Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Vet Res ; 53(1): 107, 2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36510312

ABSTRACT

Bovine respiratory syncytial virus (BRSV) is a major cause of respiratory disease in cattle. Genomic sequencing can resolve phylogenetic relationships between virus populations, which can be used to infer transmission routes and potentially inform the design of biosecurity measures. Sequencing of short (<2000 nt) segments of the 15 000-nt BRSV genome has revealed geographic and temporal clustering of BRSV populations, but insufficient variation to distinguish viruses collected from herds infected close together in space and time. This study investigated the potential for whole-genome sequencing to reveal sufficient genomic variation for inferring transmission routes between herds. Next-generation sequencing (NGS) data were generated from experimental infections and from natural outbreaks in Jämtland and Uppsala counties in Sweden. Sufficient depth of coverage for analysis of consensus and sub-consensus sequence diversity was obtained from 47 to 20 samples respectively. Few (range: 0-6 polymorphisms across the six experiments) consensus-level polymorphisms were observed along experimental transmissions. A much higher level of diversity (146 polymorphic sites) was found among the consensus sequences from the outbreak samples. The majority (144/146) of polymorphisms were between rather than within counties, suggesting that consensus whole-genome sequences show insufficient spatial resolution for inferring direct transmission routes, but might allow identification of outbreak sources at the regional scale. By contrast, within-sample diversity was generally higher in the experimental than the outbreak samples. Analyses to infer known (experimental) and suspected (outbreak) transmission links from within-sample diversity data were uninformative. In conclusion, analysis of the whole-genome sequence of BRSV from experimental samples discriminated between circulating isolates from distant areas, but insufficient diversity was observed between closely related isolates to aid local transmission route inference.


Subject(s)
Cattle Diseases , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Bovine , Cattle , Animals , Respiratory Syncytial Virus, Bovine/genetics , Phylogeny , Cattle Diseases/epidemiology , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/veterinary , Antibodies, Viral
2.
J Gen Virol ; 102(1)2021 01.
Article in English | MEDLINE | ID: mdl-33054904

ABSTRACT

Although enveloped viruses canonically mediate particle entry through virus-cell fusion, certain viruses can spread by cell-cell fusion, brought about by receptor engagement and triggering of membrane-bound, viral-encoded fusion proteins on the surface of cells. The formation of pathogenic syncytia or multinucleated cells is seen in vivo, but their contribution to viral pathogenesis is poorly understood. For the negative-strand paramyxoviruses respiratory syncytial virus (RSV) and Nipah virus (NiV), cell-cell spread is highly efficient because their oligomeric fusion protein complexes are active at neutral pH. The recently emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has also been reported to induce syncytia formation in infected cells, with the spike protein initiating cell-cell fusion. Whilst it is well established that fusion protein-specific antibodies can block particle attachment and/or entry into the cell (canonical virus neutralization), their capacity to inhibit cell-cell fusion and the consequences of this neutralization for the control of infection are not well characterized, in part because of the lack of specific tools to assay and quantify this activity. Using an adapted bimolecular fluorescence complementation assay, based on a split GFP-Renilla luciferase reporter, we have established a micro-fusion inhibition test (mFIT) that allows the identification and quantification of these neutralizing antibodies. This assay has been optimized for high-throughput use and its applicability has been demonstrated by screening monoclonal antibody (mAb)-mediated inhibition of RSV and NiV fusion and, separately, the development of fusion-inhibitory antibodies following NiV vaccine immunization in pigs. In light of the recent emergence of coronavirus disease 2019 (COVID-19), a similar assay was developed for SARS-CoV-2 and used to screen mAbs and convalescent patient plasma for fusion-inhibitory antibodies. Using mFITs to assess antibody responses following natural infection or vaccination is favourable, as this assay can be performed entirely at low biocontainment, without the need for live virus. In addition, the repertoire of antibodies that inhibit cell-cell fusion may be different to those that inhibit particle entry, shedding light on the mechanisms underpinning antibody-mediated neutralization of viral spread.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antibodies, Viral/pharmacology , COVID-19/diagnosis , Henipavirus Infections/diagnosis , High-Throughput Screening Assays , Respiratory Syncytial Virus Infections/diagnosis , Viral Fusion Proteins/antagonists & inhibitors , Animals , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/metabolism , Antibodies, Viral/isolation & purification , Antibodies, Viral/metabolism , COVID-19/immunology , COVID-19/virology , Cell Fusion , Convalescence , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Henipavirus Infections/immunology , Henipavirus Infections/virology , Humans , Immune Sera/chemistry , Luciferases/genetics , Luciferases/metabolism , Models, Molecular , Nipah Virus/immunology , Nipah Virus/pathogenicity , Protein Conformation , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus, Human/pathogenicity , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Swine , Viral Fusion Protein Inhibitors/chemistry , Viral Fusion Protein Inhibitors/metabolism , Viral Fusion Protein Inhibitors/pharmacology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology
3.
Infect Immun ; 87(6)2019 06.
Article in English | MEDLINE | ID: mdl-30962401

