Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Immunology ; 169(3): 292-308, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36726218

RESUMO

The reported enterovirus A 71 (EVA71) vaccines and immunoglobin G (IgG) antibodies have no cross-antiviral efficacy against other enterovirus A (EV-A) which caused hand, foot and mouth disease (HFMD). Here we constructed an IgM antibody (20-IgM) based on our previous discovery to address the resistance encountered by IgG-based immunotherapy. Although binding to the same conserved neutralizing epitope within the GH loop of EV-As VP1, the antiviral breath and potency of 20-IgM are still higher than its parental 20-IgG1. The 20-IgM blocks the interaction between the EV-As and its receptors, scavenger receptor class B, member 2 (SCARB2) and Kringle-containing transmembrane protein 1(KREMEN1) of the host cell. The 20-IgM also neutralizes the EV-As at the post-attachment stages, including postattachment neutralization, uncoating and RNA release inhibition after internalization. Mechanistically, the dual blockage effect of 20-IgM is dependent on both a conserved site targeting and high affinity binding. Meanwhile, 20-IgM provides cross-antiviral efficacy in EV-As orally infected neonatal ICR mice. Collectively, 20-IgM and its property exhibit excellent antiviral activity with a dual-blockage inhibitory effect at both the pre- and post-attachment stages. The finding enhances our understanding of IgM-mediated immunity and highlights the potential of IgM subtype antibodies against enterovirus infections.


Assuntos
Enterovirus Humano A , Doença de Mão, Pé e Boca , Animais , Camundongos , Anticorpos Neutralizantes , Enterovirus Humano A/química , Enterovirus Humano A/genética , Camundongos Endogâmicos ICR , Imunoglobulina G , Imunoglobulina M
2.
Nat Commun ; 13(1): 7854, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36543790

RESUMO

Coxsackievirus A16 (CVA16) causes hand, foot and mouth disease in infants and young children. However, no vaccine or anti-viral agent is currently available for CVA16. Here, the functions and working mechanisms of two CVA16-specific neutralizing monoclonal antibodies (MAbs), 9B5 and 8C4, are comprehensively investigated. Both 9B5 and 8C4 display potent neutralization in vitro and prophylactic and therapeutic efficacy in a mouse model of CVA16 infection. Mechanistically, 9B5 exerts neutralization primarily through inhibiting CVA16 attachment to cell surface via blockade of CVA16 binding to its attachment receptor, heparan sulfate, whereas 8C4 functions mainly at the post-attachment stage of CVA16 entry by interfering with the interaction between CVA16 and its uncoating receptor SCARB2. Cryo-EM studies show that 9B5 and 8C4 target distinct epitopes located at the 5-fold and 3-fold protrusions of CVA16 capsids, respectively, and exhibit differential binding preference to three forms of naturally occurring CVA16 particles. Moreover, 9B5 and 8C4 are compatible in formulating an antibody cocktail which displays the ability to prevent virus escape seen with individual MAbs. Together, our work elucidates the functional and structural basis of CVA16 antibody-mediated neutralization and protection, providing important information for design and development of effective CVA16 vaccines and antibody therapies.


Assuntos
Infecções por Coxsackievirus , Enterovirus Humano A , Enterovirus , Camundongos , Animais , Enterovirus Humano A/química , Anticorpos Neutralizantes , Capsídeo/química , Proteínas do Capsídeo/química , Enterovirus/química
3.
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
4.
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
5.
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
6.
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
7.
Nat Commun ; 11(1): 5253, 2020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-33067459

RESUMO

Enterovirus 71 (EV71)-neutralizing antibodies correlate with protection and have potential as therapeutic agents. We isolate and characterize a panel of plasmablast-derived monoclonal antibodies from an infected child whose antibody response focuses on the plateau epitope near the icosahedral 3-fold axes. Eight of a total of 19 antibodies target this epitope and three of these potently neutralize the virus. Representative neutralizing antibodies 38-1-10A and 38-3-11A both confer effective protection against lethal EV71 challenge in hSCARB2-transgenic mice. The cryo-electron microscopy structures of the EV71 virion in complex with Fab fragments of these potent and protective antibodies reveal the details of a conserved epitope formed by residues in the BC and HI loops of VP2 and the BC and HI loops of VP3 spanning the region around the 3-fold axis. Remarkably, the two antibodies interact with the epitope in quite distinct ways. These plateau-binding antibodies provide templates for promising candidate therapeutics.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Enterovirus Humano A/imunologia , Infecções por Enterovirus/imunologia , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/química , Anticorpos Antivirais/química , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Enterovirus Humano A/química , Enterovirus Humano A/genética , Infecções por Enterovirus/virologia , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Neutralização
8.
Proc Natl Acad Sci U S A ; 117(31): 18711-18718, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32690697

RESUMO

KREMEN1 (KRM1) has been identified as a functional receptor for Coxsackievirus A10 (CV-A10), a causative agent of hand-foot-and-mouth disease (HFMD), which poses a great threat to infants globally. However, the underlying mechanisms for the viral entry process are not well understood. Here we determined the atomic structures of different forms of CV-A10 viral particles and its complex with KRM1 in both neutral and acidic conditions. These structures reveal that KRM1 selectively binds to the mature viral particle above the canyon of the viral protein 1 (VP1) subunit and contacts across two adjacent asymmetry units. The key residues for receptor binding are conserved among most KRM1-dependent enteroviruses, suggesting a uniform mechanism for receptor binding. Moreover, the binding of KRM1 induces the release of pocket factor, a process accelerated under acidic conditions. Further biochemical studies confirmed that receptor binding at acidic pH enabled CV-A10 virion uncoating in vitro. Taken together, these findings provide high-resolution snapshots of CV-A10 entry and identify KRM1 as a two-in-one receptor for enterovirus infection.


Assuntos
Proteínas do Capsídeo , Enterovirus Humano A , Proteínas de Membrana , Internalização do Vírus , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Enterovirus Humano A/química , Enterovirus Humano A/metabolismo , Células HEK293 , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Modelos Moleculares , Vírion/química , Vírion/metabolismo , Desenvelopamento do Vírus
9.
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
10.
J Med Chem ; 63(3): 1233-1244, 2020 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-31939669

RESUMO

Human hand, foot, and mouth disease (HFMD) is a serious public health threat with high infection rates in children and infants who reside in Asia and the Pacific regions, and no effective drugs are currently available. Enterovirus 71 (EV71) and coxsackievirus A16 are the major etiological pathogens. Based on an essential hydrophobic pocket on the viral capsid protein VP1, we designed and synthesized a series of small molecular weight compounds as inhibitors of EV71. A potential drug candidate named NLD-22 exhibited excellent antiviral activity (with an EC50 of 5.056 nM and a 100% protection rate for mice at a dose of 20 mg/kg) and low toxicity. NLD-22 had a favorable pharmacokinetic profile. High-resolution cryo-electron microscopy structural analysis confirmed NLD-22 bound to the hydrophobic pocket in VP1 to block viral infection. In general, NLD-22 was indicated to be a promising potential drug candidate for the treatment of HFMD.


Assuntos
Antivirais/uso terapêutico , Enterovirus Humano A/efeitos dos fármacos , Doença de Mão, Pé e Boca/tratamento farmacológico , Imidazóis/uso terapêutico , Animais , Antivirais/síntese química , Antivirais/metabolismo , Antivirais/farmacocinética , Sítios de Ligação , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Desenho de Fármacos , Enterovirus Humano A/química , Feminino , Imidazóis/síntese química , Imidazóis/metabolismo , Imidazóis/farmacocinética , Cristalino/patologia , Masculino , Camundongos Endogâmicos BALB C , Microssomos Hepáticos/metabolismo , Ligação Proteica , Ratos Sprague-Dawley , Peixe-Zebra
11.
ACS Appl Mater Interfaces ; 12(3): 3363-3370, 2020 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-31845579

RESUMO

Bioorthogonal metabolic labeling through the endogenous cellular metabolic pathways (e.g., phospholipid and sugar) is a promising approach for effectively labeling live viruses. However, it remains a big challenge to label nonenveloped viruses due to lack of host-derived envelopes. Herein, a novel bioorthogonal labeling strategy is developed utilizing protein synthesis pathway to label and trace nonenveloped viruses. The results show that l-azidohomoalanine (Aha), an azido derivative of methionine, is more effective than azido sugars to introduce azido motifs into viral capsid proteins by substituting methionine residues during viral protein biosynthesis and assembly. The azide-modified EV71 (N3-EV71) particles are then effectively labeled with dibenzocyclooctyl (DBCO)-functionalized fluorescence probes through an in situ bioorthogonal reaction with well-preserved viral infectivity. Dual-labeled imaging clearly clarifies that EV71 virions primarily bind to scavenger receptors and are internalized through clathrin-mediated endocytosis. The viral particles are then transported into early and late endosomes where viral RNA is released in a low-pH dependent manner at about 70 min postinfection. These results first reveal viral trafficking and uncoating mechanisms, which may shed light on the pathogenesis of EV71 infection and contribute to antiviral drug discovery.


Assuntos
Enterovirus Humano A/genética , Infecções por Enterovirus/virologia , Coloração e Rotulagem/métodos , Proteínas Virais/química , Proteínas Virais/metabolismo , Animais , Endossomos/metabolismo , Endossomos/virologia , Enterovirus Humano A/química , Enterovirus Humano A/metabolismo , Infecções por Enterovirus/metabolismo , Humanos , Biossíntese de Proteínas , Proteínas Virais/genética
12.
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
13.
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
14.
Emerg Microbes Infect ; 7(1): 84, 2018 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-29743570

RESUMO

Human enteroviruses frequently cause severe diseases in children. Human enteroviruses are transmitted via the fecal-oral route and respiratory droplets, and primary replication occurs in the gastro-intestinal and respiratory tracts; however, how enteroviruses infect these sites is largely unknown. Human intestinal organoids have recently proven to be valuable tools for studying enterovirus-host interactions in the intestinal tract. In this study, we demonstrated the susceptibility of a newly developed human airway organoid model for enterovirus 71 (EV71) infection. We showed for the first time in a human physiological model that EV71 replication kinetics are strain-dependent. A glutamine at position 145 of the VP1 capsid protein was identified as a key determinant of infectivity, and residues VP1-98K and VP1-104D were identified as potential infectivity markers. The results from this study provide new insights into EV71 infectivity in the human airway epithelia and demonstrate the value of organoid technology for virus research.


Assuntos
Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Enterovirus Humano A/metabolismo , Infecções por Enterovirus/virologia , Organoides/virologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Proteínas do Capsídeo/genética , Enterovirus Humano A/química , Enterovirus Humano A/genética , Enterovirus Humano A/patogenicidade , Humanos , Cinética , Alinhamento de Sequência , Virulência , Replicação Viral
15.
Biophys J ; 114(4): 822-838, 2018 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-29490244

RESUMO

Enterovirus 71 can be a severe pathogen in small children and immunocompromised adults. Virus uncoating is a critical step in the infection of the host cell; however, the mechanisms that control this process remain poorly understood. We applied normal mode analysis and perturbation response scanning to several complexes of the virus capsid and present a coarse-graining approach to analyze the full capsid. We show that our method offers an alternative to expressing the system as a set of rigid blocks and accounts for the interconnection between nodes within each subunit and protein interfaces across the capsid. In our coarse-grained approach, the modes associated with capsid expansion are captured in the first three nondegenerate modes and correspond to the changes observed in structural studies of the virus. We show that the resolution of the analysis may be modified without losing information on the global motions leading to uncoating. Perturbation response scanning revealed that a protomer cannot serve as a functional unit to explain deformations of the capsid. Instead, we define a pentamer as the minimum functional unit to investigate changes within the capsid. From the modal analysis and perturbation response scanning, we locate a hotspot region surrounding the fivefold axis. The range of the effect of these single, hotspot residues extend to 140 Å. The perturbation of internal capsid residues in this region displayed greatest propensity to capsid expansion, thus indicating the significant role that the RNA genome may play in triggering uncoating.


Assuntos
Capsídeo/química , Capsídeo/fisiologia , Enterovirus Humano A/química , Enterovirus Humano A/fisiologia , Desenvelopamento do Vírus , Humanos , Modelos Moleculares , Conformação Molecular
16.
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
17.
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
18.
Nat Commun ; 8(1): 505, 2017 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-28894095

RESUMO

Coxsackievirus A6 (CVA6) has recently emerged as a major cause of hand, foot and mouth disease in children worldwide but no vaccine is available against CVA6 infections. Here, we demonstrate the isolation of two forms of stable CVA6 particles-procapsid and A-particle-with excellent biochemical stability and natural antigenicity to serve as vaccine candidates. Despite the presence (in A-particle) or absence (in procapsid) of capsid-RNA interactions, the two CVA6 particles have essentially identical atomic capsid structures resembling the uncoating intermediates of other enteroviruses. Our near-atomic resolution structure of CVA6 A-particle complexed with a neutralizing antibody maps an immune-dominant neutralizing epitope to the surface loops of VP1. The structure-guided cell-based inhibition studies further demonstrate that these loops could serve as excellent targets for designing anti-CVA6 vaccines.Coxsackievirus A6 (CVA6) causes hand, foot and mouth disease in children. Here the authors present the CVA6 procapsid and A-particle cryo-EM structures and identify an immune-dominant neutralizing epitope, which can be exploited for vaccine development.


