Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Virulence ; 15(1): 2329447, 2024 12.
Article in English | MEDLINE | ID: mdl-38548679

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that regulate the post-transcriptional expression of target genes. Virus-encoded miRNAs play an important role in the replication of viruses, modulate gene expression in both the virus and host, and affect their persistence and immune evasion in hosts. This renders viral miRNAs as potential targets for therapeutic applications, especially against pathogenic viruses that infect humans and animals. Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic RNA virus that causes severe disease in both humans and livestock. High mortality among newborn lambs and abortion storms are key characteristics of an RVF outbreak. To date, limited information is available on RVFV-derived miRNAs. In this study, computational methods were used to analyse the RVFV genome for putative pre-miRNA genes, which were then analysed for the presence of mature miRNAs. We detected 19 RVFV-encoded miRNAs and identified their potential mRNAs targets in sheep (Ovis aries), the most susceptible host. The identification of significantly enriched O. aries genes in association with RVFV miRNAs will help elucidate the molecular mechanisms underlying RVFV pathogenesis and potentially uncover novel drug targets for RVFV.


Subject(s)
Culicidae , MicroRNAs , Rift Valley Fever , Rift Valley fever virus , Humans , Pregnancy , Female , Animals , Sheep/genetics , Rift Valley fever virus/genetics , Rift Valley Fever/genetics , Rift Valley Fever/epidemiology , Culicidae/genetics , Disease Outbreaks , MicroRNAs/genetics
2.
Methods Mol Biol ; 2733: 101-113, 2024.
Article in English | MEDLINE | ID: mdl-38064029

ABSTRACT

Rift Valley fever virus (RVFV) is an important mosquito-borne virus that can cause severe disease manifestations in humans including ocular damage, vision loss, late-onset encephalitis, and hemorrhagic fever. In ruminants, RVFV can cause high mortality rates in young animals and high rates of abortion in pregnant animals resulting in an enormous negative impact on the economy of affected regions. To date, no licensed vaccines in humans or anti-RVFV therapeutics for animal or human use are available. The development of reverse genetics has facilitated the generation of recombinant infectious viruses that serve as powerful tools for investigating the molecular biology and pathogenesis of RVFV. Infectious recombinant RVFV can be rescued entirely from cDNAs containing predetermined mutations in their genomes to investigate virus-host interactions and mechanisms of pathogenesis and generate live-attenuated vaccines. In this chapter, we will describe the experimental procedures for the implementation of RVFV reverse genetics.


Subject(s)
Rift Valley Fever , Rift Valley fever virus , Animals , Humans , Rift Valley fever virus/genetics , Rift Valley Fever/genetics , Rift Valley Fever/prevention & control , Reverse Genetics , Vaccines, Attenuated/genetics , Mutation
3.
Sci Adv ; 9(28): eadh2264, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37450601

ABSTRACT

Rift Valley fever virus (RVFV) is an emerging arbovirus found in Africa. While RVFV is pantropic and infects many cells and tissues, viral replication and necrosis within the liver play a critical role in mediating severe disease. The low-density lipoprotein receptor-related protein 1 (Lrp1) is a recently identified host factor for cellular entry and infection by RVFV. The biological significance of Lrp1, including its role in hepatic disease in vivo, however, remains to be determined. Because Lrp1 has a high expression level in hepatocytes, we developed a mouse model in which Lrp1 is specifically deleted in hepatocytes to test how the absence of liver Lrp1 expression affects RVF pathogenesis. Mice lacking Lrp1 expression in hepatocytes showed minimal RVFV replication in the liver, longer time to death, and altered clinical signs toward neurological disease. In contrast, RVFV infection levels in other tissues showed no difference between the two genotypes. Therefore, Lrp1 is essential for RVF hepatic disease in mice.


Subject(s)
Rift Valley Fever , Rift Valley fever virus , Animals , Mice , Rift Valley Fever/genetics , Rift Valley fever virus/genetics , Africa , Hepatocytes , Low Density Lipoprotein Receptor-Related Protein-1/genetics
4.
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
5.
Viruses ; 14(9)2022 09 17.
Article in English | MEDLINE | ID: mdl-36146870

ABSTRACT

Rift Valley fever virus (RVFV) is a pathogenic human and livestock RNA virus that poses a significant threat to public health and biosecurity. During RVFV infection, the atypical kinase RIOK3 plays important roles in the innate immune response. Although its exact functions in innate immunity are not completely understood, RIOK3 has been shown to be necessary for mounting an antiviral interferon (IFN) response to RVFV in epithelial cells. Furthermore, after immune stimulation, the splicing pattern for RIOK3 mRNA changes markedly, and RIOK3's dominant alternatively spliced isoform, RIOK3 X2, exhibits an opposite effect on the IFN response by dampening it. Here, we further investigate the roles of RIOK3 and its spliced isoform in other innate immune responses to RVFV, namely the NFκB-mediated inflammatory response. We find that while RIOK3 is important for negatively regulating this inflammatory pathway, its alternatively spliced isoform, RIOK3 X2, stimulates it. Overall, these data demonstrate that both RIOK3 and its X2 isoform have unique roles in separate innate immune pathways that respond to RVFV infection.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Rift Valley Fever , Rift Valley fever virus , Animals , Antiviral Agents/metabolism , Humans , Immunity, Innate , Interferons/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Rift Valley Fever/genetics
6.
Viruses ; 14(2)2022 02 08.
Article in English | MEDLINE | ID: mdl-35215938

ABSTRACT

Rift Valley fever (RVF) is a zoonotic disease caused by RVF Phlebovirus (RVFV). The RVFV MP-12 vaccine strain is known to exhibit residual virulence in the case of a deficient interferon type 1 response. The hypothesis of this study is that virus replication and severity of lesions induced by the MP-12 strain in immunocompromised mice depend on the specific function of the disturbed pathway. Therefore, 10 strains of mice with deficient innate immunity (B6-IFNARtmAgt, C.129S7(B6)-Ifngtm1Ts/J, B6-TLR3tm1Flv, B6-TLR7tm1Aki, NOD/ShiLtJ), helper T-cell- (CD4tm1Mak), cytotoxic T-cell- (CD8atm1Mak), B-cell- (Igh-Jtm1DhuN?+N2), combined T- and B-cell- (NU/J) and combined T-, B-, natural killer (NK) cell- and macrophage-mediated immunity (NOD.Cg-PrkdcscidIl2rgtm1WjI/SzJ (NSG) mice) were subcutaneously infected with RVFV MP-12. B6-IFNARtmAgt mice were the only strain to develop fatal disease due to RVFV-induced severe hepatocellular necrosis and apoptosis. Notably, no clinical disease and only mild multifocal hepatocellular necrosis and apoptosis were observed in NSG mice, while immunohistochemistry detected the RVFV antigen in the liver and the brain. No or low virus expression and no lesions were observed in the other mouse strains. Conclusively, the interferon type 1 response is essential for early control of RVFV replication and disease, whereas functional NK cells, macrophages and lymphocytes are essential for virus clearance.


Subject(s)
Adaptive Immunity , Immunity, Innate , Rift Valley Fever/immunology , Rift Valley fever virus/physiology , Animals , Apoptosis , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Liver/immunology , Liver/virology , Macrophages/immunology , Macrophages/virology , Male , Mice , Mice, Inbred NOD , Rift Valley Fever/genetics , Rift Valley Fever/physiopathology , Rift Valley Fever/virology , Rift Valley fever virus/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology
7.
Viruses ; 13(3)2021 02 26.
Article in English | MEDLINE | ID: mdl-33652597

ABSTRACT

In recent years, transcriptome profiling studies have identified changes in host splicing patterns caused by viral invasion, yet the functional consequences of the vast majority of these splicing events remain uncharacterized. We recently showed that the host splicing landscape changes during Rift Valley fever virus MP-12 strain (RVFV MP-12) infection of mammalian cells. Of particular interest, we observed that the host mRNA for Rio Kinase 3 (RIOK3) was alternatively spliced during infection. This kinase has been shown to be involved in pattern recognition receptor (PRR) signaling mediated by RIG-I like receptors to produce type-I interferon. Here, we characterize RIOK3 as an important component of the interferon signaling pathway during RVFV infection and demonstrate that RIOK3 mRNA expression is skewed shortly after infection to produce alternatively spliced variants that encode premature termination codons. This splicing event plays a critical role in regulation of the antiviral response. Interestingly, infection with other RNA viruses and transfection with nucleic acid-based RIG-I agonists also stimulated RIOK3 alternative splicing. Finally, we show that specifically stimulating alternative splicing of the RIOK3 transcript using a morpholino oligonucleotide reduced interferon expression. Collectively, these results indicate that RIOK3 is an important component of the mammalian interferon signaling cascade and its splicing is a potent regulatory mechanism capable of fine-tuning the host interferon response.


Subject(s)
Alternative Splicing/genetics , Protein Serine-Threonine Kinases/genetics , Rift Valley Fever/genetics , Rift Valley fever virus/genetics , Animals , Cell Line , Chlorocebus aethiops , Gene Expression Profiling/methods , HEK293 Cells , Humans , Interferon Type I/genetics , Rift Valley Fever/virology , Signal Transduction/genetics , Vero Cells , Viral Nonstructural Proteins/genetics , Virulence Factors/genetics , Virus Replication/genetics
8.
Mamm Genome ; 32(1): 30-37, 2021 02.
Article in English | MEDLINE | ID: mdl-33420513

ABSTRACT

Rift Valley fever (RVF) is an emerging viral zoonosis that primarily affects ruminants and humans. We have previously shown that wild-derived MBT/Pas mice are highly susceptible to RVF virus and that part of this phenotype is controlled by a locus located on distal Chromosome 11. Using congenic strains, we narrowed down the critical interval to a 530 kb region containing five protein-coding genes among which Rnf213 emerged as a potential candidate. We generated Rnf213-deficient mice by CRISPR/CAS9 on the C57BL/6 J background and showed that they were significantly more susceptible to RVF than control mice, with an average survival time post-infection reduced from 7 to 4 days. The human RNF213 gene had been associated with the cerebrovascular Moyamoya disease (MMD or MYMY) but the inactivation of this gene in the mouse resulted only in mild anomalies of the neovascularization. This study provides the first evidence that the Rnf213 gene may also impact the resistance to infectious diseases such as RVF.


Subject(s)
Adenosine Triphosphatases/genetics , Disease Resistance/genetics , Host-Pathogen Interactions/genetics , Rift Valley Fever/genetics , Rift Valley Fever/virology , Rift Valley fever virus/physiology , Ubiquitin-Protein Ligases/genetics , Animals , CRISPR-Cas Systems , Chromosome Mapping , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
Sci Rep ; 10(1): 8734, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32457349

ABSTRACT

Infection of mice with Rift Valley fever virus (RVFV) reproduces major pathological features of severe human disease, notably the early-onset hepatitis and delayed-onset encephalitis. We previously reported that the Rvfs2 locus from the susceptible MBT/Pas strain reduces survival time after RVFV infection. Here, we used BALB/cByJ (BALB) mice congenic for Rvfs2 (C.MBT-Rvfs2) to investigate the pathophysiological mechanisms impacted by Rvfs2. Clinical, biochemical and histopathological features indicated similar liver damage in BALB and C.MBT-Rvfs2 mice until day 5 after infection. However, while C.MBT-Rvfs2 mice succumbed from acute liver injury, most BALB mice recovered and died later of encephalitis. Hepatocytes of BALB infected liver proliferated actively on day 6, promoting organ regeneration and recovery from liver damage. By comparison with C.MBT-Rvfs2, BALB mice had up to 100-fold lower production of infectious virions in the peripheral blood and liver, strongly decreased RVFV protein in liver and reduced viral replication in primary cultured hepatocytes, suggesting that the BALB Rvfs2 haplotype limits RVFV pathogenicity through decreased virus replication. Moreover, bone marrow chimera experiments showed that both hematopoietic and non-hematopoietic cells are required for the protective effect of the BALB Rvfs2 haplotype. Altogether, these results indicate that Rvfs2 controls critical events which allow survival to RVFV-induced hepatitis.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Genetic Loci , Hepatitis/mortality , Infectious Encephalitis/mortality , Rift Valley Fever/genetics , Rift Valley fever virus/pathogenicity , Animals , Cell Proliferation , Disease Models, Animal , Disease Susceptibility , Hepatitis/virology , Humans , Infectious Encephalitis/virology , Liver/cytology , Liver/virology , Male , Mice , Mice, Congenic , Mice, Inbred BALB C , Rift Valley Fever/complications , Rift Valley Fever/mortality
10.
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
11.
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
12.
Sci Rep ; 7(1): 7096, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28769107

ABSTRACT

Rift Valley fever virus (RVFV) leads to varied clinical manifestations in animals and in humans that range from moderate fever to fatal illness, suggesting that host immune responses are important determinants of the disease severity. We investigated the immune basis for the extreme susceptibility of MBT/Pas mice that die with mild to acute hepatitis by day 3 post-infection compared to more resistant BALB/cByJ mice that survive up to a week longer. Lower levels of neutrophils observed in the bone marrow and blood of infected MBT/Pas mice are unlikely to be causative of increased RVFV susceptibility as constitutive neutropenia in specific mutant mice did not change survival outcome. However, whereas MBT/Pas mice mounted an earlier inflammatory response accompanied by higher amounts of interferon (IFN)-α in the serum compared to BALB/cByJ mice, they failed to prevent high viral antigen load. Several immunological alterations were uncovered in infected MBT/Pas mice compared to BALB/cByJ mice, including low levels of leukocytes that expressed type I IFN receptor subunit 1 (IFNAR1) in the blood, spleen and liver, delayed leukocyte activation and decreased percentage of IFN-γ-producing leukocytes in the blood. These observations are consistent with the complex mode of inheritance of RVFV susceptibility in genetic studies.


Subject(s)
Immunity, Innate , Rift Valley Fever/immunology , Rift Valley Fever/virology , Rift Valley fever virus/immunology , Animals , Antigens, Viral/immunology , Disease Models, Animal , Disease Susceptibility , Hepatitis, Viral, Animal/genetics , Hepatitis, Viral, Animal/immunology , Hepatitis, Viral, Animal/virology , Leukocyte Count , Liver/immunology , Male , Mice , Mice, Inbred BALB C , Neutrophils/immunology , Neutrophils/metabolism , Rift Valley Fever/genetics , Rift Valley Fever/pathology , Spleen/immunology
13.
Acta Trop ; 169: 43-50, 2017 May.
Article in English | MEDLINE | ID: mdl-28126462

ABSTRACT

Culex quinquefasciatus (Diptera: Culicidae) is a vector of many pathogens and parasites of humans, as well as domestic and wild animals. In urban and semi-urban Asian countries, Cx. quinquefasciatus is a main vector of nematodes causing lymphatic filariasis. In the African region, it vectors the Rift Valley fever virus, while in the USA it transmits West Nile, St. Louis encephalitis and Western equine encephalitis virus. In this study, DNA barcoding was used to explore the genetic variation of Cx. quinquefasciatus populations from 88 geographical regions. We presented a comprehensive approach analyzing the effectiveness of two gene markers, i.e. CO1 and 16S rRNA. The high threshold genetic divergence of CO1 (0.47%) gene was reported as an ideal marker for molecular identification of this mosquito vector. Furthermore, null substitutions were lower in CO1 if compared to 16S rRNA, which influenced its differentiating potential among Indian haplotypes. NJ tree was well supported with high branch values for CO1 gene than 16S rRNA, indicating ideal genetic differentiation among haplotypes. TCS haplotype network revealed 14 distinct clusters. The intra- and inter-population polymorphism were calculated among the global and Indian Cx. quinquefasciatus lineages. The genetic diversity index Tajima' D showed negative values for all the 4 intra-population clusters (G2-4, G10). Fu's FS showed negative value for G10 cluster, which was significant and indicated recent population expansion. However, the G2-G4 (i.e. Indian lineages) had positive values, suggesting a bottleneck effect. Overall, our research firstly shed light on the genetic differences among the haplotypes of Cx. quinquefasciatus species complex, adding basic knowledge to the molecular ecology of this important mosquito vector.


Subject(s)
Culex/genetics , DNA Barcoding, Taxonomic/methods , Filariasis/epidemiology , Genetic Variation , Haplotypes/genetics , Insect Vectors/genetics , Rift Valley Fever/epidemiology , Rift Valley fever virus/pathogenicity , Animals , Filariasis/genetics , Humans , India , RNA, Ribosomal, 16S/genetics , Rift Valley Fever/genetics , Rift Valley fever virus/genetics
14.
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
15.
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
16.
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
17.
PLoS One ; 11(1): e0147027, 2016.
Article in English | MEDLINE | ID: mdl-26783758

ABSTRACT

Rift Valley fever Virus (RVFV), a negative-stranded RNA virus, is the etiological agent of the vector-borne zoonotic disease, Rift Valley fever (RVF). In both humans and livestock, protective immunity can be achieved through vaccination. Earlier and more recent vaccine trials in cattle and sheep demonstrated a strong neutralizing antibody and total IgG response induced by the RVF vaccine, authentic recombinant MP-12 (arMP-12). From previous work, protective immunity in sheep and cattle vaccinates normally occurs from 7 to 21 days after inoculation with arMP-12. While the serology and protective response induced by arMP-12 has been studied, little attention has been paid to the underlying molecular and genetic events occurring prior to the serologic immune response. To address this, we isolated RNA from whole blood of vaccinated calves over a time course of 21 days before and after vaccination with arMP-12. The time course RNAs were sequenced by RNASeq and bioinformatically analyzed. Our results revealed time-dependent activation or repression of numerous gene ontologies and pathways related to the vaccine induced immune response and its regulation. Additional bioinformatic analyses identified a correlative relationship between specific host immune response genes and protective immunity prior to the detection of protective serum neutralizing antibody responses. These results contribute an important proof of concept for identifying molecular and genetic components underlying the immune response to RVF vaccination and protection prior to serologic detection.


Subject(s)
Cattle Diseases/genetics , Gene Expression Profiling/methods , Rift Valley Fever/genetics , Rift Valley fever virus/immunology , Sequence Analysis, RNA/methods , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Cattle , Cattle Diseases/immunology , Cattle Diseases/prevention & control , Cattle Diseases/virology , Computational Biology/methods , Gene Expression Regulation , Gene Regulatory Networks , Rift Valley Fever/immunology , Rift Valley Fever/prevention & control , Seroconversion , Time Factors
18.
J Hered ; 106(6): 728-33, 2015.
Article in English | MEDLINE | ID: mdl-26546799

ABSTRACT

The Rift Valley Fever virus (RVFV) presents an epidemic and epizootic threat in sub-Saharan Africa, Egypt, and the Arabian Peninsula, and has furthermore recently gained attention as a potential weapon of bioterrorism due to its ability to infect both livestock and humans. Inbred rat strains show similar characteristic responses to the disease as humans and livestock, making them a suitable model species. Previous studies had indicated differences in susceptibility to RVFV hepatic disease among various rat strains, including a higher susceptibility of Wistar-Furth (WF) compared to a more resistant Lewis (LEW) strain. Further study revealed that this resistance trait exhibits the pattern of a major dominant gene inherited in Mendelian fashion. A genome scan of a congenic WF.LEW strain, created from the susceptible WF and resistant LEW strains and itself resistant to infection with RVFV, revealed 2 potential regions for the location of the gene, 1 on chromosome 3 and the other on chromosome 9. Through backcrossing of WF.LEW rats to WF rats, genotyping offspring using SNPs and microsatellites, and viral challenges of 3 N1 litters, we have mapped the gene to the distal end of chromosome 3.


Subject(s)
Chromosome Mapping , Disease Resistance/genetics , Rift Valley Fever/genetics , Animals , Animals, Congenic , Crosses, Genetic , Female , Genes, Dominant , Genetic Markers , Genotype , Haplotypes , Male , Microsatellite Repeats , Polymorphism, Single Nucleotide , Rats , Rats, Inbred Lew , Rats, Inbred WF , Rift Valley fever virus , Sequence Analysis, DNA
19.
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
20.
Sci Rep ; 5: 9492, 2015 Mar 25.
Article in English | MEDLINE | ID: mdl-25804974

ABSTRACT

Rift Valley fever (RVF) is a zoonotic and vector-borne disease, mainly present in Africa, which represents a threat to human health, animal health and production. South Africa has experienced three major RVF epidemics (1950-51, 1973-75 and 2008-11). Due to data scarcity, no previous study has quantified risk factors associated with RVF epidemics in animals in South Africa. Using the 2008-11 epidemic datasets, a retrospective longitudinal study was conducted to identify and quantify spatial and temporal environmental factors associated with RVF incidence. Cox regressions with a Besag model to account for the spatial effects were fitted to the data. Coefficients were estimated by Bayesian inference using integrated nested Laplace approximation. An increase in vegetation density was the most important risk factor until 2010. In 2010, increased temperature was the major risk factor. In 2011, after the large 2010 epidemic wave, these associations were reversed, potentially confounded by immunity in animals, probably resulting from earlier infection and vaccination. Both vegetation density and temperature should be considered together in the development of risk management strategies. However, the crucial need for improved access to data on population at risk, animal movements and vaccine use is highlighted to improve model predictions.


Subject(s)
Epidemics , Rift Valley Fever/epidemiology , Humans , Rift Valley Fever/genetics , Risk Factors , South Africa
SELECTION OF CITATIONS
SEARCH DETAIL
...