ABSTRACT

The Gram-negative bacterium Mannheimia haemolytica is the primary bacterial species associated with bovine respiratory disease (BRD) and is responsible for significant economic losses to livestock industries worldwide. Healthy cattle are frequently colonized by commensal serotype A2 strains, but disease is usually caused by pathogenic strains of serotype A1. For reasons that are poorly understood, a transition occurs within the respiratory tract and a sudden explosive proliferation of serotype A1 bacteria leads to the onset of pneumonic disease. Very little is known about the interactions of M. haemolytica with airway epithelial cells of the respiratory mucosa which might explain the different abilities of serotype A1 and A2 strains to cause disease. In the present study, host-pathogen interactions in the bovine respiratory tract were mimicked using a novel differentiated bovine bronchial epithelial cell (BBEC) infection model. In this model, differentiated BBECs were inoculated with serotype A1 or A2 strains of M. haemolytica and the course of infection followed over a 5-day period by microscopic assessment and measurement of key proinflammatory mediators. We have demonstrated that serotype A1, but not A2, M. haemolytica invades differentiated BBECs by transcytosis and subsequently undergoes rapid intracellular replication before spreading to adjacent cells and causing extensive cellular damage. Our findings suggest that the explosive proliferation of serotype A1 M. haemolytica that occurs within the bovine respiratory tract prior to the onset of pneumonic disease is potentially due to bacterial invasion of, and rapid proliferation within, the mucosal epithelium. The discovery of this previously unrecognized mechanism of pathogenesis is important because it will allow the serotype A1-specific virulence determinants responsible for invasion to be identified and thereby provide opportunities for the development of new strategies for combatting BRD aimed at preventing early colonization and infection of the bovine respiratory tract.


Subject(s)
Epithelial Cells/microbiology , Mannheimia haemolytica/pathogenicity , Pasteurellosis, Pneumonic/microbiology , Animals , Bronchi/cytology , Bronchi/microbiology , Cattle , Mannheimia haemolytica/growth & development , Mannheimia haemolytica/physiology , Respiratory System/microbiology , Virulence
4.
J Virol ; 92(8)2018 04 15.
Article in English | MEDLINE | ID: mdl-29386289

ABSTRACT

African swine fever virus (ASFV) causes an acute hemorrhagic fever in domestic pigs, with high socioeconomic impact. No vaccine is available, limiting options for control. Although live attenuated ASFV can induce up to 100% protection against lethal challenge, little is known of the antigens which induce this protective response. To identify additional ASFV immunogenic and potentially protective antigens, we cloned 47 viral genes in individual plasmids for gene vaccination and in recombinant vaccinia viruses. These antigens were selected to include proteins with different functions and timing of expression. Pools of up to 22 antigens were delivered by DNA prime and recombinant vaccinia virus boost to groups of pigs. Responses of immune lymphocytes from pigs to individual recombinant proteins and to ASFV were measured by interferon gamma enzyme-linked immunosorbent spot (ELISpot) assays to identify a subset of the antigens that consistently induced the highest responses. All 47 antigens were then delivered to pigs by DNA prime and recombinant vaccinia virus boost, and pigs were challenged with a lethal dose of ASFV isolate Georgia 2007/1. Although pigs developed clinical and pathological signs consistent with acute ASFV, viral genome levels were significantly reduced in blood and several lymph tissues in those pigs immunized with vectors expressing ASFV antigens compared with the levels in control pigs.IMPORTANCE The lack of a vaccine limits the options to control African swine fever. Advances have been made in the development of genetically modified live attenuated ASFV that can induce protection against challenge. However, there may be safety issues relating to the use of these in the field. There is little information about ASFV antigens that can induce a protective immune response against challenge. We carried out a large screen of 30% of ASFV antigens by delivering individual genes in different pools to pigs by DNA immunization prime and recombinant vaccinia virus boost. The responses in immunized pigs to these individual antigens were compared to identify the most immunogenic. Lethal challenge of pigs immunized with a pool of antigens resulted in reduced levels of virus in blood and lymph tissues compared to those in pigs immunized with control vectors. Novel immunogenic ASFV proteins have been identified for further testing as vaccine candidates.


Subject(s)
African Swine Fever Virus/immunology , African Swine Fever/immunology , Immunization, Secondary , Vaccines, DNA/immunology , Vaccinia virus/immunology , Viral Proteins/immunology , African Swine Fever/genetics , African Swine Fever/prevention & control , African Swine Fever Virus/genetics , Animals , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Swine , Vaccines, DNA/genetics , Vaccinia virus/genetics , Viral Proteins/genetics
5.
J Fish Biol ; 95(3): 855-869, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31219181

ABSTRACT

Here, we present a gillnet survey of Lake Liambezi a 370 km2 shallow ephemeral floodplain lake situated in north-eastern Namibia, which is fed irregularly by the upper Zambezi and Kwando Rivers during years of high flooding. The lake dried up in 1985 and, with the exception of sporadic minor annual inundation events, remained dry until 2007. We describe the temporal succession of fish species over an 8 year period from initial inundation 2007 to maturation in 2014. The succession of the fish community did not follow the typical pattern of opportunistic strategists during colonisation, to periodic strategists that are eventually succeeded by equilibrium strategists. Instead, the evolution of the fish community was characterised by three distinct phases. The first phase involved the inundation and colonisation of the lake in 2007, followed by its decline until the floods that filled the lake in 2009. During this phase the lake was colonised by fishes from the adjacent upper Zambezi and Chobe River floodplains. Fish communities predominantly comprised floodplain specialists including the barbs Enteromius paludinosus and Enteromius poechii, the mormyrid Marcusenius altisambesi and catfishes Schilbe intermedius and Clarias gariepinus. The filling of the lake in the March 2009 floods marked the beginning of the second, successional phase. The barbs declined in abundance and the alestid Rhabdalestes maunensis underwent explosive population growth between 2009 and 2010, but populations crashed equally rapidly and were replaced by Brycinus lateralis which, together with S. intermedius went on to dominate the fish community 2011-2014. Larger, slower growing tilapiine cichlids increased steadily in abundance and became the dominant components in a 2700 t y-1 artisanal fishery that developed on the lake. The fish community in the ephemeral Lake Liambezi is clearly influenced by numerous factors including connectivity, lake level fluctuations, competition and the effects of fishing, which may disrupt typical succession processes in floodplain ecosystems.


Subject(s)
Biodiversity , Fishes/classification , Fishes/physiology , Floods , Lakes , Animals , Namibia , Population Dynamics
6.
J Gen Virol ; 98(7): 1587-1599, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28714847

ABSTRACT

Bovine respiratory syncytial virus (BRSV) is an important cause of respiratory disease in young cattle and is closely related to human RSV (HRSV), which causes severe respiratory disease in infants and the elderly. The RSV genome encodes a small hydrophobic (SH) protein with viroporin activity. Previous studies have shown that recombinant BRSV lacking the SH gene (rBRSVΔSH) is attenuated in the lungs, but not in the upper respiratory tract, of calves and mucosal vaccination with rBRSVΔSH induced long-lasting protective immunity. Attenuation of rBRSVΔSH may be due to the ability of this virus to induce an early innate response as rBRSVΔSH induces higher levels of pro-inflammatory cytokines than wild-type (wt) rBRSV. In this study, we investigated the effects of the BRSV SH protein on NF-κB p65 phosphorylation, a master step in the regulation of pro-inflammatory cytokines. Expression of SH resulted in the inhibition of NF-κB p65 phosphorylation in response to BRSV infection and extracellular lipopolysaccharide, and a reduction in the production of pro-inflammatory cytokines. In contrast, rBRSVΔSH does not inhibit NF-κB p65 phosphorylation in bovine antigen-presenting cells, including monocytes, macrophages and dendritic cells, resulting in increased expression of pro-inflammatory cytokines and increased activation of T cells compared to cells infected with wt BRSV. These findings highlight an important role for the BRSV SH protein in immune modulation.


Subject(s)
Cytokines/metabolism , Dendritic Cells/immunology , Macrophages/immunology , Monocytes/immunology , Respiratory Syncytial Virus, Bovine/metabolism , Retroviridae Proteins, Oncogenic/immunology , Transcription Factor RelA/metabolism , Animals , Cattle , Cattle Diseases/immunology , Cattle Diseases/virology , Cell Line , Dendritic Cells/metabolism , Dendritic Cells/virology , Humans , Lipopolysaccharides/metabolism , Lymphocyte Activation/immunology , Macrophages/metabolism , Macrophages/virology , Mice , Monocytes/metabolism , Monocytes/virology , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation , RAW 264.7 Cells , Respiratory Syncytial Virus, Bovine/genetics , Respiratory Syncytial Virus, Bovine/immunology , Retroviridae Proteins, Oncogenic/genetics , Retroviridae Proteins, Oncogenic/metabolism , T-Lymphocytes/immunology
7.
J Virol ; 90(11): 5485-5498, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27009962

ABSTRACT

UNLABELLED: Human respiratory syncytial virus (hRSV) vaccine development has received new impetus from structure-based studies of its main protective antigen, the fusion (F) glycoprotein. Three soluble forms of F have been described: monomeric, trimeric prefusion, and trimeric postfusion. Most human neutralizing antibodies recognize epitopes found exclusively in prefusion F. Although prefusion F induces higher levels of neutralizing antibodies than does postfusion F, postfusion F can also induce protection against virus challenge in animals. However, the immunogenicity and protective efficacy of the three forms of F have not hitherto been directly compared. Hence, BALB/c mice were immunized with a single dose of the three proteins adjuvanted with CpG and challenged 4 weeks later with virus. Serum antibodies, lung virus titers, weight loss, and pulmonary pathology were evaluated after challenge. Whereas small amounts of postfusion F were sufficient to protect mice, larger amounts of monomeric and prefusion F proteins were required for protection. However, postfusion and monomeric F proteins were associated with more pathology after challenge than was prefusion F. Antibodies induced by all doses of prefusion F, in contrast to other F protein forms, reacted predominantly with the prefusion F conformation. At high doses, prefusion F also induced the highest titers of neutralizing antibodies, and all mice were protected, yet at low doses of the immunogen, these antibodies neutralized virus poorly, and mice were not protected. These findings should be considered when developing new hRSV vaccine candidates. IMPORTANCE: Protection against hRSV infection is afforded mainly by neutralizing antibodies, which recognize mostly epitopes found exclusively in the viral fusion (F) glycoprotein trimer, folded in its prefusion conformation, i.e., before activation for membrane fusion. Although prefusion F is able to induce high levels of neutralizing antibodies, highly stable postfusion F (found after membrane fusion) is also able to induce neutralizing antibodies and protect against infection. In addition, a monomeric form of hRSV F that shares epitopes with prefusion F was recently reported. Since each of the indicated forms of hRSV F may have advantages and disadvantages for the development of safe and efficacious subunit vaccines, a direct comparison of the immunogenic properties and protective efficacies of the different forms of hRSV F was made in a mouse model. The results obtained show important differences between the noted immunogens that should be borne in mind when considering the development of hRSV vaccines.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Viruses/chemistry , Respiratory Syncytial Viruses/immunology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/immunology , Adjuvants, Immunologic , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Dose-Response Relationship, Immunologic , Epitopes/immunology , Female , Humans , Immunization , Immunogenicity, Vaccine , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/immunology , Protein Conformation , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/isolation & purification , Viral Fusion Proteins/administration & dosage
8.
J Gen Virol ; 97(10): 2703-2718, 2016 10.
Article in English | MEDLINE | ID: mdl-27528389

ABSTRACT

Dendritic cells (DC) are central to the initiation of immune responses, and various approaches have been used to target vaccines to DC in order to improve immunogenicity. Cannulation of lymphatic vessels allows for the collection of DC that migrate from the skin. These migrating DC are involved in antigen uptake and presentation following vaccination. Human replication-deficient adenovirus (AdV) 5 is a promising vaccine vector for delivery of recombinant antigens. Although the mechanism of AdV attachment and penetration has been extensively studied in permissive cell lines, few studies have addressed the interaction of AdV with DC. In this study, we investigated the interaction of bovine skin-migrating DC and replication-deficient AdV-based vaccine vectors. We found that, despite lack of expression of Coxsackie B-Adenovirus Receptor and other known adenovirus receptors, AdV readily enters skin-draining DC via an actin-dependent endocytosis. Virus exit from endosomes was pH independent, and neutralizing antibodies did not prevent virus entry but did prevent virus translocation to the nucleus. We also show that combining adenovirus with adjuvant increases the absolute number of intracellular virus particles per DC but not the number of DC containing intracellular virus. This results in increased trans-gene expression and antigen presentation. We propose that, in the absence of Coxsackie B-Adenovirus Receptor and other known receptors, AdV5-based vectors enter skin-migrating DC using actin-dependent endocytosis which occurs in skin-migrating DC, and its relevance to vaccination strategies and vaccine vector targeting is discussed.


Subject(s)
Actins/immunology , Adenovirus Infections, Human/virology , Adenoviruses, Human/physiology , Dendritic Cells/virology , Genetic Vectors/physiology , Phagocytosis , Skin/virology , Adenovirus Infections, Human/immunology , Adenovirus Infections, Human/physiopathology , Adenoviruses, Human/genetics , Animals , Cattle , Cell Movement , Dendritic Cells/immunology , Endocytosis , Genetic Vectors/genetics , Humans , Skin/cytology , Skin/immunology , Transduction, Genetic
9.
Vet Res ; 47: 20, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26796101

ABSTRACT

Peste des petits ruminants virus (PPRV) causes an economically important disease of sheep and goats, primarily in developing countries. It is becoming the object of intensive international control efforts. Current vaccines do not allow vaccinated and infected animals to be distinguished (no DIVA capability). We have previously shown that recombinant, replication-defective, adenovirus expressing the PPRV H glycoprotein (AdH) gives full protection against wild type PPRV challenge. We have now tested lower doses of the vaccine, as well as AdH in combination with a similar construct expressing the PPRV F glycoprotein (AdF). We show here that, in a local breed of goat in a country where PPR disease is common (Kenya), as little as 10(7) pfu of AdH gives significant protection against PPRV challenge, while a vaccine consisting of 10(8) pfu of each of AdH and AdF gives apparently sterile protection. These findings underline the utility of these constructs as DIVA vaccines for use in PPR control.


Subject(s)
Goat Diseases/prevention & control , Peste-des-Petits-Ruminants/prevention & control , Peste-des-petits-ruminants virus , Viral Vaccines/immunology , Adenoviridae , Animals , Antibodies, Viral/blood , Antibody Specificity , Chlorocebus aethiops , Glycoproteins/immunology , Goat Diseases/virology , Goats , Nucleocapsid Proteins/immunology , Vero Cells , Viremia
10.
J Immunol ; 193(1): 208-22, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24890724

ABSTRACT

In humans and mice, γδ T cells represent <5% of the total circulating lymphocytes. In contrast, the γδ T cell compartment in ruminants accounts for 15-60% of the total circulating mononuclear lymphocytes. Despite the existence of CD4(+)CD25(high) Foxp3(+) T cells in the bovine system, these are neither anergic nor suppressive. We present evidence showing that bovine γδ T cells are the major regulatory T cell subset in peripheral blood. These γδ T cells spontaneously secrete IL-10 and proliferate in response to IL-10, TGF-ß, and contact with APCs. IL-10-expressing γδ T cells inhibit Ag-specific and nonspecific proliferation of CD4(+) and CD8(+) T cells in vitro. APC subsets expressing IL-10 and TFG-ß regulate proliferation of γδ T cells producing IL-10. We propose that γδ T cells are a major regulatory T cell population in the bovine system.


Subject(s)
Interleukin-10/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Antigens/immunology , Cattle , Cell Proliferation , Mice , T-Lymphocytes, Regulatory/cytology
SELECTION OF CITATIONS
SEARCH DETAIL