Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Viruses ; 14(2)2022 02 03.
Article in English | MEDLINE | ID: mdl-35215909

ABSTRACT

Enteroviruses (EVs) represent a substantial concern to global health. Here, we present the cryo-EM structure of a non-human enterovirus, EV-F4, isolated from the Australian brushtail possum to assess the structural diversity of these picornaviruses. The capsid structure, determined to ~3 Å resolution by single particle analysis, exhibits a largely smooth surface, similar to EV-F3 (formerly BEV-2). Although the cellular receptor is not known, the absence of charged residues on the outer surface of the canyon suggest a different receptor type than for EV-F3. Density for the pocket factor is clear, with the entrance to the pocket being smaller than for other enteroviruses.


Subject(s)
Enterovirus Infections/veterinary , Enterovirus/ultrastructure , Trichosurus/virology , Animals , Australia , Capsid/metabolism , Capsid/ultrastructure , Cryoelectron Microscopy , Enterovirus/genetics , Enterovirus/isolation & purification , Enterovirus/metabolism , Enterovirus Infections/virology
2.
Cell Host Microbe ; 29(3): 448-462.e5, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33539764

ABSTRACT

Enterovirus uncoating receptors bind at the surface depression ("canyon") that encircles each capsid vertex causing the release of a host-derived lipid called "pocket factor" that is buried in a hydrophobic pocket formed by the major viral capsid protein, VP1. Coxsackievirus and adenovirus receptor (CAR) is a universal uncoating receptor of group B coxsackieviruses (CVB). Here, we present five high-resolution cryoEM structures of CVB representing different stages of virus infection. Structural comparisons show that the CAR penetrates deeper into the canyon than other uncoating receptors, leading to a cascade of events: collapse of the VP1 hydrophobic pocket, high-efficiency release of the pocket factor and viral uncoating and genome release under neutral pH, as compared with low pH. Furthermore, we identified a potent therapeutic antibody that can neutralize viral infection by interfering with virion-CAR interactions, destabilizing the capsid and inducing virion disruption. Together, these results define the structural basis of CVB cell entry and antibody neutralization.


Subject(s)
Cryoelectron Microscopy , Enterovirus/metabolism , Enterovirus/ultrastructure , Animals , Antibodies, Neutralizing , Capsid/metabolism , Capsid Proteins/ultrastructure , Enterovirus B, Human/metabolism , Enterovirus Infections/immunology , Enterovirus Infections/metabolism , Enterovirus Infections/virology , Female , Mice , Mice, Inbred BALB C , Models, Molecular , Protein Interaction Domains and Motifs , Receptors, Virus , Virion/metabolism , Virion/ultrastructure , Virus Uncoating
3.
PLoS Pathog ; 16(12): e1009146, 2020 12.
Article in English | MEDLINE | ID: mdl-33370422

ABSTRACT

Picornaviruses are important viral pathogens, but despite extensive study, the assembly process of their infectious virions is still incompletely understood, preventing the development of anti-viral strategies targeting this essential part of the life cycle. We report the identification, via RNA SELEX and bioinformatics, of multiple RNA sites across the genome of a typical enterovirus, enterovirus-E (EV-E), that each have affinity for the cognate viral capsid protein (CP) capsomer. Many of these sites are evolutionarily conserved across known EV-E variants, suggesting they play essential functional roles. Cryo-electron microscopy was used to reconstruct the EV-E particle at ~2.2 Å resolution, revealing extensive density for the genomic RNA. Relaxing the imposed symmetry within the reconstructed particles reveals multiple RNA-CP contacts, a first for any picornavirus. Conservative mutagenesis of the individual RNA-contacting amino acid side chains in EV-E, many of which are conserved across the enterovirus family including poliovirus, is lethal but does not interfere with replication or translation. Anti-EV-E and anti-poliovirus aptamers share sequence similarities with sites distributed across the poliovirus genome. These data are consistent with the hypothesis that these RNA-CP contacts are RNA Packaging Signals (PSs) that play vital roles in assembly and suggest that the RNA PSs are evolutionarily conserved between pathogens within the family, augmenting the current protein-only assembly paradigm for this family of viruses.


Subject(s)
Capsid Proteins/metabolism , Enterovirus/physiology , RNA, Viral/genetics , Virus Assembly/physiology , Amino Acid Sequence , Capsid Proteins/ultrastructure , Cryoelectron Microscopy , Enterovirus/ultrastructure , RNA, Viral/ultrastructure
4.
Cell Host Microbe ; 27(2): 249-261.e5, 2020 02 12.
Article in English | MEDLINE | ID: mdl-32027857

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing , Enterovirus A, Human/immunology , Hand, Foot and Mouth Disease/virology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/ultrastructure , Antibodies, Viral/immunology , Antibodies, Viral/ultrastructure , Capsid Proteins/immunology , Cell Line , Cryoelectron Microscopy , Enterovirus/immunology , Enterovirus/ultrastructure , Enterovirus A, Human/ultrastructure , Hand, Foot and Mouth Disease/immunology , Hand, Foot and Mouth Disease/prevention & control , Humans , Mice , Viral Vaccines/immunology , Virion/immunology
5.
PLoS Biol ; 17(6): e3000281, 2019 06.
Article in English | MEDLINE | ID: mdl-31185007

ABSTRACT

Rhino- and enteroviruses are important human pathogens, against which no antivirals are available. The best-studied inhibitors are "capsid binders" that fit in a hydrophobic pocket of the viral capsid. Employing a new class of entero-/rhinovirus inhibitors and by means of cryo-electron microscopy (EM), followed by resistance selection and reverse genetics, we discovered a hitherto unknown druggable pocket that is formed by viral proteins VP1 and VP3 and that is conserved across entero-/rhinovirus species. We propose that these inhibitors stabilize a key region of the virion, thereby preventing the conformational expansion needed for viral RNA release. A medicinal chemistry effort resulted in the identification of analogues targeting this pocket with broad-spectrum activity against Coxsackieviruses B (CVBs) and compounds with activity against enteroviruses (EV) of groups C and D, and even rhinoviruses (RV). Our findings provide novel insights in the biology of the entry of entero-/rhinoviruses and open new avenues for the design of broad-spectrum antivirals against these pathogens.


Subject(s)
Capsid Proteins/ultrastructure , Capsid/drug effects , Capsid/ultrastructure , Amino Acid Sequence/genetics , Amino Acids/genetics , Antigens, Viral , Antiviral Agents , Binding Sites , Capsid/metabolism , Capsid Proteins/metabolism , Cryoelectron Microscopy/methods , Drug Development/methods , Enterovirus/drug effects , Enterovirus/ultrastructure , Humans , Models, Molecular , Molecular Conformation , Rhinovirus/drug effects , Rhinovirus/ultrastructure , Viral Proteins/chemistry , Viral Proteins/ultrastructure , Virion/genetics
6.
PLoS One ; 14(1): e0210553, 2019.
Article in English | MEDLINE | ID: mdl-30650163

ABSTRACT

Enterovirus 71 (EV71) has emerged as a neurological virus causing life-threatening diseases in young children and infants. Although EV71 vaccines in development have presented promising results in several clinical trials, the identified key antigen for improving the broad protective efficacy of EV71 vaccines has not been well investigated. In this report, we show that different multiplicities of infection (MOIs) of the B4(E59) virus significantly affect EV71 vaccine production in a serum-free microcarrier bioreactor system. The antigens produced from high MOIs of 10-1 and 10-2 exhibited higher yield and more infectious full particle (FP) contents in the EV71 vaccines than those produced with low MOIs of 10-4 and 10-6, leading to better cross-neutralizing efficacy. The C4(E36) neutralization results showed that only antisera raised from EV71 FPs provided substantial neutralizing titers against C4(E36), whereas empty particles (EPs) of EV71 conferred no efficacy. Competitive ELISA showed that anti-FP mainly binds to FPs and that 20% of antibodies bind to EPs, whereas most anti-EP binds EPs, with only 10% antibodies binding to FPs. VP1-adsorbed anti-FP lost most of the virus neutralization efficiency, suggesting that the VP1 subunit of FP is the major immunogenic antigen determining the ability of the EV71 vaccine to elicit cross-neutralizing antibodies against EV71 virus subtypes. These findings demonstrate that the high-MOI production approach is significantly correlated with FP productivity, thereby improving the cross-neutralization efficacy of an EV71 vaccine and providing the basis for a better vaccine design against widespread EV71 viruses.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Enterovirus/genetics , Enterovirus/immunology , Virion/immunology , Animals , Antibody Specificity , Chlorocebus aethiops , Enterovirus/ultrastructure , Genotype , Humans , Mice, Inbred BALB C , Neutralization Tests , Vero Cells , Viral Vaccines/immunology , Virion/ultrastructure
7.
Nat Microbiol ; 4(3): 414-419, 2019 03.
Article in English | MEDLINE | ID: mdl-30531980

ABSTRACT

Enterovirus 71 (EV71) is a common cause of hand, foot and mouth disease-a disease endemic especially in the Asia-Pacific region1. Scavenger receptor class B member 2 (SCARB2) is the major receptor of EV71, as well as several other enteroviruses responsible for hand, foot and mouth disease, and plays a key role in cell entry2. The isolated structures of EV71 and SCARB2 are known3-6, but how they interact to initiate infection is not. Here, we report the EV71-SCARB2 complex structure determined at 3.4 Å resolution using cryo-electron microscopy. This reveals that SCARB2 binds EV71 on the southern rim of the canyon, rather than across the canyon, as predicted3,7,8. Helices 152-163 (α5) and 183-193 (α7) of SCARB2 and the viral protein 1 (VP1) GH and VP2 EF loops of EV71 dominate the interaction, suggesting an allosteric mechanism by which receptor binding might facilitate the low-pH uncoating of the virus in the endosome/lysosome. Remarkably, many residues within the binding footprint are not conserved across SCARB2-dependent enteroviruses; however, a conserved proline and glycine seem to be key residues. Thus, although the virus maintains antigenic variability even within the receptor-binding footprint, the identification of binding 'hot spots' may facilitate the design of receptor mimic therapeutics less likely to quickly generate resistance.


Subject(s)
Enterovirus/metabolism , Host Microbial Interactions , Lysosomal Membrane Proteins/chemistry , Receptors, Scavenger/chemistry , Viral Proteins/chemistry , Cryoelectron Microscopy , Enterovirus/ultrastructure , Humans , Virus Attachment
8.
Nat Commun ; 9(1): 4985, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30478256

ABSTRACT

Coxsackievirus A10 (CVA10), a human type-A Enterovirus (HEV-A), can cause diseases ranging from hand-foot-and-mouth disease to polio-myelitis-like disease. CVA10, together with some other HEV-As, utilizing the molecule KREMEN1 as an entry receptor, constitutes a KREMEN1-dependent subgroup within HEV-As. Currently, there is no vaccine or antiviral therapy available for treating diseases caused by CVA10. The atomic-resolution structure of the CVA10 virion, which is within the KREMEN1-dependent subgroup, shows significant conformational differences in the putative receptor binding sites and serotype-specific epitopes, when compared to the SCARB2-dependent subgroup of HEV-A, such as EV71, highlighting specific differences between the sub-groups. We also report two expanded structures of CVA10, an empty particle and uncoating intermediate at atomic resolution, as well as a medium-resolution genome structure reconstructed using a symmetry-mismatch method. Structural comparisons coupled with previous results, reveal an ordered signal transmission process for enterovirus uncoating, converting exo-genetic receptor-attachment inputs into a generic RNA release mechanism.


Subject(s)
Enterovirus/chemistry , Receptors, Virus/metabolism , Virus Uncoating , Animals , Capsid/metabolism , Cryoelectron Microscopy , Enterovirus/ultrastructure , Enterovirus Infections/pathology , Enterovirus Infections/virology , Genome, Viral , Mice , Models, Molecular
9.
Proc Natl Acad Sci U S A ; 114(30): 8017-8022, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28696310

ABSTRACT

Rhinoviruses (RVs) are the major causes of common colds in humans. They have a nonenveloped, icosahedral capsid surrounding a positive-strand RNA genome. Here we report that the antigen-binding (Fab) fragment of a neutralizing antibody (C5) can trigger genome release from RV-B14 to form emptied particles and neutralize virus infection. Using cryo-electron microscopy, structures of the C5 Fab in complex with the full and emptied particles have been determined at 2.3 Å and 3.0 Å resolution, respectively. Each of the 60 Fab molecules binds primarily to a region on viral protein 3 (VP3). Binding of the C5 Fabs to RV-B14 results in significant conformational changes around holes in the capsid through which the viral RNA might exit. These results are so far the highest resolution view of an antibody-virus complex and elucidate a mechanism whereby antibodies neutralize RVs and related viruses by inducing virus uncoating.


Subject(s)
Enterovirus/physiology , Virus Uncoating , Antibodies, Neutralizing/metabolism , Enterovirus/ultrastructure , HeLa Cells , Humans
10.
Respir Res ; 18(1): 84, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28472984

ABSTRACT

BACKGROUND: The Rhinovirus C (RV-C), first identified in 2006, produce high symptom burdens in children and asthmatics, however, their primary target host cell in the airways remains unknown. Our primary hypotheses were that RV-C target ciliated airway epithelial cells (AECs), and that cell specificity is determined by restricted and high expression of the only known RV-C cell-entry factor, cadherin related family member 3 (CDHR3). METHODS: RV-C15 (C15) infection in differentiated human bronchial epithelial cell (HBEC) cultures was assessed using immunofluorescent and time-lapse epifluorescent imaging. Morphology of C15-infected differentiated AECs was assessed by immunohistochemistry. RESULTS: C15 produced a scattered pattern of infection, and infected cells were shed from the epithelium. The percentage of cells infected with C15 varied from 1.4 to 14.7% depending on cell culture conditions. Infected cells had increased staining for markers of ciliated cells (acetylated-alpha-tubulin [aat], p < 0.001) but not markers of goblet cells (wheat germ agglutinin or Muc5AC, p = ns). CDHR3 expression was increased on ciliated epithelial cells, but not other epithelial cells (p < 0.01). C15 infection caused a 27.4% reduction of ciliated cells expressing CDHR3 (p < 0.01). During differentiation of AECs, CDHR3 expression progressively increased and correlated with both RV-C binding and replication. CONCLUSIONS: The RV-C only replicate in ciliated AECs in vitro, leading to infected cell shedding. CDHR3 expression positively correlates with RV-C binding and replication, and is largely confined to ciliated AECs. Our data imply that factors regulating differentiation and CDHR3 production may be important determinants of RV-C illness severity.


Subject(s)
Bronchi/cytology , Bronchi/virology , Enterovirus/physiology , Epithelial Cells/cytology , Epithelial Cells/virology , Virus Internalization , Virus Replication/physiology , Cells, Cultured , Cilia/physiology , Cilia/ultrastructure , Cilia/virology , Enterovirus/ultrastructure , Humans , Virus Shedding/physiology
11.
J Virol ; 91(8)2017 04 15.
Article in English | MEDLINE | ID: mdl-28148783

ABSTRACT

Beta-propiolactone (BPL) is an inactivating agent that is widely used in the vaccine industry. However, its effects on vaccine protein antigens and its mechanisms of action remain poorly understood. Here we present cryo-electron microscopy (cryo-EM) structures of BPL-treated coxsackievirus A16 (CVA16) mature virions and procapsids at resolutions of 3.9 Å and 6.5 Å, respectively. Notably, both particles were found to adopt an expanded conformation resembling the 135S-like uncoating intermediate, with characteristic features including an opened 2-fold channel, the externalization of the N terminus of VP1 capsid protein, and the absence of pocket factor. However, major neutralizing epitopes are very well preserved on these particles. Further biochemical analyses revealed that BPL treatment impairs the abilities of CVA16 particles to bind to the attachment receptor heparan sulfate and to a conformation-dependent monoclonal antibody in a BPL dose-dependent manner, indicating that BPL is able to modify surface-exposed amino acid residues. Taken together, our results demonstrate that BPL treatment may induce alteration of the overall structure and surface properties of a nonenveloped viral capsid, thus revealing a novel mode of action of BPL.IMPORTANCE Beta-propiolactone (BPL) is commonly used as an inactivating reagent to produce viral vaccines. It is recognized that BPL inactivates viral infectivity through modification of viral nucleic acids. However, its effect on viral proteins remains largely unknown. Here, we present high-resolution cryo-EM structures of BPL-treated coxsackievirus A16 (CVA16) mature virions and procapsids, which reveals an expanded overall conformation and characteristic features that are typical for the 135S-like uncoating intermediate. We further show that the BPL concentration affects the binding of inactivated CVA16 particles to their receptor/antibody. Thus, BPL treatment can alter the overall structure and surface properties of viral capsids, which may lead to antigenic and immunogenic variations. Our findings provide important information for future development of BPL-inactivated vaccines.


Subject(s)
Capsid/drug effects , Capsid/ultrastructure , Disinfectants/pharmacology , Enterovirus/drug effects , Enterovirus/ultrastructure , Propiolactone/pharmacology , Virus Inactivation , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Capsid/immunology , Cryoelectron Microscopy , Enterovirus/immunology
13.
Proc Natl Acad Sci U S A ; 113(32): 8997-9002, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27511920

ABSTRACT

Isolates of rhinovirus C (RV-C), a recently identified Enterovirus (EV) species, are the causative agents of severe respiratory infections among children and are linked to childhood asthma exacerbations. The RV-C have been refractory to structure determination because they are difficult to propagate in vitro. Here, we report the cryo-EM atomic structures of the full virion and native empty particle (NEP) of RV-C15a. The virus has 60 "fingers" on the virus outer surface that probably function as dominant immunogens. Because the NEPs also display these fingers, they may have utility as vaccine candidates. A sequence-conserved surface depression adjacent to each finger forms a likely binding site for the sialic acid on its receptor. The RV-C, unlike other EVs, are resistant to capsid-binding antiviral compounds because the hydrophobic pocket in VP1 is filled with multiple bulky residues. These results define potential molecular determinants for designing antiviral therapeutics and vaccines.


Subject(s)
Asthma/etiology , Enterovirus/ultrastructure , Virion/ultrastructure , Asthma/virology , Conserved Sequence , Cryoelectron Microscopy , Glycosylation , Humans , Receptors, Virus/chemistry
14.
Virus Res ; 210: 165-8, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26272672

ABSTRACT

Atypical hand, foot, and mouth disease (aHFMD) outbreaks have been frequently reported worldwide in recent years. It is believed that coxsackievirus A6 (CA6) is the major pathogen for aHFMD. Studies regarding CA6 infection are limited and the genetic mechanism for the high pathogenicity of some new CA6 variants is still unclear. Infectious clones are powerful tools for studying the genetic mechanisms of RNA viruses. In this study, we describe the construction of a full-length cDNA clone of CA6 strain TW-2007-00141. The whole genome of CA6 was amplified in a single step and ligated into a plasmid vector through an efficient cloning method, Gibson assembly. The whole genome sequence of CA6 strain TW-2007-00141 was determined and phylogenetic analysis indicated that it shared a high degree of similarity (≥94%) with the CA6 strains found in Taiwan in 2009. The infectious clone of CA6 viruses were recovered by transfection into 293FT cells and showed similar biological properties to the parental virus. Viral particles were purified by CsCl isopycnic centrifugation, and two types of viral particles were observed under transmission electron microscopy. The rescued virus showed high virulence in one-day-old suckling mice. This clone may be useful for establishing animal models for the evaluation of CA6 vaccine efficiency in future.


Subject(s)
Coxsackievirus Infections/pathology , Coxsackievirus Infections/virology , Enterovirus/pathogenicity , Animals , Animals, Newborn , Cloning, Molecular , Cluster Analysis , Disease Models, Animal , Enterovirus/genetics , Enterovirus/isolation & purification , Enterovirus/ultrastructure , Genome, Viral , Humans , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Molecular Sequence Data , Nucleic Acid Amplification Techniques , Phylogeny , Plasmids , RNA, Viral/genetics , Reverse Genetics , Sequence Analysis, DNA , Sequence Homology , Taiwan , Virion/ultrastructure , Virulence
15.
Trends Microbiol ; 23(4): 183-4, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25748799

ABSTRACT

All (+)RNA viruses replicate on distinct membranous domains; however, how they induce and maintain their unique lipid composition is largely unknown. Two recent studies reveal that enteroviruses harness the PI4P-cholestrol exchange cycle driven by OSBP1 protein and PI4 kinase(s), and that blocking the dynamic lipid flow inhibits virus replication.


Subject(s)
Enterovirus/physiology , Picornaviridae/physiology , Rhinovirus/physiology , Virus Replication , Cell Membrane/metabolism , Cholesterol/metabolism , Enterovirus/ultrastructure , Homeostasis , Lipid Metabolism , Lipids/biosynthesis , Phosphatidylinositol Phosphates/metabolism , Picornaviridae/ultrastructure , Receptors, Steroid/metabolism , Rhinovirus/ultrastructure , Virus Replication/drug effects
17.
Proc Natl Acad Sci U S A ; 111(4): 1277-81, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24474748

ABSTRACT

Development of precise protocols for accurate site-specific conjugation of monodisperse inorganic nanoparticles to biological material is one of the challenges in contemporary bionanoscience and nanomedicine. We report here a successful site-specific covalent conjugation of functionalized atomically monodisperse gold clusters with 1.5-nm metal cores to viral surfaces. Water-soluble Au102(para-mercaptobenzoic acid)44 clusters, functionalized by maleimide linkers to target cysteines of viral capsid proteins, were synthesized and conjugated to enteroviruses echovirus 1 and coxsackievirus B3. Quantitative analysis of transmission electron microscopy images and the known virus structures showed high affinity and mutual ordering of the bound gold clusters on the viral surface and a clear correlation between the clusters and the targeted cysteine sites close to the viral surface. Infectivity of the viruses was not compromised by loading of several tens of gold clusters per virus. These advances allow for future investigations of the structure-function relations of enteroviruses and enterovirus-related virus-like particles, including their entry mechanisms into cells and uncoating in cellular endosomes.


Subject(s)
Capsid , Enterovirus/physiology , Gold/chemistry , Metal Nanoparticles , Cell Line , Enterovirus/ultrastructure , Microscopy, Electron, Transmission
18.
J Struct Biol ; 185(3): 427-39, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24333899

ABSTRACT

Fitting of atomic components into electron cryo-microscopy (cryoEM) density maps is routinely used to understand the structure and function of macromolecular machines. Many fitting methods have been developed, but a standard protocol for successful fitting and assessment of fitted models has yet to be agreed upon among the experts in the field. Here, we created and tested a protocol that highlights important issues related to homology modelling, density map segmentation, rigid and flexible fitting, as well as the assessment of fits. As part of it, we use two different flexible fitting methods (Flex-EM and iMODfit) and demonstrate how combining the analysis of multiple fits and model assessment could result in an improved model. The protocol is applied to the case of the mature and empty capsids of Coxsackievirus A7 (CAV7) by flexibly fitting homology models into the corresponding cryoEM density maps at 8.2 and 6.1Å resolution. As a result, and due to the improved homology models (derived from recently solved crystal structures of a close homolog - EV71 capsid - in mature and empty forms), the final models present an improvement over previously published models. In close agreement with the capsid expansion observed in the EV71 structures, the new CAV7 models reveal that the expansion is accompanied by ∼5° counterclockwise rotation of the asymmetric unit, predominantly contributed by the capsid protein VP1. The protocol could be applied not only to viral capsids but also to many other complexes characterised by a combination of atomic structure modelling and cryoEM density fitting.


Subject(s)
Capsid/ultrastructure , Cryoelectron Microscopy/methods , Enterovirus/ultrastructure , Models, Molecular , Molecular Dynamics Simulation , Protein Conformation , Software
19.
Cell Death Dis ; 4: e803-9, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24030155

