Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 288
Filter
1.
J Virol ; 95(14): e0043321, 2021 06 24.
Article in English | MEDLINE | ID: mdl-33952645

ABSTRACT

Negeviruses are a group of insect-specific viruses (ISVs) that have been found in many arthropods. Their presence in important vector species led us to examine their interactions with arboviruses during coinfections. Wild-type negeviruses reduced the replication of several alphaviruses during coinfections in mosquito cells. Negev virus (NEGV) isolates were also used to express green fluorescent protein (GFP) and anti-chikungunya virus (CHIKV) antibody fragments during coinfections with CHIKV. NEGV expressing anti-CHIKV antibody fragments was able to further reduce replication of CHIKV during coinfections, while reductions of CHIKV with NEGV expressing GFP were similar to titers with wild-type NEGV alone. These results are the first to show that negeviruses induce superinfection exclusion of arboviruses and to demonstrate a novel approach to deliver antiviral antibody fragments with paratransgenic ISVs. The ability to inhibit arbovirus replication and express exogenous proteins in mosquito cells makes negeviruses a promising platform for control of arthropod-borne pathogens. IMPORTANCE Negeviruses are a group of insect-specific viruses (ISVs), viruses known to infect only insects. They have been discovered over a wide geographical and species range. Their ability to infect mosquito species that transmit dangerous arboviruses makes negeviruses a candidate for a pathogen control platform. Coinfections of mosquito cells with a negevirus and an alphavirus demonstrated that negeviruses can inhibit the replication of alphaviruses. Additionally, modifying Negev virus (NEGV) to express a fragment of an anti-CHIKV antibody further reduced the replication of CHIKV in coinfected cells. This is the first evidence to demonstrate that negeviruses can inhibit the replication of important arboviruses in mosquito cells. The ability of a modified NEGV to drive the expression of antiviral proteins also highlights a method for negeviruses to target specific pathogens and limit the incidence of vector-borne diseases.


Subject(s)
Alphavirus/physiology , Insect Viruses/physiology , Virus Replication , Aedes/virology , Animals , Cells, Cultured , Chikungunya virus/physiology , Chlorocebus aethiops , Culex/virology , O'nyong-nyong Virus/physiology , Semliki forest virus/physiology , Vero Cells
2.
PLoS Pathog ; 17(5): e1009603, 2021 05.
Article in English | MEDLINE | ID: mdl-34019569

ABSTRACT

The positive-sense, single-stranded RNA alphaviruses pose a potential epidemic threat. Understanding the complex interactions between the viral and the host cell proteins is crucial for elucidating the mechanisms underlying successful virus replication strategies and for developing specific antiviral interventions. Here we present the first comprehensive protein-protein interaction map between the proteins of Semliki Forest Virus (SFV), a mosquito-borne member of the alphaviruses, and host cell proteins. Among the many identified cellular interactors of SFV proteins, the enrichment of factors involved in translation and nonsense-mediated mRNA decay (NMD) was striking, reflecting the virus' hijacking of the translation machinery and indicating viral countermeasures for escaping NMD by inhibiting NMD at later time points during the infectious cycle. In addition to observing a general inhibition of NMD about 4 hours post infection, we also demonstrate that transient expression of the SFV capsid protein is sufficient to inhibit NMD in cells, suggesting that the massive production of capsid protein during the SFV reproduction cycle is responsible for NMD inhibition.


Subject(s)
Alphavirus Infections/virology , Capsid Proteins/metabolism , Host-Pathogen Interactions , Nonsense Mediated mRNA Decay/genetics , Semliki forest virus/physiology , Capsid Proteins/genetics , HeLa Cells , Humans , Semliki forest virus/genetics , Virus Replication
3.
Arch Virol ; 166(4): 1015-1033, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33582855

ABSTRACT

Multiple sclerosis (MS) is a common inflammatory demyelinating disease of the central nervous system. Although the etiology of MS is unknown, genetics and environmental factors, such as infections, play a role. Viral infections of mice have been used as model systems to study this demyelinating disease of humans. Three viruses that have long been studied in this capacity are Theiler's murine encephalomyelitis virus, mouse hepatitis virus, and Semliki Forest virus. This review describes the viruses themselves, the infection process, the disease caused by infection and its accompanying pathology, and the model systems and their usefulness in studying MS.


Subject(s)
Disease Models, Animal , Multiple Sclerosis/pathology , Multiple Sclerosis/virology , RNA Virus Infections/pathology , RNA Virus Infections/virology , Animals , Central Nervous System/pathology , Central Nervous System/physiology , Central Nervous System/virology , Humans , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Murine hepatitis virus/pathogenicity , Murine hepatitis virus/physiology , RNA Virus Infections/immunology , RNA Virus Infections/physiopathology , Semliki forest virus/pathogenicity , Semliki forest virus/physiology , Theilovirus/pathogenicity , Theilovirus/physiology
4.
Science ; 371(6528)2021 01 29.
Article in English | MEDLINE | ID: mdl-33243852

ABSTRACT

Inflammasomes function as intracellular sensors of pathogen infection or cellular perturbation and thereby play a central role in numerous diseases. Given the high abundance of NLRP1 in epithelial barrier tissues, we screened a diverse panel of viruses for inflammasome activation in keratinocytes. We identified Semliki Forest virus (SFV), a positive-strand RNA virus, as a potent activator of human but not murine NLRP1B. SFV replication and the associated formation of double-stranded (ds) RNA was required to engage the NLRP1 inflammasome. Moreover, delivery of long dsRNA was sufficient to trigger activation. Biochemical studies revealed that NLRP1 binds dsRNA through its leucine-rich repeat domain, resulting in its NACHT domain gaining adenosine triphosphatase activity. Altogether, these results establish human NLRP1 as a direct sensor for dsRNA and thus RNA virus infection.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Host-Pathogen Interactions/immunology , Immunity, Innate , Inflammasomes/immunology , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Alphavirus Infections/immunology , Animals , Apoptosis Regulatory Proteins/chemistry , HEK293 Cells , Humans , Hydrolysis , Inflammasomes/metabolism , Keratinocytes/immunology , Keratinocytes/metabolism , Keratinocytes/virology , Mice , Mice, Transgenic , NLR Proteins , Protein Domains , Semliki forest virus/immunology , Semliki forest virus/physiology , Virus Replication
5.
Cell Death Dis ; 11(1): 48, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31969562

ABSTRACT

Oncolytic viruses have the potential to induce immunogenic cell death (ICD) that may provoke potent and long-lasting anti-cancer immunity. Here we aimed to characterize the ICD-inducing ability of wild-type Adenovirus (Ad), Semliki Forest virus (SFV) and Vaccinia virus (VV). We did so by investigating the cell death and immune-activating properties of virus-killed tumor cells. Ad-infection of tumor cells primarily activates autophagy, but also activate events of necroptotic and pyroptotic cell death. SFV infection on the other hand primarily activates immunogenic apoptosis while VV activates necroptosis. All viruses mediated lysis of tumor cells leading to the release of danger-associated molecular patterns, triggering of phagocytosis and maturation of dendritic cells (DCs). However, only SFV-infected tumor cells triggered significant T helper type 1 (Th1)-cytokine release by DCs and induced antigen-specific T-cell activation. Our results elucidate cell death processes activated upon Ad, SFV, and VV infection and their potential to induce T cell-mediated anti-tumor immune responses. This knowledge provides important insight for the choice and design of therapeutically successful virus-based immunotherapies.


Subject(s)
Adenoviridae/physiology , Immunogenic Cell Death , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Oncolytic Virotherapy , Semliki forest virus/physiology , Vaccinia virus/physiology , Animals , Apoptosis , Autophagy , Cell Line , Cell Survival , Dendritic Cells/metabolism , Epitopes/immunology , Humans , Inflammasomes/metabolism , Necroptosis , Neoplasms/pathology , T-Lymphocytes/immunology
6.
PLoS Pathog ; 15(6): e1007842, 2019 06.
Article in English | MEDLINE | ID: mdl-31199850

ABSTRACT

G3BP-1 and -2 (hereafter referred to as G3BP) are multifunctional RNA-binding proteins involved in stress granule (SG) assembly. Viruses from diverse families target G3BP for recruitment to replication or transcription complexes in order to block SG assembly but also to acquire pro-viral effects via other unknown functions of G3BP. The Old World alphaviruses, including Semliki Forest virus (SFV) and chikungunya virus (CHIKV) recruit G3BP into viral replication complexes, via an interaction between FGDF motifs in the C-terminus of the viral non-structural protein 3 (nsP3) and the NTF2-like domain of G3BP. To study potential proviral roles of G3BP, we used human osteosarcoma (U2OS) cell lines lacking endogenous G3BP generated using CRISPR-Cas9 and reconstituted with a panel of G3BP1 mutants and truncation variants. While SFV replicated with varying efficiency in all cell lines, CHIKV could only replicate in cells expressing G3BP1 variants containing both the NTF2-like and the RGG domains. The ability of SFV to replicate in the absence of G3BP allowed us to study effects of different domains of the protein. We used immunoprecipitation to demonstrate that that both NTF2-like and RGG domains are necessary for the formation a complex between nsP3, G3BP1 and the 40S ribosomal subunit. Electron microscopy of SFV-infected cells revealed that formation of nsP3:G3BP1 complexes via the NTF2-like domain was necessary for clustering of cytopathic vacuoles (CPVs) and that the presence of the RGG domain was necessary for accumulation of electron dense material containing G3BP1 and nsP3 surrounding the CPV clusters. Clustered CPVs also exhibited localised high levels of translation of viral mRNAs as detected by ribopuromycylation staining. These data confirm that G3BP is a ribosomal binding protein and reveal that alphaviral nsP3 uses G3BP to concentrate viral replication complexes and to recruit the translation initiation machinery, promoting the efficient translation of viral mRNAs.


Subject(s)
Carrier Proteins/metabolism , Chikungunya Fever/metabolism , Chikungunya virus/physiology , DNA Helicases/metabolism , Peptide Chain Initiation, Translational , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Semliki forest virus/physiology , Virus Replication , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Chikungunya Fever/genetics , Chikungunya Fever/pathology , Cricetinae , DNA Helicases/genetics , Humans , Poly-ADP-Ribose Binding Proteins/genetics , Protein Domains , RNA Helicases/genetics , RNA Recognition Motif Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Binding Proteins , Ribosome Subunits, Small, Eukaryotic/genetics , Ribosome Subunits, Small, Eukaryotic/metabolism
7.
J Virol ; 93(16)2019 08 15.
Article in English | MEDLINE | ID: mdl-31142668

ABSTRACT

Dynamin GTPases, best known for their role in membrane fission of endocytic vesicles, provide a target for viruses to be exploited during endocytic uptake. Recently, we found that entry of herpes simplex virus 1 (HSV-1) into skin cells depends on dynamin, although our results supported that viral internalization occurs via both direct fusion with the plasma membrane and via endocytic pathways. To further explore the role of dynamin for efficient HSV-1 entry, we utilized conditional dynamin 1 and dynamin 2 double-knockout (DKO) fibroblasts as an experimental tool. Strikingly, HSV-1 entered control and DKO fibroblasts with comparable efficiencies. For comparison, we infected DKO cells with Semliki Forest virus, which is known to adopt clathrin-mediated endocytosis as its internalization pathway, and observed efficient virus entry. These results support the notion that the DKO cells provide alternative pathways for viral uptake. Treatment of cells with the dynamin inhibitor dynasore confirmed that HSV-1 entry depended on dynamin in the control fibroblasts. As expected, dynasore did not interfere with viral entry into DKO cells. Electron microscopy of HSV-1-infected cells suggests viral entry after fusion with the plasma membrane and by endocytosis in both dynamin-expressing and dynamin-deficient cells. Infection at low temperatures where endocytosis is blocked still resulted in HSV-1 entry, although at a reduced level, which suggests that nonendocytic pathways contribute to successful entry. Overall, our results strengthen the impact of dynamin for HSV-1 entry, as only cells that adapt to the lack of dynamin allow dynamin-independent entry.IMPORTANCE The human pathogen herpes simplex virus 1 (HSV-1) can adapt to a variety of cellular pathways to enter cells. In general, HSV-1 is internalized by fusion of its envelope with the plasma membrane or by endocytic pathways, which reflects the high adaptation to differences in its target cells. The challenges are to distinguish whether multiple or only one of these internalization pathways leads to successful entry and, furthermore, to identify the mode of viral uptake. In this study, we focused on dynamin, which promotes endocytic vesicle fission, and explored how the presence and absence of dynamin can influence viral entry. Our results support the idea that HSV-1 entry into mouse embryonic fibroblasts depends on dynamin; however, depletion of dynamin still allows efficient viral entry, suggesting that alternative pathways present upon dynamin depletion can accomplish viral internalization.


Subject(s)
Dynamin II/genetics , Dynamin I/genetics , Fibroblasts/metabolism , Fibroblasts/virology , Herpes Simplex/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Virus Internalization , Animals , Cells, Cultured , Endocytosis , Gene Knockdown Techniques , Genetic Predisposition to Disease , Host-Pathogen Interactions/genetics , Humans , Mice , Semliki forest virus/physiology
8.
Viruses ; 10(5)2018 05 18.
Article in English | MEDLINE | ID: mdl-29783708

ABSTRACT

Alphaviruses are mosquito-transmitted RNA viruses which generally cause acute disease including mild febrile illness, rash, arthralgia, myalgia and more severely, encephalitis. In the mouse, peripheral infection with Semliki Forest virus (SFV) results in encephalitis. With non-virulent strains, infectious virus is detectable in the brain, by standard infectivity assays, for around ten days. As we have shown previously, in severe combined immunodeficient (SCID) mice, infectious virus is detectable for months in the brain. Here we show that in MHC-II-/- mice, with no functional CD4 T-cells, infectious virus is also detectable in the brain for long periods. In contrast, in the brains of CD8-/- mice, virus RNA persists but infectious virus is not detectable. In SCID mice infected with SFV, repeated intraperitoneal administration of anti-SFV immune serum rapidly reduced the titer of infectious virus in the brain to undetectable, however virus RNA persisted. Repeated intraperitoneal passive transfer of immune serum resulted in maintenance of brain virus RNA, with no detectable infectious virus, for several weeks. When passive antibody transfer was stopped, antibody levels declined and infectious virus was again detectable in the brain. In aged immunocompetent mice, previously infected with SFV, immunosuppression of antibody responses many months after initial infection also resulted in renewed ability to detect infectious virus in the brain. In summary, antiviral antibodies control and determine whether infectious virus is detectable in the brain but immune responses cannot clear this infection from the brain. Functional virus RNA capable of generating infectious virus persists and if antibody levels decline, infectious virus is again detectable.


Subject(s)
Brain/virology , Encephalitis, Viral/immunology , Encephalitis, Viral/virology , RNA, Viral/immunology , Semliki forest virus/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Brain/immunology , Cell Line , Cricetinae , Cyclophosphamide/pharmacology , Immune Sera/immunology , Immune Sera/isolation & purification , Immunity, Cellular/drug effects , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred Strains , Mice, SCID , RNA, Viral/genetics , Semliki forest virus/growth & development , Semliki forest virus/physiology , T-Lymphocytes/immunology , Viral Plaque Assay
9.
Viruses ; 10(5)2018 04 27.
Article in English | MEDLINE | ID: mdl-29702546

ABSTRACT

Infection by Chikungunya virus (CHIKV) of the Old World alphaviruses (family Togaviridae) in humans can cause arthritis and arthralgia. The virus encodes four non-structural proteins (nsP) (nsP1, nsp2, nsP3 and nsP4) that act as subunits of the virus replicase. These proteins also interact with numerous host proteins and some crucial interactions are mediated by the unstructured C-terminal hypervariable domain (HVD) of nsP3. In this study, a human cell line expressing EGFP tagged with CHIKV nsP3 HVD was established. Using quantitative proteomics, it was found that CHIKV nsP3 HVD can bind cytoskeletal proteins, including CD2AP, SH3KBP1, CAPZA1, CAPZA2 and CAPZB. The interaction with CD2AP was found to be most evident; its binding site was mapped to the second SH3 ligand-like element in nsP3 HVD. Further assessment indicated that CD2AP can bind to nsP3 HVDs of many different New and Old World alphaviruses. Mutation of the short binding element hampered the ability of the virus to establish infection. The mutation also abolished ability of CD2AP to co-localise with nsP3 and replication complexes of CHIKV; the same was observed for Semliki Forest virus (SFV) harbouring a similar mutation. Similar to CD2AP, its homolog SH3KBP1 also bound the identified motif in CHIKV and SFV nsP3.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alphavirus/physiology , Amino Acid Motifs/genetics , Binding Sites/genetics , Cytoskeletal Proteins/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Alphavirus/genetics , Animals , CapZ Actin Capping Protein/metabolism , Cell Line , Chikungunya virus/genetics , Chikungunya virus/physiology , Cricetinae , Host-Pathogen Interactions , Humans , Mutation , Protein Binding , Semliki forest virus/genetics , Semliki forest virus/physiology , Viral Nonstructural Proteins/genetics , Virus Replication/genetics
10.
J Virol ; 92(14)2018 07 15.
Article in English | MEDLINE | ID: mdl-29695431

ABSTRACT

Polyprotein processing has an important regulatory role in the life cycle of positive-strand RNA viruses. In the case of alphaviruses, sequential cleavage of the nonstructural polyprotein (ns-polyprotein) at three sites eventually yields four mature nonstructural proteins (nsPs) that continue working in complex to replicate viral genomic RNA and transcribe subgenomic RNA. Recognition of cleavage sites by viral nsP2 protease is guided by short sequences upstream of the scissile bond and, more importantly, by the spatial organization of the replication complex. In this study, we analyzed the consequences of the artificially accelerated processing of the Semliki Forest virus ns-polyprotein. It was found that in mammalian cells, not only the order but also the correct timing of the cleavage events is essential for the success of viral replication. Analysis of the effects of compensatory mutations in rescued viruses as well as in vitro translation and trans-replicase assays corroborated our findings and revealed the importance of the V515 residue in nsP2 for recognizing the P4 position in the nsP1/nsP2 cleavage site. We also extended our conclusions to Sindbis virus by analyzing the properties of the hyperprocessive variant carrying the N614D mutation in nsP2. We conclude that the sequence of the nsP1/nsP2 site in alphaviruses is under selective pressure to avoid the presence of sequences that are recognized too efficiently and would otherwise lead to premature cleavage at this site before completion of essential tasks of RNA synthesis or virus-induced replication complex formation. Even subtle changes in the ns-polyprotein processing pattern appear to lead to virus attenuation.IMPORTANCE The polyprotein expression strategy is a cornerstone of alphavirus replication. Three sites within the ns-polyprotein are recognized by the viral nsP2 protease and cleaved in a defined order. Specific substrate targeting is achieved by the recognition of the short sequence upstream of the scissile bond and a correct macromolecular assembly of ns-polyprotein. Here, we highlighted the importance of the timeliness of proteolytic events, as an additional layer of regulation of efficient virus replication. We conclude that, somewhat counterintuitively, the cleavage site sequences at the nsP1/nsP2 and nsP2/nsP3 junctions are evolutionarily selected to be recognized by protease inefficiently, to avoid premature cleavages that would be detrimental for the assembly and functionality of the replication complex. Understanding the causes and consequences of viral polyprotein processing events is important for predicting the properties of mutant viruses and should be helpful for the development of better vaccine candidates and understanding potential mechanisms of resistance to protease inhibitors.


Subject(s)
Alphavirus Infections/virology , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Proteolysis , Semliki forest virus/physiology , Viral Nonstructural Proteins/metabolism , Alphavirus Infections/metabolism , Cells, Cultured , Genome, Viral , Kidney/virology , Mutation , RNA, Viral , Viral Nonstructural Proteins/genetics , Virus Replication
11.
PLoS Pathog ; 14(1): e1006835, 2018 01.
Article in English | MEDLINE | ID: mdl-29377936

ABSTRACT

Virus reprogramming of cellular metabolism is recognised as a critical determinant for viral growth. While most viruses appear to activate central energy metabolism, different viruses have been shown to rely on alternative mechanisms of metabolic activation. Whether related viruses exploit conserved mechanisms and induce similar metabolic changes is currently unclear. In this work we investigate how two alphaviruses, Semliki Forest virus and Ross River virus, reprogram host metabolism and define the molecular mechanisms responsible. We demonstrate that in both cases the presence of a YXXM motif in the viral protein nsP3 is necessary for binding to the PI3K regulatory subunit p85 and for activating AKT. This leads to an increase in glucose metabolism towards the synthesis of fatty acids, although additional mechanisms of metabolic activation appear to be involved in Ross River virus infection. Importantly, a Ross River virus mutant that fails to activate AKT has an attenuated phenotype in vivo, suggesting that viral activation of PI3K/AKT contributes to virulence and disease.


Subject(s)
Alphavirus Infections/metabolism , Alphavirus Infections/virology , Alphavirus/physiology , Glucose/metabolism , Host-Pathogen Interactions , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Alphavirus/pathogenicity , Animals , Cells, Cultured , Cricetinae , Enzyme Activation , Glycolysis/physiology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Ross River virus/physiology , Semliki forest virus/physiology , Virulence
12.
PLoS Pathog ; 14(1): e1006815, 2018 01.
Article in English | MEDLINE | ID: mdl-29370307

ABSTRACT

Introduced transinfections of the inherited bacteria Wolbachia can inhibit transmission of viruses by Aedes mosquitoes, and in Ae. aegypti are now being deployed for dengue control in a number of countries. Only three Wolbachia strains from the large number that exist in nature have to date been introduced and characterized in this species. Here novel Ae. aegypti transinfections were generated using the wAlbA and wAu strains. In its native Ae. albopictus, wAlbA is maintained at lower density than the co-infecting wAlbB, but following transfer to Ae. aegypti the relative strain density was reversed, illustrating the strain-specific nature of Wolbachia-host co-adaptation in determining density. The wAu strain also reached high densities in Ae. aegypti, and provided highly efficient transmission blocking of dengue and Zika viruses. Both wAu and wAlbA were less susceptible than wMel to density reduction/incomplete maternal transmission resulting from elevated larval rearing temperatures. Although wAu does not induce cytoplasmic incompatibility (CI), it was stably combined with a CI-inducing strain as a superinfection, and this would facilitate its spread into wild populations. Wolbachia wAu provides a very promising new option for arbovirus control, particularly for deployment in hot tropical climates.


Subject(s)
Aedes/microbiology , Aedes/virology , Antibiosis/physiology , Dengue Virus , Infectious Disease Transmission, Vertical/prevention & control , Wolbachia/physiology , Alphavirus Infections/microbiology , Alphavirus Infections/transmission , Alphavirus Infections/virology , Animals , Dengue Virus/pathogenicity , Female , Infectious Disease Transmission, Vertical/veterinary , Inheritance Patterns , Male , Pest Control, Biological , Semliki forest virus/pathogenicity , Semliki forest virus/physiology , Viral Load
13.
mBio ; 8(6)2017 11 07.
Article in English | MEDLINE | ID: mdl-29114027

ABSTRACT

Alphaviruses are members of a group of small enveloped RNA viruses that includes important human pathogens such as Chikungunya virus and the equine encephalitis viruses. The virus membrane is covered by a lattice composed of 80 spikes, each a trimer of heterodimers of the E2 and E1 transmembrane proteins. During virus endocytic entry, the E1 glycoprotein mediates the low-pH-dependent fusion of the virus membrane with the endosome membrane, thus initiating virus infection. While much is known about E1 structural rearrangements during membrane fusion, it is unclear how the E1/E2 dimer dissociates, a step required for the fusion reaction. A recent Alphavirus cryo-electron microscopy reconstruction revealed a previously unidentified D subdomain in the E2 ectodomain, close to the virus membrane. A loop within this region, here referred to as the D-loop, contains two highly conserved histidines, H348 and H352, which were hypothesized to play a role in dimer dissociation. We generated Semliki Forest virus mutants containing the single and double alanine substitutions H348A, H352A, and H348/352A. The three D-loop mutations caused a reduction in virus growth ranging from 1.6 to 2 log but did not significantly affect structural protein biosynthesis or transport, dimer stability, virus fusion, or specific infectivity. Instead, growth reduction was due to inhibition of a late stage of virus assembly at the plasma membrane. The virus particles that are produced show reduced thermostability compared to the wild type. We propose the E2 D-loop as a key region in establishing the E1-E2 contacts that drive glycoprotein lattice formation and promote Alphavirus budding from the plasma membrane.IMPORTANCEAlphavirus infection causes severe and debilitating human diseases for which there are no effective antiviral therapies or vaccines. In order to develop targeted therapeutics, detailed molecular understanding of the viral entry and exit mechanisms is required. In this report, we define the role of the E2 protein juxtamembrane D-loop, which contains highly conserved histidine residues at positions 348 and 352. These histidines do not play an important role in virus fusion and infection. However, mutation of the D-loop histidines causes significant decreases in the assembly and thermostability of Alphavirus particles. Our results suggest that the E2 D-loop interacts with the E1 protein to promote Alphavirus budding.


Subject(s)
Membrane Glycoproteins/chemistry , Semliki forest virus/physiology , Viral Envelope Proteins/chemistry , Virus Release , Animals , Cell Line , Cricetinae , Histidine/chemistry , Membrane Fusion , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Models, Molecular , Mutation , Nucleocapsid/metabolism , Nucleocapsid/ultrastructure , Protein Conformation , Protein Multimerization , Semliki forest virus/genetics , Semliki forest virus/metabolism , Semliki forest virus/ultrastructure , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Assembly , Virus Internalization
14.
Immunol Lett ; 190: 221-232, 2017 10.
Article in English | MEDLINE | ID: mdl-28851629

ABSTRACT

Development of a vaccine targeting human immunodeficiency virus-1 subtype C (HIV-1C) is an important public health priority in regions with a high prevalence of the clade C virus. The present study demonstrates the immunogenicity of recombinant Semliki Forest virus (SFV)-based virus-like replicon particles (VRPs) expressing Indian HIV-1C env/gag/polRT genes. Immunization of mice with recombinant VRPs in a homologous prime-boost protocol, either individually or in combination, elicited significant antigen-specific IFN-γ T cell responses as detected by the ELISPOT assay. Additionally, Gag-specific TNF-α secreting CD8+ and CD4+ T cells and Env-specific IL-2 secreting T cells were also elicited by mice immunized with Gag and Env constructs, respectively, as estimated by intracellular cytokine staining assay. Moreover, an HIV Pol-specific TNF-α response was elicited in mice immunized with a combination of the three VRP constructs. Furthermore, HIV-1C Gag and Env-specific binding antibodies were elicited as verified by gp120 ELISA and p24 Gag ELISA, respectively. The immunogenicity of VRPs was found to be higher as compared to that of RNA replicons and VRPs may therefore be promising preventive and therapeutic candidate vaccines for the control and management of HIV/AIDS.


Subject(s)
AIDS Vaccines/immunology , HIV Infections/immunology , HIV-1/immunology , Semliki forest virus/physiology , Virion/immunology , AIDS Vaccines/genetics , Animals , Female , Fusion Proteins, gag-pol/genetics , Gene Products, env/genetics , Genetic Vectors , HIV Antibodies/blood , HIV Antigens/genetics , Humans , Mice , Replicon/genetics , Vaccination , Vaccines, DNA
15.
Open Biol ; 6(7)2016 Jul.
Article in English | MEDLINE | ID: mdl-27383630

ABSTRACT

Recent findings have highlighted the role of the Old World alphavirus non-structural protein 3 (nsP3) as a host defence modulator that functions by disrupting stress granules, subcellular phase-dense RNA/protein structures formed upon environmental stress. This disruption mechanism was largely explained through nsP3-mediated recruitment of the host G3BP protein via two tandem FGDF motifs. Here, we present the 1.9 Å resolution crystal structure of the NTF2-like domain of G3BP-1 in complex with a 25-residue peptide derived from Semliki Forest virus nsP3 (nsP3-25). The structure reveals a poly-complex of G3BP-1 dimers interconnected through the FGDF motifs in nsP3-25. Although in vitro and in vivo binding studies revealed a hierarchical interaction of the two FGDF motifs with G3BP-1, viral growth curves clearly demonstrated that two intact FGDF motifs are required for efficient viral replication. Chikungunya virus nsP3 also binds G3BP dimers via a hierarchical interaction, which was found to be critical for viral replication. These results highlight a conserved molecular mechanism in host cell modulation.


Subject(s)
DNA Helicases/chemistry , DNA Helicases/metabolism , Poly-ADP-Ribose Binding Proteins/chemistry , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/chemistry , RNA Helicases/metabolism , RNA Recognition Motif Proteins/chemistry , RNA Recognition Motif Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Semliki forest virus/physiology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Amino Acid Motifs , Cell Line , Chikungunya virus/metabolism , Chikungunya virus/physiology , Conserved Sequence , Crystallography, X-Ray , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Semliki forest virus/metabolism , Virus Replication
16.
Immunity ; 44(6): 1455-69, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27332734

ABSTRACT

Aedes aegypti mosquitoes are responsible for transmitting many medically important viruses such as those that cause Zika and dengue. The inoculation of viruses into mosquito bite sites is an important and common stage of all mosquito-borne virus infections. We show, using Semliki Forest virus and Bunyamwera virus, that these viruses use this inflammatory niche to aid their replication and dissemination in vivo. Mosquito bites were characterized by an edema that retained virus at the inoculation site and an inflammatory influx of neutrophils that coordinated a localized innate immune program that inadvertently facilitated virus infection by encouraging the entry and infection of virus-permissive myeloid cells. Neutrophil depletion and therapeutic blockade of inflammasome activity suppressed inflammation and abrogated the ability of the bite to promote infection. This study identifies facets of mosquito bite inflammation that are important determinants of the subsequent systemic course and clinical outcome of virus infection.


Subject(s)
Arbovirus Infections/immunology , Bunyamwera virus/physiology , Inflammation/immunology , Insect Bites and Stings/immunology , Neutrophils/immunology , Semliki forest virus/physiology , Virus Replication , Animals , Cell Movement , Cells, Cultured , Culicidae/immunology , Humans , Immunity, Innate , Inflammasomes/metabolism , Inflammation/virology , Insect Bites and Stings/virology , Mice , Neutrophils/virology
17.
Traffic ; 17(9): 997-1013, 2016 09.
Article in English | MEDLINE | ID: mdl-27219333

ABSTRACT

Interferon inducible transmembrane proteins (IFITMs) are broad-spectrum antiviral factors. In cell culture the entry of many enveloped viruses, including orthomyxo-, flavi-, and filoviruses, is inhibited by IFITMs, though the mechanism(s) involved remain unclear and may vary between viruses. We demonstrate that Sindbis and Semliki Forest virus (SFV), which both use endocytosis and acid-induced membrane fusion in early endosomes to infect cells, are restricted by the early endosomal IFITM3. The late endosomal IFITM2 is less restrictive and the plasma membrane IFITM1 does not inhibit normal infection by either virus. IFITM3 inhibits release of the SFV capsid into the cytosol, without inhibiting binding, internalization, trafficking to endosomes or low pH-induced conformational changes in the envelope glycoprotein. Infection by SFV fusion at the cell surface was inhibited by IFITM1, but was equally inhibited by IFITM3. Furthermore, an IFITM3 mutant (Y20A) that is localized to the plasma membrane inhibited infection by cell surface fusion more potently than IFITM1. Together, these results indicate that IFITMs, in particular IFITM3, can restrict alphavirus infection by inhibiting viral fusion with cellular membranes. That IFITM3 can restrict SFV infection by fusion at the cell surface equivalently to IFITM1 suggests that IFITM3 has greater antiviral potency against SFV.


Subject(s)
Antigens, Differentiation/metabolism , Endosomes/metabolism , Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism , Semliki forest virus/physiology , Sindbis Virus/physiology , A549 Cells , Alphavirus Infections/metabolism , Alphavirus Infections/virology , Antigens, Differentiation/genetics , Endocytosis/physiology , Endosomes/virology , Humans , Membrane Proteins/genetics , RNA-Binding Proteins/genetics , Virus Internalization
18.
J Gen Virol ; 97(6): 1395-1407, 2016 06.
Article in English | MEDLINE | ID: mdl-26953094

ABSTRACT

During virus multiplication, the viral genome is recognized and recruited for replication based on specific cis-acting elements. Here, we dissected the important cis-acting sequence elements in Semliki Forest virus RNA by using a trans-replication system. As the viral replicase is expressed from a separate plasmid, the template RNA can be freely modified in this system. We show that the cis-acting element at the beginning of the non-structural protein 1 (nsP1) coding region together with the end of the 3' UTR are the minimal requirements for minus-strand synthesis. To achieve a high level of replication, the native 5' UTR was also needed. The virus-induced membranous replication compartments (spherules) were only detected when a replication-competent template was present with an active replicase and minus strands were produced. No translation could be detected from the minus strands, suggesting that they are segregated from the cytoplasm. Minus strands could not be recruited directly to initiate the replication process. Thus, there is only one defined pathway for replication, starting with plus-strand recognition followed by concomitant spherule formation and minus-strand synthesis.


Subject(s)
RNA, Viral/biosynthesis , RNA, Viral/genetics , Semliki forest virus/genetics , Semliki forest virus/physiology , Virus Replication , Animals , Cell Line , Cricetinae , Protein Binding , Protein Biosynthesis , RNA-Dependent RNA Polymerase/metabolism , Transcription, Genetic
19.
J Virol ; 90(9): 4289-97, 2016 May.
Article in English | MEDLINE | ID: mdl-26865714

ABSTRACT

UNLABELLED: Alphaviruses are small enveloped RNA viruses that infect cells via clathrin-mediated endocytosis and low-pH-triggered fusion in the early endosome. Using a small interfering RNA (siRNA) screen in human cells, we previously identified TSPAN9 as a host factor that promotes infection by the alphaviruses Sindbis virus (SINV), Semliki Forest virus (SFV), and chikungunya virus (CHIKV). Depletion of TSPAN9 specifically decreases SFV membrane fusion in endosomes. TSPAN9 is a member of the tetraspanin family of multipass membrane proteins, but its cellular function is currently unknown. Here we used U-2 OS cells stably overexpressing TSPAN9 to show that TSPAN9 is localized at the plasma membrane and in early and late endosomes. Internalized SFV particles colocalized with TSPAN9 in vesicles early during infection. Depletion of TSPAN9 led to reductions in the amounts of the late endosomal proteins LAMP1 and CD63 and an increase in the amount of LAMP2. However, TSPAN9 depletion did not alter the delivery of SFV to early endosomes or change their pH or protease activity. Comparative studies showed that TSPAN9 depletion strongly inhibited infection by several viruses that fuse in early endosomes (SFV, SINV, CHIKV, and vesicular stomatitis virus [VSV]), while viruses that fuse in the late endosome (recombinant VSV-Lassa and VSV-Junin), including an SFV point mutant with a lower pH threshold for fusion (SFV E2 T12I), were relatively resistant. Our data suggest that TSPAN9 modulates the early endosome compartment to make it more permissive for membrane fusion of early-penetrating viruses. IMPORTANCE: Alphaviruses are spread by mosquitoes and can cause serious human diseases such as arthritis and encephalitis. Recent outbreaks of CHIKV infection are responsible for millions of cases of acute illness and long-term complications. There are no vaccines or antiviral treatments for these important human pathogens. Alphaviruses infect host cells by utilizing the endocytic machinery of the cell and fusing their membrane with that of the endosome. Although the mechanism of virus-membrane fusion is well studied, we still know relatively little about the host cell proteins that are involved in alphavirus entry. Here we characterized the role of the host membrane protein TSPAN9 in alphavirus infection. TSPAN9 was localized to early endosomes containing internalized alphavirus, and depletion of TSPAN9 inhibited virus fusion with the early endosome membrane. In contrast, infection of viruses that enter through the late endosome was relatively resistant to TSPAN9 depletion, suggesting an important role for TSPAN9 in the early endosome.


Subject(s)
Alphavirus Infections/genetics , Alphavirus Infections/virology , Alphavirus/physiology , Endosomes/virology , Host-Pathogen Interactions , Tetraspanins/genetics , Virus Internalization , Alphavirus Infections/metabolism , Cell Line , Cholesterol/metabolism , Endocytosis , Endosomes/metabolism , Gene Expression , Humans , Hydrogen-Ion Concentration , Intracellular Space , Peptide Hydrolases/metabolism , Protein Transport , Semliki forest virus/physiology , Tetraspanins/metabolism , Vesicular Transport Proteins/metabolism
20.
J Virol ; 90(3): 1687-92, 2016 02 01.
Article in English | MEDLINE | ID: mdl-26581991

ABSTRACT

The alphaviruses induce membrane invaginations known as spherules as their RNA replication sites. Here, we show that inactivation of any function (polymerase, helicase, protease, or membrane association) essential for RNA synthesis also prevents the generation of spherule structures in a Semliki Forest virus trans-replication system. Mutants capable of negative-strand synthesis, including those defective in RNA capping, gave rise to spherules. Recruitment of RNA to membranes in the absence of spherule formation was not detected.


Subject(s)
Cell Membrane/metabolism , RNA, Viral/metabolism , Semliki forest virus/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...