Assuntos
Anticorpos Neutralizantes/química , Microscopia Crioeletrônica/métodos , Enterovirus Humano A/química , Animais , Capsídeo/química , Enterovirus Humano A/imunologia , Enterovirus Humano A/patogenicidade , Epitopos , Humanos , Imageamento Tridimensional , Camundongos , Proteínas Virais/química , Proteínas Virais/metabolismo , Vacinas Virais/química , Vacinas Virais/imunologia
19.
Microb Pathog ; 111: 292-297, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28867629

RESUMO

Hand, Foot and mouth disease (HFMD) is a common disease with high infectivity for children, and enterovirus 71 (EV71) is one of the main pathogens to cause the type of illness. Therefore, the aim of this study was to propose a rapid and effective technique for detecting EV71 directly based on the mechanism of biological intermolecular force by using atomic force microscopy (AFM). At first, we coated EV71 particles on the mica surface and made the EV71 antibodies (anti-EV71) fixed on the AFM tip by means of several chemical procedures. Then, AFM chemically modified tip was applied to measure the unbinding forces between EV71 and anti-EV71 by contact mode. Finally, by using AFM imaging calculating software, the EV71 particle size (mean±SD) was 31.36±3.87 nm (n = 200) and this result was concordance with previous literature. Besides, the force (mean±SD) between EV71 antigen and antibody complex was 336.9±64.7 pN. The force (mean±SD) between anti-EV71 and non-specific specimens was 47.1±15.1 pN and was significantly smaller (P < 0.05). Apparently, the results show that we can precisely identify EV71 infection among the samples by measuring the force magnitude and observing the occurrence of EV71/anti-EV71 unbinding events. Therefore, the combination of AFM system and the chemically modified tip has the potential to be a rapid and effective method for detecting EV71 directly.


Assuntos
Anticorpos Antivirais/análise , Antígenos Virais/análise , Técnicas Biossensoriais/métodos , Enterovirus Humano A/isolamento & purificação , Infecções por Enterovirus/virologia , Microscopia de Força Atômica/métodos , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Técnicas Biossensoriais/instrumentação , Enterovirus Humano A/química , Enterovirus Humano A/imunologia , Humanos , Microscopia de Força Atômica/instrumentação
20.
Emerg Microbes Infect ; 6(7): e65, 2017 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-28745308

RESUMO

Coxsackievirus A16 (CVA16) is one of the major pathogens responsible for hand, foot and mouth disease, which affects more than two million children in the Asian-Pacific region annually. Previous studies have shown that scavenger receptor B2 is a functional receptor for CVA16 that facilitates the uncoating process. However, it remains unclear whether other receptors are required for efficient CVA16 infection. In this study, by using a variety of assays we demonstrated that CVA16 utilizes surface heparan sulfate glycosaminoglycans as its attachment receptor. We further showed that five surface-exposed positively charged residues located in a cluster at the five-fold vertex of the virion are critical to heparan sulfate binding and cellular attachment of CVA16. Among the five residues, the arginine at position 166 (R166) of VP1 capsid protein appeared to be the most important for the interaction between CVA16 and heparan sulfate. Alanine substitution at this site (R166A) almost completely abolished heparan sulfate binding and cellular attachment of the virus. Our work achieves insight into the early events of CVA16 infection, thereby providing information that may facilitate the rational design of antiviral drugs and vaccines against CVA16 infection.


Assuntos
Enterovirus Humano A/fisiologia , Heparitina Sulfato/metabolismo , Receptores Virais/metabolismo , Ligação Viral , Alanina , Substituição de Aminoácidos , Animais , Arginina , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Chlorocebus aethiops , Enterovirus Humano A/química , Heparitina Sulfato/química , Humanos , Receptores Virais/química , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA