Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Virol ; 96(3): e0173221, 2022 02 09.
Article in English | MEDLINE | ID: mdl-34787452

ABSTRACT

The chikungunya virus has spread globally with a remarkably high attack rate. Infection causes arthralgic sequelae that can last for years. Nevertheless, there are no specific drugs or vaccines to contain the virus. Understanding the biology of the virus, such as its replication cycle, is a powerful tool to identify new drugs and comprehend virus-host interactions. Even though the chikungunya virus has been known for a long time (it was first described in 1952), many aspects of the replication cycle remain unclear. Furthermore, part of the cycle is based on observations of other alphaviruses. In this study, we used electron and scanning microscopy, as well as biological assays, to analyze and investigate the stages of the chikungunya virus replication cycle. Based on our data, we found infection cellular activities other than those usually described for the chikungunya virus replication cycle, i.e., we show particles enveloping intracellularly without budding in a membrane-delimited morphogenesis area, and we also observed virion release by membrane protrusions. Our work provides novel details regarding the biology of chikungunya virus and fills gaps in our knowledge of its replication cycle. These findings may contribute to a better understanding of virus-host interactions and support the development of antivirals. IMPORTANCE The understanding of virus biology is essential to containing virus dissemination, and exploring the virus replication cycle is a powerful tool to do this. There are many points in the biology of the chikungunya virus that need to be clarified, especially regarding its replication cycle. Our incomplete understanding of chikungunya virus infection stages is based on studies with other alphaviruses. We systematized the chikungunya virus replication cycle using microscopic imaging in the order of infection stages, as follows: entry, replication, protein synthesis, assembly/morphogenesis, and release. The imaging evidence shows novel points in the replication cycle of enveloping without budding, as well as particle release by cell membrane protrusion.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/physiology , Chikungunya virus/ultrastructure , Virus Physiological Phenomena , Virus Replication , Animals , Cells, Cultured , Chlorocebus aethiops , Cytopathogenic Effect, Viral , Vacuoles/ultrastructure , Vero Cells , Virus Release
2.
Front Immunol ; 12: 655743, 2021.
Article in English | MEDLINE | ID: mdl-33868299

ABSTRACT

Chikungunya fever is an acute infectious disease that is mediated by the mosquito-transmitted chikungunya virus (CHIKV), for which no licensed vaccines are currently available. Here, we explored several immunization protocols and investigated their immunity and protective effects in mice, with DNA- and virus-like particle (VLP)- vaccines, both alone and in combination. Both DNA and VLP vaccine candidates were developed and characterized, which express CHIKV structural genes (C-E3-E2-6K-E1). Mice were immunized twice, with different protocols, followed by immunological detection and CHIKV Ross challenge. The highest antigen-specific IgG and neutralizing activity were induced by DNA and VLP co-immunization, while the highest cellular immunity was induced by DNA vaccination alone. Although all vaccine groups could protect mice from lethal CHIKV challenge, demonstrated as reduced viral load in various tissues, without weight loss, mice co-immunized with DNA and VLP exhibited the mildest histopathological changes and lowest International Harmonization of Nomenclature and Diagnostic Criteria (INHAND) scores, in comparison to mice with either DNA or VLP vaccination alone. We concluded that co-immunization with DNA and VLP is a promising strategy to inducing better protective immunity against CHIKV infection.


Subject(s)
Chikungunya Fever/immunology , Chikungunya virus/immunology , Immunization , Vaccines, DNA/immunology , Vaccines, Virus-Like Particle/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cell Line , Chikungunya Fever/prevention & control , Chikungunya Fever/virology , Chikungunya virus/ultrastructure , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunization/methods , Mice , Neutralization Tests , Outcome Assessment, Health Care , Vaccines, DNA/administration & dosage , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/ultrastructure , Viral Load , Viral Vaccines/administration & dosage
3.
Nature ; 589(7843): 615-619, 2021 01.
Article in English | MEDLINE | ID: mdl-33328629

ABSTRACT

Positive-sense single-stranded RNA viruses, such as coronaviruses, flaviviruses and alphaviruses, carry out transcription and replication inside virus-induced membranous organelles within host cells1-7. The remodelling of the host-cell membranes for the formation of these organelles is coupled to the membrane association of viral replication complexes and to RNA synthesis. These viral niches allow for the concentration of metabolites and proteins for the synthesis of viral RNA, and prevent the detection of this RNA by the cellular innate immune system8. Here we present the cryo-electron microscopy structure of non-structural protein 1 (nsP1) of the alphavirus chikungunya virus, which is responsible for RNA capping and membrane binding of the viral replication machinery. The structure shows the enzyme in its active form, assembled in a monotopic membrane-associated dodecameric ring. The structure reveals the structural basis of the coupling between membrane binding, oligomerization and allosteric activation of the capping enzyme. The stoichiometry-with 12 active sites in a single complex-redefines viral replication complexes as RNA synthesis reactors. The ring shape of the complex implies it has a role in controlling access to the viral organelle and ensuring the exit of properly capped viral RNA. Our results provide high-resolution information about the membrane association of the replication machinery of positive-sense single-stranded RNA viruses, and open up avenues for the further characterization of viral replication on cell membranes and the generation of antiviral agents.


Subject(s)
Cell Membrane/metabolism , Chikungunya virus/growth & development , Chikungunya virus/ultrastructure , Cryoelectron Microscopy , RNA Caps/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Catalytic Domain , Cell Line , Cell Membrane/chemistry , Chikungunya virus/chemistry , Chikungunya virus/genetics , Models, Molecular , RNA Caps/chemistry , RNA, Viral/biosynthesis , RNA, Viral/chemistry , RNA, Viral/metabolism , Viral Nonstructural Proteins/ultrastructure
4.
Virus Res ; 272: 197732, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31445103

ABSTRACT

To visually examine the early phase of chikungunya virus (CHIKV) infection in target cells, we constructed a virus-like particle (VLP) in which the envelope protein E1 is fused with green fluorescent protein (GFP). This chikungunya VLP-GFP (CHIK-VLP-EGFP), purified by density gradient fractionation, was observed as 60-70 nm-dia. particles and was detected as tiny puncta of fluorescence in the cells. CHIK-VLP-EGFP showed binding properties similar to those of the wild-type viruses. Most of the fluorescence signals that had bound on Vero cells disappeared within 30 min at 37 °C, but not in the presence of anti-CHIKV neutralizing serum or an endosomal acidification inhibitor (bafilomycin A1), suggesting that the loss of fluorescence signals is due to the disassembly of the viral envelope following the internalization of CHIK-VLP-EGFP. In addition to these results, the fluorescence signals disappeared in highly susceptible Vero and U251MG cells but not in poorly susceptible A549 cells. Thus, CHIK-VLP-EGFP is a useful tool to examine the effects of the CHIKV neutralizing antibodies and antiviral compounds that are effective in the entry phase of CHIKV.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/physiology , Genes, Reporter , Green Fluorescent Proteins/genetics , Virus Replication , Animals , Cells, Cultured , Chikungunya virus/ultrastructure , Chlorocebus aethiops , Gene Expression , Genetic Vectors/genetics , Models, Biological , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Internalization
5.
Cell ; 177(7): 1725-1737.e16, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31080061

ABSTRACT

Mxra8 is a receptor for multiple arthritogenic alphaviruses that cause debilitating acute and chronic musculoskeletal disease in humans. Herein, we present a 2.2 Å resolution X-ray crystal structure of Mxra8 and 4 to 5 Å resolution cryo-electron microscopy reconstructions of Mxra8 bound to chikungunya (CHIKV) virus-like particles and infectious virus. The Mxra8 ectodomain contains two strand-swapped Ig-like domains oriented in a unique disulfide-linked head-to-head arrangement. Mxra8 binds by wedging into a cleft created by two adjacent CHIKV E2-E1 heterodimers in one trimeric spike and engaging a neighboring spike. Two binding modes are observed with the fully mature VLP, with one Mxra8 binding with unique contacts. Only the high-affinity binding mode was observed in the complex with infectious CHIKV, as viral maturation and E3 occupancy appear to influence receptor binding-site usage. Our studies provide insight into how Mxra8 binds CHIKV and creates a path for developing alphavirus entry inhibitors.


Subject(s)
Chikungunya virus/chemistry , Membrane Proteins/chemistry , Viral Envelope Proteins/chemistry , Chikungunya virus/metabolism , Chikungunya virus/ultrastructure , Cryoelectron Microscopy , HEK293 Cells , Humans , Membrane Proteins/metabolism , Protein Domains , Viral Envelope Proteins/metabolism
6.
Viruses ; 10(10)2018 10 18.
Article in English | MEDLINE | ID: mdl-30340365

ABSTRACT

The transmission cycle of chikungunya virus (CHIKV) requires that mosquito vectors get persistently infected with the virus, following its oral acqsuisition from a vertebrate host. The mosquito midgut is the initial organ that gets infected with orally acquired CHIKV. Following its replication in the midgut epithelium, the virus exits the midgut and infects secondary tissues including the salivary glands before being transmitted to another host. Here, we investigate the pattern of CHIKV dissemination from the midgut of Aedes aegypti at the ultrastructural level. Bloodmeal ingestion caused overstretching of the midgut basal lamina (BL), which was disrupted in areas adjacent to muscles surrounding the midgut as shown by scanning electron microscopy (SEM). Using both transmission electron microscopy (TEM) and focused ion beam scanning electron microscopy (FIB-SEM) to analyze midgut preparations, mature chikungunya (CHIK) virions were found accumulating at the BL and within strands of the BL at 24⁻32 h post-infectious bloodmeal (pibm). From 48 h pibm onwards, virions no longer congregated at the BL and became dispersed throughout the basal labyrinth of the epithelial cells. Ingestion of a subsequent, non-infectious bloodmeal caused mature virions to congregate again at the midgut BL. Our study suggests that CHIKV needs a single replication cycle in the midgut epithelium before mature virions directly traverse the midgut BL during a relatively narrow time window, within 48 h pibm.


Subject(s)
Aedes/virology , Chikungunya virus/ultrastructure , Mosquito Vectors/virology , Aedes/growth & development , Aedes/physiology , Animals , Basement Membrane/ultrastructure , Basement Membrane/virology , Chikungunya virus/genetics , Chikungunya virus/isolation & purification , Chikungunya virus/physiology , Female , Gastrointestinal Tract/ultrastructure , Gastrointestinal Tract/virology , Microscopy, Electron, Transmission , Mosquito Vectors/growth & development , Mosquito Vectors/physiology , Salivary Glands/ultrastructure , Salivary Glands/virology
7.
PLoS Negl Trop Dis ; 12(7): e0006693, 2018 07.
Article in English | MEDLINE | ID: mdl-30063703

ABSTRACT

BACKGROUND: Chikungunya virus (CHIKV) is the most common alphavirus infecting humans worldwide, causing acute and chronically debilitating arthralgia at a great economic expense. METHODOLOGY/PRINCIPAL FINDINGS: To facilitate our study of CHIKV, we generated a mCherry tagged replication-competent chimeric virus, CHIKV 37997-mCherry. Single particle cryoEM demonstrated icosahedral organization of the chimeric virus and the display of mCherry proteins on virus surface. CHIKV 37997-mCherry is attenuated in both IFNαR knockout and wild-type mice. Strong anti-CHIKV and anti-mCherry antibody responses were induced in CHIKV 37997-mCherry infected mice. CONCLUSIONS/SIGNIFICANCE: Our work suggests that chimeric alphaviruses displaying foreign antigen can serve as vaccines against both aphaviruses and other pathogens and diseases.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/physiology , Luminescent Proteins/metabolism , Viral Envelope Proteins/metabolism , Virus Replication , Animals , Chikungunya virus/genetics , Chikungunya virus/ultrastructure , Cryoelectron Microscopy , Female , Fluorescence , Humans , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Red Fluorescent Protein
8.
Proc Natl Acad Sci U S A ; 114(52): 13703-13707, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29203665

ABSTRACT

Cleavage of the alphavirus precursor glycoprotein p62 into the E2 and E3 glycoproteins before assembly with the nucleocapsid is the key to producing fusion-competent mature spikes on alphaviruses. Here we present a cryo-EM, 6.8-Å resolution structure of an "immature" Chikungunya virus in which the cleavage site has been mutated to inhibit proteolysis. The spikes in the immature virus have a larger radius and are less compact than in the mature virus. Furthermore, domains B on the E2 glycoproteins have less freedom of movement in the immature virus, keeping the fusion loops protected under domain B. In addition, the nucleocapsid of the immature virus is more compact than in the mature virus, protecting a conserved ribosome-binding site in the capsid protein from exposure. These differences suggest that the posttranslational processing of the spikes and nucleocapsid is necessary to produce infectious virus.


Subject(s)
Chikungunya virus/chemistry , Chikungunya virus/ultrastructure , Glycoproteins/chemistry , Viral Envelope Proteins/chemistry , Chikungunya virus/metabolism , Cryoelectron Microscopy , Glycoproteins/metabolism , Protein Domains , Protein Structure, Secondary , Viral Envelope Proteins/metabolism
9.
Science ; 358(6363): 663-667, 2017 11 03.
Article in English | MEDLINE | ID: mdl-29097548

ABSTRACT

The Rift Valley fever virus (RVFV) is transmitted by infected mosquitoes, causing severe disease in humans and livestock across Africa. We determined the x-ray structure of the RVFV class II fusion protein Gc in its postfusion form and in complex with a glycerophospholipid (GPL) bound in a conserved cavity next to the fusion loop. Site-directed mutagenesis and molecular dynamics simulations further revealed a built-in motif allowing en bloc insertion of the fusion loop into membranes, making few nonpolar side-chain interactions with the aliphatic moiety and multiple polar interactions with lipid head groups upon membrane restructuring. The GPL head-group recognition pocket is conserved in the fusion proteins of other arthropod-borne viruses, such as Zika and chikungunya viruses, which have recently caused major epidemics worldwide.


Subject(s)
Cell Membrane/virology , Glycerophospholipids/chemistry , Rift Valley fever virus/chemistry , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Chikungunya virus/chemistry , Chikungunya virus/ultrastructure , Cholesterol/chemistry , Conserved Sequence , Crystallography, X-Ray , Humans , Livestock/virology , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Conformation , Rift Valley fever virus/genetics , Rift Valley fever virus/ultrastructure , Viral Fusion Proteins/genetics , Viral Fusion Proteins/ultrastructure , Zika Virus/chemistry , Zika Virus/ultrastructure
10.
Bull World Health Organ ; 95(7): 517-525I, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28670016

ABSTRACT

OBJECTIVE: To analyse the proportions of protein identity between Zika virus and dengue, Japanese encephalitis, yellow fever, West Nile and chikungunya viruses as well as polymorphism between different Zika virus strains. METHODS: We used published protein sequences for the Zika virus and obtained protein sequences for the other viruses from the National Center for Biotechnology Information (NCBI) protein database or the NCBI virus variation resource. We used BLASTP to find regions of identity between viruses. We quantified the identity between the Zika virus and each of the other viruses, as well as within-Zika virus polymorphism for all amino acid k-mers across the proteome, with k ranging from 6 to 100. We assessed accessibility of protein fragments by calculating the solvent accessible surface area for the envelope and nonstructural-1 (NS1) proteins. FINDINGS: In total, we identified 294 Zika virus protein fragments with both low proportion of identity with other viruses and low levels of polymorphisms among Zika virus strains. The list includes protein fragments from all Zika virus proteins, except NS3. NS4A has the highest number (190 k-mers) of protein fragments on the list. CONCLUSION: We provide a candidate list of protein fragments that could be used when developing a sensitive and specific serological test to detect previous Zika virus infections.


Subject(s)
Chikungunya virus/ultrastructure , Databases, Protein , Flavivirus/ultrastructure , Animals , Dengue Virus/ultrastructure , Encephalitis Virus, Japanese/ultrastructure , Humans , Insect Vectors , Polymorphism, Genetic , West Nile virus/ultrastructure , Yellow fever virus/ultrastructure , Zika Virus/ultrastructure
11.
Nat Med ; 23(2): 192-199, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27991917

ABSTRACT

Traditionally, vaccine development involves tradeoffs between immunogenicity and safety. Live-attenuated vaccines typically offer rapid and durable immunity but have reduced safety when compared to inactivated vaccines. In contrast, the inability of inactivated vaccines to replicate enhances safety at the expense of immunogenicity, often necessitating multiple doses and boosters. To overcome these tradeoffs, we developed the insect-specific alphavirus, Eilat virus (EILV), as a vaccine platform. To address the chikungunya fever (CHIKF) pandemic, we used an EILV cDNA clone to design a chimeric virus containing the chikungunya virus (CHIKV) structural proteins. The recombinant EILV/CHIKV was structurally identical at 10 Å to wild-type CHIKV, as determined by single-particle cryo-electron microscopy, and it mimicked the early stages of CHIKV replication in vertebrate cells from attachment and entry to viral RNA delivery. Yet the recombinant virus remained completely defective for productive replication, providing a high degree of safety. A single dose of EILV/CHIKV produced in mosquito cells elicited rapid (within 4 d) and long-lasting (>290 d) neutralizing antibodies that provided complete protection in two different mouse models. In nonhuman primates, EILV/CHIKV elicited rapid and robust immunity that protected against viremia and telemetrically monitored fever. Our EILV platform represents the first structurally native application of an insect-specific virus in preclinical vaccine development and highlights the potential application of such viruses in vaccinology.


Subject(s)
Alphavirus/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Chikungunya Fever/prevention & control , Chikungunya virus/immunology , Immunogenicity, Vaccine/immunology , Insect Viruses/immunology , Viral Vaccines/immunology , Alphavirus/ultrastructure , Animals , Cell Line , Chikungunya virus/ultrastructure , Chimera , Cryoelectron Microscopy , Culicidae/virology , Female , Flow Cytometry , Macaca fascicularis , Male , Mice , Microscopy, Electron , Microscopy, Fluorescence , Vaccines, Attenuated/immunology , Vaccines, Inactivated/immunology , Virus Replication
12.
J Virol Methods ; 238: 70-76, 2016 12.
Article in English | MEDLINE | ID: mdl-27751950

ABSTRACT

Transmission electron microscopy can be used to observe the ultrastructure of viruses and other microbial pathogens with nanometer resolution. In a transmission electron microscope (TEM), the image is created by passing an electron beam through a specimen with contrast generated by electron scattering from dense elements in the specimen. Viruses do not normally contain dense elements, so a negative stain that places dense heavy metal salts around the sample is added to create a dark border. To prepare a virus sample for a negative stain transmission electron microscopy, a virus suspension is applied to a TEM grid specimen support, which is a 3mm diameter fragile specimen screen coated with a few nanometers of plastic film. Then, deionized (dI) water rinses and a negative stain solution are applied to the grid. All infectious viruses must be handled in a biosafety cabinet (BSC) and many require a biocontainment laboratory environment. Staining viruses in biosafety levels (BSL) 3 and 4 is especially challenging because the support grids are small, fragile, and easily moved by air currents. In this study we evaluated a new device for negative staining viruses called mPrep/g capsule. It is a capsule that holds up to two TEM grids during all processing steps and for storage after staining is complete. This study reports that the mPrep/g capsule method is valid and effective to negative stain virus specimens, especially in high containment laboratory environments.


Subject(s)
Containment of Biohazards , Microscopy, Electron, Transmission/methods , Negative Staining/methods , Specimen Handling/methods , Viruses/ultrastructure , Chikungunya virus/ultrastructure , Containment of Biohazards/methods , Ebolavirus/ultrastructure , Microscopy, Electron, Transmission/instrumentation , Microscopy, Electron, Transmission/standards , Viruses/isolation & purification
13.
Sci Rep ; 6: 21406, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26892458

ABSTRACT

Chikungunya virus (CHIKV) is a re-emerging arbovirus known to cause chronic myalgia and arthralgia and is now considered endemic in countries across Asia and Africa. The tissue tropism of CHIKV infection in humans remains, however, ill-defined. Due to the fact that myositis is commonly observed in most patients infected with CHIKV, we sought to develop a clinically relevant cellular model to better understand the pathogenesis of CHIKV infection. In this study, primary human skeletal muscle myoblasts (HSMM) were established as a novel human primary cell line that is highly permissive to CHIKV infection, with maximal amounts of infectious virions observed at 16 hours post infection. Genome-wide microarray profiling analyses were subsequently performed to identify and map genes that are differentially expressed upon CHIKV infection. Infection of HSMM cells with CHIKV resulted in altered expressions of host genes involved in skeletal- and muscular-associated disorders, innate immune responses, cellular growth and death, host metabolism and virus replication. Together, this study has shown the establishment of a clinically relevant primary human cell model that paves the way for the further analysis of host factors and their involvement in the various stages of CHIKV replication cycle and viral pathogenesis.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/physiology , Host-Pathogen Interactions , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/virology , Apoptosis , Chikungunya Fever/genetics , Chikungunya virus/ultrastructure , Cluster Analysis , Computational Biology/methods , Disease Resistance/genetics , Disease Resistance/immunology , Disease Susceptibility , Gene Expression Profiling , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Primary Cell Culture , Reproducibility of Results , Viral Tropism , Virus Replication
14.
J Virol ; 90(3): 1169-77, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26537684

ABSTRACT

UNLABELLED: Chikungunya virus is a positive-stranded RNA alphavirus. Structures of chikungunya virus-like particles in complex with strongly neutralizing antibody Fab fragments (8B10 and 5F10) were determined using cryo-electron microscopy and X-ray crystallography. By fitting the crystallographically determined structures of these Fab fragments into the cryo-electron density maps, we show that Fab fragments of antibody 8B10 extend radially from the viral surface and block receptor binding on the E2 glycoprotein. In contrast, Fab fragments of antibody 5F10 bind the tip of the E2 B domain and lie tangentially on the viral surface. Fab 5F10 fixes the B domain rigidly to the surface of the virus, blocking exposure of the fusion loop on glycoprotein E1 and therefore preventing the virus from becoming fusogenic. Although Fab 5F10 can neutralize the wild-type virus, it can also bind to a mutant virus without inhibiting fusion or attachment. Although the mutant virus is no longer able to propagate by extracellular budding, it can, however, enter the next cell by traveling through junctional complexes without being intercepted by a neutralizing antibody to the wild-type virus, thus clarifying how cell-to-cell transmission can occur. IMPORTANCE: Alphaviral infections are transmitted mainly by mosquitoes. Chikungunya virus (CHIKV), which belongs to the Alphavirus genus, has a wide distribution in the Old World that has expanded in recent years into the Americas. There are currently no vaccines or drugs against alphaviral infections. Therefore, a better understanding of CHIKV and its associated neutralizing antibodies will aid in the development of effective treatments.


Subject(s)
Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Chikungunya virus/immunology , Chikungunya virus/ultrastructure , Virosomes/immunology , Virosomes/ultrastructure , Chikungunya virus/chemistry , Chikungunya virus/physiology , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Immunoglobulin Fab Fragments/metabolism , Models, Molecular , Protein Binding , Virosomes/chemistry , Virus Attachment
16.
PLoS One ; 9(9): e108169, 2014.
Article in English | MEDLINE | ID: mdl-25265335

ABSTRACT

Chikungunya virus (CHIKV) is becoming a global concern due to the increasing number of outbreaks throughout the world and the absence of any CHIKV-specific vaccine or treatment. Virus-like particles (VLPs) are multistructured proteins that mimic the organization and conformation of native viruses but lack the viral genome. They are noninfectious and potentially safer vaccine candidates. Recent studies demonstrated that the yield of CHIKV VLPs varies depending on the strains, despite the 95% amino acid similarity of the strains. This might be due to the codon usage, since protein expression is differently controlled by different organisms. We optimized the region encoding CHIKV structural proteins, C-E3-E2-6k-E1, inserted it into a mammalian expression vector, and used the resulting construct to transfect 293 cells. We detected 50-kDa proteins corresponding to E1 and/or E2 in the cell lysate and the supernatant. Transmission electron microscopy revealed spherical particles with a 50- to 60-nm diameter in the supernatant that resembled the native CHIKV virions. The buoyant density of the VLPs was 1.23 g/mL, and the yield was 20 µg purified VLPs per 108 cells. The VLPs aggregated when mixed with convalescent sera from chikungunya patients, indicating that their antigenicity is similar to that of native CHIKV. Antibodies elicited with the VLPs were capable of detecting native CHIKV, demonstrating that the VLPs retain immunogenicity similar to that of the native virion. These results indicated that CHIKV VLPs are morphologically, antigenically, and immunologically similar to the native CHIKV, suggesting that they have potential for use in chikungunya vaccines.


Subject(s)
Chikungunya virus/ultrastructure , Virion/ultrastructure , Antibodies, Viral/analysis , Antigens, Viral/analysis , Base Sequence , Chikungunya virus/immunology , DNA Primers , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Microscopy, Electron, Transmission , Microscopy, Immunoelectron
17.
Elife ; 2: e00435, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23577234

ABSTRACT

A 5.3 Å resolution, cryo-electron microscopy (cryoEM) map of Chikungunya virus-like particles (VLPs) has been interpreted using the previously published crystal structure of the Chikungunya E1-E2 glycoprotein heterodimer. The heterodimer structure was divided into domains to obtain a good fit to the cryoEM density. Differences in the T = 4 quasi-equivalent heterodimer components show their adaptation to different environments. The spikes on the icosahedral 3-fold axes and those in general positions are significantly different, possibly representing different phases during initial generation of fusogenic E1 trimers. CryoEM maps of neutralizing Fab fragments complexed with VLPs have been interpreted using the crystal structures of the Fab fragments and the VLP structure. Based on these analyses the CHK-152 antibody was shown to stabilize the viral surface, hindering the exposure of the fusion-loop, likely neutralizing infection by blocking fusion. The CHK-9, m10 and m242 antibodies surround the receptor-attachment site, probably inhibiting infection by blocking cell attachment. DOI:http://dx.doi.org/10.7554/eLife.00435.001.


Subject(s)
Antibodies, Neutralizing/ultrastructure , Antibodies, Viral/ultrastructure , Chikungunya virus/ultrastructure , Vaccines, Virus-Like Particle/ultrastructure , Viral Envelope Proteins/ultrastructure , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Binding Sites , Chikungunya virus/immunology , Chikungunya virus/metabolism , Chikungunya virus/pathogenicity , Cryoelectron Microscopy , Crystallography, X-Ray , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fab Fragments/ultrastructure , Models, Molecular , Protein Binding , Protein Conformation , Vaccines, Virus-Like Particle/metabolism , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Virion/immunology , Virion/metabolism , Virion/ultrastructure , Virus Internalization
18.
Pathol Biol (Paris) ; 57(2): 127-32, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18456435

ABSTRACT

Responsible for a massive outbreak in the Indian Ocean in 2005-2006, the chikungunya virus is also reemerging in India where it has already infected over a million persons. Imported cases of the disease are reported in Asia, USA, and Europe, where a small epidemic occurred, due to transmission by local mosquitoes. Chikungunya virus is an alphavirus (Togaviridae family) that usually induces an acute illness characterized by fever, rash, and painful, incapacitating arthralgia a few days after being bitten by an infected mosquito, but recurrent joint pains are frequent. Unusual severe forms of the disease are also being reported that emphasize the importance of close monitoring of arboviruses in more fragile populations, such as the elderly and the newborns. Alphaviruses have generally been studied out of their epidemic context, leading to a large knowledge of their molecular features, and a much narrower understanding of their epidemiology and induced pathogenesis. Deciphering chikungunya virus specific molecular features as well as how the virus interacts with its vector and with its host are key to foresee, prevent and manage future epidemics, as well as prevent, treat or cure chikungunya disease.


Subject(s)
Alphavirus Infections/epidemiology , Alphavirus Infections/pathology , Chikungunya virus/physiology , Alphavirus Infections/transmission , Alphavirus Infections/veterinary , Animals , Chikungunya virus/genetics , Chikungunya virus/ultrastructure , Culicidae/virology , Disease Outbreaks , Genome, Viral , Humans , RNA, Viral/genetics , RNA, Viral/isolation & purification , Vertebrates/virology
19.
PLoS Pathog ; 3(6): e89, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17604450

ABSTRACT

An unprecedented epidemic of chikungunya virus (CHIKV) infection recently started in countries of the Indian Ocean area, causing an acute and painful syndrome with strong fever, asthenia, skin rash, polyarthritis, and lethal cases of encephalitis. The basis for chikungunya disease and the tropism of CHIKV remain unknown. Here, we describe the replication characteristics of recent clinical CHIKV strains. Human epithelial and endothelial cells, primary fibroblasts and, to a lesser extent, monocyte-derived macrophages, were susceptible to infection and allowed viral production. In contrast, CHIKV did not replicate in lymphoid and monocytoid cell lines, primary lymphocytes and monocytes, or monocyte-derived dendritic cells. CHIKV replication was cytopathic and associated with an induction of apoptosis in infected cells. Chloroquine, bafilomycin-A1, and short hairpin RNAs against dynamin-2 inhibited viral production, indicating that viral entry occurs through pH-dependent endocytosis. CHIKV was highly sensitive to the antiviral activity of type I and II interferons. These results provide a general insight into the interaction between CHIKV and its mammalian host.


Subject(s)
Alphavirus Infections/virology , Chikungunya virus/pathogenicity , Communicable Diseases, Emerging/virology , Virus Replication , Alphavirus Infections/epidemiology , Chikungunya virus/ultrastructure , Communicable Diseases, Emerging/epidemiology , Cytopathogenic Effect, Viral , Endothelial Cells/pathology , Endothelial Cells/virology , Epithelial Cells/pathology , Epithelial Cells/virology , Humans , Indian Ocean Islands
SELECTION OF CITATIONS
SEARCH DETAIL
...