Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Virol ; 96(24): e0136722, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36448797

ABSTRACT

Coxsackievirus A9 (CVA9), an enterovirus, is a common cause of pediatric aseptic meningitis and neonatal sepsis. During cell entry, enterovirus capsids undergo conformational changes leading to expansion, formation of large pores, externalization of VP1 N termini, and loss of the lipid factor from VP1. Factors such as receptor binding, heat, and acidic pH can trigger capsid expansion in some enteroviruses. Here, we show that fatty acid-free bovine serum albumin or neutral endosomal ionic conditions can independently prime CVA9 for expansion and genome release. Our results showed that CVA9 treatment with albumin or endosomal ions generated a heterogeneous population of virions, which could be physically separated by asymmetric flow field flow fractionation and computationally by cryo-electron microscopy (cryo-EM) and image processing. We report cryo-EM structures of CVA9 A-particles obtained by albumin or endosomal ion treatment and a control nonexpanded virion to 3.5, 3.3, and 2.9 Å resolution, respectively. Whereas albumin promoted stable expanded virions, the endosomal ionic concentrations induced unstable CVA9 virions which easily disintegrated, losing their genome. Loss of most of the VP4 molecules and exposure of negatively charged amino acid residues in the capsid's interior after expansion created a repulsive viral RNA-capsid interface, aiding genome release. IMPORTANCE Coxsackievirus A9 (CVA9) is a common cause of meningitis and neonatal sepsis. The triggers and mode of action of RNA release into the cell unusually do not require receptor interaction. Rather, a slow process in the endosome, independent of low pH, is required. Here, we show by biophysical separation, cryogenic electron microscopy, and image reconstruction that albumin and buffers mimicking the endosomal ion composition can separately and together expand and prime CVA9 for uncoating. Furthermore, we show in these expanded particles that VP4 is present at only ~10% of the occupancy found in the virion, VP1 is externalized, and the genome is repelled by the negatively charged, repulsive inner surface of the capsid that occurs due to the expansion. Thus, we can now link observations from cell biology of infection with the physical processes that occur in the capsid to promote genome uncoating.


Subject(s)
Cations , Enterovirus B, Human , Humans , Albumins/pharmacology , Capsid Proteins/metabolism , Cations/pharmacology , Cryoelectron Microscopy , Endosomes/metabolism , Enterovirus B, Human/drug effects , Enterovirus B, Human/genetics , Enterovirus B, Human/ultrastructure , Enterovirus Infections/pathology , Enterovirus Infections/virology , RNA/metabolism , Virion/drug effects , Virion/metabolism , Virion/ultrastructure , Genome, Viral
2.
J Virol ; 96(9): e0010522, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35442060

ABSTRACT

Coxsackie virus B5 (CVB5), a main serotype in human Enterovirus B (EVB), can cause severe viral encephalitis and aseptic meningitis among infants and children. Currently, there is no approved vaccine or antiviral therapy available against CVB5 infection. Here, we determined the atomic structures of CVB5 in three forms: mature full (F) particle (2.73 Å), intermediate altered (A) particle (2.81 Å), and procapsid empty (E) particle (2.95 Å). Structural analysis of F particle of CVB5 unveiled similar structures of "canyon," "puff," and "knob" as those other EV-Bs. We observed structural rearrangements that are alike during the transition from F to A particle, indicative of similar antigenicity, cell entry, and uncoating mechanisms shared by all EV-Bs. Further comparison of structures and sequences among all structure-known EV-Bs revealed that while the residues targeted by neutralizing MAbs are diversified and drive the evolution of EV-Bs, the relative conserved residues recognized by uncoating receptors could serve as the basis for the development of antiviral vaccines and therapeutics. IMPORTANCE As one of the main serotypes in Enterovirus B, CVB5 has been commonly reported in recent years. The atomic structures of CVB5 shown here revealed classical features found in EV-Bs and the structural rearrangement occurring during particle expansion and uncoating. Also, structure- and sequence-based comparison between CVB5 and other structure-known EV-Bs screened out key domains important for viral evolution and survival. All these provide insights into the development of vaccine and therapeutics for EV-Bs.


Subject(s)
Enterovirus B, Human , Biological Evolution , Capsid/chemistry , Coxsackievirus Infections/virology , Enterovirus B, Human/chemistry , Enterovirus B, Human/genetics , Enterovirus B, Human/ultrastructure , Humans , Protein Domains
3.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35046043

ABSTRACT

Receptor usage defines cell tropism and contributes to cell entry and infection. Coxsackievirus B (CVB) engages coxsackievirus and adenovirus receptor (CAR), and selectively utilizes the decay-accelerating factor (DAF; CD55) to infect cells. However, the differential receptor usage mechanism for CVB remains elusive. This study identified VP3-234 residues (234Q/N/V/D/E) as critical population selection determinants during CVB3 virus evolution, contributing to diverse binding affinities to CD55. Cryoelectron microscopy (cryo-EM) structures of CD55-binding/nonbinding isolates and their complexes with CD55 or CAR were obtained under both neutral and acidic conditions, and the molecular mechanism of VP3-234 residues determining CD55 affinity/specificity for naturally occurring CVB3 strains was elucidated. Structural and biochemical studies in vitro revealed the dynamic entry process of CVB3 and the function of the uncoating receptor CAR with different pH preferences. This work provides detailed insight into the molecular mechanism of CVB infection and contributes to an in-depth understanding of enterovirus attachment receptor usage.


Subject(s)
CD55 Antigens/metabolism , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/virology , Enterovirus B, Human/physiology , Host-Pathogen Interactions , Receptors, Virus/metabolism , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Enterovirus B, Human/ultrastructure , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Receptors, Virus/chemistry , Structure-Activity Relationship , Virus Attachment
4.
Viruses ; 13(9)2021 09 14.
Article in English | MEDLINE | ID: mdl-34578416

ABSTRACT

Coxsackievirus B3 (CVB3) belongs to the enteroviruses, which are a well-known cause of acute and chronic myocarditis, primarily infecting cardiac myocytes. As primary human cardiomyocytes are difficult to obtain, viral myocarditis is quite frequently studied in vitro in different non-cardiac and cardiac-like cell lines. Recently, cardiomyocytes that have been differentiated from human-induced pluripotent stem cells have been described as a new model system to study CVB3 infection. Here, we compared iCell® Cardiomyocytes with other cell lines that are commonly used to study CVB3 infection regarding their susceptibility and patterns of infection and the mode of cell death. iCell® Cardiomyocytes, HeLa cells, HL-1 cells and H9c2 cells were infected with CVB3 (Nancy strain). The viral load, CVB3 RNA genome localization, VP1 expression (including the intracellular localization), cellular morphology and the expression of cell death markers were compared. The various cell lines clearly differed in their permissiveness to CVB3 infection, patterns of infection, viral load, and mode of cell death. When studying the mode of cell death of CVB3-infected iCell® Cardiomyocytes in more detail, especially regarding the necroptosis key players RIPK1 and RIPK3, we found that RIPK1 is cleaved during CVB3 infection. iCell® Cardiomyocytes represent well the natural host of CVB3 in the heart and are thus the most appropriate model system to study molecular mechanisms of CVB3-induced myocarditis in vitro. Doubts are raised about the suitability of commonly used cell lines such as HeLa cells, HL-1 cells and H9c2 cells to evaluate molecular pathways and processes occurring in vivo in enteroviral myocarditis.


Subject(s)
Cell Line/virology , Enterovirus B, Human/physiology , Models, Biological , Myocarditis/virology , Myocytes, Cardiac/virology , Animals , Capsid Proteins/biosynthesis , Cell Death , Chlorocebus aethiops , Enterovirus B, Human/ultrastructure , HeLa Cells , Humans , In Vitro Techniques , Mice , Peptide Hydrolases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Vero Cells , Viral Load , Virus Replication
5.
Commun Biol ; 4(1): 250, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637854

ABSTRACT

Enteroviruses pose a persistent and widespread threat to human physical health, with no specific treatments available. Small molecule capsid binders have the potential to be developed as antivirals that prevent virus attachment and entry into host cells. To aid with broad-range drug development, we report here structures of coxsackieviruses B3 and B4 bound to different interprotomer-targeting capsid binders using single-particle cryo-EM. The EM density maps are beyond 3 Å resolution, providing detailed information about interactions in the ligand-binding pocket. Comparative analysis revealed the residues that form a conserved virion-stabilizing network at the interprotomer site, and showed the small molecule properties that allow anchoring in the pocket to inhibit virus disassembly.


Subject(s)
Antiviral Agents/pharmacology , Capsid Proteins/metabolism , Capsid/drug effects , Enterovirus B, Human/drug effects , Virus Assembly/drug effects , Animals , Antiviral Agents/metabolism , Binding Sites , Capsid/metabolism , Capsid/ultrastructure , Capsid Proteins/ultrastructure , Cell Line , Chlorocebus aethiops , Cryoelectron Microscopy , Drug Development , Enterovirus B, Human/metabolism , Enterovirus B, Human/ultrastructure , Ligands , Molecular Docking Simulation , Protein Conformation
6.
Nat Commun ; 11(1): 4421, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32887891

ABSTRACT

Receptor usage that determines cell tropism and drives viral classification closely correlates with the virus structure. Enterovirus B (EV-B) consists of several subgroups according to receptor usage, among which echovirus 30 (E30), a leading causative agent for human aseptic meningitis, utilizes FcRn as an uncoating receptor. However, receptors for many EVs remain unknown. Here we analyzed the atomic structures of E30 mature virion, empty- and A-particles, which reveals serotype-specific epitopes and striking conformational differences between the subgroups within EV-Bs. Of these, the VP1 BC loop markedly distinguishes E30 from other EV-Bs, indicative of a role as a structural marker for EV-B. By obtaining cryo-electron microscopy structures of E30 in complex with its receptor FcRn and CD55 and comparing its homologs, we deciphered the underlying molecular basis for receptor recognition. Together with experimentally derived viral receptor identifications, we developed a structure-based in silico algorithm to inform a rational prediction for EV receptor usage.


Subject(s)
Antigen-Antibody Complex/ultrastructure , Enterovirus B, Human/ultrastructure , Antigens, Viral/ultrastructure , CD55 Antigens/immunology , Cryoelectron Microscopy , Enterovirus B, Human/immunology , Epitopes/ultrastructure , Humans , Receptors, Fc/immunology , Virion/ultrastructure
7.
Nat Commun ; 10(1): 1138, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30850609

ABSTRACT

Viruses from the genus Enterovirus are important human pathogens. Receptor binding or exposure to acidic pH in endosomes converts enterovirus particles to an activated state that is required for genome release. However, the mechanism of enterovirus uncoating is not well understood. Here, we use cryo-electron microscopy to visualize virions of human echovirus 18 in the process of genome release. We discover that the exit of the RNA from the particle of echovirus 18 results in a loss of one, two, or three adjacent capsid-protein pentamers. The opening in the capsid, which is more than 120 Å in diameter, enables the release of the genome without the need to unwind its putative double-stranded RNA segments. We also detect capsids lacking pentamers during genome release from echovirus 30. Thus, our findings uncover a mechanism of enterovirus genome release that could become target for antiviral drugs.


Subject(s)
Capsid/ultrastructure , Enterovirus B, Human/ultrastructure , Genome, Viral , RNA, Viral/genetics , Virion/ultrastructure , Virus Uncoating/genetics , Animals , Capsid/chemistry , Chlorocebus aethiops , Cryoelectron Microscopy , Enterovirus B, Human/genetics , Epithelial Cells/ultrastructure , Epithelial Cells/virology , Humans , Hydrogen-Ion Concentration , Molecular Dynamics Simulation , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , RNA, Viral/chemistry , Virion/genetics
8.
Sci Rep ; 8(1): 33, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29311608

ABSTRACT

Enteroviruses (EVs) are common RNA viruses that cause diseases ranging from rash to paralytic poliomyelitis. For example, EV-A and EV-C viruses cause hand-foot and mouth disease and EV-B viruses cause encephalitis and myocarditis, which can result in severe morbidity and mortality. While new vaccines and treatments for EVs are under development, methods for studying and diagnosing EV infections are still limited and therefore new diagnostic tools are required. Our aim was to produce and characterize new antibodies that work in multiple applications and detect EVs in tissues and in vitro. Rats were immunized with Coxsackievirus B1 capsid protein VP1 and hybridomas were produced. Hybridoma clones were selected based on their reactivity in different immunoassays. The most promising clone, 3A6, was characterized and it performed well in multiple techniques including ELISA, immunoelectron microscopy, immunocyto- and histochemistry and in Western blotting, detecting EVs in infected cells and tissues. It recognized several EV-Bs and also the EV-C representative Poliovirus 3, making it a broad-spectrum EV specific antibody. The 3A6 rat monoclonal antibody can help to overcome some of the challenges faced with commonly used EV antibodies: it enables simultaneous use of mouse-derived antibodies in double staining and it is useful in murine models.


Subject(s)
Antibodies, Monoclonal/immunology , Capsid Proteins/immunology , Enterovirus B, Human/immunology , Animals , Antibodies, Neutralizing/immunology , Capsid Proteins/chemistry , Enterovirus B, Human/classification , Enterovirus B, Human/ultrastructure , Enterovirus Infections/immunology , Enterovirus Infections/virology , Enzyme-Linked Immunosorbent Assay , Epitopes , Humans , Immunohistochemistry , Mice , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains/immunology , Rats
9.
Nanoscale ; 7(41): 17457-67, 2015 Nov 07.
Article in English | MEDLINE | ID: mdl-26440968

ABSTRACT

Visualization and tracking of viruses without compromising their functionality is crucial in order to understand virus targeting to cells and tissues, and to understand the subsequent subcellular steps leading to virus uncoating and replication. Enteroviruses are important human pathogens causing a vast number of acute infections, and are also suggested to contribute to the development of chronic diseases like type I diabetes. Here, we demonstrate a novel method to target site-specifically the hydrophobic pocket of enteroviruses. A probe, a derivative of Pleconaril, was developed and conjugated to various labels that enabled the visualization of enteroviruses under light and electron microscopes. The probe mildly stabilized the virus particle by increasing the melting temperature by 1-3 degrees, and caused a delay in the uncoating of the virus in the cellular endosomes, but could not however inhibit the receptor binding, cellular entry or infectivity of the virus. The hydrophobic pocket binding moiety of the probe was shown to bind to echovirus 1 particle by STD and tr-NOESY NMR methods. Furthermore, binding to echovirus 1 and Coxsackievirus A9, and to a lesser extent to Coxsackie virus B3 was verified by using a gold nanocluster labeled probe by TEM analysis. Molecular modelling suggested that the probe fits the hydrophobic pockets of EV1 and CVA9, but not of CVB3 as expected, correlating well with the variations in the infectivity and stability of the virus particles. EV1 conjugated to the fluorescent dye labeled probe was efficiently internalized into the cells. The virus-fluorescent probe conjugate accumulated in the cytoplasmic endosomes and caused infection starting from 6 hours onwards. Remarkably, before and during the time of replication, the fluorescent probe was seen to leak from the virus-positive endosomes and thus separate from the capsid proteins that were left in the endosomes. These results suggest that, like the physiological hydrophobic content, the probe may be released upon virus uncoating. Our results collectively thus show that the gold and fluorescently labeled probes may be used to track and visualize the studied enteroviruses during the early phases of infection opening new avenues to follow virus uncoating in cells.


Subject(s)
Enterovirus B, Human/chemistry , Fluorescent Dyes/chemistry , Oxadiazoles/chemistry , Cell Line, Tumor , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/pathology , Enterovirus B, Human/metabolism , Enterovirus B, Human/ultrastructure , Gold/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Oxazoles
10.
J Virol ; 88(15): 8504-13, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24850734

ABSTRACT

UNLABELLED: In recent decades, Raman spectroscopy has entered the biological and medical fields. It enables nondestructive analysis of structural details at the molecular level and has been used to study viruses and their constituents. Here, we used Raman spectroscopy to study echovirus 1 (EV1), a small, nonenveloped human pathogen, in two different uncoating states induced by heat treatments. Raman signals of capsid proteins and RNA genome were observed from the intact virus, the uncoating intermediate, and disrupted virions. Transmission electron microscopy data revealed general structural changes between the studied particles. Compared to spectral characteristics of proteins in the intact virion, those of the proteins of the heat-treated particles indicated reduced α-helix content with respect to ß-sheets and coil structures. Changes observed in tryptophan and tyrosine signals suggest an increasingly hydrophilic environment around these residues. RNA signals revealed a change in the environment of the genome and in its conformation. The ionized-carbonyl vibrations showed small changes between the intact virion and the uncoating intermediate, which points to cleavage of salt bridges in the protein structure during the uncoating process. In conclusion, our data reveal distinguishable Raman signatures of the intact, intermediate, and disrupted EV1 particles. These changes indicate structural, chemical, and solute-solvent alterations in the genome and in the capsid proteins and lay the essential groundwork for investigating the uncoating of EV1 and related viruses in real time. IMPORTANCE: In order to combat virus infection, we need to know the details of virus uncoating. We present here the novel Raman signatures for opened and intact echovirus 1. This gives hope that the signatures may be used in the near future to evaluate the ambient conditions in endosomes leading to virus uncoating using, e.g., coherent anti-Stokes Raman spectroscopy (CARS) imaging. These studies will complement structural studies on virus uncoating. In addition, Raman/CARS imaging offers the possibility of making dynamic live measurements in vitro and in cells which are impossible to measure by, for example, cryo-electron tomography. Furthermore, as viral Raman spectra can be overwhelmed with various contaminants, our study is highly relevant in demonstrating the importance of sample preparation for Raman spectroscopy in the field of virology.


Subject(s)
Enterovirus B, Human/chemistry , Enterovirus B, Human/physiology , RNA, Viral/analysis , Spectrum Analysis, Raman , Viral Proteins/analysis , Virus Uncoating , Animals , Chlorocebus aethiops , Enterovirus B, Human/radiation effects , Enterovirus B, Human/ultrastructure , Hot Temperature , Microscopy, Electron, Transmission , Vero Cells , Virion/chemistry , Virion/ultrastructure
11.
J Virol ; 88(10): 5755-65, 2014 May.
Article in English | MEDLINE | ID: mdl-24623425

ABSTRACT

UNLABELLED: The coxsackievirus and adenovirus receptor (CAR) has been identified as the cellular receptor for group B coxsackieviruses, including serotype 3 (CVB3). CAR mediates infection by binding to CVB3 and catalyzing conformational changes in the virus that result in formation of the altered, noninfectious A-particle. Kinetic analyses show that the apparent first-order rate constant for the inactivation of CVB3 by soluble CAR (sCAR) at physiological temperatures varies nonlinearly with sCAR concentration. Cryo-electron microscopy (cryo-EM) reconstruction of the CVB3-CAR complex resulted in a 9.0-Šresolution map that was interpreted with the four available crystal structures of CAR, providing a consensus footprint for the receptor binding site. The analysis of the cryo-EM structure identifies important virus-receptor interactions that are conserved across picornavirus species. These conserved interactions map to variable antigenic sites or structurally conserved regions, suggesting a combination of evolutionary mechanisms for receptor site preservation. The CAR-catalyzed A-particle structure was solved to a 6.6-Šresolution and shows significant rearrangement of internal features and symmetric interactions with the RNA genome. IMPORTANCE: This report presents new information about receptor use by picornaviruses and highlights the importance of attaining at least an ∼9-Šresolution for the interpretation of cryo-EM complex maps. The analysis of receptor binding elucidates two complementary mechanisms for preservation of the low-affinity (initial) interaction of the receptor and defines the kinetics of receptor-catalyzed conformational change to the A-particle.


Subject(s)
Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Enterovirus B, Human/physiology , Enterovirus B, Human/ultrastructure , Virus Attachment , Amino Acid Sequence , Binding Sites , Cryoelectron Microscopy , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Virion/metabolism , Virion/ultrastructure , Virus Inactivation
13.
J Virol ; 86(23): 12571-81, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22973031

ABSTRACT

The coxsackievirus-adenovirus receptor (CAR) and decay-accelerating factor (DAF) have been identified as cellular receptors for coxsackievirus B3 (CVB3). The first described DAF-binding isolate was obtained during passage of the prototype strain, Nancy, on rhabdomyosarcoma (RD) cells, which express DAF but very little CAR. Here, the structure of the resulting variant, CVB3-RD, has been solved by X-ray crystallography to 2.74 Å, and a cryo-electron microscopy reconstruction of CVB3-RD complexed with DAF has been refined to 9.0 Å. This new high-resolution structure permits us to correct an error in our previous view of DAF-virus interactions, providing a new footprint of DAF that bridges two adjacent protomers. The contact sites between the virus and DAF clearly encompass CVB3-RD residues recently shown to be required for binding to DAF; these residues interact with DAF short consensus repeat 2 (SCR2), which is known to be essential for virus binding. Based on the new structure, the mode of the DAF interaction with CVB3 differs significantly from the mode reported previously for DAF binding to echoviruses.


Subject(s)
CD55 Antigens/chemistry , Enterovirus B, Human/ultrastructure , Models, Molecular , Protein Conformation , Receptors, Virus/chemistry , CD55 Antigens/metabolism , Cell Line, Tumor , Cryoelectron Microscopy , Crystallization , Crystallography, X-Ray , Humans , Receptors, Virus/metabolism
14.
J Virol ; 85(12): 5718-32, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21471247

ABSTRACT

Enteroviruses, including coxsackieviruses, exhibit significant tropism for the central nervous system, and these viruses are commonly associated with viral meningitis and encephalitis. Previously, we described the ability of coxsackievirus B3 (CVB3) to infect proliferating neuronal progenitor cells located in the neonatal subventricular zone and persist in the adult murine central nervous system (CNS). Here, we demonstrate that cultured murine neurospheres, which comprise neural stem cells and their progeny at different stages of development, were highly susceptible to CVB3 infection. Neurospheres, or neural progenitor and stem cells (NPSCs), isolated from neonatal C57BL/6 mice, supported high levels of infectious virus production and high viral protein expression levels following infection with a recombinant CVB3 expressing enhanced green fluorescent protein (eGFP) protein. Similarly, NPSCs isolated from neonatal actin-promoter-GFP transgenic mice (actin-GFP NPSCs) were highly susceptible to infection with a recombinant CVB3 expressing DsRed (Discosoma sp. red fluorescent protein). Both nestin-positive and NG2(+) progenitor cells within neurospheres were shown to preferentially express high levels of viral protein as soon as 24 h postinfection (p.i.). By day 3 p.i., viral protein expression and viral titers increased dramatically in NPSCs with resultant cytopathic effects (CPE) and eventual cell death. In contrast, reduced viral replication, lower levels of CPE, and diminished viral protein expression levels were observed in NPSCs differentiated for 5 or 16 days in the presence of fetal bovine serum (FBS). Despite the presence of CPE and high levels of cell death following early CVB3 infection, surviving neurospheres were readily observed and continued to express detectable levels of viral protein as long as 37 days after initial infection. Also, CVB3 infection of actin-GFP NPSCs increased the percentage of cells expressing neuronal class III ß-tubulin following their differentiation in the presence of FBS. These results suggest that neural stem cells may be preferentially targeted by CVB3 and that neurogenic regions of the CNS may support persistent viral replication in the surviving host. In addition, normal progenitor cell differentiation may be altered in the host following infection.


Subject(s)
Cell Differentiation , Enterovirus B, Human/physiology , Enterovirus B, Human/pathogenicity , Neural Stem Cells/virology , Animals , Cells, Cultured , Cytopathogenic Effect, Viral , Enterovirus B, Human/genetics , Enterovirus B, Human/ultrastructure , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Neural Stem Cells/cytology , Neural Stem Cells/ultrastructure , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Red Fluorescent Protein
15.
J Virol ; 83(9): 4216-26, 2009 May.
Article in English | MEDLINE | ID: mdl-19225001

ABSTRACT

We performed a comparative analysis of the internalization mechanisms used by three viruses causing important vesicular diseases in animals. Swine vesicular disease virus (SVDV) internalization was inhibited by treatments that affected clathrin-mediated endocytosis and required traffic through an endosomal compartment. SVDV particles were found in clathrin-coated pits by electron microscopy and colocalized with markers of early endosomes by confocal microscopy. SVDV infectivity was significantly inhibited by drugs that raised endosomal pH. When compared to foot-and-mouth disease virus (FMDV), which uses clathrin-mediated endocytosis, the early step of SVDV was dependent on the integrity of microtubules. SVDV-productive endocytosis was more sensitive to plasma membrane cholesterol extraction than that of FMDV, and differential cell signaling requirements for virus infection were also found. Vesicular stomatitis virus, a model virus internalized by clathrin-mediated endocytosis, was included as a control of drug treatments. These results suggest that different clathrin-mediated routes are responsible for the internalization of these viruses.


Subject(s)
Enterovirus B, Human/metabolism , Foot-and-Mouth Disease Virus/metabolism , Virus Internalization , Animals , Cell Line , Cholesterol/metabolism , Clathrin/metabolism , Cricetinae , Cytoskeleton/metabolism , Endocytosis , Enterovirus B, Human/ultrastructure , Foot-and-Mouth Disease Virus/ultrastructure , Microscopy, Electron, Transmission , Signal Transduction , Vesiculovirus/metabolism
16.
Curr Top Microbiol Immunol ; 323: 67-87, 2008.
Article in English | MEDLINE | ID: mdl-18357766

ABSTRACT

The coxsackievirus and adenovirus receptor (CAR) has been studied extensively since its identification and isolation in 1997. The CAR is an immunoglobulin superfamily protein with two extracellular Ig-like domains, a single membrane-spanning sequence, and a significant cytoplasmic domain. It is structurally and functionally similar to the junctional adhesion molecules. The amino terminal domain, distal from the membrane, has been structurally characterized alone, bound to the adenovirus fiber knob, and, in full-length CAR, docked in the canyon structure of the coxsackievirus capsid. Although the past decade has produced a burst of new knowledge about CAR, significant questions concerning its function in normal physiology and coxsackievirus-related pathology remain unanswered.


Subject(s)
Coxsackievirus Infections/virology , Enterovirus B, Human/metabolism , Membrane Proteins/metabolism , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Animals , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Enterovirus B, Human/ultrastructure , Humans , Protein Conformation , Protein Structure, Tertiary , Receptors, Virus/genetics
17.
J Virol ; 81(23): 12927-35, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17804498

ABSTRACT

Many entero-, parecho-, and rhinoviruses use immunoglobulin (Ig)-like receptors that bind into the viral canyon and are required to initiate viral uncoating during infection. However, some of these viruses use an alternative or additional receptor that binds outside the canyon. Both the coxsackievirus-adenovirus receptor (CAR), an Ig-like molecule that binds into the viral canyon, and decay-accelerating factor (DAF) have been identified as cellular receptors for coxsackievirus B3 (CVB3). A cryoelectron microscopy reconstruction of a variant of CVB3 complexed with DAF shows full occupancy of the DAF receptor in each of 60 binding sites. The DAF molecule bridges the canyon, blocking the CAR binding site and causing the two receptors to compete with one another. The binding site of DAF on CVB3 differs from the binding site of DAF on the surface of echoviruses, suggesting independent evolutionary processes.


Subject(s)
CD55 Antigens/metabolism , Enterovirus B, Human/metabolism , Receptors, Virus/metabolism , Virus Attachment , CD55 Antigens/chemistry , CD55 Antigens/ultrastructure , Cryoelectron Microscopy , Enterovirus B, Human/chemistry , Enterovirus B, Human/ultrastructure , Models, Molecular , Protein Binding , Receptors, Virus/chemistry , Receptors, Virus/ultrastructure
18.
J Biol Chem ; 279(12): 11632-8, 2004 Mar 19.
Article in English | MEDLINE | ID: mdl-14701832

ABSTRACT

Integrins are cell surface receptors for several microbial pathogens including echovirus 1 (EV1), a picornavirus. Cryo-electron microscopy revealed that the functional domain (alpha(2)I) of human alpha(2)beta(1) integrin binds to a surface depression on the EV1 capsid. This three-dimensional structure of EV1 bound to alpha(2)I domain provides the first structural details of an integrin interacting with a picornavirus. The model indicates that alpha(2)beta(1) integrin cannot simultaneously bind both EV1 and the physiological ligand collagen. Compared with collagen binding to the alpha(2)I domain, the virus binds with a 10-fold higher affinity but in vitro uncoating of EV1 was not observed as a result of attachment of alpha(2)I. A molecular model, constructed on the basis of the EV1-integrin complex, shows that multiple alpha(2)beta(1) heterodimers can bind at adjacent sites around the virus 5-fold symmetry axes without steric hindrance. In agreement with this, virus attachment to alpha(2)beta(1) integrin on the cell surface was found to result in integrin clustering, which can give rise to signaling and facilitate the initiation of the viral entry process that takes place via caveolae-mediated endocytosis.


Subject(s)
Enterovirus B, Human/physiology , Integrin alpha2/physiology , Capsid/metabolism , Cell Membrane/metabolism , Cryoelectron Microscopy , Enterovirus B, Human/ultrastructure , Enterovirus Infections/physiopathology , Humans , Integrin alpha2/chemistry , Integrin alpha2/metabolism , Microscopy, Electron, Scanning , Protein Conformation , Receptors, Virus/physiology
19.
J Biol Chem ; 279(9): 8325-32, 2004 Feb 27.
Article in English | MEDLINE | ID: mdl-14634014

ABSTRACT

Echovirus type 12 (EV12), an Enterovirus of the Picornaviridae family, uses the complement regulator decay-accelerating factor (DAF, CD55) as a cellular receptor. We have calculated a three-dimensional reconstruction of EV12 bound to a fragment of DAF consisting of short consensus repeat domains 3 and 4 from cryo-negative stain electron microscopy data (EMD code 1057). This shows that, as for an earlier reconstruction of the related echovirus type 7 bound to DAF, attachment is not within the viral canyon but occurs close to the 2-fold symmetry axes. Despite this general similarity our reconstruction reveals a receptor interaction that is quite different from that observed for EV7. Fitting of the crystallographic co-ordinates for DAF(34) and EV11 into the reconstruction shows a close agreement between the crystal structure of the receptor fragment and the density for the virus-bound receptor, allowing unambiguous positioning of the receptor with respect to the virion (PDB code 1UPN). Our finding that the mode of virus-receptor interaction in EV12 is distinct from that seen for EV7 raises interesting questions regarding the evolution and biological significance of the DAF binding phenotype in these viruses.


Subject(s)
CD55 Antigens/metabolism , CD55 Antigens/ultrastructure , Enterovirus B, Human/metabolism , Enterovirus B, Human/ultrastructure , Binding Sites , Computer Simulation , Crystallization , Crystallography, X-Ray , Image Processing, Computer-Assisted , Microscopy, Electron , Models, Molecular
20.
J Virol ; 77(9): 5475-86, 2003 May.
Article in English | MEDLINE | ID: mdl-12692248

ABSTRACT

Swine vesicular disease virus (SVDV) is an Enterovirus of the family Picornaviridae that causes symptoms indistinguishable from those of foot-and-mouth disease virus. Phylogenetic studies suggest that it is a recently evolved genetic sublineage of the important human pathogen coxsackievirus B5 (CBV5), and in agreement with this, it has been shown to utilize the coxsackie and adenovirus receptor (CAR) for cell entry. The 3.0-A crystal structure of strain UK/27/72 SVDV (highly virulent) reveals the expected similarity in core structure to those of other picornaviruses, showing most similarity to the closest available structure to CBV5, that of coxsackievirus B3 (CBV3). Features that help to cement together and rigidify the protein subunits are extended in this virus, perhaps explaining its extreme tolerance of environmental factors. Using the large number of capsid sequences available for both SVDV and CBV5, we have mapped the amino acid substitutions that may have occurred during the supposed adaptation of SVDV to a new host onto the structure of SVDV and a model of the SVDV/CAR complex generated by reference to the cryo-electron microscopy-visualized complex of CBV3 and CAR. The changes fall into three clusters as follows: one lines the fivefold pore, a second maps to the CAR-binding site and partially overlaps the site for decay accelerating factor (DAF) to bind to echovirus 7 (ECHO7), and the third lies close to the fivefold axis, where the low-density lipoprotein receptor binds to the minor group of rhinoviruses. Later changes in SVDV (post-1971) map to the first two clusters and may, by optimizing recognition of a pig CAR and/or DAF homologue, have improved the adaptation of the virus to pigs.


Subject(s)
Adaptation, Physiological , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Capsid Proteins/chemistry , Capsid Proteins/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Cryoelectron Microscopy , Crystallography, X-Ray , Enterovirus B, Human/chemistry , Enterovirus B, Human/pathogenicity , Enterovirus B, Human/ultrastructure , Humans , Models, Molecular , Molecular Sequence Data , Receptors, Virus/metabolism , Swine , Swine Vesicular Disease/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...