29 research outputs found

    Comparison of Rift Valley fever virus replication in North American livestock and wildlife cell lines

    Get PDF
    Citation: Gaudreault, N. N., Indran, S. V., Bryant, P. K., Richt, J. A., & Wilson, W. C. (2015). Comparison of Rift Valley fever virus replication in North American livestock and wildlife cell lines. Frontiers in Microbiology, 6(JUN). doi:10.3389/fmicb.2015.00664Rift Valley fever virus (RVFV) causes disease outbreaks across Africa and the Arabian Peninsula, resulting in high morbidity and mortality among young domestic livestock, frequent abortions in pregnant animals, and potentially severe or fatal disease in humans. The possibility of RVFV spreading to the United States or other countries worldwide is of significant concern to animal and public health, livestock production, and trade. The mechanism for persistence of RVFV during inter-epidemic periods may be through mosquito transovarial transmission and/or by means of a wildlife reservoir. Field investigations in endemic areas and previous in vivo studies have demonstrated that RVFV can infect a wide range of animals, including indigenous wild ruminants of Africa. Yet no predominant wildlife reservoir has been identified, and gaps in our knowledge of RVFV permissive hosts still remain. In North America, domestic goats, sheep, and cattle are susceptible hosts for RVFV and several competent vectors exist. Wild ruminants such as deer might serve as a virus reservoir and given their abundance, wide distribution, and overlap with livestock farms and human populated areas could represent an important risk factor. The objective of this study was to assess a variety of cell lines derived from North American livestock and wildlife for susceptibility and permissiveness to RVFV. Results of this study suggest that RVFV could potentially replicate in native deer species such as white-tailed deer, and possibly a wide range of non-ruminant animals. This work serves to guide and support future animal model studies and risk model assessment regarding this high-consequence zoonotic pathogen. © 2015 Gaudreault, Indran, Bryant, Richt and Wilson

    Whole genome sequencing and phylogenetic analysis of \u3ci\u3eBluetongue virus\u3c/i\u3e serotype 2 strains isolated in the Americas including a novel strain from the western United States

    Get PDF
    Bluetongue is a potentially fatal arboviral disease of domestic and wild ruminants that is characterized by widespread edema and tissue necrosis. Bluetongue virus (BTV) serotypes 10, 11, 13, and 17 occur throughout much of the United States, whereas serotype 2 (BTV-2) was previously only detected in the southeastern United States. Since 1998, 10 other BTV serotypes have also been isolated from ruminants in the southeastern United States. In 2010, BTV-2 was identified in California for the first time, and preliminary sequence analysis indicated that the virus isolate was closely related to BTV strains circulating in the southeastern United States. In the current study, the whole genome sequence of the California strain of BTV-2 was compared with those of other BTV-2 strains in the Americas. The results of the analysis suggest co-circulation of genetically distinct viruses in the southeastern United States, and further suggest that the 2010 western isolate is closely related to southeastern strains of BTV. Although it remains uncertain as to how this novel virus was translocated to California, the findings of the current study underscore the need for ongoing surveillance of this economically important livestock disease

    A Recombinant Rift Valley Fever Virus Glycoprotein Subunit Vaccine Confers Full Protection against Rift Valley Fever Challenge in Sheep

    Get PDF
    Citation: Faburay, B., Wilson, W. C., Gaudreault, N. N., Davis, A. S., Shivanna, V., Bawa, B., . . . Richt, J. A. (2016). A Recombinant Rift Valley Fever Virus Glycoprotein Subunit Vaccine Confers Full Protection against Rift Valley Fever Challenge in Sheep. Scientific Reports, 6, 12. doi:10.1038/srep27719Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic pathogen causing disease outbreaks in Africa and the Arabian Peninsula. The virus has great potential for transboundary spread due to the presence of competent vectors in non-endemic areas. There is currently no fully licensed vaccine suitable for use in livestock or humans outside endemic areas. Here we report the evaluation of the efficacy of a recombinant subunit vaccine based on the RVFV Gn and Gc glycoproteins. In a previous study, the vaccine elicited strong virus neutralizing antibody responses in sheep and was DIVA (differentiating naturally infected from vaccinated animals) compatible. In the current efficacy study, a group of sheep (n=5) was vaccinated subcutaneously with the glycoprotein-based subunit vaccine candidate and then subjected to heterologous challenge with the virulent Kenya-128B-15 RVFV strain. The vaccine elicited high virus neutralizing antibody titers and conferred complete protection in all vaccinated sheep, as evidenced by prevention of viremia, fever and absence of RVFV-associated histopathological lesions. We conclude that the subunit vaccine platform represents a promising strategy for the prevention and control of RVFV infections in susceptible hosts

    Evaluation of a viral DNA-protein immunization strategy against African swine fever in domestic pigs

    Get PDF
    African swine fever virus (ASFV) causes serious disease in domestic pigs for which there is no vaccine currently available. ASFV is a large DNA virus that encodes for more than 150 proteins, thus making the identification of viral antigens that induce a protective immune response difficult. Based on the functional roles of several ASFV proteins found in previous studies, we selected combinations of ASFV recombinant proteins and pcDNAs-expressing ASFV genes, to analyze their ability to induce humoral and cellular immune responses in pigs. Pigs were immunized using a modified prime-boost approach with combinations of previously selected viral DNA and proteins, resulting in induction of antibodies and specific cell-mediated immune response, measured by IFN-γ ELISpots. The ability of antibodies from pigs immunized with various combinations of ASFV-specific antigens to neutralize infection in vitro, and antigen-specific activation of the cellular immune response were analyzed.U.S. Department of Homeland Security under Grant Award Number DHS-2010-ST-061-AG0001 for the Center of Excellence for Emerging and Zoonotic Animal Disease (CEEZAD) and the State of Kansas National Bio and Agro-Defense Facility (NBAF

    Experimental infection of calves by two genetically-distinct strains of rift valley fever virus

    Get PDF
    Citation: Wilson, W. C., Davis, A. S., Gaudreault, N. N., Faburay, B., Trujillo, J. D., Shivanna, V., . . . Richt, J. A. (2016). Experimental infection of calves by two genetically-distinct strains of rift valley fever virus. Viruses, 8(5). doi:10.3390/v8050145Additional Authors: McVey, D. S.Recent outbreaks of Rift Valley fever in ruminant livestock, characterized by mass abortion and high mortality rates in neonates, have raised international interest in improving vaccine control strategies. Previously, we developed a reliable challenge model for sheep that improves the evaluation of existing and novel vaccines in sheep. This sheep model demonstrated differences in the pathogenesis of Rift Valley fever virus (RVFV) infection between two genetically-distinct wild-type strains of the virus, Saudi Arabia 2001 (SA01) and Kenya 2006 (Ken06). Here, we evaluated the pathogenicity of these two RVFV strains in mixed breed beef calves. There was a transient increase in rectal temperatures with both virus strains, but this clinical sign was less consistent than previously reported with sheep. Three of the five Ken06-infected animals had an early-onset viremia, one day post-infection (dpi), with viremia lasting at least three days. The same number of SA01-infected animals developed viremia at 2 dpi, but it only persisted through 3 dpi in one animal. The average virus titer for the SA01-infected calves was 1.6 logs less than for the Ken06-infected calves. Calves, inoculated with either strain, seroconverted by 5 dpi and showed time-dependent increases in their virus-neutralizing antibody titers. Consistent with the results obtained in the previous sheep study, elevated liver enzyme levels, more severe liver pathology and higher virus titers occurred with the Ken06 strain as compared to the SA01 strain. These results demonstrate the establishment of a virulent challenge model for vaccine evaluation in calves. © 2016 by the authors; licensee MDPI, Basel, Switzerland

    Persistence of African Swine Fever Virus in Feed and Feed Mill Environment over Time after Manufacture of Experimentally Inoculated Feed

    Get PDF
    To reduce the risk of disease from harmful feed-based pathogens, some feed manufacturers quarantine high-risk ingredients prior to their inclusion in feed. Data exist that confirms this practice is effective, but to our knowledge there is no information about porcine pathogen survival in mill environments. The objective of this study was to determine survival of African swine fever virus (ASFV) in swine feed and on mill surfaces after manufacture of experimentally inoculated swine feed. A pilot-scale feed mill was placed within a biosecurity level (BSL) 3 facility to manufacture batches of feed. The priming batch, Batch 1, was ASFV-free feed and was followed with Batch 2 which was experimentally inoculated with ASFV (5.6 × 104 TCID50/gram). Four subsequent ASFV-free batches were then manufactured (Batch 3-6). After each batch of feed, 10 feed samples were aseptically collected in a double ‘X’ pattern. During feed manufacturing, 24 steel coupons were placed on the floor of the manufacturing area and feed dust was allowed to settle onto them overnight. Once feed manufacturing was completed, feed samples and steel coupons were stored at room temperature. On the day of (day 0) and d 3, 7, 14, 28, 60, 90, and 180 after feed manufacturing, feed samples and 3 steel coupons were randomly selected, taken out of storage, and analyzed for ASFV DNA. For feed samples there was a statistically significant (P = 0.023) batch × day interaction for log10 genomic copies per gram of feed, and a marginal statistical significance (P = 0.072) for batch × day interaction for cycle threshold (Ct) values. This indicates that the batch of feed and days held at room temperature impacted the amount of the detectable ASFV DNA in feed samples. There was no evidence (P = 0.433) of ASFV degradation on environmental coupons over the 180-d storage period. This study found that quarantine time can help reduce, but not eliminate ASFV DNA in feed over time. Surprisingly, ASFV DNA is detectable on feed manufacturing surfaces for at least 180 days

    Prevalence and Distribution of African Swine Fever Virus in Swine Feed After Mixing and Feed Batch Sequencing

    Get PDF
    As the United States maintains trade with countries where African swine fever virus (ASFV) is endemic, it is critical to have methods that can detect and mitigate the risk of ASFV in potentially contaminated feed or ingredients. Therefore, the objectives of this study were to 1) evaluate feed batch sequencing as a mitigation technique for ASFV contamination in a feed mill, and 2) determine if a feed sampling method could identify ASFV following experimental inoculation. Batches of feed were manufactured in a BSL-3Ag room at Kansas State University’s Biosafety Research Institute in Manhattan, KS. First, the pilot feed manufacturing system mixed, conveyed, and discharged an ASFV-free diet. Next, a diet was manufactured using the same equipment, but contained feed inoculated with ASFV for a final concentration of 5.6 × 104 TCID50/g. Then, four subsequent ASFV-free batches of feed were manufactured. After discharging each batch into a biohazard tote, 10 samples were collected in a double ‘X’ pattern. Samples were analyzed using a qPCR assay specific for the ASFV p72 gene to determine the cycle threshold (Ct) and log10 genomic copy number (CN)/g of feed. Batch of feed affected the qPCR Ct values (P \u3c 0.0001) and the log10 genomic CN/g (P \u3c 0.0001) content of feed. Feed samples obtained after manufacturing the ASFV-contaminated diet contained the greatest (P \u3c 0.05) amounts of ASFV p72 DNA across all criteria. Quantity of ASFV p72 DNA decreased sequentially as additional batches of initially ASFV-free feed were manufactured, but it was still detectable after batch sequence 4, suggesting cross contamination between batches. This subsampling method was able to identify ASFV genetic material in feed samples using the PCR assay specific for the ASFV p72 gene. In summary, sequencing batches of feed decreases concentration of ASFV contamination in feed, but does not eliminate it. Bulk ingredients or feed can be accurately evaluated for ASFV contamination by collecting 10 evenly distributed subsamples, representing 0.05% of the volume of the container, using the sampling method described herein

    Effect of mixing and feed batch sequencing on the prevalence and distribution of African swine fever virus in swine feed

    Get PDF
    It is critical to have methods that can detect and mitigate the risk of African swine fever virus (ASFV) in potentially contaminated feed or ingredients bound for the United States. The purpose of this work was to evaluate feed batch sequencing as a mitigation technique for ASFV contamination in a feed mill, and to determine if a feed sampling method could identify ASFV following experimental inoculation. Batches of feed were manufactured in a BSL-3Ag room at Kansas State University's Biosafety Research Institute in Manhattan, Kansas. First, the pilot feed manufacturing system mixed, conveyed, and discharged an ASFV-free diet. Next, a diet was manufactured using the same equipment, but contained feed inoculated with ASFV for final concentration of 5.6 × 104 TCID50/g. Then, four subsequent ASFV-free batches of feed were manufactured. After discharging each batch into a collection container, 10 samples were collected in a double ‘X’ pattern. Samples were analysed using a qPCR assay for ASFV p72 gene then the cycle threshold (Ct) and Log10 genomic copy number (CN)/g of feed were determined. The qPCR Ct values (p < .0001) and the Log10 genomic CN/g (p < .0001) content of feed samples were impacted based on the batch of feed. Feed samples obtained after manufacturing the ASFV-contaminated diet contained the greatest amounts of ASFV p72 DNA across all criteria (p < .05). Quantity of ASFV p72 DNA decreased sequentially as additional batches of feed were manufactured, but was still detectable after batch sequence 4. This subsampling method was able to identify ASFV genetic material in feed samples using p72 qPCR. In summary, sequencing batches of feed decreases concentration of ASFV contamination in feed, but does not eliminate it. Bulk ingredients can be accurately evaluated for ASFV contamination by collecting 10 subsamples using the sampling method described herein. Future research is needed to evaluate if different mitigation techniques can reduce ASFV feed contamination

    Evaluating the distribution of African swine fever virus within a feed mill environment following manufacture of inoculated feed

    Get PDF
    11 Pág. Centro de Investigación en Sanidad Animal (CISA)It is critical to understand the role feed manufacturing may have regarding potential African swine fever virus (ASFV) transmission, especially given the evidence that feed and/or ingredients may be potential vectors. The objective of the study was to evaluate the distribution of ASFV in a feed mill following manufacture of contaminated feed. To accomplish this, a pilot-scale feed mill consisting of a mixer, bucket elevator, and spouting was constructed in a BSL-3Ag facility. First, a batch of ASFV-free feed was manufactured, followed by a batch of feed that had an ASFV-contaminated ingredient added to feed, which was then mixed and discharged from the equipment. Subsequently, four additional ASFV-free batches of feed were manufactured using the same equipment. Environmental swabs from 18 locations within the BSL-3Ag room were collected after each batch of feed was discharged. The locations of the swabs were categorized into four zones: 1) feed contact surface, 2) non-feed contact surface 1 meter from feed, and 4) transient surfaces. Environmental swabs were analyzed using a qPCR specific for the ASFV p72 gene and reported as genomic copy number (CN)/mL of environmental swab processing buffer. Genomic copies were transformed with a log10 function for statistical analysis. There was no evidence of a zone × batch interaction for log10 genomic CN/mL (P = 0.625) or cycle threshold (Ct) value (P = 0.608). Sampling zone impacted the log10 p72 genomic CN/mL (P < 0.0001) and Ct values (P < 0.0001), with a greater amount of viral genome detected on transient surfaces compared to other surfaces (P < 0.05). This study illustrates that once ASFV enters the feed mill environment it becomes widespread and movement of people can significantly contribute to the spread of ASFV in a feed mill environment.Funding for this work was obtained from the NBAF Transition Funds from the state of Kansas (JAR), the National Pork Board under award number 20-018 (CKJ), the Department of Homeland Security Center of Excellence for Emerging and Zoonotic Animal Diseases under grant number HSHQDC 16-A-B0006 (JAR), and the AMP Core of the NIGMS COBRE Center on Emerging and Zoonotic Infectious Diseases (CEZID) under award number P20GM13044 (JAR)Peer reviewe

    Investigation of Bovine Herpesvirus 1 (BHV-1) Encoded Infected Cell Protein 0 (bICP0)

    Get PDF
    Bovine herpesvirus 1 (BHV-1) is a significant pathogen of cattle. Following acute infection, BHV-1 establishes a latent infection that persists for the life of the infected host. Stress induced factors cause the virus to reactivate from latency, resulting in virus transmission and transient immune suppression. BHV-1 encoded bICP0 is expressed early and constitutively throughout productive infection. bICP0 is critical for efficient viral replication, virulence, and reactivation in cattle because it stimulates viral transcription and interferes with innate immune responses. bICP0 potentially interacts with a variety of proteins to activate viral gene expression and inhibit innate antiviral defenses. bICP0 localizes to promyelocytic leukemia (PML) protein-containing nuclear domains, which are associated with antiviral activity and commonly targeted for disruption by a wide variety of viruses. The zinc RING finger motif within bICP0 plays a critical role in the biological functions of bICP0, and possesses intrinsic E3 ubiquitin ligase activity that is important for polyubiquitination and subsequent degradation of proteins. Results from these studies demonstrated mutations within the zinc RING finger increased bICP0 protein levels, presumably due to disruption of the ability of bICP0 to induce its own ubiquitination. Sequences at its C-terminus are also important for regulating the half-life of bICP0. BHV-1 infection and bICP0 expression alone reduced PML protein levels. Unexpectedly, bICP0 mutants that localized primarily in the cytoplasm also induced PML degradation. bICP0 was readily detected in the cytoplasm of low passage bovine cells at later times post infection, suggesting bICP0 induces degradation of cytoplasmic isoforms of PML to promote viral replication. Identification of bICP0-interacting proteins was also investigated to further elucidate the mechanisms underlying bICP0 functions. The ability of BHV-1 and a bICP0 zinc RING finger mutant to grow in oncogenic cells was also examined to determine if defects in viral growth of the mutant could be relieved in cells with potential defects in their interferon response. Collectively, studies presented in this dissertation determine that nuclear and cytoplasmic bICP0 have functions that promote productive infection. Advisor: Clinton Jone
    corecore