Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
PLoS One ; 16(2): e0246695, 2021.
Article in English | MEDLINE | ID: mdl-33600498

ABSTRACT

Bovine Respiratory Syncytial virus (BRSV) is one of the major infectious agents in the etiology of the bovine respiratory disease complex. BRSV causes a respiratory syndrome in calves, which is associated with severe bronchiolitis. In this study we describe the effect of treatment with antiviral fusion protein inhibitor (FPI) and ibuprofen, on gene expression in lung tissue of calves infected with BRSV. Calves infected with BRSV are an excellent model of human RSV in infants: we hypothesized that FPI in combination with ibuprofen would provide the best therapeutic intervention for both species. The following experimental treatment groups of BRSV infected calves were used: 1) ibuprofen day 3-10, 2) ibuprofen day 5-10, 3) placebo, 4) FPI day 5-10, 5) FPI and ibuprofen day 5-10, 6) FPI and ibuprofen day 3-10. All calves were infected with BRSV on day 0. Daily clinical evaluation with monitoring of virus shedding by qRT-PCR was conducted. On day10 lung tissue with lesions (LL) and non-lesional (LN) was collected at necropsy, total RNA extracted, and RNA sequencing performed. Differential gene expression analysis was conducted with Gene ontology (GO) and KEGG pathway enrichment analysis. The most significant differential gene expression in BRSV infected lung tissues was observed in the comparison of LL with LN; oxidative stress and cell damage was especially noticeable. Innate and adaptive immune functions were reduced in LL. As expected, combined treatment with FPI and Ibuprofen, when started early, made the most difference in gene expression patterns in comparison with placebo, especially in pathways related to the innate and adaptive immune response in both LL and LN. Ibuprofen, when used alone, negatively affected the antiviral response and caused higher virus loads as shown by increased viral shedding. In contrast, when used with FPI Ibuprofen enhanced the specific antiviral effect of FPI, due to its ability to reduce the damaging effect of prostanoids and oxidative stress.


Subject(s)
Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus, Bovine/genetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antiviral Agents/pharmacokinetics , Cattle , Cattle Diseases/immunology , Cyclooxygenase Inhibitors/pharmacology , Gene Expression/drug effects , Gene Expression/genetics , Gene Expression Profiling/methods , Ibuprofen/pharmacology , Lung/cytology , Lung/pathology , Lung/virology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus, Bovine/pathogenicity , Transcriptome/drug effects , Transcriptome/genetics , Virus Shedding/drug effects
2.
Virulence ; 12(1): 404-414, 2021 12.
Article in English | MEDLINE | ID: mdl-33470171

ABSTRACT

Bovine respiratory syncytial virus (BRSV) is a clinically important causative agent of acute respiratory diseases in postweaning calves and feedlot cattle and causes numerous economic losses to the cattle industry. In June 2018, an outbreak of an acute respiratory disease occurred among 4- to 10-month-old calves on three intensive beef cattle farms in Heilongjiang Province, Northeast China, with a 27.42% morbidity rate (329/1200) and a > 25% mortality rate (85/329). Using next-generation sequencing, we comprehensively analyzed microbial diversity in the lung samples of the diseased cattle and found that the causative agent of this epidemic outbreak is mainly a bovine orthopneumovirus named BRSV strain DQ. We then isolated and confirmed the virus by RT-PCR and an indirect immunofluorescence assay. Phylogenetic analysis of genes G, F, N, NS1, NS2, and SH of BRSV strain DQ showed that this strain shares the highest genetic similarity with strains USII/S1, 15489, V41, and NY487834 belonging to subgroup III of BRSV. This is the first report of subgroup III strain of BRSV presence in China. Heilongjiang Province is a major cattle-breeding province in China; therefore, it is necessary to test for BRSV in the cattle trade and to conduct region-extended epidemiological surveillance for BRSV in China.


Subject(s)
Cattle Diseases/virology , Phylogeny , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus, Bovine/classification , Respiratory Syncytial Virus, Bovine/genetics , Acute Disease/epidemiology , Animals , Antibodies, Viral/blood , Cattle , Cattle Diseases/epidemiology , China/epidemiology , Disease Outbreaks , High-Throughput Nucleotide Sequencing , Lung/pathology , Lung/virology , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Bovine/isolation & purification , Respiratory Syncytial Virus, Bovine/pathogenicity
3.
Sci Rep ; 8(1): 3021, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445124

ABSTRACT

Human respiratory syncytial virus (HRSV) is a leading cause of severe acute lower respiratory tract infection in infants and children worldwide. Bovine RSV (BRSV) is closely related to HRSV and a significant cause of morbidity in young cattle. BRSV infection in calves displays many similarities to RSV infection in humans, including similar age dependency and disease pathogenesis. Polyanhydride nanoparticle-based vaccines (i.e., nanovaccines) have shown promise as adjuvants and vaccine delivery vehicles due to their ability to promote enhanced immunogenicity through the route of administration, provide sustained antigen exposure, and induce both antibody- and cell-mediated immunity. Here, we developed a novel, mucosal nanovaccine that encapsulates the post-fusion F and G glycoproteins from BRSV into polyanhydride nanoparticles and determined the efficacy of the vaccine against RSV infection using a neonatal calf model. Calves receiving the BRSV-F/G nanovaccine exhibited reduced pathology in the lungs, reduced viral burden, and decreased virus shedding compared to unvaccinated control calves, which correlated with BRSV-specific immune responses in the respiratory tract and peripheral blood. Our results indicate that the BRSV-F/G nanovaccine is highly immunogenic and, with optimization, has the potential to significantly reduce the disease burden associated with RSV infection in both humans and animals.


Subject(s)
Polyanhydrides/pharmacology , Respiratory Syncytial Virus, Bovine/drug effects , Respiratory Syncytial Virus, Bovine/immunology , Animals , Animals, Newborn/immunology , Antibodies, Viral/immunology , Antibody Formation , Cattle , Cattle Diseases/immunology , Female , Immunity, Cellular , Lung/pathology , Male , Nanoparticles/therapeutic use , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Bovine/pathogenicity , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/pathogenicity , Vaccination/methods
4.
BMC Vet Res ; 14(1): 53, 2018 Feb 27.
Article in English | MEDLINE | ID: mdl-29482563

ABSTRACT

BACKGROUND: Bovine respiratory disease (BRD) remains among the leading causes of death of cattle internationally. The objective of this study was to identify risk factors associated with exposure to BRD pathogens during the peri-weaning period (day (d)-14 to d 14 relative to weaning at 0) in dairy bull calves using serological responses to these pathogens as surrogate markers of exposure. Clinically normal Holstein-Friesian and Jersey breed bull calves (n = 72) were group housed in 4 pens using a factorial design with calves of different breeds and planes of nutrition in each pen. Intrinsic, management and clinical data were collected during the pre-weaning (d - 56 to d - 14) period. Calves were gradually weaned over 14 days (d - 14 to d 0). Serological analysis for antibodies against key BRD pathogens (BRSV, BPI3V, BHV-1, BHV-4, BCoV, BVDV and H. somni) was undertaken at d - 14 and d 14. Linear regression models (for BVDV, BPI3V, BHV-1, BHV-4, BCoV and H. somni) and a single mixed effect random variable model (for BRSV) were used to identify risk factors for changes in antibody levels to these pathogens. RESULTS: BRSV was the only pathogen which demonstrated clustering by pen. Jersey calves experienced significantly lower changes in BVDV S/P than Holstein-Friesian calves. Animals with a high maximum respiratory score (≥8) recorded significant increases in H. somni S/P during the peri-weaning period when compared to those with respiratory scores of ≤3. Haptoglobin levels of between 1.32 and 1.60 mg/ml at d - 14 were significantly associated with decreases in BHV-1 S/N during the peri-weaning period. Higher BVDV S/P ratios at d - 14 were significantly correlated with increased changes in serological responses to BHV-4 over the peri-weaning period. CONCLUSIONS: Haptoglobin may have potential as a predictor of exposure to BHV-1. BRSV would appear to play a more significant role at the 'group' rather than 'individual animal' level. The significant associations between the pre-weaning levels of antibodies to certain BRD pathogens and changes in the levels of antibodies to the various pathogens during the peri-weaning period may reflect a cohort of possibly genetically linked 'better responders' among the study population.


Subject(s)
Bovine Respiratory Disease Complex/etiology , Animals , Animals, Newborn , Bovine Respiratory Disease Complex/virology , Cattle , Coronavirus, Bovine/pathogenicity , Herpesvirus 1, Bovine/pathogenicity , Herpesvirus 4, Bovine/pathogenicity , Male , Parainfluenza Virus 3, Bovine/pathogenicity , Respiratory Syncytial Virus, Bovine/pathogenicity , Risk Factors , Weaning
5.
PLoS One ; 12(3): e0173845, 2017.
Article in English | MEDLINE | ID: mdl-28282443

ABSTRACT

The bovine paranasal sinuses are a group of complex cavernous air-filled spaces, lined by respiratory epithelium, the exact function of which is unclear. While lesions affecting these sinuses are occasionally reported in cattle, their microbial flora has not been defined. Furthermore, given that the various bacterial and viral pathogens causing bovine respiratory disease (BRD) persist within herds, we speculated that the paranasal sinuses may serve as a refuge for such infectious agents. The paranasal sinuses of clinically normal cattle (n = 99) and of cattle submitted for post-mortem examination (PME: n = 34) were examined by microbial culture, PCR and serology to include bacterial and viral pathogens typically associated with BRD: Mycoplasma bovis, Histophilus somni, Mannheimia haemolytica and Pasteurella multocida, bovine respiratory syncytial virus (BRSV) and bovine parainfluenza-3 virus (BPIV-3). Overall, the paranasal sinuses were either predominantly sterile or did not contain detectable microbes (83.5%: 94.9% of clinically normal and 50.0% of cattle submitted for PME). Bacteria, including BRD causing pathogens, were identified in relatively small numbers of cattle (<10%). While serology indicated widespread exposure of both clinically normal and cattle submitted for PME to BPIV-3 and BRSV (seroprevalences of 91.6% and 84.7%, respectively), PCR identified BPIV-3 in only one animal. To further explore these findings we investigated the potential role of the antimicrobial molecule nitric oxide (NO) within paranasal sinus epithelium using immunohistochemistry. Expression of the enzyme responsible for NO synthesis, inducible nitric oxide synthase (iNOS), was detected to varying degrees in 76.5% of a sub-sample of animals suggesting production of this compound plays a similar protective role in the bovine sinus as it does in humans.


Subject(s)
Bovine Respiratory Disease Complex/virology , Microbiota , Nitric Oxide/metabolism , Paranasal Sinuses/microbiology , Animals , Bacteria/genetics , Bacteria/pathogenicity , Bovine Respiratory Disease Complex/microbiology , Cattle , Cross-Sectional Studies , Epithelial Cells/metabolism , Female , Male , Microbiota/genetics , Nitric Oxide Synthase Type II/metabolism , Parainfluenza Virus 3, Bovine/genetics , Parainfluenza Virus 3, Bovine/isolation & purification , Parainfluenza Virus 3, Bovine/pathogenicity , Paranasal Sinuses/metabolism , Respiratory Syncytial Virus, Bovine/genetics , Respiratory Syncytial Virus, Bovine/isolation & purification , Respiratory Syncytial Virus, Bovine/pathogenicity
6.
PLoS One ; 11(2): e0148551, 2016.
Article in English | MEDLINE | ID: mdl-26859677

ABSTRACT

Our previous studies showed that bovine respiratory syncytial virus (BRSV) followed by Histophilus somni causes more severe bovine respiratory disease and a more permeable alveolar barrier in vitro than either agent alone. However, microarray analysis revealed the treatment of bovine alveolar type 2 (BAT2) epithelial cells with H. somni concentrated culture supernatant (CCS) stimulated up-regulation of four antiviral protein genes as compared with BRSV infection or dual treatment. This suggested that inhibition of viral infection, rather than synergy, may occur if the bacterial infection occurred before the viral infection. Viperin (or radical S-adenosyl methionine domain containing 2--RSAD2) and ISG15 (IFN-stimulated gene 15--ubiquitin-like modifier) were most up-regulated. CCS dose and time course for up-regulation of viperin protein levels were determined in treated bovine turbinate (BT) upper respiratory cells and BAT2 lower respiratory cells by Western blotting. Treatment of BAT2 cells with H. somni culture supernatant before BRSV infection dramatically reduced viral replication as determined by qRT PCR, supporting the hypothesis that the bacterial infection may inhibit viral infection. Studies of the role of the two known H. somni cytotoxins showed that viperin protein expression was induced by endotoxin (lipooligosaccharide) but not by IbpA, which mediates alveolar permeability and H. somni invasion. A naturally occurring IbpA negative asymptomatic carrier strain of H. somni (129Pt) does not cause BAT2 cell retraction or permeability of alveolar cell monolayers, so lacks virulence in vitro. To investigate initial steps of pathogenesis, we showed that strain 129Pt attached to BT cells and induced a strong viperin response in vitro. Thus colonization of the bovine upper respiratory tract with an asymptomatic carrier strain lacking virulence may decrease viral infection and the subsequent enhancement of bacterial respiratory infection in vivo.


Subject(s)
Antiviral Agents/metabolism , Haemophilus somnus/physiology , Respiratory Mucosa/microbiology , Respiratory Mucosa/virology , Respiratory Syncytial Virus, Bovine/physiology , Respiratory Syncytial Virus, Bovine/pathogenicity , Animals , Bacterial Adhesion , Cattle , Cells, Cultured , Haemophilus Infections/complications , Haemophilus Infections/microbiology , Haemophilus Infections/virology , Haemophilus somnus/genetics , Haemophilus somnus/pathogenicity , Proteins/genetics , Proteins/metabolism , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus Infections/microbiology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Bovine/genetics , Up-Regulation , Virulence , Virulence Factors/biosynthesis , Virulence Factors/genetics , Virus Replication
7.
J Vet Intern Med ; 29(3): 770-80, 2015.
Article in English | MEDLINE | ID: mdl-25929158

ABSTRACT

Bovine respiratory disease (BRD) is an economically important disease of cattle and continues to be an intensely studied topic. However, literature summarizing the time between pathogen exposure and clinical signs, shedding, and seroconversion is minimal. A structured literature review of the published literature was performed to determine cattle responses (time from pathogen exposure to clinical signs, shedding, and seroconversion) in challenge models using common BRD viral and bacterial pathogens. After review a descriptive analysis of published studies using common BRD pathogen challenge studies was performed. Inclusion criteria were single pathogen challenge studies with no treatment or vaccination evaluating outcomes of interest: clinical signs, shedding, and seroconversion. Pathogens of interest included: bovine viral diarrhea virus (BVDV), bovine herpesvirus type 1 (BHV-1), parainfluenza-3 virus, bovine respiratory syncytial virus, Mannheimia haemolytica, Mycoplasma bovis, Pastuerella multocida, and Histophilus somni. Thirty-five studies and 64 trials were included for analysis. The median days to the resolution of clinical signs after BVDV challenge was 15 and shedding was not detected on day 12 postchallenge. Resolution of BHV-1 shedding resolved on day 12 and clinical signs on day 12 postchallenge. Bovine respiratory syncytial virus ceased shedding on day 9 and median time to resolution of clinical signs was on day 12 postchallenge. M. haemolytica resolved clinical signs 8 days postchallenge. This literature review and descriptive analysis can serve as a resource to assist in designing challenge model studies and potentially aid in estimation of duration of clinical disease and shedding after natural pathogen exposure.


Subject(s)
Bovine Respiratory Disease Complex/microbiology , Animals , Bovine Respiratory Disease Complex/virology , Cattle/microbiology , Cattle/virology , Diarrhea Viruses, Bovine Viral/pathogenicity , Herpesvirus 1, Bovine/pathogenicity , Mannheimia haemolytica/pathogenicity , Mycoplasma bovis/pathogenicity , Parainfluenza Virus 3, Bovine/pathogenicity , Pasteurella multocida/pathogenicity , Respiratory Syncytial Virus, Bovine/pathogenicity
8.
PLoS One ; 9(6): e100392, 2014.
Article in English | MEDLINE | ID: mdl-24945377

ABSTRACT

The development of safe and effective vaccines against both bovine and human respiratory syncytial viruses (BRSV, HRSV) to be used in the presence of RSV-specific maternally-derived antibodies (MDA) remains a high priority in human and veterinary medicine. Herein, we present safety and efficacy results from a virulent BRSV challenge of calves with MDA, which were immunized with one of three vaccine candidates that allow serological differentiation of infected from vaccinated animals (DIVA): an SH gene-deleted recombinant BRSV (ΔSHrBRSV), and two subunit (SU) formulations based on HRSV-P, -M2-1, and -N recombinant proteins displaying BRSV-F and -G epitopes, adjuvanted by either oil emulsion (Montanide ISA71VG, SUMont) or immunostimulating complex matrices (AbISCO-300, SUAbis). Whereas all control animals developed severe respiratory disease and shed high levels of virus following BRSV challenge, ΔSHrBRSV-immunized calves demonstrated almost complete clinical and virological protection five weeks after a single intranasal vaccination. Although mucosal vaccination with ΔSHrBRSV failed to induce a detectable immunological response, there was a rapid and strong anamnestic mucosal BRSV-specific IgA, virus neutralizing antibody and local T cell response following challenge with virulent BRSV. Calves immunized twice intramuscularly, three weeks apart with SUMont were also well protected two weeks after boost. The protection was not as pronounced as that in ΔSHrBRSV-immunized animals, but superior to those immunized twice subcutaneously three weeks apart with SUAbis. Antibody responses induced by the subunit vaccines were non-neutralizing and not directed against BRSV F or G proteins. When formulated as SUMont but not as SUAbis, the HRSV N, P and M2-1 proteins induced strong systemic cross-protective cell-mediated immune responses detectable already after priming. ΔSHrBRSV and SUMont are two promising DIVA-compatible vaccines, apparently inducing protection by different immune responses that were influenced by vaccine-composition, immunization route and regimen.


Subject(s)
Antibodies, Viral/immunology , Genes, Viral , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus, Bovine/genetics , Respiratory Syncytial Virus, Bovine/immunology , Respiratory Syncytial Virus, Human/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Cattle , Epitopes/chemistry , Epitopes/immunology , Gene Deletion , Humans , Lung/immunology , Lung/pathology , Lung/virology , Lymph Nodes/pathology , Lymphocytes/immunology , Molecular Sequence Data , Respiratory Syncytial Virus Infections/blood , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Bovine/pathogenicity , Respiratory Syncytial Virus, Human/immunology , Species Specificity , Vaccination , Vaccines, Subunit/adverse effects , Vaccines, Subunit/immunology , Viral Load , Virulence
9.
Clin Vaccine Immunol ; 21(7): 997-1004, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24828093

ABSTRACT

Bovine respiratory syncytial virus (BRSV) and human respiratory syncytial virus (HRSV) are major causes of respiratory disease in calves and children, respectively, and are priorities for vaccine development. We previously demonstrated that an experimental vaccine, BRSV-immunostimulating complex (ISCOM), is effective in calves with maternal antibodies. The present study focuses on the antigenic characterization of this vaccine for the design of new-generation subunit vaccines. The results of our study confirmed the presence of membrane glycoprotein (G), fusion glycoprotein (F), and nucleoprotein (N) proteins in the ISCOMs, and this knowledge was extended by the identification of matrix (M), M2-1, phosphoprotein (P), small hydrophobic protein (SH) and of cellular membrane proteins, such as the integrins αVß1, αVß3, and α3ß1. The quantity of the major protein F was 4- to 5-fold greater than that of N (∼77 µg versus ∼17 µg/calf dose), whereas G, M, M2-1, P, and SH were likely present in smaller amounts. The polymerase (L), M2-2, nonstructural 1 (NS1), and NS2 proteins were not detected, suggesting that they are not essential for protection. Sera from the BRSV-ISCOM-immunized calves contained high titers of IgG antibody specific for F, G, N, and SH. Antibody responses against M and P were not detected; however, this does not exclude their role in protective T-cell responses. The absence of immunopathological effects of the cellular proteins, such as integrins, needs to be further confirmed, and their possible contribution to adjuvant functions requires elucidation. This work suggests that a combination of several surface and internal proteins should be included in subunit RSV vaccines and identifies absent proteins as potential candidates for differentiating infected from vaccinated animals.


Subject(s)
Antibodies, Viral/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Bovine/immunology , Vaccines, Subunit/immunology , Animals , Antibody Formation , Cattle , Humans , Immunoglobulin G/immunology , Integrins/immunology , Membrane Glycoproteins/immunology , Nucleoproteins/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Bovine/pathogenicity , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus, Human/pathogenicity , Vaccination , Viral Fusion Proteins/immunology
10.
J Gen Virol ; 95(Pt 6): 1244-1254, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24700100

ABSTRACT

Bovine respiratory syncytial virus (BRSV) causes inflammation and obstruction of the small airways, leading to severe respiratory disease in young calves. The virus is closely related to human (H)RSV, a major cause of bronchiolitis and pneumonia in young children. The ability to manipulate the genome of RSV has provided opportunities for the development of stable, live attenuated RSV vaccines. The role of the SH protein in the pathogenesis of BRSV was evaluated in vitro and in vivo using a recombinant (r)BRSV in which the SH gene had been deleted. Infection of bovine epithelial cells and monocytes with rBRSVΔSH, in vitro, resulted in an increase in apoptosis, and higher levels of TNF-α and IL-1ß compared with cells infected with parental, wild-type (WT) rBRSV. Although replication of rBRSVΔSH and WT rBRSV, in vitro, were similar, the replication of rBRSVΔSH was moderately reduced in the lower, but not the upper, respiratory tract of experimentally infected calves. Despite the greater ability of rBRSVΔSH to induce pro-inflammatory cytokines, in vitro, the pulmonary inflammatory response in rBRSVΔSH-infected calves was significantly reduced compared with that in calves inoculated with WT rBRSV, 6 days previously. Virus lacking SH appeared to be as immunogenic and effective in inducing resistance to virulent virus challenge, 6 months later, as the parental rBRSV. These findings suggest that rBRSVΔSH may be an ideal live attenuated virus vaccine candidate, combining safety with a high level of immunogenicity.


Subject(s)
Cytokines/biosynthesis , Genes, Viral , Respiratory Syncytial Virus, Bovine/genetics , Respiratory Syncytial Virus, Bovine/immunology , Animals , Apoptosis , Cattle , Cattle Diseases/immunology , Cattle Diseases/prevention & control , Cattle Diseases/virology , Gene Deletion , Humans , Immunity, Mucosal , Inflammation Mediators/metabolism , Interleukin-1beta/biosynthesis , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Bovine/pathogenicity , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus, Human/pathogenicity , Respiratory System/virology , Retroviridae Proteins, Oncogenic/genetics , Retroviridae Proteins, Oncogenic/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Virulence/genetics , Virulence/immunology
11.
Infect Immun ; 81(7): 2592-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23649093

ABSTRACT

Our previous studies showed that Histophilus somni and bovine respiratory syncytial virus (BRSV) act synergistically in vivo to cause more severe bovine respiratory disease than either agent alone causes. Since H. somni surface and secreted immunoglobulin binding protein A (IbpA) causes retraction of bovine alveolar type 2 (BAT2) cells and invasion between BAT2 cells in vitro, we investigated mechanisms of BRSV-plus-H. somni infection at the alveolar barrier. BRSV treatment of BAT2 cells prior to treatment with IbpA-rich H. somni concentrated culture supernatant (CCS) resulted in increased BAT2 cell rounding and retraction compared to those with either treatment alone. This mimicked the increased alveolar cell thickening in calves experimentally infected with BRSV followed by H. somni compared to that in calves infected with BRSV or H. somni alone. BRSV-plus-H. somni CCS treatment of BAT2 cells also enhanced paracellular migration. The effect of matrix metalloproteinases (MMPs) was investigated as well because microarray analysis revealed that treatment with BRSV plus H. somni synergistically upregulated BAT2 cell expression of mmp1 and mmp3 compared to that in cells treated with either agent alone. Enzyme-linked immunosorbent assay (ELISA) confirmed that MMP1 and MMP3 protein levels were similarly upregulated. In collagen I and collagen IV (targets for MMP1 and MMP3, respectively) substrate zymography, digestion was increased with supernatants from dually treated BAT2 cells compared with those from singly treated cells. Enhanced breakdown of collagen IV in the basal lamina and of fibrillar collagen I in the adjacent interstitium in the dual infection may facilitate dissemination of H. somni infection.


Subject(s)
Pasteurellaceae Infections/pathology , Pasteurellaceae/pathogenicity , Pulmonary Alveoli/pathology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus, Bovine/pathogenicity , Animals , Bacterial Proteins/metabolism , Cattle , Cell Movement , Cell Shape , Cells, Cultured , Coinfection/metabolism , Coinfection/microbiology , Coinfection/pathology , Coinfection/virology , Collagen Type I/metabolism , Collagen Type IV/metabolism , Culture Media/metabolism , Enzyme Assays , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Enzymologic , Host-Pathogen Interactions , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Movement , Oligonucleotide Array Sequence Analysis , Pasteurellaceae/metabolism , Pasteurellaceae Infections/metabolism , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/microbiology , Pulmonary Alveoli/virology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus, Bovine/metabolism , Up-Regulation
12.
Comp Immunol Microbiol Infect Dis ; 35(3): 253-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22410266

ABSTRACT

Bovine respiratory syncytial virus (BRSV) is a respiratory pathogen of cattle that causes severe disease in calves alone and as one of several viruses and bacteria that cause bovine respiratory disease complex. Like human RSV this virus modulates the immune response to avoid stimulation of a vibrant CD8+ T cytotoxic cell response and instead promotes a Th2 response. The Th2 skew sometimes results in the production of IgE antibodies and depresses production of the Th1 cytokine interferon γ. Innate immune cells have a pivotal role in guiding the adaptive response to BRSV, with selective secretion of cytokines by pulmonary dendritic cells. Here we review some of the pertinent observations on immune responses to BRSV infection and vaccination and illustrate how experimental infection models have been used to elucidate the immunopathogenesis of BRSV infection. Recent experiments using intranasal vaccination and/or immune modulation with DNA based adjuvants show promise for effective vaccination by the stimulation of Th1 T cell responses.


Subject(s)
Cattle Diseases/immunology , Cattle/virology , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus, Bovine/immunology , Vaccination/veterinary , Animals , Cattle/immunology , Cattle Diseases/pathology , Cattle Diseases/virology , Dendritic Cells/immunology , Immunity, Cellular , Immunoglobulin E/immunology , Interferon-gamma/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Bovine/pathogenicity , Th1 Cells/immunology
14.
Vet Clin North Am Food Anim Pract ; 26(2): 323-33, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20619187

ABSTRACT

Bovine respiratory syncytial virus (BRSV) is a major cause of respiratory disease and a major contributor to the bovine respiratory disease (BRD) complex. BRSV infects the upper and lower respiratory tract and is shed in nasal secretions. The close relatedness of BRSV to human respiratory syncytial virus (HRSV) has allowed researchers to use BRSV and HRSV to elucidate the mechanisms by which these viruses induce disease. Attempted vaccine production using formalin-inactivated vaccine resulted in exacerbated disease when infants became exposed to HRSV. Cattle vaccinated with formalin-inactivated virus had enhanced disease when inoculated with BRSV. This article discusses various aspects of BRSV, its epidemiology, pathogenesis, diagnostic tests, immunity, and vaccination.


Subject(s)
Bovine Respiratory Disease Complex/prevention & control , Bovine Respiratory Disease Complex/virology , Cattle Diseases/prevention & control , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus, Bovine/immunology , Animals , Bovine Respiratory Disease Complex/epidemiology , Bovine Respiratory Disease Complex/pathology , Cattle , Cattle Diseases/epidemiology , Cattle Diseases/pathology , Female , Male , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Bovine/pathogenicity , Time Factors , Treatment Outcome
15.
J Gen Virol ; 91(Pt 10): 2497-506, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20554798

ABSTRACT

The severity of respiratory syncytial virus (RSV) infections appears to differ with age in both humans and bovines. A primary RSV infection in naïve infants and in young calves runs a more severe course when they are 1-6 months old than in their first month of life. The relative lack of clinical signs in the first month of age may be due to high levels of maternally derived neutralizing antibodies or low exposure to infectious virus. This study examined whether age-dependent differences in the pathogenesis of bovine RSV (bRSV) between neonatal and young calves may be due to differences in age-dependent immunocompetence. To study the effect of age and immune parameters on bRSV disease in neonatal and young calves, neonatal (1-day-old) calves without maternally derived antibodies were infected experimentally with bRSV and the severity of disease and immune responses were evaluated in comparison with disease in similar 6-week-old infected calves. Neonatal calves had more extensive virus replication and lung consolidation, but lower pro-inflammatory [in particular tumour necrosis factor alpha (TNF-α)] responses, specific humoral immune responses, lung neutrophilic infiltration and clinical signs of disease than 6-week-old calves. The lack of correlation between virus replication and clinical signs suggests an important role of pro-inflammatory cytokines, in particular TNF-α, in the disease. The capacity to produce pro-inflammatory TNF-α appeared to increase with age, and may explain the age-dependent differences in RSV pathogenesis.


Subject(s)
Cattle Diseases/immunology , Cattle Diseases/pathology , Immunocompetence , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus, Bovine/immunology , Respiratory Syncytial Virus, Bovine/pathogenicity , Age Factors , Animals , Animals, Newborn , Antibodies, Viral/immunology , Cattle , Cytokines/immunology , Cytokines/metabolism , Lung/immunology , Lung/pathology , Neutrophils/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Severity of Illness Index
16.
Vet Immunol Immunopathol ; 136(1-2): 55-64, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20207014

ABSTRACT

Alveolar macrophages (AMvarphis) secrete regulatory molecules that are believed to be critical in maintaining normal lung homeostasis. However, in response to activating signals, AMvarphis have been shown to become highly phagocytic cells capable of secreting significant levels of pro-inflammatory cytokines. There is evidence to suggest that susceptibility of Mvarphi subpopulations to viral infection, and their subsequent cytokine/chemokine response, is dependent on age of the host. In the present study, we compared bovine respiratory syncytial virus (BRSV) replication and induction of cytokine responses in neonatal ovine AMvarphis to those cells isolated from adult animals. While neonatal AMvarphis could be infected with BRSV, viral replication was limited as previously shown for AMvarphis from mature animals. Interestingly, following BRSV infection, peak mRNA levels of IL-1beta and IL-8 in neonatal AMvarphi were several fold higher than levels induced in adult AMvarphis. In addition, peak mRNA expression for the cytokines examined occurred at earlier time points in neonatal AMvarphis compared to adult AMvarphis. However, the data indicated that viral replication was not required for the induction of specific cytokines in either neonatal or adult AMvarphis. TLR3 and TLR4 agonists induced significantly higher levels of cytokine transcripts than BRSV in both neonatal and adult AMvarphis. It was recently proposed that immaturity of the neonatal immune system extends from production of pro-inflammatory cytokines to regulation of such responses. Differential regulation of cytokines in neonatal AMvarphis compared to adult AMvarphis in response to RSV could be a contributory factor to more severe clinical episodes seen in neonates.


Subject(s)
Cytokines/genetics , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus, Bovine/immunology , Respiratory Syncytial Virus, Bovine/pathogenicity , Sheep Diseases/genetics , Sheep Diseases/immunology , Toll-Like Receptors/agonists , Animals , Base Sequence , Cattle , DNA Primers/genetics , Gene Expression , In Vitro Techniques , Interleukin-1beta/genetics , Interleukin-8/genetics , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Bovine/genetics , Respiratory Syncytial Virus, Bovine/physiology , Sheep , Sheep Diseases/virology , Virus Replication
17.
Vet Res ; 38(2): 153-80, 2007.
Article in English | MEDLINE | ID: mdl-17257568

ABSTRACT

Bovine respiratory syncytial virus (BRSV) belongs to the pneumovirus genus within the family Paramyxoviridae and is a major cause of respiratory disease in young calves. BRSV is enveloped and contains a negative sense, single-stranded RNA genome encoding 11 proteins. The virus replicates predominantly in ciliated respiratory epithelial cells but also in type II pneumocytes. It appears to cause little or no cytopathology in ciliated epithelial cell cultures in vitro, suggesting that much of the pathology is due to the host's response to virus infection. RSV infection induces an array of pro-inflammatory chemokines and cytokines that recruit neutrophils, macrophages and lymphocytes to the respiratory tract resulting in respiratory disease. Although the mechanisms responsible for induction of these chemokines and cytokines are unclear, studies on the closely related human (H)RSV suggest that activation of NF-kappaB via TLR4 and TLR3 signalling pathways is involved. An understanding of the mechanisms by which BRSV is able to establish infection and induce an inflammatory response has been facilitated by advances in reverse genetics, which have enabled manipulation of the virus genome. These studies have demonstrated an important role for the non-structural proteins in anti-interferon activity, a role for a virokinin, released during proteolytic cleavage of the fusion protein, in the inflammatory response and a role for the SH and the secreted form of the G protein in establishing pulmonary infection. Knowledge gained from these studies has also provided the opportunity to develop safe, stable, live attenuated virus vaccine candidates.


Subject(s)
Cattle Diseases/virology , Cattle/virology , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus, Bovine/physiology , Animals , Cattle Diseases/epidemiology , Cattle Diseases/immunology , Cattle Diseases/prevention & control , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Bovine/genetics , Respiratory Syncytial Virus, Bovine/immunology , Respiratory Syncytial Virus, Bovine/pathogenicity
18.
Anim Health Res Rev ; 8(2): 207-13, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18218161

ABSTRACT

Bovine respiratory syncytial virus (BRSV) causes severe respiratory disease in young cattle. Much like the human respiratory syncytial virus, BRSV induces immunomodulation in the infected host, favoring a Th2 response. Several groups have demonstrated IgE responses to BRSV proteins during infection and particularly in response to vaccination with formalin-inactivated vaccine in the field and experimentally. Newer vaccine modalities that favor a shift to Th1 cytokine production have provided promising results. Infection with BRSV is a major contributor to the multi-pathogen disease, bovine respiratory disease complex. This review stresses the unique immunomodulatory aspects of BRSV infection, vaccination and its interaction with the host's immune system.


Subject(s)
Cattle Diseases/immunology , Cattle Diseases/virology , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Bovine/pathogenicity , Animals , Antibodies, Viral/biosynthesis , Cattle , Host-Pathogen Interactions , Immunologic Factors , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Bovine/immunology , Vaccination/veterinary
19.
Vet Microbiol ; 118(3-4): 161-8, 2006 Dec 20.
Article in English | MEDLINE | ID: mdl-16959444

ABSTRACT

Bovine respiratory syncytial virus (BRSV), a member of the subfamily Pneumovirinae, family Paramyxoviridae, is a major cause of respiratory disorders in young cattle. A number of studies were conducted to validate a reliable animal model for the infection, since BRSV inoculation on the natural host is costly and often unsuccessful. Unfortunately, after inoculation of BRSV in Balb/C mice, viral replication may be detected; however, evident pathological alterations are absent on the experimentally infected animals. In order to establish a mice model that could be used further for preliminary studies of pathological and immunological aspects of BRSV infection, three mice inbred lineages (Balb/C, A/J and C57BL6), possessing different genetic backgrounds, were tested about its susceptibility to the inoculation with BRSV. Animals were inoculated through the nasal and ocular routes and were observed after inoculation. At 7 days post-inoculation (dpi) animals were necropsied and virological (virus isolation and viral nucleic acid amplification) as well as histopathological examinations were performed. A/J and C57BL6 showed interstitial pneumonia, when compared to the Balb/C group. These findings shows that mice may constitute a suitable model for the study of BRSV infections, depending on the mice strain used for experimental inoculations.


Subject(s)
Cattle Diseases/virology , Disease Models, Animal , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus, Bovine/pathogenicity , Animals , Cattle , DNA, Viral/isolation & purification , Immunohistochemistry/veterinary , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Respiratory Syncytial Virus Infections/virology , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sensitivity and Specificity , Species Specificity
20.
J Gen Virol ; 87(Pt 6): 1659-1667, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16690931

ABSTRACT

The BRSV fusion (F) protein is cleaved at two furin consensus sequence sites, resulting in the generation of disulphide-linked F1 and F2 subunits and the release of an intervening peptide of 27 amino acids (pep27), which is converted into a biologically active tachykinin (virokinin). The role of the virokinin and the importance of one of the furin cleavage sites, FCS-2 [RA(R/K)R109], in the pathogenesis of BRSV infection and in the subsequent development of immunity was studied in gnotobiotic calves infected with a recombinant BRSV (rBRSV) lacking pep27 (rBRSVdelta p27) or with rBRSV108/109, which contains two amino acid substitutions in FCS-2 (RANN109). Although replication of the mutant viruses and the parental wild-type (WT) rBRSV in the lungs was similar, the extent of gross and microscopic lesions induced by the mutant viruses was less than that induced by WT rBRSV. Furthermore, the numbers of eosinophils in the lungs of calves infected with the mutant viruses were significantly less than that in calves infected with WT virus. These observations suggest a role for the virokinin in the pathogenesis of BRSV infection. Following mucosal immunization with rBRSVdelta p27, the levels of BRSV-specific serum antibodies were similar to those induced by WT virus. In contrast, the level of neutralizing antibodies induced by rBRSV108/109 was 10-fold lower than that induced by WT virus. Nevertheless, resistance to BRSV challenge induced by the mutant and WT viruses was similar, suggesting that neither pep27 nor FCS-2 plays a major role in the induction of protective immunity.


Subject(s)
Cattle Diseases/immunology , Mutation , Pneumonia/veterinary , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus, Bovine/pathogenicity , Viral Fusion Proteins/genetics , Amino Acid Sequence , Animals , Cattle , Cattle Diseases/virology , Cells, Cultured , Furin/metabolism , Germ-Free Life , Immunization , Molecular Sequence Data , Pneumonia/immunology , Pneumonia/physiopathology , Recombination, Genetic , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Bovine/genetics , Respiratory Syncytial Virus, Bovine/immunology , Tachykinins/metabolism , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...