the interaction mechanism between SCARB2 and EV71, and is also helpful for further clinical treatment research of HFMD.


Assuntos
Enterovirus Humano A/metabolismo , Proteínas de Membrana Lisossomal/imunologia , Receptores Depuradores/imunologia , Animais , Anticorpos Antivirais , Sequência de Bases , Linhagem Celular Tumoral , Enterovirus Humano A/química , Enterovirus Humano A/imunologia , Ensaio de Imunoadsorção Enzimática , Doença de Mão, Pé e Boca/metabolismo , Humanos , Proteínas de Membrana Lisossomal/química , Proteínas de Membrana Lisossomal/metabolismo , Camundongos , Testes de Neutralização , Ligação Proteica , Receptores Depuradores/química , Receptores Depuradores/metabolismo , Proteínas Recombinantes , Vacinas Virais/imunologia
7.
Emerg Microbes Infect ; 5: e60, 2016 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-27329847

RESUMO

Enterovirus 71 (EV71) is a neurotrophic virus that causes hand, foot and mouth disease (HFMD) and occasional neurological infection among children. It infects primate cells but not rodent cells, primarily due to the incompatibility between the virus and the expressed form of its receptor, scavenger receptor class B member 2 (SCARB2) protein, on rodent cells (mSCARB2). We previously generated adapted strains (EV71:TLLm and EV71:TLLmv) that were shown to productively infect primate and rodent cell lines and whose genomes exhibited a multitude of non-synonymous mutations compared with the EV71:BS parental virus. In this study, we aimed to identify mutations that are necessary for productive infection of murine cells by EV71:BS. Using reverse genetics and site-directed mutagenesis, we constructed EV71 infectious clones with specific mutations that generated amino acid substitutions in the capsid VP1 and VP2 proteins. We subsequently assessed the infection induced by clone-derived viruses (CDVs) in mouse embryonic fibroblast NIH/3T3 and murine neuroblastoma Neuro-2a cell lines. We found that the CDV:BS-VP1(K98E,E145A,L169F) with three substitutions in the VP1 protein-K98E, E145A and L169F-productively infected both mouse cell lines for at least three passages of the virus in murine cells. Moreover, the virus gained the ability to utilize the mSCARB2 protein to infect murine cell lines. These results demonstrate that the three VP1 residues cooperate to effectively interact with the mSCARB2 protein on murine cells and permit the virus to infect murine cells. Gain-of-function studies similar to the present work provide valuable insight into the mutational trajectory required for EV71 to infect new host cells previously non-susceptible to infection.


Assuntos
Aminoácidos , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Enterovirus Humano A/química , Enterovirus Humano A/fisiologia , Mutação , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Antígenos CD36/genética , Antígenos CD36/metabolismo , Proteínas do Capsídeo/química , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Enterovirus Humano A/genética , Proteínas de Membrana Lisossomal/genética , Proteínas de Membrana Lisossomal/metabolismo , Camundongos , Mutagênese Sítio-Dirigida , Células NIH 3T3 , Genética Reversa , Células Vero
8.
J Virol ; 90(10): 5141-5151, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26962213

RESUMO

UNLABELLED: The NF-κB signaling network, which is an ancient signaling pathway, plays a pivotal role in innate immunity and constitutes a first line of defense against invading pathogens, including viruses. However, numerous viruses possess evolved strategies to antagonize the activation of the NF-κB signaling pathway. Our previous study demonstrated that the nonstructural protein 2C of enterovirus 71 (EV71), which is the major pathogen of hand, foot, and mouth disease, inhibits tumor necrosis factor alpha (TNF-α)-mediated activation of NF-κB by suppressing IκB kinase ß (IKKß) phosphorylation. Nevertheless, the mechanism underlying the inhibition of IKKß phosphorylation by EV71 2C remains largely elusive. We demonstrate that EV71 2C interacts with all isoforms of the protein phosphatase 1 (PP1) catalytic subunit (the PP1α, PP1ß, and PP1γ isoforms) through PP1-docking motifs. EV71 2C has no influence on the subcellular localization of PP1. In addition, the PP1-binding-deficient EV71 2C mutant 3E3L nearly completely lost the ability to suppress IKKß phosphorylation and NF-κB activation was markedly restored in the mutant, thereby indicating that PP1 binding is efficient for EV71 2C-mediated inhibition of IKKß phosphorylation and NF-κB activation. We further demonstrate that 2C forms a complex with PP1 and IKKß to dephosphorylate IKKß. Notably, we reveal that other human enteroviruses, including poliovirus (PV), coxsackie A virus 16 (CVA16), and coxsackie B virus 3 (CVB3), use 2C proteins to recruit PP1, leading to the inhibition of IKKß phosphorylation. Our findings indicate that enteroviruses exploit a novel mechanism to inhibit IKKß phosphorylation by recruiting PP1 and IKKß to form a complex through 2C proteins, which ultimately results in the inhibition of the NF-κB signaling pathway. IMPORTANCE: The innate antiviral immunity system performs an essential function in recognizing and eliminating invading viruses. Enteroviruses include a number of important human pathogens, including poliovirus (PV), EV71, and coxsackieviruses (CVs). As 2C is the most conserved and complex nonstructural protein of enteroviruses, its biological function is largely unclear, whereas the 2A and 3C proteinases of enteroviruses are well characterized. We reveal that EV71 2C forms a complex with PP1 and IKKß to maintain IKKß in an unphosphorylated and inactive state, resulting in the inactivation of the TNF-α-mediated NF-κB signaling pathway. We provide evidence that the 2C proteins of the enteroviruses PV, CVA16, and CVB3 suppress IKKß phosphorylation through the same mechanism involving PP1. We demonstrate that enteroviruses exploit a novel mechanism involving PP1 to regulate innate antiviral immunity, and our findings may be particularly important for understanding the pathogenicity of enteroviruses.