ABSTRACT

Our previous studies have shown that the inhibition of phosphatidylinositol 3-kinase (PI3K) or mTOR complex 1 can obviously promote the Coxsackievirus B3 (CVB3)-induced apoptosis of HeLa cells by regulating the expression of proapoptotic factors. To further illustrate it, Homo sapiens eIF4E-binding protein 1 (4EBP1), p70S6 kinase (p70S6K), Akt1 and Akt2 were transfected to HeLa cells, respectively. And then, we established the stable transfected cell lines. Next, after CVB3 infection, apoptosis in different groups was determined by flow cytometry; the expressions of Bim, Bax, caspase-9 and caspase-3 were examined by real-time fluorescence quantitative PCR and western blot analysis; the expression of CVB3 mRNA and viral capsid protein VP1 were also analyzed by real-time fluorescence quantitative PCR, western blot analysis and immunofluorescence, respectively. At the meantime, CVB3 replication was observed by transmission electron microscope. We found that CVB3-induced cytopathic effect and apoptosis in transfected groups were more obvious than that in controls. Unexpectedly, apoptosis rate in Akt1 group was higher than others at the early stage after viral infection and decreased with the viral-infected time increasing, which was opposite to other groups. Compared with controls, the expression of CVB3 mRNA was increased at 3, 6, 12 and 24 h postinfection (p. i.) in all groups. At the meantime, VP1 expression in 4EBP1 group was higher than control during the process of infection, while the expressions in the other groups were change dynamically. Moreover, overexpression of 4EBP1 did not affect the mRNA expressions of Bim, Bax, caspase-9 and caspase-3; while protein expressions of Bim and Bax were decreased, the self-cleavages of caspase-9 and caspase-3 were stimulated. Meanwhile, overexpression of p70S6K blocked the CVB3-induced Bim, Bax and caspase-9 expressions but promoted the self-cleavage of caspase-9. In the Akt1 group, it is noteworthy that the expressions of Bim protein were higher than controls at 3 and 6 h p. i. but lower at 24 h p. i., and the expression of Bax protein were higher at 6 and 24 h p. i., while their mRNA expressions were all decreased. Furthermore, overexpression of Akt1 stimulated the procaspase-9 and procaspase-3 expression but blocked their self-cleavages. Overexpression of Akt2, however, had little effect on Bim, Bax and caspase-3, while prevented caspase-9 from self-cleavage at the late stage of CVB3 infection. As stated above, our results demonstrated that overexpression of 4EBP1, p70S6K, Akt1 or Akt2 could promote the CVB3-induced apoptosis in diverse degree via different mediating ways in viral replication and proapoptotic factors in BcL-2 and caspase families. As 4EBP1, p70S6K and Akt are the important substrates of PI3K and mammalian target of rapamycin (mTOR), we further illustrated the role of PI3K/Akt/mTOR signaling pathway in the process of CVB3-induced apoptosis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Enterovirus/physiology , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Cycle Proteins , Cytopathogenic Effect, Viral , Enterovirus/genetics , Enterovirus/ultrastructure , Gene Expression Regulation, Viral , HeLa Cells , Humans , Membrane Proteins/metabolism , Models, Biological , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection , Viral Proteins/metabolism , bcl-2-Associated X Protein/metabolism
20.
Klin Mikrobiol Infekc Lek ; 19(3): 85-90, 2013 Sep.
Article in Czech | MEDLINE | ID: mdl-24579450

ABSTRACT

OBJECTIVE: In aseptic neuroinfections, the etiology is usually known in 50-70% of cases. The aim was to increase the rates using electron microscopy (EM) and virus isolation in cell cultures. MATERIAL AND METHODS: The prospective study included 34 patients with aseptic neuroinfections hospitalized at the Department of Infectious Diseases in Ostrava fromJuly to November 2012. EM examined cerebrospinal fluid of all patients and virus isolation in tissue cultures was performed in all cerebrospinal fluid samples. Cerebrospinal fluid was examined by polymerase chain reaction for enteroviruses in 30 patients and for herpes simplex virus 1 and 2 in 29 patients. Detection of antibodies against Borrelia burgdorferi and tick-borne encephalitis was performed in all 34 patients. RESULTS: Possible etiological agents were discovered in 31 out of 34 patients (91%), with one agent being found in 23 patients (68%) and two agents being detected in 8 patients (24%). EM revealed the agents in 26 patients and virus isolation was successful in 10 patients. EM was the only method to identify 10 agents. A group of 23 patients with a single agent detected included 14 patients with enteroviral meningitis, 4 patients with Lyme borreliosis and 4 patients with tick-borne encephalitis; EM detected an undefined virus in the last patient. An unusual group of 8 patients with two agents detected comprised 5 patients with enteroviruses and spirochetes, 2 patients with tick-borne encephalitis and undefined viruses and 1 patient with a spirochete and an undetermined virus. CONCLUSION: EM can aid in explaining the etiology of aseptic neuroinfections. However, the clinical interpretation of results remains problematic, such as detection of unknown viruses or two possible agents in 8 out of 34 patients.


Subject(s)
Borrelia burgdorferi/isolation & purification , Encephalitis, Tick-Borne/diagnosis , Enterovirus Infections/diagnosis , Lyme Disease/diagnosis , Meningitis, Viral/diagnosis , Borrelia burgdorferi/ultrastructure , Encephalitis, Tick-Borne/virology , Enterovirus/isolation & purification , Enterovirus/ultrastructure , Enterovirus Infections/virology , Humans , Lyme Disease/virology , Meningitis, Viral/virology , Microscopy, Electron , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL