Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Front Vet Sci ; 7: 554305, 2020.
Article in English | MEDLINE | ID: mdl-33088833

ABSTRACT

Inactivated, wild-type foot-and-mouth disease virus (FMDV) vaccines are currently used to control FMD around the world. These traditional FMD vaccines are produced using large quantities of infectious, virulent, wild-type FMD viruses, with the associated risk of virus escape from manufacturing facilities or incomplete inactivation during the vaccine formulation process. While higher quality vaccines produced from wild-type FMDV are processed to reduce non-structural antigens, there is still a risk that small amounts of non-structural proteins may be present in the final product. A novel, antigenically marked FMD-LL3B3D vaccine platform under development by Zoetis, Inc. and the USDA-ARS, consists of a highly attenuated virus platform containing negative antigenic markers in the conserved non-structural proteins 3Dpol and 3B that render resultant vaccines fully DIVA compatible. This vaccine platform allows for the easy exchange of capsid coding sequences to create serotype-specific vaccines. Here we demonstrate the efficacy of the inactivated FMD-LL3B3D-A24 Cruzeiro vaccine in cattle against wild-type challenge with A24 Cruzerio. A proprietary adjuvant system was used to formulate the vaccines that conferred effective protection at low doses while maintaining the DIVA compatibility. In contrast to wild-type FMDV, the recombinant FMD-LL3B3D mutant viruses have been shown to induce no clinical signs of FMD and no shedding of virus in cattle or pigs when inoculated as a live virus. The FMD-LL3B3D vaccine platform, currently undergoing development in the US, provides opportunities for safer vaccine production with full DIVA compatibility in support of global FMDV control and eradication initiatives.

2.
Vaccine ; 37(42): 6221-6231, 2019 09 30.
Article in English | MEDLINE | ID: mdl-31493951

ABSTRACT

Foot-and-mouth disease (FMD) is a highly contagious viral infection of cloven hooved animals that continues to cause economic disruption in both endemic countries or when introduced into a formally FMD free country. Vaccines that protect against clinical disease and virus shedding are critical to control FMD. The replication deficient human adenovirus serotype 5 (Ad5) vaccine vector expressing empty FMD virus (FMDV) capsid, AdtFMD, is a promising new vaccine platform. With no shedding or spreading of viral vector detected in field trials, this vaccine is very safe to manufacture, as there is no requirement for high containment faciitites. Here, we describe three studies assessing the proportion of animals protected from clinical vesicular disease (foot lesions) following live-FMDV challenge by intradermolingual inoculation at 6 or 9 months following a single vaccination with the commercial AdtFMD vaccine, provisionally licensed for cattle in the United States. Further, we tested the effect of vaccination route (transdermal, intramuscular, subcutaneous) on clinical outcome and humoral immunity. Results demonstrate that a single dose vaccination in cattle with the commercial vaccine vector expressing capsid proteins of the FMDV strain A24 Cruzeiro (Adt.A24), induced protection against clinical FMD at 6 months (100% transdermal, 80% intramuscular, and 60% subcutaneous) that waned by 9 months post-vaccination (33% transdermal and 20% intramuscular). Post-vaccination serum from immunized cattle (all studies) generally contained FMDV specific neutralizing antibodies by day 14. Anti-FMDV antibody secreting cells are detected in peripheral blood early following vaccination, but are absent after 28 days post-vaccination. Thus, the decay in antibody mediated immunity over time is likely a function of FMDV-specific antibody half-life. These data reveal the short time span of anti-FMDV antibody secreting cells (ASCs) and important performance characteristics of needle-free vaccination with a recombinant vectored subunit vaccine for FMDV.


Subject(s)
Cattle Diseases/prevention & control , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/prevention & control , Vaccination/veterinary , Vaccines, Subunit/immunology , Viral Vaccines/immunology , Adenoviruses, Human/genetics , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Capsid Proteins/immunology , Cattle , Cattle Diseases/virology , Genetic Vectors , Immunity, Humoral/immunology , Vaccines, Synthetic/immunology
3.
PLoS One ; 14(12): e0227061, 2019.
Article in English | MEDLINE | ID: mdl-31891626

ABSTRACT

Foot-and-mouth disease virus (FMDV) is capable of infecting all cloven-hoofed domestic livestock species, including cattle, pigs, goats, and sheep. However, in contrast to cattle and pigs, the pathogenesis of FMDV in small ruminants has been incompletely elucidated. The objective of the current investigation was to characterize tissue- and cellular tropism of early and late stages of FMDV infection in sheep following three different routes of simulated natural virus exposure. Extensive post-mortem harvest of tissue samples at pre-determined time points during early infection (24 and 48 hours post infection) demonstrated that tissues specifically susceptible to primary FMDV infection included the paraepiglottic- and palatine tonsils, as well as the nasopharyngeal mucosa. Additionally, experimental aerosol inoculation of sheep led to substantial virus replication in the lungs at 24-48 hours post-inoculation. During persistent infection (35 days post infection), the paraepiglottic- and palatine tonsils were the only tissues from which infectious FMDV was recovered. This is strikingly different from cattle, in which persistent FMDV infection has consistently been located to the nasopharyngeal mucosa. Analysis of tissue sections by immunomicroscopy revealed a strict epithelial tropism during both early and late phases of infection as FMDV was consistently localized to cytokeratin-expressing epithelial cells. This study expands upon previous knowledge of FMDV pathogenesis in sheep by providing detailed information on the temporo-anatomic distribution of FMDV in ovine tissues. Findings are discussed in relation to similar investigations previously performed in cattle and pigs, highlighting similarities and differences in FMDV pathogenesis across natural host species.


Subject(s)
Adenoids/virology , Foot-and-Mouth Disease Virus/pathogenicity , Foot-and-Mouth Disease/virology , Palatine Tonsil/virology , Sheep/virology , Adenoids/pathology , Animals , Cattle , Foot-and-Mouth Disease/pathology , Foot-and-Mouth Disease Virus/isolation & purification , Male , Palatine Tonsil/pathology , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Species Specificity , Swine , Virulence , Virus Replication
4.
Front Microbiol ; 9: 1891, 2018.
Article in English | MEDLINE | ID: mdl-30158915

ABSTRACT

Vesicular stomatitis virus (VSV) causes sporadic outbreaks of vesicular disease in the southwestern United States. The intrinsic characteristics of epidemic strains associated with these outbreaks are poorly understood. In this study, we report the distinctive genomic and biological characteristics of an epidemic (NJ0612NME6) strain of VSV compared with an endemic (NJ0806VCB) strain. Genomic comparisons between the two strains revealed a total of 111 nucleotide differences (23 non-synonymous) with potentially relevant replacements located in the P, G, and L proteins. When tested in experimentally infected pigs, a natural host of VSV, the epidemic strain caused higher fever and an increased number of vesicular lesions compared to pigs infected with the endemic strain. Pigs infected with the epidemic strain showed decreased systemic antiviral activity (type I - IFN), lower antibody levels, higher levels of interleukin 6, and lower levels of tumor necrosis factor during the acute phase of disease compared to pigs infected with the endemic strain. Furthermore, we document the existence of an RNAemia phase in pigs experimentally infected with VSV and explored the cause for the lack of recovery of infectious virus from blood. Finally, the epidemic strain was shown to be more efficient in down-regulating transcription of IRF-7 in primary porcine macrophages. Collectively, the data shows that the epidemic strain of VSV we tested has an enhanced ability to modulate the innate immune response of the vertebrate host. Further studies are needed to examine other epidemic strains and what contributions a phenotype of increased virulence might have on the transmission of VSV during epizootics.

5.
Virology ; 516: 115-126, 2018 03.
Article in English | MEDLINE | ID: mdl-29346074

ABSTRACT

Deletions within the 3A coding region of foot-and-mouth disease virus (FMDV) are associated with decreased virulence in cattle; however, the mechanisms are unknown. We compared experimental infection of cattle with virulent FMDV O1Campos (O1Ca) and a mutant derivative (O1Ca∆3A) lacking residues 87-106 of 3A. Unexpectedly, primary infection of the nasopharyngeal mucosa was similar for both viruses. However, while O1Ca caused viremia and fulminant clinical disease, O1Ca∆3A infection was subclinical and aviremic. There were no differences in expression of anti-viral cytokines in nasopharyngeal tissues between the groups, suggesting attenuation by O1Ca∆3A was a consequence of reduced replication efficiency in bovine cells, rather than a difference in the host response. These results demonstrated that although deletion in 3A of FMDV confers a clinically attenuated phenotype in cattle, the deletion does not prevent subclinical infection. These findings have implications for field scenarios involving outbreaks with apparently host-limited strains of FMDV.


Subject(s)
Cattle Diseases/virology , Foot-and-Mouth Disease Virus/physiology , Foot-and-Mouth Disease/virology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Animals , Cattle , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/pathogenicity , Sequence Deletion , Virulence , Virus Replication
6.
J Virol ; 91(22)2017 11 15.
Article in English | MEDLINE | ID: mdl-28878081

ABSTRACT

The foot-and-mouth disease virus (FMDV) afflicts livestock in more than 80 countries, limiting food production and global trade. Production of foot-and-mouth disease (FMD) vaccines requires cytosolic expression of the FMDV 3C protease to cleave the P1 polyprotein into mature capsid proteins, but the FMDV 3C protease is toxic to host cells. To identify less-toxic isoforms of the FMDV 3C protease, we screened 3C mutants for increased transgene output in comparison to wild-type 3C using a Gaussia luciferase reporter system. The novel point mutation 3C(L127P) increased yields of recombinant FMDV subunit proteins in mammalian and bacterial cells expressing P1-3C transgenes and retained the ability to process P1 polyproteins from multiple FMDV serotypes. The 3C(L127P) mutant produced crystalline arrays of FMDV-like particles in mammalian and bacterial cells, potentially providing a practical method of rapid, inexpensive FMD vaccine production in bacteria.IMPORTANCE The mutant FMDV 3C protease L127P significantly increased yields of recombinant FMDV subunit antigens and produced virus-like particles in mammalian and bacterial cells. The L127P mutation represents a novel advancement for economical FMD vaccine production.


Subject(s)
Amino Acid Substitution , Cysteine Endopeptidases/immunology , Foot-and-Mouth Disease Virus/immunology , Mutation, Missense , Viral Proteins/immunology , Viral Vaccines/immunology , 3C Viral Proteases , Animals , Cysteine Endopeptidases/genetics , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease Virus/genetics , HEK293 Cells , Humans , Viral Proteins/genetics , Viral Vaccines/genetics
7.
Virol J ; 14(1): 89, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28464897

ABSTRACT

BACKGROUND: Understanding the mechanisms of attenuation and virulence of foot-and-mouth disease virus (FMDV) in the natural host species is critical for development of next-generation countermeasures such as live-attenuated vaccines. Functional genomics analyses of FMDV have identified few virulence factors of which the leader proteinase (Lpro) is the most thoroughly investigated. Previous work from our laboratory has characterized host factors in cattle inoculated with virulent FMDV and attenuated mutant strains with transposon insertions within Lpro. METHODS: In the current study, the characteristics defining virulence of FMDV in cattle were further investigated by comparing the pathogenesis of a mutant, attenuated strain (FMDV-Mut) to the parental, virulent virus from which the mutant was derived (FMDV-WT). The only difference between the two viruses was an insertion mutation in the inter-AUG region of the leader proteinase of FMDV-Mut. All cattle were infected by simulated-natural, aerosol inoculation. RESULTS: Both viruses were demonstrated to establish primary infection in the nasopharyngeal mucosa with subsequent dissemination to the lungs. Immunomicroscopic localization of FMDV antigens indicated that both viruses infected superficial epithelial cells of the nasopharynx and lungs. The critical differences between the two viruses were a more rapid establishment of infection by FMDV-WT and quantitatively greater virus loads in secretions and infected tissues compared to FMDV-Mut. The slower replicating FMDV-Mut established a subclinical infection that was limited to respiratory epithelial sites, whereas the faster replication of FMDV-WT facilitated establishment of viremia, systemic dissemination of infection, and clinical disease. CONCLUSION: The mutant FMDV was capable of achieving all the same early pathogenesis landmarks as FMDV-WT, but was unable to establish systemic infection. The precise mechanism of attenuation remains undetermined; but current data suggests that the impaired replication of the mutant is more responsible for attenuation than differences in host immunological factors. These results complement previous studies by providing data of high-granularity describing tissue-specific tropism of FMDV and by demonstrating microscopic localization of virulent and attenuated clones of the same field-strain FMDV.


Subject(s)
Cattle Diseases/virology , Foot-and-Mouth Disease Virus/pathogenicity , Foot-and-Mouth Disease/virology , Virulence , Aerosols , Animals , Cattle , Epithelial Cells/pathology , Epithelial Cells/virology , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/pathology , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/growth & development , Foot-and-Mouth Disease Virus/isolation & purification , Lung/virology , Mutagenesis, Insertional , Nasopharynx/pathology , Nasopharynx/virology , RNA, Viral/isolation & purification , Vaccines, Attenuated/immunology , Viral Structural Proteins , Virulence Factors , Virus Replication
8.
Front Vet Sci ; 3: 105, 2016.
Article in English | MEDLINE | ID: mdl-27917386

ABSTRACT

Understanding the quantitative characteristics of a pathogen's capability to transmit during distinct phases of infection is important to enable accurate predictions of the spread and impact of a disease outbreak. In the current investigation, the potential for transmission of foot-and-mouth disease virus (FMDV) during the incubation (preclinical) period of infection was investigated in seven groups of pigs that were sequentially exposed to a group of donor pigs that were infected by simulated-natural inoculation. Contact-exposed pigs were comingled with infected donors through successive 8-h time slots spanning from 8 to 64 h post-inoculation (hpi) of the donor pigs. The transition from latent to infectious periods in the donor pigs was clearly defined by successful transmission of foot-and-mouth disease (FMD) to all contact pigs that were exposed to the donors from 24 hpi and later. This onset of infectiousness occurred concurrent with detection of viremia, but approximately 24 h prior to the first appearance of clinical signs of FMD in the donors. Thus, the latent period of infection ended approximately 24 h before the end of the incubation period. There were significant differences between contact-exposed groups in the time elapsed from virus exposure to the first detection of FMDV shedding, viremia, and clinical lesions. Specifically, the onset and progression of clinical FMD were more rapid in pigs that had been exposed to the donor pigs during more advanced phases of disease, suggesting that these animals had received a higher effective challenge dose. These results demonstrate transmission and dissemination of FMD within groups of pigs during the incubation period of infection. Furthermore, these findings suggest that under current conditions, shedding of FMDV in oropharyngeal fluids is a more precise proxy for FMDV infectiousness than clinical signs of infection. These findings may impact modeling of the propagation of FMD outbreaks that initiate in pig holdings and should be considered when designing FMD control strategies.

9.
PLoS One ; 11(9): e0162750, 2016.
Article in English | MEDLINE | ID: mdl-27643611

ABSTRACT

In order to investigate the mechanisms of persistent foot-and-mouth disease virus (FMDV) infection in cattle, transcriptome alterations associated with the FMDV carrier state were characterized using a bovine whole-transcriptome microarray. Eighteen cattle (8 vaccinated with a recombinant FMDV A vaccine, 10 non-vaccinated) were challenged with FMDV A24 Cruzeiro, and the gene expression profiles of nasopharyngeal tissues collected between 21 and 35 days after challenge were compared between 11 persistently infected carriers and 7 non-carriers. Carriers and non-carriers were further compared to 2 naïve animals that had been neither vaccinated nor challenged. At a controlled false-discovery rate of 10% and a minimum difference in expression of 50%, 648 genes were differentially expressed between FMDV carriers and non-carriers, and most (467) had higher expression in carriers. Among these, genes associated with cellular proliferation and the immune response-such as chemokines, cytokines and genes regulating T and B cells-were significantly overrepresented. Differential gene expression was significantly correlated between non-vaccinated and vaccinated animals (biological correlation +0.97), indicating a similar transcriptome profile across these groups. Genes related to prostaglandin E2 production and the induction of regulatory T cells were overexpressed in carriers. In contrast, tissues from non-carrier animals expressed higher levels of complement regulators and pro-apoptotic genes that could promote virus clearance. Based on these findings, we propose a working hypothesis for FMDV persistence in nasopharyngeal tissues of cattle, in which the virus may be maintained by an impairment of apoptosis and the local suppression of cell-mediated antiviral immunity by inducible regulatory T cells.


Subject(s)
Cattle Diseases/immunology , Cattle/virology , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/immunology , Immunity, Cellular , Transcriptome , Animals , Apoptosis , Cattle/genetics , Cattle/immunology , Cattle Diseases/genetics , Cattle Diseases/prevention & control , Cattle Diseases/virology , Foot-and-Mouth Disease/genetics , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/virology , Gene Expression Profiling , Immunity, Humoral , Immunity, Innate , Nasopharynx/immunology , Nasopharynx/metabolism , Nasopharynx/virology , Vaccination , Vaccines, Synthetic/immunology , Viral Vaccines/immunology
10.
BMC Vet Res ; 12: 205, 2016 Sep 15.
Article in English | MEDLINE | ID: mdl-27634113

ABSTRACT

BACKGROUND: In order to investigate host factors associated with the establishment of persistent foot-and-mouth disease virus (FMDV) infection, the systemic response to vaccination and challenge was studied in 47 steers. Eighteen steers that had received a recombinant FMDV A vaccine 2 weeks earlier and 29 non-vaccinated steers were challenged by intra-nasopharyngeal deposition of FMDV A24. For up to 35 days after challenge, host factors including complete blood counts with T lymphocyte subsets, type I/III interferon (IFN) activity, neutralizing and total FMDV-specific antibody titers in serum, as well as antibody-secreting cells (in 6 non-vaccinated animals) were characterized in the context of viral infection dynamics. RESULTS: Vaccination generally induced a strong antibody response. There was a transient peak of FMDV-specific serum IgM in non-vaccinated animals after challenge, while IgM levels in vaccinated animals did not increase further. Both groups had a lasting increase of specific IgG and neutralizing antibody after challenge. Substantial systemic IFN activity in non-vaccinated animals coincided with viremia, and no IFN or viremia was detected in vaccinated animals. After challenge, circulating lymphocytes decreased in non-vaccinated animals, coincident with viremia, IFN activity, and clinical disease, whereas lymphocyte and monocyte counts in vaccinated animals were unaffected by vaccination but transiently increased after challenge. The CD4(+)/CD8(+) T cell ratio in non-vaccinated animals increased during acute infection, driven by an absolute decrease of CD8(+) cells. CONCLUSIONS: The incidence of FMDV persistence was 61.5 % in non-vaccinated and 54.5 % in vaccinated animals. Overall, the systemic factors examined were not associated with the FMDV carrier/non-carrier divergence; however, significant differences were identified between responses of non-vaccinated and vaccinated cattle.


Subject(s)
Foot-and-Mouth Disease Virus/physiology , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease/virology , Viral Vaccines/immunology , Adenoviridae , Animals , Carrier State , Cattle , Cattle Diseases , Enzyme-Linked Immunosorbent Assay/veterinary , Enzyme-Linked Immunospot Assay/veterinary , Female , Foot-and-Mouth Disease/immunology , Genetic Vectors , Male , Vaccination , Vaccines, Synthetic
11.
Virus Res ; 221: 8-14, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27182007

ABSTRACT

African swine fever virus (ASFV) produces a contagious disease of domestic pigs that results in severe economic consequences to the swine industry. Control of the disease has been hampered by the unavailability of vaccines. We recently reported the development of two experimental vaccine strains (ASFV-G-Δ9GL and ASFV-G-ΔMGF) based on the attenuation of the highly virulent and epidemiologically relevant Georgia2007 isolate. Deletion of the 9GL gene or six genes of the MGF360/505 group produced two attenuated ASFV strains which were able to confer protection to animals when challenged with the virulent parental virus. Both viruses, although efficient in inducing protection, present concerns regarding their safety. In an attempt to solve this problem we developed a novel virus strain, ASFV-G-Δ9GL/ΔMGF, based on the deletion of all genes deleted in ASFV-G-Δ9GL and ASFV-G-ΔMGF. ASFV-G-Δ9GL/ΔMGF is the first derivative of a highly virulent ASFV field strain subjected to a double round of recombination events seeking to sequentially delete specific genes. ASFV-G-Δ9GL/ΔMGF showed a decreased ability to replicate in primary swine macrophage cultures relative to that of ASFV-G and ASFV-G-ΔMGF but similar to that of ASFV-G-Δ9GL. ASFV-G-Δ9GL/ΔMGF was attenuated when intramuscularly inoculated into swine, even at doses as high as 10(6) HAD50. Animals infected with doses ranging from 10(2) to 10(6) HAD50 did not present detectable levels of virus in blood at any time post-infection and they did not develop detectable levels of anti-ASFV antibodies. Importantly, ASFV-G-Δ9GL/ΔMGF does not induce protection against challenge with the virulent parental ASFV-G isolate. Results presented here suggest caution towards approaches involving genomic manipulations when developing rationally designed ASFV vaccine strains.


Subject(s)
African Swine Fever Virus/genetics , African Swine Fever Virus/pathogenicity , African Swine Fever/pathology , African Swine Fever/virology , Sequence Deletion , Viral Proteins/genetics , Viral Vaccines/immunology , African Swine Fever/prevention & control , African Swine Fever Virus/immunology , African Swine Fever Virus/physiology , Animals , Antibodies, Viral/blood , Cells, Cultured , Georgia , Injections, Intramuscular , Macrophages/virology , Recombination, Genetic , Swine , Treatment Outcome , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virulence , Virus Replication
12.
J Virol ; 90(14): 6344-64, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27147736

ABSTRACT

UNLABELLED: The pathogenesis of persistent foot-and-mouth disease virus (FMDV) infection was investigated in 46 cattle that were either naive or had been vaccinated using a recombinant, adenovirus-vectored vaccine 2 weeks before challenge. The prevalence of FMDV persistence was similar in both groups (62% in vaccinated cattle, 67% in nonvaccinated cattle), despite vaccinated cattle having been protected from clinical disease. Analysis of antemortem infection dynamics demonstrated that the subclinical divergence between FMDV carriers and animals that cleared the infection had occurred by 10 days postinfection (dpi) in vaccinated cattle and by 21 dpi in nonvaccinated animals. The anatomic distribution of virus in subclinically infected, vaccinated cattle was restricted to the pharynx throughout both the early and the persistent phases of infection. In nonvaccinated cattle, systemically disseminated virus was cleared from peripheral sites by 10 dpi, while virus selectively persisted within the nasopharynx of a subset of animals. The quantities of viral RNA shed in oropharyngeal fluid during FMDV persistence were similar in vaccinated and nonvaccinated cattle. FMDV structural and nonstructural proteins were localized to follicle-associated epithelium of the dorsal soft palate and dorsal nasopharynx in persistently infected cattle. Host transcriptome analysis of tissue samples processed by laser capture microdissection indicated suppression of antiviral host factors (interferon regulatory factor 7, CXCL10 [gamma interferon-inducible protein 10], gamma interferon, and lambda interferon) in association with persistent FMDV. In contrast, during the transitional phase of infection, the level of expression of IFN-λ mRNA was higher in follicle-associated epithelium of animals that had cleared the infection. This work provides novel insights into the intricate mechanisms of FMDV persistence and contributes to further understanding of this critical aspect of FMDV pathogenesis. IMPORTANCE: The existence of a prolonged, asymptomatic carrier state is a political impediment for control and potential eradication of foot-and-mouth disease (FMD). When FMD outbreaks occur, they are often extinguished by massive depopulation of livestock due to the fear that some animals may have undiagnosed subclinical infection, despite uncertainty over the biological relevance of FMD virus (FMDV) persistence. The work described here elucidates aspects of the FMDV carrier state in cattle which may facilitate identification and/or abrogation of asymptomatic FMDV infection. The divergence between animals that clear infection and those that develop persistent infection was demonstrated to occur earlier than previously established. The host antiviral response in tissues maintaining persistent FMDV was downregulated, whereas upregulation of IFN-λ mRNA was found in the epithelium of cattle that had recently cleared the infection. This suggests that the clearing of FMDV infection is associated with an enhanced mucosal antiviral response, whereas FMDV persistence is associated with suppression of the host antiviral response.


Subject(s)
Carrier State/veterinary , Cattle Diseases/virology , Foot-and-Mouth Disease Virus/immunology , Foot-and-Mouth Disease/transmission , Pharynx/virology , Vaccination/veterinary , Vaccines, Synthetic/administration & dosage , Animals , Carrier State/immunology , Carrier State/virology , Cattle , Cattle Diseases/immunology , Cattle Diseases/prevention & control , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/prevention & control , RNA, Viral/genetics , Viral Vaccines/administration & dosage , Virus Replication
13.
Vet Clin Pathol ; 45(1): 110-5, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26802284

ABSTRACT

BACKGROUND: Immunophenotyping of blood lymphocytes by flow cytometry is important in infectious disease research. In animal experiments and other longitudinal studies, the processing, prompt staining, and analysis of fresh samples is a logistical challenge and daily assay variation can confound data interpretation. OBJECTIVE: This study examined the feasibility of cryopreservation and deferred analysis of bovine peripheral blood T lymphocytes from normal or infected animals. METHODS: Peripheral blood mononuclear cells were collected from 4 naïve Holstein steers and 4 steers infected with foot-and-mouth-disease virus serotype Asia1. Identical aliquots were labeled and analyzed immediately, labeled for deferred analysis, or stored at -70°C or over liquid nitrogen for up to 3 weeks before labeling. RESULTS: Freezing of unlabeled cells induced statistically significant changes in phenotypic recognition. In infected animals, the γδ T-cell population increased by 28% and CD8(+) αßT cells by 32%, while total CD3(+) cells decreased by 16%, and CD4(+) αßT cells decreased by 12%. Subsequent storage of frozen cells for the duration of the study, however, had no significant effect. There was less than 20% relative change in subpopulation sizes, and storage at -70°C or over liquid nitrogen was equivalent. CONCLUSIONS: Depending on the objectives and practical limitations of a study, deferred labeling of peripheral blood lymphocytes can be a viable option. Although frozen storage of lymphocytes can introduce some artifactual distortion of relative cell populations, frozen cells can be maintained in storage until all samples in a longitudinal study can be analyzed in batch under standardized conditions and without introducing further bias.


Subject(s)
Cattle Diseases/blood , Cattle/blood , Cryopreservation/veterinary , Foot-and-Mouth Disease/blood , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cattle Diseases/immunology , Cryopreservation/methods , Cryopreservation/standards , Feasibility Studies , Flow Cytometry/veterinary , Foot-and-Mouth Disease Virus/immunology , Freezing , Immunophenotyping/veterinary , Leukocytes, Mononuclear/immunology , Longitudinal Studies , Male , Specimen Handling/veterinary
14.
PLoS One ; 11(1): e0146445, 2016.
Article in English | MEDLINE | ID: mdl-26735130

ABSTRACT

Since the early 2000s outbreaks of foot-and-mouth disease (FMD) have been described in several previously FMD-free Asian nations, including the Republic of Korea (South Korea). One outbreak with FMD virus (FDMV) serotype A and two with serotype O occurred in South Korea in 2010/2011. The causative viruses belonged to lineages that had been spreading in South East Asia, far East and East Asia since 2009 and presented a great threat to the countries in that region. Most FMDV strains infect ruminants and pigs, as it happened during the outbreaks of FMDV serotype O in South Korea. Contrastingly, the strain of serotype A affected only ruminants. Based upon these findings, the intention of the work described in the current report was to characterize and compare the infectivity, virulence and transmission of both strains under laboratory conditions in cattle and pigs, by direct inoculation and contact exposure. As expected, FMDV serotype O was highly virulent in both cattle and swine by contact exposure and direct inoculation. Surprisingly, FMDV serotype A was highly virulent in swine, but was less infectious in cattle by contact exposure to infected swine or cattle. Interestingly, similar quantities of aerosolized FMDV RNA were detected during experiments with viruses of serotypes O and A. Specific virus-host interaction of A/SKR/2010 could affect the transmission of this strain to cattle, and this may explain in part the limited spread of the serotype A epizootic.


Subject(s)
Cattle Diseases/virology , Disease Outbreaks , Foot-and-Mouth Disease Virus/pathogenicity , Foot-and-Mouth Disease/virology , Animals , Cattle , Cattle Diseases/epidemiology , Cell Line , Foot-and-Mouth Disease/epidemiology , Foot-and-Mouth Disease/transmission , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/isolation & purification , Male , RNA, Viral/genetics , Republic of Korea , Sus scrofa , Virulence , Virus Shedding
15.
Am J Vet Res ; 77(1): 98-107, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26709943

ABSTRACT

Abnormal body temperature is a major indicator of disease; infrared thermography (IRT) can assess changes in body surface temperature quickly and remotely. This technology can be applied to a myriad of diseases of various etiologies across a wide range of host species in veterinary medicine. It is used to monitor the physiologic status of individual animals, such as measuring feed efficiency or diagnosing pregnancy. Infrared thermography has applications in the assessment of animal welfare, and has been used to detect soring in horses and monitor stress responses. This review addresses the variety of uses for IRT in veterinary medicine, including disease detection, physiologic monitoring, welfare assessment, and potential future applications.


Subject(s)
Thermography/veterinary , Animal Welfare , Animals , Body Temperature/physiology , Infrared Rays , Monitoring, Physiologic/veterinary
16.
PLoS One ; 10(11): e0143666, 2015.
Article in English | MEDLINE | ID: mdl-26599543

ABSTRACT

A time-course pathogenesis study was performed to compare and contrast primary foot-and-mouth disease virus (FMDV) infection following simulated-natural (intra-nasopharyngeal) virus exposure of cattle that were non-vaccinated or vaccinated using a recombinant adenovirus-vectored FMDV vaccine. FMDV genome and infectious virus were detected during the initial phase of infection in both categories of animals with consistent predilection for the nasopharyngeal mucosa. A rapid progression of infection with viremia and widespread dissemination of virus occurred in non-vaccinated animals whilst vaccinated cattle were protected from viremia and clinical FMD. Analysis of micro-anatomic distribution of virus during early infection by lasercapture microdissection localized FMDV RNA to follicle-associated epithelium of the nasopharyngeal mucosa in both groups of animals, with concurrent detection of viral genome in nasopharyngeal MALT follicles in vaccinated cattle only. FMDV structural and non-structural proteins were detected in epithelial cells of the nasopharyngeal mucosa by immunomicroscopy 24 hours after inoculation in both non-vaccinated and vaccinated steers. Co-localization of CD11c+/MHC II+ cells with viral protein occurred early at primary infection sites in vaccinated steers while similar host-virus interactions were observed at later time points in non-vaccinated steers. Additionally, numerous CD8+/CD3- host cells, representing presumptive natural killer cells, were observed in association with foci of primary FMDV infection in the nasopharyngeal mucosa of vaccinated steers but were absent in non-vaccinated steers. Immunomicroscopic evidence of an activated antiviral response at primary infection sites of vaccinated cattle was corroborated by a relative induction of interferon -α, -ß, -γ and -λ mRNA in micro-dissected samples of nasopharyngeal mucosa. Although vaccination protected cattle from viremia and clinical FMD, there was subclinical infection of epithelial cells of the nasopharyngeal mucosa that could enable shedding and long-term persistence of infectious virus. Additionally, these data indicate different mechanisms within the immediate host response to infection between non-vaccinated and vaccinated cattle.


Subject(s)
Cattle Diseases/immunology , Cattle Diseases/pathology , Foot-and-Mouth Disease Virus/pathogenicity , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/pathology , Animals , Cattle , Cattle Diseases/prevention & control , Foot-and-Mouth Disease/prevention & control , Foot-and-Mouth Disease Virus/immunology , Vaccination
17.
PLoS One ; 10(5): e0125698, 2015.
Article in English | MEDLINE | ID: mdl-25996935

ABSTRACT

Tissues obtained post-mortem from cattle persistently infected with foot-and-mouth disease virus (FMDV) were analyzed to characterize the tissue-specific localization of FMDV and partial transcriptome profiles for selected immunoregulatory cytokines. Analysis of 28 distinct anatomic sites from 21 steers infected with FMDV serotype A, O or SAT2, had the highest prevalence of overall viral detection in the dorsal nasopharynx (80.95%) and dorsal soft palate (71.43%). FMDV was less frequently detected in laryngeal mucosal tissues, oropharyngeal mucosal sites, and lymph nodes draining the pharynx. Immunomicroscopy indicated that within persistently infected mucosal tissues, FMDV antigens were rarely detectable within few epithelial cells in regions of mucosa-associated lymphoid tissue (MALT). Transcriptome analysis of persistently infected pharyngeal tissues by qRT-PCR for 14 cytokine genes indicated a general trend of decreased mRNA levels compared to uninfected control animals. Although, statistically significant differences were not observed, greatest suppression of relative expression (RE) was identified for IP-10 (RE = 0.198), IFN-ß (RE = 0.269), IL-12 (RE = 0.275), and IL-2 (RE = 0.312). Increased relative expression was detected for IL-6 (RE = 2.065). Overall, this data demonstrates that during the FMDV carrier state in cattle, viral persistence is associated with epithelial cells of the nasopharynx in the upper respiratory tract and decreased levels of mRNA for several immunoregulatory cytokines in the infected tissues.


Subject(s)
Cattle Diseases/virology , Cytokines/metabolism , Foot-and-Mouth Disease Virus/isolation & purification , Foot-and-Mouth Disease/metabolism , Foot-and-Mouth Disease/virology , Nasopharynx/virology , Animals , Antigens, Viral/immunology , Cattle , Cattle Diseases/genetics , Cattle Diseases/immunology , Cattle Diseases/metabolism , Cytokines/genetics , Foot-and-Mouth Disease/genetics , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease Virus/immunology , Gene Expression , Immunohistochemistry , RNA, Messenger/genetics , RNA, Viral/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Respiratory Mucosa/virology
18.
Vet Microbiol ; 178(1-2): 50-60, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-25937316

ABSTRACT

Within-host infection dynamics of a recent field isolate of foot-and-mouth disease virus (FMDV), serotype O, topotype South East Asia, lineage Myamar'98 were evaluated in sheep using four different systems for virus exposure. Two novel, simulated natural, inoculation systems consisting of intra-nasopharyngeal (INP) deposition and aerosol inoculation were evaluated in comparison with two conventional systems: coronary band inoculation and direct contact exposure. All four exposure systems were efficient in generating consistently severe, generalized FMD with synchronous clinical characteristics within exposure groups, indicating that this Myanmar98 strain is highly virulent in sheep. Clinical and virological dynamics were similarly rapid following INP- and coronary band inoculation, with both systems leading to significantly earlier detection of virus shedding when compared to aerosol inoculation and contact exposure. The data presented herein support application of the two optimized simulated natural inoculation systems as valid alternatives to conventionally used exposure systems for studies of FMDV pathogenesis and vaccinology in sheep. Furthermore, the data suggest that targeted exposure of the ovine pharynx is highly efficient for generating consistent FMDV infection, which supports critical involvement of this anatomic region as a site of primary virus replication in sheep.


Subject(s)
Environmental Exposure , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease/physiopathology , Foot-and-Mouth Disease/virology , Sheep Diseases/physiopathology , Sheep Diseases/virology , Sheep, Domestic , Aerosols , Animals , Myanmar , Nasopharynx/virology , Serogroup , Sheep
20.
PLoS One ; 9(9): e106859, 2014.
Article in English | MEDLINE | ID: mdl-25184288

ABSTRACT

A time-course study was performed to elucidate the early events of foot-and-mouth disease virus (FMDV) infection in pigs subsequent to simulated natural, intra-oropharyngeal, inoculation. The earliest detectable event was primary infection in the lingual and paraepiglottic tonsils at 6 hours post inoculation (hpi) characterized by regional localization of viral RNA, viral antigen, and infectious virus. At this time FMDV antigen was localized in cytokeratin-positive epithelial cells and CD172a-expressing leukocytes of the crypt epithelium of the paraepiglottic tonsils. De novo replication of FMDV was first detected in oropharyngeal swab samples at 12 hpi and viremia occurred at 18-24 hpi, approximately 24 hours prior to the appearance of vesicular lesions. From 12 through 78 hpi, microscopic detection of FMDV was consistently localized to cytokeratin-positive cells within morphologically characteristic segments of oropharyngeal tonsil crypt epithelium. During this period, leukocyte populations expressing CD172a, SLA-DQ class II and/or CD8 were found in close proximity to infected epithelial cells, but with little or no co-localization with viral proteins. Similarly, M-cells expressing cytokeratin-18 did not co-localize with FMDV proteins. Intra-epithelial micro-vesicles composed of acantholytic epithelial cells expressing large amounts of structural and non-structural FMDV proteins were present within crypts of the tonsil of the soft palate during peak clinical infection. These findings inculpate the paraepiglottic tonsils as the primary site of FMDV infection in pigs exposed via the gastrointestinal tract. Furthermore, the continuing replication of FMDV in the oropharyngeal tonsils during viremia and peak clinical infection with no concurrent amplification of virus occurring in the lower respiratory tract indicates that these sites are the major source of shedding of FMDV from pigs.


Subject(s)
Adenoids , Foot-and-Mouth Disease Virus/physiology , Foot-and-Mouth Disease , Swine Diseases , Virus Replication , Adenoids/metabolism , Adenoids/pathology , Adenoids/virology , Animals , CD8-Positive T-Lymphocytes/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Foot-and-Mouth Disease/metabolism , Foot-and-Mouth Disease/pathology , Histocompatibility Antigens Class II/metabolism , Male , Swine , Swine Diseases/metabolism , Swine Diseases/pathology , Swine Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...