Assuntos
Proteínas de Transporte/metabolismo , Enterovirus/metabolismo , Quinase I-kappa B/metabolismo , NF-kappa B/metabolismo , Proteína Fosfatase 1/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas de Transporte/genética , Enterovirus/química , Enterovirus/genética , Enterovirus Humano A/química , Enterovirus Humano A/metabolismo , Enterovirus Humano B/metabolismo , Células HeLa , Humanos , Fosforilação , Poliovirus/química , Poliovirus/metabolismo , Ligação Proteica , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Proteínas não Estruturais Virais/genética
9.
Antiviral Res ; 129: 58-66, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26899790

RESUMO

Childhood exanthema caused by different serotypes of coxsackievirus (CV-A) and enterovirus A71 (EV-A71) has become a serious global health problem; it is commonly known as hand, foot, and mouth disease (HFMD). Current EV-A71 vaccine clinical trials have demonstrated that human antibody responses generated by EV-A71 vaccinations do not cross-neutralize coxsackievirus A16 (CV-A16). An effective multivalent HFMD vaccine is urgently needed. From molecular epidemiological studies in Southeast Asia, CV-A6 and CV-A10 are commonly found in HFMD outbreaks. In this study, CV-A6 and CV-A10 were individually cultured in rhabdomyosarcoma (RD) cells grown in medium containing serum, harvested and concentrated. In viral downstream purification, two viral fractions were separated by sucrose gradient zonal ultracentrifugation and detected using a SDS-PAGE analysis and a virus infectivity assay. These two viral fractions were formalin-inactivated, and only the infectious particle fraction was found to be capable of inducing CV-A serotype-specific neutralizing antibody responses in animal immunogenicity studies. These mouse and rabbit antisera also failed to cross-neutralize EV-A71 and CV-A16 infections. Only a combination of formalin-inactivated EV-A71, CV-A6, CV-A10 and CV-A16 multivalent vaccine candidates elicited cross-neutralizing antibody responses in both mouse and rabbit immunogenicity studies. The current results certainly provide important information for multivalent HFMD vaccine development.


Assuntos
Anticorpos Antivirais/imunologia , Enterovirus Humano A/imunologia , Vacinas Virais/imunologia , Vírion/imunologia , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/sangue , Antígenos Virais/química , Antígenos Virais/imunologia , Reações Cruzadas , Enterovirus Humano A/química , Enterovirus Humano A/isolamento & purificação , Enterovirus Humano A/ultraestrutura , Infecções por Enterovirus/imunologia , Genótipo , Doença de Mão, Pé e Boca/imunologia , Doença de Mão, Pé e Boca/virologia , Camundongos , Coelhos , Alinhamento de Sequência , Vacinação , Vacinas de Produtos Inativados/imunologia , Tropismo Viral , Vírion/química , Vírion/isolamento & purificação
10.
J Virol ; 88(6): 3114-26, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24352461

RESUMO

UNLABELLED: Human enterovirus 71 (EV71) is the major causative agent of severe hand-foot-and-mouth diseases (HFMD) in young children, and structural characterization of EV71 during its life cycle can aid in the development of therapeutics against HFMD. Here, we present the atomic structures of the full virion and an uncoating intermediate of a clinical EV71 C4 strain to illustrate the structural changes in the full virion that lead to the formation of the uncoating intermediate prepared for RNA release. Although the VP1 N-terminal regions observed to penetrate through the junction channel at the quasi-3-fold axis in the uncoating intermediate of coxsackievirus A16 were not observed in the EV71 uncoating intermediate, drastic conformational changes occur in this region, as has been observed in all capsid proteins. Additionally, the RNA genome interacts with the N-terminal extensions of VP1 and residues 32 to 36 of VP3, both of which are situated at the bottom of the junction. These observations highlight the importance of the junction for genome release. Furthermore, EV71 uncoating is associated with apparent rearrangements and expansion around the 2- and 5-fold axes without obvious changes around the 3-fold axes. Therefore, these structures enabled the identification of hot spots for capsid rearrangements, which led to the hypothesis that the protomer interface near the junction and the 2-fold axis permits the opening of large channels for the exit of polypeptides and viral RNA, which is an uncoating mechanism that is likely conserved in enteroviruses. IMPORTANCE: Human enterovirus 71 (EV71) is the major causative agent of severe hand-foot-and-mouth diseases (HFMD) in young children. EV71 contains an RNA genome protected by an icosahedral capsid shell. Uncoating is essential in EV71 life cycle, which is characterized by conformational changes in the capsid to facilitate RNA release into host cell. Here we present the atomic structures of the full virion and an uncoating intermediate of a clinical C4 strain of EV71. Structural analysis revealed drastic conformational changes associated with uncoating in all the capsid proteins near the junction at the quasi-3-fold axis and protein-RNA interactions at the bottom of the junction in the uncoating intermediate. Significant capsid rearrangements also occur at the icosahedral 2- and 5-fold axes but not at the 3-fold axis. Taking the results together, we hypothesize that the junction and nearby areas are hot spots for capsid breaches for the exit of polypeptides and viral RNA during uncoating.


Assuntos
Capsídeo/química , Enterovirus Humano A/química , Doença de Mão, Pé e Boca/virologia , Capsídeo/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Cristalização , Enterovirus Humano A/genética , Enterovirus Humano A/metabolismo , Humanos , Modelos Moleculares
11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 28(9): 956-9, 2012 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-22980661

RESUMO

AIM: To express recombinant structural protein VP1 of enterovirus 71 (EV71) in E.coli and prepare anti-VP1 monoclonal antibodies (mAbs). METHODS: With P1 gene as a template, EV71 VP1 gene was amplified by PCR and cloned into the expression vector pET-32a(+). After transformation into E.coli TG1, the positive clones were screened and sequenced. The recombinant plasmid was then transformed into BLgold (DE3) and the recombinant protein in inclusion bodies was induced by IPTG and detected by SDS-PAGE. After the inclusion bodies were solubilized with 6 mol/L guanidine hydrochloride, we purified VP1 protein using Ni-NTA affinity chromatography, and then immunized the mice with it to prepare the mAbs against VP1 protein. RESULTS: Recombinant VP1 protein was expressed in E.coli. We obtained totally 24 VP1 monoclonal hybridoma cell strains, of which one was EV71 positive determined by Western blotting and five were positive for IFA. CONCLUSION: These mAbs are valuable reagents for the development of new vaccines and detection kits for EV71.


Assuntos
Anticorpos Monoclonais/biossíntese , Enterovirus Humano A/imunologia , Proteínas Recombinantes/biossíntese , Proteínas Estruturais Virais/biossíntese , Vacinas Virais/imunologia , Animais , Anticorpos Monoclonais/imunologia , Enterovirus Humano A/química , Enterovirus Humano A/isolamento & purificação , Escherichia coli/genética , Camundongos , Proteínas Estruturais Virais/imunologia
12.
J Biomed Sci ; 19: 73, 2012 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-22889374

RESUMO

BACKGROUND: Enterovirus 71 (EV71) is a highly infectious agent that plays an etiological role in hand, foot, and mouth disease. It is associated with severe neurological complications and has caused significant mortalities in recent large-scale outbreaks. Currently, no effective vaccine or specific clinical therapy is available against EV71. METHODS: Unmodified 21 nucleotide small interfering RNAs (siRNAs) and classic 2'-modified (2'-O-methylation or 2'-fluoro modification) siRNAs were designed to target highly conserved 5' untranslated region (UTR) of the EV71 genome and employed as anti-EV71 agents. Real-time TaqMan RT-PCR, western blot analysis and plaque assays were carried out to evaluate specific viral inhibition by the siRNAs. RESULTS: Transfection of rhabdomyosarcoma (RD) cells with siRNAs targeting the EV71 genomic 5' UTR significantly delayed and alleviated the cytopathic effects of EV71 infection, increased cell viability in EV71-infected RD cells. The inhibitory effect on EV71 replication was sequence-specific and dosage-dependent, with significant corresponding decreases in viral RNA, VP1 protein and viral titer. Appropriate 2'-modified siRNAs exhibited similar RNA interference (RNAi) activity with dramatically increased serum stability in comparison with unmodified counterparts. CONCLUSION: Sequences were identified within the highly conserved 5' UTR that can be targeted to effectively inhibit EV71 replication through RNAi strategies. Appropriate 2'-modified siRNAs provide a promising approach to optimizing siRNAs to overcome barriers on RNAi-based antiviral therapies for broader administration.


Assuntos
Enterovirus Humano A/genética , Infecções por Enterovirus , RNA Interferente Pequeno/genética , Replicação Viral/genética , Regiões 5' não Traduzidas/genética , Linhagem Celular , Sequência Conservada , Enterovirus Humano A/química , Infecções por Enterovirus/genética , Infecções por Enterovirus/terapia , Infecções por Enterovirus/virologia , Humanos , RNA Interferente Pequeno/química , Rabdomiossarcoma/genética , Rabdomiossarcoma/virologia , Transfecção
13.
J Virol ; 86(1): 542-52, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22031931

RESUMO

Human enterovirus 71 (HEV71) is the causative agent of hand, foot, and mouth disease and associated acute neurological disease. At present, little is known about the genetic determinants of HEV71 neurovirulence. Studies of related enteroviruses have indicated that the untranslated regions (UTRs), which control virus-directed translation and replication, also exert significant influence on neurovirulence. We used an infectious cDNA clone of a subgenogroup B3 strain to construct and characterize chimeras with 5'- and 3'-UTR modifications. Replacement of the entire HEV71 5' UTR with that of human rhinovirus 2 (HRV2) resulted in a small reduction in growth efficiency in cells of both nonneuronal (rhabdomyosarcoma) and neuronal (SH-SY5Y) origin due to reduced translational efficiency. However, the introduction of a 17-nucleotide deletion into the proximal region of the 3' UTR significantly decreased the growth of HEV71-HRV2 in SH-SY5Y cells. This observation is similar to that made with stem-loop domain Z (SLD Z)-deleted coxsackievirus B3-HRV2 5'-UTR chimeras reported previously and provides the first evidence of a potentially functional SLD Z in the 3' UTR in human enterovirus A species viruses. We further showed that the cell-specific growth impairment was caused by the synergistic effects of cis-acting UTR control elements on different stages of the virus life cycle. These chimeras will further improve our understanding of the control of HEV71 replication and its relationship to neurovirulence.


Assuntos
Enterovirus Humano A/crescimento & desenvolvimento , Doença de Mão, Pé e Boca/virologia , Regiões não Traduzidas , Sequência de Bases , Linhagem Celular , DNA Viral/química , DNA Viral/genética , Enterovirus Humano A/química , Enterovirus Humano A/genética , Enterovirus Humano A/fisiologia , Regulação Viral da Expressão Gênica , Humanos , Dados de Sequência Molecular , Mutação , Conformação de Ácido Nucleico , Biossíntese de Proteínas , Especificidade da Espécie , Proteínas Virais/genética , Proteínas Virais/metabolismo , Cultura de Vírus , Replicação Viral
14.
J Virol ; 85(19): 10021-30, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21813612

RESUMO

EV71 is the primary pathogenic cause of hand-foot-mouth disease (HFMD), but an effective antiviral drug currently is unavailable. Rupintrivir, an inhibitor against human rhinovirus (HRV), has potent antiviral activities against EV71. We determined the high-resolution crystal structures of the EV71 3C(pro)/rupintrivir complex, showing that although rupintrivir interacts with EV71 3C(pro) similarly to HRV 3C(pro), the C terminus of the inhibitor cannot accommodate the leaving-group pockets of EV71 3C(pro). Our structures reveal that EV71 3C(pro) possesses a surface-recessive S2' pocket that is not present in HRV 3C(pro) that contributes to the additional substrate binding affinity. Combined with mutagenic studies, we demonstrated that catalytic Glu71 is irreplaceable for maintaining the overall architecture of the active site and, most importantly, the productive conformation of catalytic His40. We discovered the role of a previously uncharacterized residue, Arg39 of EV71 3C(pro), that can neutralize the negative charge of Glu71, which may subsequently assist deprotonation of His40 during proteolysis.


Assuntos
Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Enterovirus Humano A/química , Isoxazóis/química , Isoxazóis/metabolismo , Pirrolidinonas/química , Pirrolidinonas/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Proteases Virais 3C , Sequência de Aminoácidos , Cristalografia por Raios X , Cisteína Endopeptidases/genética , Enterovirus Humano A/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fenilalanina/análogos & derivados , Ligação Proteica , Estrutura Terciária de Proteína , Valina/análogos & derivados , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA