Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Host Microbe ; 27(2): 249-261.e5, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-32027857

RESUMO

Hand, foot, and mouth disease is a common childhood illness primarily caused by coxsackievirus A16 (CVA16), for which there are no current vaccines or treatments. We identify three CVA16-specific neutralizing monoclonal antibodies (nAbs) with therapeutic potential: 18A7, 14B10, and NA9D7. We present atomic structures of these nAbs bound to all three viral particle forms-the mature virion, A-particle, and empty particle-and show that each Fab can simultaneously occupy the mature virion. Additionally, 14B10 or NA9D7 provide 100% protection against lethal CVA16 infection in a neonatal mouse model. 18A7 binds to a non-conserved epitope present in all three particles, whereas 14B10 and NA9D7 recognize broad protective epitopes but only bind the mature virion. NA9D7 targets an immunodominant site, which may overlap the receptor-binding site. These findings indicate that CVA16 vaccines should be based on mature virions and that these antibodies could be used to discriminate optimal virion-based immunogens.


Assuntos
Anticorpos Neutralizantes , Enterovirus Humano A/imunologia , Doença de Mão, Pé e Boca/virologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/ultraestrutura , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/ultraestrutura , Proteínas do Capsídeo/imunologia , Linhagem Celular , Microscopia Crioeletrônica , Enterovirus/imunologia , Enterovirus/ultraestrutura , Enterovirus Humano A/ultraestrutura , Doença de Mão, Pé e Boca/imunologia , Doença de Mão, Pé e Boca/prevenção & controle , Humanos , Camundongos , Vacinas Virais/imunologia , Vírion/imunologia
2.
Nat Commun ; 11(1): 38, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31911601

RESUMO

Coxsackievirus A10 (CV-A10) is responsible for an escalating number of severe infections in children, but no prophylactics or therapeutics are currently available. KREMEN1 (KRM1) is the entry receptor for the largest receptor-group of hand-foot-and-mouth disease causing viruses, which includes CV-A10. We report here structures of CV-A10 mature virus alone and in complex with KRM1 as well as of the CV-A10 A-particle. The receptor spans the viral canyon with a large footprint on the virus surface. The footprint has some overlap with that seen for the neonatal Fc receptor complexed with enterovirus E6 but is larger and distinct from that of another enterovirus receptor SCARB2. Reduced occupancy of a particle-stabilising pocket factor in the complexed virus and the presence of both unbound and expanded virus particles suggests receptor binding initiates a cascade of conformational changes that produces expanded particles primed for viral uncoating.


Assuntos
Enterovirus Humano A/fisiologia , Infecções por Enterovirus/metabolismo , Vírus da Febre Aftosa/fisiologia , Doença de Mão, Pé e Boca/metabolismo , Proteínas de Membrana/metabolismo , Receptores Virais/metabolismo , Enterovirus Humano A/química , Enterovirus Humano A/genética , Enterovirus Humano A/ultraestrutura , Infecções por Enterovirus/genética , Infecções por Enterovirus/virologia , Vírus da Febre Aftosa/genética , Doença de Mão, Pé e Boca/genética , Doença de Mão, Pé e Boca/virologia , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Receptores Virais/química , Receptores Virais/genética , Desenvelopamento do Vírus
3.
Nucleic Acids Res ; 48(3): 1392-1405, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31863580

RESUMO

The enterovirus 71 (EV71) 3Dpol is an RNA-dependent RNA polymerase (RdRP) that plays the central role in the viral genome replication, and is an important target in antiviral studies. Here, we report a crystal structure of EV71 3Dpol elongation complex (EC) at 1.8 Å resolution. The structure reveals that the 5'-end guanosine of the downstream RNA template interacts with a fingers domain pocket, with the base sandwiched by H44 and R277 side chains through hydrophobic stacking interactions, and these interactions are still maintained after one in-crystal translocation event induced by nucleotide incorporation, implying that the pocket could regulate the functional properties of the polymerase by interacting with RNA. When mutated, residue R277 showed an impact on virus proliferation in virological studies with residue H44 having a synergistic effect. In vitro biochemical data further suggest that mutations at these two sites affect RNA binding, EC stability, but not polymerase catalytic rate (kcat) and apparent NTP affinity (KM,NTP). We propose that, although rarely captured by crystallography, similar surface pocket interaction with nucleobase may commonly exist in nucleic acid motor enzymes to facilitate their processivity. Potential applications in antiviral drug and vaccine development are also discussed.


Assuntos
Enterovirus Humano A/ultraestrutura , Complexos Multiproteicos/ultraestrutura , Conformação Proteica , RNA Polimerase Dependente de RNA/ultraestrutura , Antivirais/química , Sítios de Ligação , Cristalografia por Raios X , Enterovirus Humano A/química , Enterovirus Humano A/genética , Genoma Viral , Humanos , Modelos Moleculares , Complexos Multiproteicos/química , Nucleotídeos/química , RNA Viral/química , RNA Viral/ultraestrutura , RNA Polimerase Dependente de RNA/química , Replicação Viral/genética
4.
PLoS Pathog ; 14(1): e1006778, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29324904

RESUMO

A close relative of poliovirus, enterovirus 71 (EV71) is regarded as an important neurotropic virus of serious public health concern. EV71 causes Hand, Foot and Mouth Disease and has been associated with neurological complications in young children. Our limited understanding of the mechanisms involved in its neuropathogenesis has hampered the development of effective therapeutic options. Here, using a two-dimensional proteomics approach combined with mass spectrometry, we have identified a unique panel of host proteins that were differentially and dynamically modulated during EV71 infection of motor-neuron NSC-34 cells, which are found at the neuromuscular junctions where EV71 is believed to enter the central nervous system. Meta-analysis with previously published proteomics studies in neuroblastoma or muscle cell lines revealed minimal overlapping which suggests unique host-pathogen interactions in NSC-34 cells. Among the candidate proteins, we focused our attention on prohibitin (PHB), a protein that is involved in multiple cellular functions and the target of anti-cancer drug Rocaglamide (Roc-A). We demonstrated that cell surface-expressed PHB is involved in EV71 entry into neuronal cells specifically, while membrane-bound mitochondrial PHB associates with the virus replication complex and facilitates viral replication. Furthermore, Roc-A treatment of EV71-infected neuronal cells reduced significantly virus yields. However, the inhibitory effect of Roc-A on PHB in NSC-34 cells was not through blocking the CRAF/MEK/ERK pathway as previously reported. Instead, Roc-A treated NSC-34 cells had lower mitochondria-associated PHB and lower ATP levels that correlated with impaired mitochondria integrity. In vivo, EV71-infected mice treated with Roc-A survived longer than the vehicle-treated animals and had significantly lower virus loads in their spinal cord and brain, whereas virus titers in their limb muscles were comparable to controls. Together, this study uncovers PHB as the first host factor that is specifically involved in EV71 neuropathogenesis and a potential drug target to limit neurological complications.


Assuntos
Membrana Celular/virologia , Enterovirus Humano A/fisiologia , Interações Hospedeiro-Patógeno , Membranas Mitocondriais/virologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/virologia , Proteínas Repressoras/metabolismo , Animais , Antivirais/uso terapêutico , Benzofuranos/uso terapêutico , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Enterovirus Humano A/efeitos dos fármacos , Enterovirus Humano A/patogenicidade , Enterovirus Humano A/ultraestrutura , Infecções por Enterovirus/tratamento farmacológico , Infecções por Enterovirus/metabolismo , Infecções por Enterovirus/patologia , Infecções por Enterovirus/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/ultraestrutura , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/ultraestrutura , Proibitinas , Proteômica/métodos , Interferência de RNA , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Organismos Livres de Patógenos Específicos , Análise de Sobrevida , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
5.
J Virol ; 92(2)2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29093091

RESUMO

Coxsackievirus A6 (CVA6) has recently emerged as one of the predominant causative agents of hand, foot, and mouth disease (HFMD). The structure of the CVA6 mature viral particle has not been solved thus far. Our previous work shows that recombinant virus-like particles (VLPs) of CVA6 represent a promising CVA6 vaccine candidate. Here, we report the first cryo-electron microscopy (cryo-EM) structure of the CVA6 VLP at 3.0-Å resolution. The CVA6 VLP exhibits the characteristic features of enteroviruses but presents an open channel at the 2-fold axis and an empty, collapsed VP1 pocket, which is broadly similar to the structures of the enterovirus 71 (EV71) VLP and coxsackievirus A16 (CVA16) 135S expanded particle, indicating that the CVA6 VLP is in an expanded conformation. Structural comparisons reveal that two common salt bridges within protomers are maintained in the CVA6 VLP and other viruses of the Enterovirus genus, implying that these salt bridges may play a critical role in enteroviral protomer assembly. However, there are apparent structural differences among the CVA6 VLP, EV71 VLP, and CVA16 135S particle in the surface-exposed loops and C termini of subunit proteins, which are often antigenic sites for enteroviruses. By immunological assays, we identified two CVA6-specific linear B-cell epitopes (designated P42 and P59) located at the GH loop and the C-terminal region of VP1, respectively, in agreement with the structure-based prediction of antigenic sites. Our findings elucidate the structural basis and important antigenic sites of the CVA6 VLP as a strong vaccine candidate and also provide insight into enteroviral protomer assembly.IMPORTANCE Coxsackievirus A6 (CVA6) is becoming one of the major pathogens causing hand, foot, and mouth disease (HFMD), leading to significant morbidity and mortality in children and adults. However, no vaccine is currently available to prevent CVA6 infection. Our previous work shows that recombinant virus-like particles (VLPs) of CVA6 are a promising CVA6 vaccine candidate. Here, we present a 3.0-Å structure of the CVA6 VLP determined by cryo-electron microscopy. The overall architecture of the CVA6 VLP is similar to those of the expanded structures of enterovirus 71 (EV71) and coxsackievirus A16 (CVA16), but careful structural comparisons reveal significant differences in the surface-exposed loops and C termini of each capsid protein of these particles. In addition, we identified two CVA6-specific linear B-cell epitopes and mapped them to the GH loop and the C-terminal region of VP1, respectively. Collectively, our findings provide a structural basis and important antigenic information for CVA6 VLP vaccine development.


Assuntos
Microscopia Crioeletrônica , Enterovirus Humano A/química , Enterovirus Humano A/ultraestrutura , Epitopos/química , Vírion/química , Vírion/ultraestrutura , Sequência de Aminoácidos , Animais , Capsídeo/química , Capsídeo/imunologia , Proteínas do Capsídeo/química , Proteínas do Capsídeo/imunologia , Enterovirus Humano A/imunologia , Mapeamento de Epitopos , Epitopos/imunologia , Epitopos de Linfócito B/química , Epitopos de Linfócito B/imunologia , Humanos , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Conformação Proteica , Células Sf9 , Vírion/imunologia
6.
PLoS Pathog ; 12(3): e1005454, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26938634

RESUMO

Enterovirus 71 (EV71) is the main pathogen responsible for hand, foot and mouth disease with severe neurological complications and even death in young children. We have recently identified a highly potent anti-EV71 neutralizing monoclonal antibody, termed D5. Here we investigated the structural basis for recognition of EV71 by the antibody D5. Four three-dimensional structures of EV71 particles in complex with IgG or Fab of D5 were reconstructed by cryo-electron microscopy (cryo-EM) single particle analysis all at subnanometer resolutions. The most critical EV71 mature virion-Fab structure was resolved to a resolution of 4.8 Å, which is rare in cryo-EM studies of virus-antibody complex so far. The structures reveal a bivalent binding pattern of D5 antibody across the icosahedral 2-fold axis on mature virion, suggesting that D5 binding may rigidify virions to prevent their conformational changes required for subsequent RNA release. Moreover, we also identified that the complementary determining region 3 (CDR3) of D5 heavy chain directly interacts with the extremely conserved VP1 GH-loop of EV71, which was validated by biochemical and virological assays. We further showed that D5 is indeed able to neutralize a variety of EV71 genotypes and strains. Moreover, D5 could potently confer protection in a mouse model of EV71 infection. Since the conserved VP1 GH-loop is involved in EV71 binding with its uncoating receptor, the scavenger receptor class B, member 2 (SCARB2), the broadly neutralizing ability of D5 might attribute to its inhibition of EV71 from binding SCARB2. Altogether, our results elucidate the structural basis for the binding and neutralization of EV71 by the broadly neutralizing antibody D5, thereby enhancing our understanding of antibody-based protection against EV71 infection.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Enterovirus Humano A/imunologia , Modelos Moleculares , Animais , Sequência de Bases , Chlorocebus aethiops , Microscopia Crioeletrônica , Enterovirus Humano A/química , Enterovirus Humano A/genética , Enterovirus Humano A/ultraestrutura , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Camundongos , Conformação Molecular , Dados de Sequência Molecular , Mutação , Receptores Depuradores/metabolismo , Alinhamento de Sequência , Análise de Sequência de DNA , Células Vero , Vírion/química
7.
Virus Genes ; 52(2): 189-94, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26837894

RESUMO

Exosomes are small secreted cellular vesicles for intercellular communications which contain proteins, mRNAs, and microRNAs (miRNAs). Recent studies have shown that exosomes play an important role in the transmission of infectious agents including hepatitis C virus, human immunodeficiency virus, and so on. However, the role of exosomes in the transfer of enterovirus 71 (EV71) between host cells remains unknown. In this study, we show that the exosomes derived from EV71-infected rhabdomyosarcoma cells contain EV71 RNA and capsid protein VP1, determined by quantitative reverse transcription-PCR (QRT-PCR) and Western blot analysis. The shedding of exosomes containing virus can establish a productive infection in human neuroblastoma cell line (SK-N-SH). A comparative analysis of neutralization by EV71-specific immunoglobulins showed different levels of neutralization of exosomes-mediated infection compared with free virus. In conclusion, exosomes from EV71-infected cells may play an important role in virus dissemination and are partially resisted to antibody neutralization. Our results suggest that there is an exosomal route of EV71 transmission infection.


Assuntos
Enterovirus Humano A/fisiologia , Exossomos/virologia , Replicação Viral , Anticorpos Neutralizantes/imunologia , Transporte Biológico , Linhagem Celular Tumoral , Enterovirus Humano A/ultraestrutura , Infecções por Enterovirus/virologia , Exossomos/ultraestrutura , Humanos , Neuroblastoma
8.
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
9.
PLoS Pathog ; 11(10): e1005165, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26485389

RESUMO

The replication of enterovirus 71 (EV71) and coxsackievirus A16 (CVA16), which are the major cause of hand, foot and mouth disease (HFMD) in children, can be inhibited by the capsid binder GPP3. Here, we present the crystal structure of CVA16 in complex with GPP3, which clarifies the role of the key residues involved in interactions with the inhibitor. Based on this model, in silico docking was performed to investigate the interactions with the two next-generation capsid binders NLD and ALD, which we show to be potent inhibitors of a panel of enteroviruses with potentially interesting pharmacological properties. A meta-analysis was performed using the available structural information to obtain a deeper insight into those structural features required for capsid binders to interact effectively and also those that confer broad-spectrum anti-enterovirus activity.


Assuntos
Antivirais/farmacologia , Enterovirus Humano A/química , Enterovirus Humano A/efeitos dos fármacos , Enterovirus Humano A/ultraestrutura , Modelos Moleculares , Animais , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Infecções por Coxsackievirus/prevenção & controle , Cristalografia por Raios X , Humanos
10.
Proc Natl Acad Sci U S A ; 111(6): 2134-9, 2014 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-24469789

RESUMO

Antibodies were prepared by immunizing mice with empty, immature particles of human enterovirus 71 (EV71), a picornavirus that causes severe neurological disease in young children. The capsid structure of these empty particles is different from that of the mature virus and is similar to "A" particles encountered when picornaviruses recognize a potential host cell before genome release. The monoclonal antibody E18, generated by this immunization, induced a conformational change when incubated at temperatures between 4 °C and 37 °C with mature virus, transforming infectious virions into A particles. The resultant loss of genome that was observed by cryo-EM and a fluorescent SYBR Green dye assay inactivated the virus, establishing the mechanism by which the virus is inactivated and demonstrating that the E18 antibody has potential as an anti-EV71 therapy. The antibody-mediated virus neutralization by the induction of genome release has not been previously demonstrated. Furthermore, the present results indicate that antibodies with genome-release activity could also be produced for other picornaviruses by immunization with immature particles.


Assuntos
Anticorpos Neutralizantes/imunologia , Enterovirus Humano A/genética , Genoma Viral , Microscopia Crioeletrônica , Enterovirus Humano A/imunologia , Enterovirus Humano A/ultraestrutura , Ensaio de Placa Viral
11.
J Virol ; 87(21): 11363-70, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23946455

RESUMO

Enterovirus 71 (EV71) is a picornavirus that causes outbreaks of hand, foot, and mouth disease (HFMD), primarily in the Asia-Pacific area. Unlike coxsackievirus A16, which also causes HFMD, EV71 induces severe neuropathology leading to high fatalities, especially among children under the age of 6 years. Currently, no established vaccines or treatments are available against EV71 infection. The monoclonal antibody MA28-7 neutralizes only specific strains of EV71 that have a conserved glycine at amino acid VP1-145, a surface-exposed residue that maps to the 5-fold vertex and that has been implicated in receptor binding. The cryo-electron microscopy structure of a complex between EV71 and the Fab fragment of MA28-7 shows that only one Fab fragment occupies each 5-fold vertex. A positively charged patch, which has also been implicated in receptor binding, lies within the Fab footprint. We identify the strain-specific epitope of EV71 and discuss the possible neutralization mechanisms of the antibody.


Assuntos
Anticorpos Neutralizantes/imunologia , Enterovirus Humano A/imunologia , Epitopos/imunologia , Fragmentos Fab das Imunoglobulinas/imunologia , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/ultraestrutura , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Neutralizantes/ultraestrutura , Pré-Escolar , Microscopia Crioeletrônica , Enterovirus Humano A/química , Enterovirus Humano A/ultraestrutura , Epitopos/química , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/isolamento & purificação , Fragmentos Fab das Imunoglobulinas/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Vírion/ultraestrutura
12.
PLoS Pathog ; 9(3): e1003240, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23555253

RESUMO

Since its discovery in 1969, enterovirus 71 (EV71) has emerged as a serious worldwide health threat. This human pathogen of the picornavirus family causes hand, foot, and mouth disease, and also has the capacity to invade the central nervous system to cause severe disease and death. Upon binding to a host receptor on the cell surface, the virus begins a two-step uncoating process, first forming an expanded, altered "A-particle", which is primed for genome release. In a second step after endocytosis, an unknown trigger leads to RNA expulsion, generating an intact, empty capsid. Cryo-electron microscopy reconstructions of these two capsid states provide insight into the mechanics of genome release. The EV71 A-particle capsid interacts with the genome near the icosahedral two-fold axis of symmetry, which opens to the external environment via a channel ∼10 Šin diameter that is lined with patches of negatively charged residues. After the EV71 genome has been released, the two-fold channel shrinks, though the overall capsid dimensions are conserved. These structural characteristics identify the two-fold channel as the site where a gateway forms and regulates the process of genome release.


Assuntos
Proteínas do Capsídeo/metabolismo , Enterovirus Humano A/genética , Enterovirus Humano A/patogenicidade , Genoma Viral , Interações Hospedeiro-Patógeno , Desenvelopamento do Vírus/fisiologia , Capsídeo/fisiologia , Capsídeo/ultraestrutura , Microscopia Crioeletrônica , Endocitose , Enterovirus Humano A/metabolismo , Enterovirus Humano A/ultraestrutura , Infecções por Enterovirus/virologia , Células HeLa , Humanos , Ligação Proteica , RNA Viral/fisiologia , Receptores de Superfície Celular/metabolismo , Vírion/genética
13.
PLoS One ; 7(11): e49973, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23226233

RESUMO

BACKGROUND: Coxsackie virus A16 (CVA16) infections have become a serious public health problem in the Asia-Pacific region. It manifests most often in childhood exanthema, commonly known as hand-foot-and-mouth disease (HFMD). There are currently no vaccine or effective medical treatments available. PRINCIPAL FINDING: In this study, we describe the production, purification and characterization of CVA16 virus produced from Vero cells grown on 5 g/L Cytodex 1 microcarrier beads in a five-liter serum-free bioreactor system. The viral titer was found to be >10(6) the tissue culture's infectious dose (TCID(50)) per mL within 7 days post-infection when a multiplicity of infection (MOI) of 10(-5) was used for initial infection. Two CVA16 virus fractions were separated and detected when the harvested CVA16 viral concentrate was purified by a sucrose gradient zonal ultracentrifugation. The viral particles detected in the 24-28% sucrose fractions had low viral infectivity and RNA content. The viral particles obtained from 35-38% sucrose fractions were found to have high viral infectivity and RNA content, and composed of four viral proteins (VP1, VP2, VP3 and VP4), as shown by SDS-PAGE analyses. These two virus fractions were formalin-inactivated and only the infectious particle fraction was found to be capable of inducing CVA16-specific neutralizing antibody responses in both mouse and rabbit immunogenicity studies. But these antisera failed to neutralize enterovirus 71. In addition, rabbit antisera did not react with any peptides derived from CVA16 capsid proteins. Mouse antisera recognized a single linear immunodominant epitope of VP3 corresponding to residues 176-190. CONCLUSION: These results provide important information for cell-based CVA16 vaccine development. To eliminate HFMD, a bivalent EV71/CVA16 vaccine formulation is necessary.


Assuntos
Anticorpos Antivirais/biossíntese , Proteínas do Capsídeo/imunologia , Enterovirus Humano A/isolamento & purificação , Vírion/isolamento & purificação , Animais , Anticorpos Antivirais/imunologia , Reatores Biológicos , Proteínas do Capsídeo/genética , Centrifugação com Gradiente de Concentração , Chlorocebus aethiops , Eletroforese em Gel de Poliacrilamida , Enterovirus Humano A/crescimento & desenvolvimento , Enterovirus Humano A/imunologia , Enterovirus Humano A/ultraestrutura , Epitopos/imunologia , Doença de Mão, Pé e Boca/prevenção & controle , Humanos , Camundongos , Microscopia Eletrônica de Transmissão , Testes de Neutralização , Coelhos , Células Vero , Vírion/crescimento & desenvolvimento , Vírion/imunologia , Vírion/ultraestrutura
14.
Bing Du Xue Bao ; 28(3): 201-6, 2012 May.
Artigo em Chinês | MEDLINE | ID: mdl-22764520

RESUMO

To construct a recombinant expression plasmid Bacmid-P1-3CD containing the P1 and 3CD genes of enterovirus 71(EV71), the P1 and 3CD genes were cloned into the same baculovirus shuttle vector (Bacmid). Recombinant AcMNPV-P1-3CD was obtained by transfecting the Bacmid-P1-3CD into the insect cell line of S f9. With the IFA and Western-blot methods for identification of expression products confirmed that the target protein was expressed in interior of infected S f9 cells. Electron microscopy showed that the structural protein capsid P1 was cut by virus-encoded protease 3CD and assembled into EV71 virus like particles (VLPs) about 27nm diameter. Different values of MOI and time points of expression were compared to explore the optimal expression condition, and the results showed that the time point could be a more important factor. Then we used S f9 cells with serum-free medium in CellSTACK-10 Culture Chambers to produce EV71 VLPs in the confirmed condition. After purification of VLPs by density gradient centrifugation, we observed on SDS-PAGE profile the purified sample contained three major proteins whose molecular masses corresponded to those of VP1 (39kD), VP0 (34kD) and VP3 (26kD) as well as the intact structure, which can be greatly used for further study in protein structure and genetic engineering vaccine research.


Assuntos
Enterovirus Humano A/fisiologia , Expressão Gênica , Vírion/fisiologia , Animais , Baculoviridae/genética , Baculoviridae/metabolismo , Linhagem Celular , Enterovirus Humano A/genética , Enterovirus Humano A/isolamento & purificação , Enterovirus Humano A/ultraestrutura , Spodoptera , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vírion/genética , Vírion/isolamento & purificação , Vírion/ultraestrutura , Montagem de Vírus
15.
Science ; 336(6086): 1274, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22383808

RESUMO

Enterovirus 71 is a picornavirus associated with fatal neurological illness in infants and young children. Here, we report the crystal structure of enterovirus 71 and show that, unlike in other enteroviruses, the "pocket factor," a small molecule that stabilizes the virus, is partly exposed on the floor of the "canyon." Thus, the structure of antiviral compounds may require a hydrophilic head group designed to interact with residues at the entrance of the pocket.


Assuntos
Proteínas do Capsídeo/química , Enterovirus Humano A/química , Enterovirus Humano A/ultraestrutura , Capsídeo/química , Capsídeo/metabolismo , Capsídeo/ultraestrutura , Proteínas do Capsídeo/metabolismo , Cristalografia por Raios X , Enterovirus Humano A/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Conformação Proteica , Receptores Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA