Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 14: 1331755, 2024.
Article in English | MEDLINE | ID: mdl-38800833

ABSTRACT

The mosquito-borne Rift Valley fever virus (RVFV) from the Phenuiviridae family is a single-stranded RNA virus that causes the re-emerging zoonotic disease Rift Valley fever (RVF). Classified as a Category A agent by the NIH, RVFV infection can cause debilitating disease or death in humans and lead to devastating economic impacts by causing abortion storms in pregnant cattle. In a previous study, we showed that the host chaperone protein HSP90 is an RVFV-associated host factor that plays a critical role post viral entry, during the active phase of viral genome replication/transcription. In this study, we have elucidated the molecular mechanisms behind the regulatory effect of HSP90 during infection with RVFV. Our results demonstrate that during the early infection phase, host HSP90 associates with the viral RNA-dependent RNA polymerase (L protein) and prevents its degradation through the proteasome, resulting in increased viral replication.


Subject(s)
HSP90 Heat-Shock Proteins , Proteasome Endopeptidase Complex , Proteolysis , Rift Valley fever virus , Virus Replication , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/genetics , Rift Valley fever virus/genetics , Rift Valley fever virus/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Genome, Viral , Humans , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/genetics , Host-Pathogen Interactions , Viral Proteins/metabolism , Viral Proteins/genetics , Transcription, Genetic , Rift Valley Fever/virology , Rift Valley Fever/metabolism , Cell Line
2.
PLoS Biol ; 20(11): e3001870, 2022 11.
Article in English | MEDLINE | ID: mdl-36378688

ABSTRACT

Bunyaviruses lack a specific mechanism to ensure the incorporation of a complete set of genome segments into each virion, explaining the generation of incomplete virus particles lacking one or more genome segments. Such incomplete virus particles, which may represent the majority of particles produced, are generally considered to interfere with virus infection and spread. Using the three-segmented arthropod-borne Rift Valley fever virus as a model bunyavirus, we here show that two distinct incomplete virus particle populations unable to spread autonomously are able to efficiently complement each other in both mammalian and insect cells following co-infection. We further show that complementing incomplete virus particles can co-infect mosquitoes, resulting in the reconstitution of infectious virus that is able to disseminate to the mosquito salivary glands. Computational models of infection dynamics predict that incomplete virus particles can positively impact virus spread over a wide range of conditions, with the strongest effect at intermediate multiplicities of infection. Our findings suggest that incomplete particles may play a significant role in within-host spread and between-host transmission, reminiscent of the infection cycle of multipartite viruses.


Subject(s)
Arboviruses , Culicidae , Orthobunyavirus , Rift Valley Fever , Rift Valley fever virus , Virus Diseases , Animals , Humans , Rift Valley fever virus/genetics , Rift Valley Fever/genetics , Rift Valley Fever/metabolism , Virion/metabolism , Mammals
3.
Front Cell Infect Microbiol ; 11: 799024, 2021.
Article in English | MEDLINE | ID: mdl-35127560

ABSTRACT

Rift Valley fever virus (RVFV) is an emerging pathogen that has potential to cause severe disease in humans and domestic livestock. Propagation of RVFV strain MP-12 is negatively impacted by the actions of RIOK3, a protein involved in the cellular immune response to viral infection. During RVFV infection, RIOK3 mRNA is alternatively spliced to produce an isoform that correlates with the inhibition of interferon ß signaling. Here, we identify splicing factor TRA2-ß (also known as TRA2beta and hTRA2-ß) as a key regulator governing the relative abundance of RIOK3 splicing isoforms. Using RT-PCR and minigenes, we determined that TRA2-ß interaction with RIOK3 pre-mRNA was necessary for constitutive splicing of RIOK3 mRNA, and conversely, lack of TRA2-ß engagement led to increased alternative splicing. Expression of TRA2-ß was found to be necessary for RIOK3's antiviral effect against RVFV. Intriguingly, TRA2-ß mRNA is also alternatively spliced during RVFV infection, leading to a decrease in cellular TRA2-ß protein levels. These results suggest that splicing modulation serves as an immune evasion strategy by RVFV and/or is a cellular mechanism to prevent excessive immune response. Furthermore, the results suggest that TRA2-ß can act as a key regulator of additional steps of the innate immune response to viral infection.


Subject(s)
Rift Valley Fever , Rift Valley fever virus , Alternative Splicing , Animals , Antiviral Agents/metabolism , Humans , Immunity, Innate , Rift Valley Fever/metabolism , Rift Valley fever virus/genetics
4.
Int J Mol Sci ; 22(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374561

ABSTRACT

Rift Valley fever virus (RVFV) is a mosquito-transmitted virus from the Bunyaviridae family that causes high rates of mortality and morbidity in humans and ruminant animals. Previous studies indicated that DEAD-box helicase 17 (DDX17) restricts RVFV replication by recognizing two primary non-coding RNAs in the S-segment of the genome: the intergenic region (IGR) and 5' non-coding region (NCR). However, we lack molecular insights into the direct binding of DDX17 with RVFV non-coding RNAs and information on the unwinding of both non-coding RNAs by DDX17. Therefore, we performed an extensive biophysical analysis of the DDX17 helicase domain (DDX17135-555) and RVFV non-coding RNAs, IGR and 5' NCR. The homogeneity studies using analytical ultracentrifugation indicated that DDX17135-555, IGR, and 5' NCR are pure. Next, we performed small-angle X-ray scattering (SAXS) experiments, which suggested that DDX17 and both RNAs are homogenous as well. SAXS analysis also demonstrated that DDX17 is globular to an extent, whereas the RNAs adopt an extended conformation in solution. Subsequently, microscale thermophoresis (MST) experiments were performed to investigate the direct binding of DDX17 to the non-coding RNAs. The MST experiments demonstrated that DDX17 binds with the IGR and 5' NCR with a dissociation constant of 5.77 ± 0.15 µM and 9.85 ± 0.11 µM, respectively. As DDX17135-555 is an RNA helicase, we next determined if it could unwind IGR and NCR. We developed a helicase assay using MST and fluorescently-labeled oligos, which suggested DDX17135-555 can unwind both RNAs. Overall, our study provides direct evidence of DDX17135-555 interacting with and unwinding RVFV non-coding regions.


Subject(s)
DEAD-box RNA Helicases/metabolism , Host-Pathogen Interactions , RNA, Untranslated , RNA, Viral , Rift Valley Fever/metabolism , Rift Valley Fever/virology , Rift Valley fever virus/genetics , Adenosine Triphosphate , Animals , DEAD-box RNA Helicases/chemistry , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Structure-Activity Relationship
5.
J Virol ; 95(1)2020 12 09.
Article in English | MEDLINE | ID: mdl-33087469

ABSTRACT

Rift Valley fever virus (RVFV) is a highly pathogenic zoonotic arbovirus endemic in many African countries and the Arabian Peninsula. Animal infections cause high rates of mortality and abortion among sheep, goats, and cattle. In humans, an estimated 1 to 2% of RVFV infections result in severe disease (encephalitis, hepatitis, or retinitis) with a high rate of lethality when associated with hemorrhagic fever. The RVFV NSs protein, which is the main virulence factor, counteracts the host innate antiviral response to favor viral replication and spread. However, the mechanisms underlying RVFV-induced cytopathic effects and the role of NSs in these alterations remain for the most part unknown. In this work, we have analyzed the effects of NSs expression on the actin cytoskeleton while conducting infections with the NSs-expressing virulent (ZH548) and attenuated (MP12) strains of RVFV and the non-NSs-expressing avirulent (ZH548ΔNSs) strain, as well as after the ectopic expression of NSs. In macrophages, fibroblasts, and hepatocytes, NSs expression prevented the upregulation of Abl2 (a major regulator of the actin cytoskeleton) expression otherwise induced by avirulent infections and identified here as part of the antiviral response. The presence of NSs was also linked to an increased mobility of ZH548-infected cells compared to ZH548ΔNSs-infected fibroblasts and to strong changes in cell morphology in nonmigrating hepatocytes, with reduction of lamellipodia, cell spreading, and dissolution of adherens junctions reminiscent of the ZH548-induced cytopathic effects observed in vivo Finally, we show evidence of the presence of NSs within long actin-rich structures associated with NSs dissemination from NSs-expressing toward non-NSs-expressing cells.IMPORTANCE Rift Valley fever virus (RVFV) is a dangerous human and animal pathogen that was ranked by the World Health Organization in 2018 as among the eight pathogens of most concern for being likely to cause wide epidemics in the near future and for which there are no, or insufficient, countermeasures. The focus of this work is to address the question of the mechanisms underlying RVFV-induced cytopathic effects that participate in RVFV pathogenicity. We demonstrate here that RVFV targets cell adhesion and the actin cytoskeleton at the transcriptional and cellular level, affecting cell mobility and inducing cell shape collapse, along with distortion of cell-cell adhesion. All these effects may participate in RVFV-induced pathogenicity, facilitate virulent RVFV dissemination, and thus constitute interesting potential targets for future development of antiviral therapeutic strategies that, in the case of RVFV, as with several other emerging arboviruses, are presently lacking.


Subject(s)
Actin Cytoskeleton/genetics , Protein-Tyrosine Kinases/genetics , Rift Valley Fever/pathology , Rift Valley fever virus/pathogenicity , Viral Nonstructural Proteins/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Adhesion , Cell Line , Cell Movement , Cell Shape , Host-Pathogen Interactions , Immunity, Innate , Mice , Mutation , Protein-Tyrosine Kinases/metabolism , Rift Valley Fever/metabolism , Rift Valley Fever/virology , Rift Valley fever virus/genetics , Rift Valley fever virus/metabolism , Viral Nonstructural Proteins/genetics , Virulence Factors/genetics , Virulence Factors/metabolism , Virus Replication
6.
Nat Commun ; 11(1): 3281, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32612175

ABSTRACT

Amyloid fibrils result from the aggregation of host cell-encoded proteins, many giving rise to specific human illnesses such as Alzheimer's disease. Here we show that the major virulence factor of Rift Valley fever virus, the protein NSs, forms filamentous structures in the brain of mice and affects mortality. NSs assembles into nuclear and cytosolic disulfide bond-dependent fibrillary aggregates in infected cells. NSs structural arrangements exhibit characteristics typical for amyloids, such as an ultrastructure of 12 nm-width fibrils, a strong detergent resistance, and interactions with the amyloid-binding dye Thioflavin-S. The assembly dynamics of viral amyloid-like fibrils can be visualized in real-time. They form spontaneously and grow in an amyloid fashion within 5 hours. Together, our results demonstrate that viruses can encode amyloid-like fibril-forming proteins and have strong implications for future research on amyloid aggregation and toxicity in general.


Subject(s)
Amyloid/metabolism , Amyloidogenic Proteins/metabolism , Rift Valley Fever/metabolism , Rift Valley fever virus/metabolism , Viral Nonstructural Proteins/metabolism , Amyloid/chemistry , Amyloid/ultrastructure , Amyloidogenic Proteins/chemistry , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cell Nucleus/virology , Chlorocebus aethiops , HeLa Cells , Humans , Mice , Microscopy, Confocal , Microscopy, Electron, Transmission , Protein Aggregation, Pathological/metabolism , Rift Valley Fever/virology , Rift Valley fever virus/pathogenicity , Vero Cells , Viral Nonstructural Proteins/chemistry , Virulence , Virulence Factors
7.
J Gen Virol ; 101(7): 712-716, 2020 07.
Article in English | MEDLINE | ID: mdl-31671053

ABSTRACT

The non-structural protein NSs is the main virulence factor of Rift Valley fever virus, a major zoonotic pathogen in Africa. NSs forms large aggregates in the nucleus and impairs induction of the antiviral type I IFN system by several mechanisms, including degradation of subunit p62 of the general RNA polymerase II transcription factor TFIIH. Here, we show that depletion of the nuclear pore protein Nup98 affects the nuclear import of NSs. Nonetheless, NSs was still able to degrade TFIIH-p62 under these conditions. Depletion of Nup98, however, had a negative effect on Rift Valley fever virus multiplication. Our data thus indicate that NSs utilizes Nup98 for import into the nucleus, but also plays a general role in the viral replication cycle.


Subject(s)
Host-Pathogen Interactions , Nuclear Pore Complex Proteins/metabolism , Rift Valley fever virus/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication , Active Transport, Cell Nucleus , Cell Line , Cells, Cultured , Humans , Protein Transport , Rift Valley Fever/genetics , Rift Valley Fever/metabolism , Rift Valley Fever/virology , Virulence Factors
8.
PLoS One ; 14(5): e0217497, 2019.
Article in English | MEDLINE | ID: mdl-31136639

ABSTRACT

Rift Valley fever virus (RVFV) is a negative-sense RNA virus belonging to the Phenuiviridae family that infects both domestic livestock and humans. The NIAID has designated RVFV as a Category A priority emerging pathogen due to the devastating public health outcomes associated with epidemic outbreaks. However, there is no licensed treatment or vaccine approved for human use. Therefore it is of great interest to understand RVFV pathogenesis in infected hosts in order to facilitate creation of targeted therapies and treatment options. Here we provide insight into the host-pathogen interface in human HEK293 cells during RVFV MP-12 strain infection using high-throughput mRNA sequencing technology. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of differentially expressed genes showed robust innate immune and cytokine-mediated inflammatory pathway activation as well as alterations in pathways associated with fatty acid metabolism and extracellular matrix receptor signaling. We also analyzed the promoter regions of DEGs for patterns in transcription factor binding sites, and found several that are known to act synergistically to impact apoptosis, immunity, metabolism, and cell growth and differentiation. Lastly, we noted dramatic changes in host alternative splicing patterns in genes associated with mRNA decay and surveillance, RNA transport, and DNA repair. This study has improved our understanding of RVFV pathogenesis and has provided novel insight into pathways and signaling modules important for RVFV diagnostics and therapeutic development.


Subject(s)
Alternative Splicing , Gene Expression Profiling , Gene Expression Regulation, Viral , Rift Valley Fever/metabolism , Rift Valley fever virus/metabolism , Transcription, Genetic , HEK293 Cells , Humans
9.
PLoS One ; 13(2): e0191983, 2018.
Article in English | MEDLINE | ID: mdl-29408900

ABSTRACT

Rift Valley fever virus (RVFV) infects both ruminants and humans leading to a wide variance of pathologies dependent on host background and age. Utilizing a targeted reverse phase protein array (RPPA) to define changes in signaling cascades after in vitro infection of human cells with virulent and attenuated RVFV strains, we observed high phosphorylation of Smad transcription factors. This evolutionarily conserved family is phosphorylated by and transduces the activation of TGF-ß superfamily receptors. Moreover, we observed that phosphorylation of Smad proteins required active RVFV replication and loss of NSs impaired this activation, further corroborating the RPPA results. Gene promoter analysis of transcripts altered after RVFV infection identified 913 genes that contained a Smad-response element. Functional annotation of these potential Smad-regulated genes clustered in axonal guidance, hepatic fibrosis and cell signaling pathways involved in cellular adhesion/migration, calcium influx, and cytoskeletal reorganization. Furthermore, chromatin immunoprecipitation confirmed the presence of a Smad complex on the interleukin 1 receptor type 2 (IL1R2) promoter, which acts as a decoy receptor for IL-1 activation.


Subject(s)
Phosphoproteins/metabolism , Proteomics , Rift Valley Fever/metabolism , Smad Proteins/metabolism , Animals , Cells, Cultured , Humans , Phosphorylation , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Rift Valley fever virus/genetics , Rift Valley fever virus/physiology , Smad Proteins/genetics , Virus Replication/genetics
10.
Sci Rep ; 7(1): 14385, 2017 10 30.
Article in English | MEDLINE | ID: mdl-29085037

ABSTRACT

Rift Valley fever virus (RVFV) causes major outbreaks among livestock, characterized by "abortion storms" in which spontaneous abortion occurs in almost 100% of pregnant ruminants. Humans can also become infected with mild symptoms that can progress to more severe symptoms, such as hepatitis, encephalitis, and hemorrhagic fever. The goal of this study was to use RNA-sequencing (RNA-seq) to analyze the host transcriptome in response to RVFV infection. G2/M DNA damage checkpoint, ATM signaling, mitochondrial dysfunction, regulation of the antiviral response, and integrin-linked kinase (ILK) signaling were among the top altered canonical pathways with both the attenuated MP12 strain and the fully virulent ZH548 strain. Although several mRNA transcripts were highly upregulated, an increase at the protein level was not observed for the selected genes, which was at least partially due to the NSs dependent block in mRNA export. Inhibition of ILK signaling, which is involved in cell motility and cytoskeletal reorganization, resulted in reduced RVFV replication, indicating that this pathway is important for viral replication. Overall, this is the first global transcriptomic analysis of the human host response following RVFV infection, which could give insight into novel host responses that have not yet been explored.


Subject(s)
Rift Valley Fever/genetics , Cell Culture Techniques , Cell Cycle Checkpoints , Epithelial Cells , Humans , Protein Serine-Threonine Kinases , RNA, Messenger/genetics , Rift Valley Fever/metabolism , Rift Valley fever virus/genetics , Rift Valley fever virus/pathogenicity , Sequence Analysis, RNA , Signal Transduction , Transcriptome/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/physiology
11.
J Virol ; 91(21)2017 11 01.
Article in English | MEDLINE | ID: mdl-28794043

ABSTRACT

There is an urgent need for therapeutic development to combat infections caused by Rift Valley fever virus (RVFV), which causes devastating disease in both humans and animals. In an effort to repurpose drugs for RVFV treatment, our previous studies screened a library of FDA-approved drugs. The most promising candidate identified was the hepatocellular and renal cell carcinoma drug sorafenib. Mechanism-of-action studies indicated that sorafenib targeted a late stage in virus infection and caused a buildup of virions within cells. In addition, small interfering RNA (siRNA) knockdown studies suggested that nonclassical targets of sorafenib are important for the propagation of RVFV. Here we extend our previous findings to identify the mechanism by which sorafenib inhibits the release of RVFV virions from the cell. Confocal microscopy imaging revealed that glycoprotein Gn colocalizes and accumulates within the endoplasmic reticulum (ER) and the transport of Gn from the Golgi complex to the host cell membrane is reduced. Transmission electron microscopy demonstrated that sorafenib caused virions to be present inside large vacuoles inside the cells. p97/valosin-containing protein (VCP), which is involved in membrane remodeling in the secretory pathway and a known target of sorafenib, was found to be important for RVFV egress. Knockdown of VCP resulted in decreased RVFV replication, reduced Gn Golgi complex localization, and increased Gn ER accumulation. The intracellular accumulation of RVFV virions was also observed in cells transfected with siRNA targeting VCP. Collectively, these data indicate that sorafenib causes a disruption in viral egress by targeting VCP and the secretory pathway, resulting in a buildup of virions within dilated ER vesicles.IMPORTANCE In humans, symptoms of RVFV infection mainly include a self-limiting febrile illness. However, in some cases, infected individuals can also experience hemorrhagic fever, neurological disorders, liver failure, and blindness, which could collectively be lethal. The ability of RVFV to expand geographically outside sub-Saharan Africa is of concern, particularly to the Americas, where native mosquito species are capable of virus transmission. Currently, there are no FDA-approved therapeutics to treat RVFV infection, and thus, there is an urgent need to understand the mechanisms by which the virus hijacks the host cell machinery to replicate. The significance of our research is in identifying the cellular target of sorafenib that inhibits RVFV propagation, so that this information can be used as a tool for the further development of therapeutics used to treat RVFV infection.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Rift Valley Fever/drug therapy , Rift Valley fever virus/physiology , Secretory Pathway/drug effects , Virus Release/drug effects , Adenosine Triphosphatases/genetics , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Cell Cycle Proteins/genetics , Chlorocebus aethiops , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Niacinamide/pharmacology , Rift Valley Fever/metabolism , Rift Valley Fever/virology , Rift Valley fever virus/drug effects , Sorafenib , Tumor Cells, Cultured , Valosin Containing Protein , Vero Cells , Virion/drug effects , Virus Replication/drug effects
12.
PLoS Pathog ; 12(8): e1005800, 2016 08.
Article in English | MEDLINE | ID: mdl-27548280

ABSTRACT

The bunyavirus genome comprises a small (S), medium (M), and large (L) RNA segment of negative polarity. Although genome segmentation confers evolutionary advantages by enabling genome reassortment events with related viruses, genome segmentation also complicates genome replication and packaging. Accumulating evidence suggests that genomes of viruses with eight or more genome segments are incorporated into virions by highly selective processes. Remarkably, little is known about the genome packaging process of the tri-segmented bunyaviruses. Here, we evaluated, by single-molecule RNA fluorescence in situ hybridization (FISH), the intracellular spatio-temporal distribution and replication kinetics of the Rift Valley fever virus (RVFV) genome and determined the segment composition of mature virions. The results reveal that the RVFV genome segments start to replicate near the site of infection before spreading and replicating throughout the cytoplasm followed by translocation to the virion assembly site at the Golgi network. Despite the average intracellular S, M and L genome segments approached a 1:1:1 ratio, major differences in genome segment ratios were observed among cells. We also observed a significant amount of cells lacking evidence of M-segment replication. Analysis of two-segmented replicons and four-segmented viruses subsequently confirmed the previous notion that Golgi recruitment is mediated by the Gn glycoprotein. The absence of colocalization of the different segments in the cytoplasm and the successful rescue of a tri-segmented variant with a codon shuffled M-segment suggested that inter-segment interactions are unlikely to drive the copackaging of the different segments into a single virion. The latter was confirmed by direct visualization of RNPs inside mature virions which showed that the majority of virions lack one or more genome segments. Altogether, this study suggests that RVFV genome packaging is a non-selective process.


Subject(s)
Genome, Viral/physiology , Rift Valley fever virus/physiology , Virus Assembly/physiology , Cell Line , Humans , In Situ Hybridization, Fluorescence , Rift Valley Fever/metabolism
13.
Virol J ; 13: 118, 2016 07 02.
Article in English | MEDLINE | ID: mdl-27368371

ABSTRACT

Rift Valley fever is a mosquito-borne zoonotic disease that affects both ruminants and humans. The nonstructural (NS) protein, which is a major virulence factor for Rift Valley fever virus (RVFV), is encoded on the S-segment. Through the cullin 1-Skp1-Fbox E3 ligase complex, the NSs protein promotes the degradation of at least two host proteins, the TFIIH p62 and the PKR proteins. NSs protein bridges the Fbox protein with subsequent substrates, and facilitates the transfer of ubiquitin. The SAP30-YY1 complex also bridges the NSs protein with chromatin DNA, affecting cohesion and segregation of chromatin DNA as well as the activation of interferon-ß promoter. The presence of NSs filaments in the nucleus induces DNA damage responses and causes cell-cycle arrest, p53 activation, and apoptosis. Despite the fact that NSs proteins have poor amino acid similarity among bunyaviruses, the strategy utilized to hijack host cells are similar. This review will provide and summarize an update of recent findings pertaining to the biological functions of the NSs protein of RVFV as well as the differences from those of other bunyaviruses.


Subject(s)
Rift Valley Fever/virology , Rift Valley fever virus/isolation & purification , Viral Nonstructural Proteins/metabolism , Animals , Apoptosis , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Rift Valley Fever/genetics , Rift Valley Fever/metabolism , Rift Valley Fever/physiopathology , Rift Valley fever virus/classification , Rift Valley fever virus/genetics , Rift Valley fever virus/metabolism , Viral Nonstructural Proteins/genetics
14.
Virology ; 496: 175-185, 2016 09.
Article in English | MEDLINE | ID: mdl-27318793

ABSTRACT

Rift Valley fever (RVF) is a zoonotic disease that can cause severe illness in humans and livestock, triggering spontaneous abortion in almost 100% of pregnant ruminants. In this study, we demonstrate that signal transducer and activator of transcription 3 (STAT3) is phosphorylated on its conserved tyrosine residue (Y705) following RVFV infection. This phosphorylation was dependent on a major virulence factor, the viral nonstructural protein NSs. Loss of STAT3 had little effect on viral replication, but rather resulted in cells being more susceptible to RVFV-induced cell death. Phosphorylated STAT3 translocated to the nucleus, coinciding with inhibition of fos, jun, and nr4a2 gene expression, and the presence of STAT3 and NSs at the nr4a2 promoter. NSs was found predominantly in the cytoplasm of STAT3 null cells, indicating that STAT3 influences NSs nuclear localization. Collectively, these data demonstrate that STAT3 functions in a pro-survival capacity through modulation of NSs localization.


Subject(s)
Rift Valley Fever/metabolism , Rift Valley Fever/virology , Rift Valley fever virus/physiology , STAT3 Transcription Factor/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Chlorocebus aethiops , Humans , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Transport , Rift Valley Fever/genetics , Rift Valley fever virus/drug effects , Tyrosine/metabolism , Vero Cells , Viral Nonstructural Proteins/metabolism , Virus Replication
15.
J Virol ; 90(16): 7084-7097, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27226375

ABSTRACT

UNLABELLED: Rift Valley fever virus (RVFV) is an arbovirus within the Bunyaviridae family capable of causing serious morbidity and mortality in humans and livestock. To identify host factors involved in bunyavirus replication, we employed genome-wide RNA interference (RNAi) screening and identified 381 genes whose knockdown reduced infection. The Wnt pathway was the most represented pathway when gene hits were functionally clustered. With further investigation, we found that RVFV infection activated Wnt signaling, was enhanced when Wnt signaling was preactivated, was reduced with knockdown of ß-catenin, and was blocked using Wnt signaling inhibitors. Similar results were found using distantly related bunyaviruses La Crosse virus and California encephalitis virus, suggesting a conserved role for Wnt signaling in bunyaviral infection. We propose a model where bunyaviruses activate Wnt-responsive genes to regulate optimal cell cycle conditions needed to promote efficient viral replication. The findings in this study should aid in the design of efficacious host-directed antiviral therapeutics. IMPORTANCE: RVFV is a mosquito-borne bunyavirus that is endemic to Africa but has demonstrated a capacity for emergence in new territories (e.g., the Arabian Peninsula). As a zoonotic pathogen that primarily affects livestock, RVFV can also cause lethal hemorrhagic fever and encephalitis in humans. Currently, there are no treatments or fully licensed vaccines for this virus. Using high-throughput RNAi screening, we identified canonical Wnt signaling as an important host pathway regulating RVFV infection. The beneficial role of Wnt signaling was observed for RVFV, along with other disparate bunyaviruses, indicating a conserved bunyaviral replication mechanism involving Wnt signaling. These studies supplement our knowledge of the fundamental mechanisms of bunyavirus infection and provide new avenues for countermeasure development against pathogenic bunyaviruses.


Subject(s)
Genome, Viral/genetics , RNA Interference , Rift Valley Fever/metabolism , Rift Valley fever virus/physiology , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , A549 Cells , Animals , Blotting, Western , Cells, Cultured , Chlorocebus aethiops , HeLa Cells , High-Throughput Screening Assays , Humans , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rift Valley Fever/genetics , Rift Valley Fever/virology , Vero Cells , Virus Replication , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
16.
Virology ; 486: 88-93, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26410240

ABSTRACT

We investigated the localization of host mRNA during Rift Valley fever virus (RVFV) infection. Fluorescence in situ hybridization revealed that infection with RVFV altered the localization of host mRNA. mRNA accumulated in the nuclei of RVFV-infected but not mock-infected cells. Further, overexpression of the NSS gene, but not the N, GN or NSM genes correlated with mRNA nuclear accumulation. Nuclear accumulation of host mRNA was not observed in cells infected with a strain of RVFV lacking the gene encoding NSS, confirming that expression of NSS is likely responsible for this phenomenon.


Subject(s)
Cell Nucleus/metabolism , RNA, Messenger/metabolism , Rift Valley Fever/metabolism , Rift Valley fever virus/metabolism , Viral Nonstructural Proteins/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/genetics , Cell Nucleus/virology , HeLa Cells , Host-Pathogen Interactions , Humans , RNA, Messenger/genetics , Rift Valley Fever/genetics , Rift Valley Fever/virology , Rift Valley fever virus/genetics , Viral Nonstructural Proteins/genetics
17.
Proc Natl Acad Sci U S A ; 112(22): E2920-9, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-26038567

ABSTRACT

The mosquito-transmitted bunyavirus, Rift Valley fever virus (RVFV), is a highly successful pathogen for which there are no vaccines or therapeutics. Translational arrest is a common antiviral strategy used by hosts. In response, RVFV inhibits two well-known antiviral pathways that attenuate translation during infection, PKR and type I IFN signaling. Despite this, translational arrest occurs during RVFV infection by unknown mechanisms. Here, we find that RVFV infection triggers the decay of core translation machinery mRNAs that possess a 5'-terminal oligopyrimidine (5'-TOP) motif in their 5'-UTR, including mRNAs encoding ribosomal proteins, which leads to a decrease in overall ribosomal protein levels. We find that the RNA decapping enzyme NUDT16 selectively degrades 5'-TOP mRNAs during RVFV infection and this decay is triggered in response to mTOR attenuation via the translational repressor 4EBP1/2 axis. Translational arrest of 5'-TOPs via 4EBP1/2 restricts RVFV replication, and this increased RNA decay results in the loss of visible RNA granules, including P bodies and stress granules. Because RVFV cap-snatches in RNA granules, the increased level of 5'-TOP mRNAs in this compartment leads to snatching of these targets, which are translationally suppressed during infection. Therefore, translation of RVFV mRNAs is compromised by multiple mechanisms during infection. Together, these data present a previously unknown mechanism for translational shutdown in response to viral infection and identify mTOR attenuation as a potential therapeutic avenue against bunyaviral infection.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis/physiology , Pyrophosphatases/metabolism , RNA 5' Terminal Oligopyrimidine Sequence/physiology , Rift Valley Fever/metabolism , Rift Valley fever virus/metabolism , TOR Serine-Threonine Kinases/metabolism , Cell Cycle Proteins , Cell Line , DNA Primers/genetics , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Humans , Immunoblotting , Linear Models , Luciferases , RNA 5' Terminal Oligopyrimidine Sequence/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
PLoS One ; 10(5): e0128215, 2015.
Article in English | MEDLINE | ID: mdl-26020252

ABSTRACT

BACKGROUND: Rift Valley fever virus (RVFV) is a highly pathogenic arthropod-borne virus that has a detrimental effect on both livestock and human populations. While there are several diagnostic methodologies available for RVFV detection, many are not sensitive enough to diagnose early infections. Furthermore, detection may be hindered by high abundant proteins such as albumin. Previous findings have shown that Nanotrap particles can be used to significantly enhance detection of various small analytes of low abundance. We have expanded upon this repertoire to show that this simple and efficient sample preparation technology can drastically improve the detection of the RVFV nucleoprotein (NP), the most abundant and widely used viral protein for RVFV diagnostics. RESULTS: After screening multiple Nanotrap particle architectures, we found that one particle, NT45, was optimal for RVFV NP capture, as demonstrated by western blotting. NT45 significantly enhanced detection of the NP at levels undetectable without the technology. Importantly, we demonstrated that Nanotrap particles are capable of concentrating NP in a number of matrices, including infected cell lysates, viral supernatants, and animal sera. Specifically, NT45 enhanced detection of NP at various viral titers, multiplicity of infections, and time points. Our most dramatic results were observed in spiked serum samples, where high abundance serum proteins hindered detection of NP without Nanotrap particles. Nanotrap particles allowed for sample cleanup and subsequent detection of RVFV NP. Finally, we demonstrated that incubation of our samples with Nanotrap particles protects the NP from degradation over extended periods of time (up to 120 hours) and at elevated temperatures (at 37ºC). CONCLUSION: This study demonstrates that Nanotrap particles are capable of drastically lowering the limit of detection for RVFV NP by capturing, concentrating, and preserving RVFV NP in clinically relevant matrices. These studies can be extended to a wide range of pathogens and their analytes of diagnostic interest.


Subject(s)
Nanoparticles/chemistry , Nucleoproteins/chemistry , Rift Valley fever virus/chemistry , Viral Proteins/chemistry , Animals , Chlorocebus aethiops , Humans , Nanoparticles/metabolism , Nucleoproteins/metabolism , Rift Valley Fever/diagnosis , Rift Valley Fever/metabolism , Rift Valley fever virus/metabolism , Vero Cells , Viral Proteins/metabolism
19.
Virology ; 449: 270-86, 2014 Jan 20.
Article in English | MEDLINE | ID: mdl-24418562

ABSTRACT

Rift Valley fever virus (RVFV) infection is often associated with pronounced liver damage. Previously, our studies revealed altered host phospho-signaling responses (NFκB, MAPK and DNA damage responses) in RVFV infected epithelial cells that correlated with a cellular stress response. Here, we report that RVFV infection of liver cells leads to an increase in reactive oxygen species (ROS). Our data suggests the presence of the viral protein NSs in the mitochondria of infected cells, hence contributing to early increase in ROS. Increased ROS levels correlated with activation of NFκB (p65) and p53 responses, which in conjunction with infection, was also reflected as macromolecular rearrangements observed using size fractionation of protein lysates. Additionally, we documented an increase in cytokine expression and pro-apoptotic gene expression with infection, which was reversed with antioxidant treatment. Collectively, we identified ROS and oxidative stress as critical contributors to apoptosis of liver cells during RVFV infection.


Subject(s)
Apoptosis , Liver/cytology , Reactive Oxygen Species/metabolism , Rift Valley Fever/metabolism , Rift Valley fever virus/physiology , Transcription Factor RelA/metabolism , Tumor Suppressor Protein p53/metabolism , Humans , Liver/metabolism , Liver/virology , Oxidative Stress , Rift Valley Fever/physiopathology , Rift Valley Fever/virology , Transcription Factor RelA/genetics , Tumor Suppressor Protein p53/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication
20.
J Virol ; 88(6): 3464-73, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24403578

ABSTRACT

UNLABELLED: The nonstructural protein NSs is the main virulence factor of Rift Valley fever virus (RVFV; family Bunyaviridae, genus Phlebovirus), a serious pathogen of livestock and humans in Africa. RVFV NSs blocks transcriptional upregulation of antiviral type I interferons (IFN) and destroys the general transcription factor TFIIH subunit p62 via the ubiquitin/proteasome pathway. Here, we identified a subunit of E3 ubiquitin ligases, F-box protein FBXO3, as a host cell interactor of NSs. Small interfering RNA (siRNA)-mediated depletion of FBXO3 rescued p62 protein levels in RVFV-infected cells and elevated IFN transcription by 1 order of magnitude. NSs interacts with the full-length FBXO3 protein as well as with a truncated isoform that lacks the C-terminal acidic and poly(R)-rich domains. These isoforms are present in both the nucleus and the cytoplasm. NSs exclusively removes the nuclear pool of full-length FBXO3, likely due to consumption during the degradation process. F-box proteins form the variable substrate recognition subunit of the so-called SCF ubiquitin ligases, which also contain the constant components Skp1, cullin 1 (or cullin 7), and Rbx1. siRNA knockdown of Skp1 also protected p62 from degradation, suggesting involvement in NSs action. However, knockdown of cullin 1, cullin 7, or Rbx1 could not rescue p62 degradation by NSs. Our data show that the enzymatic removal of p62 via the host cell factor FBXO3 is a major mechanism of IFN suppression by RVFV. IMPORTANCE: Rift Valley fever virus is a serious emerging pathogen of animals and humans. Its main virulence factor, NSs, enables unhindered virus replication by suppressing the antiviral innate immune system. We identified the E3 ubiquitin ligase FBXO3 as a novel host cell interactor of NSs. NSs recruits FBXO3 to destroy the general host cell transcription factor TFIIH-p62, resulting in suppression of the transcriptional upregulation of innate immunity.


Subject(s)
F-Box Proteins/metabolism , Phosphoproteins/metabolism , Rift Valley Fever/metabolism , Rift Valley fever virus/metabolism , Transcription Factors, TFII/metabolism , Viral Nonstructural Proteins/metabolism , Virulence Factors/metabolism , Cell Line , F-Box Proteins/genetics , Humans , Phosphoproteins/genetics , Proteolysis , Rift Valley Fever/enzymology , Rift Valley Fever/genetics , Rift Valley Fever/virology , Rift Valley fever virus/genetics , Transcription Factor TFIIH , Transcription Factors, TFII/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Viral Nonstructural Proteins/genetics , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...