Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Viruses ; 14(1)2022 01 06.
Article in English | MEDLINE | ID: mdl-35062301

ABSTRACT

Human respiratory syncytial virus (hRSV) infection brings a wide spectrum of clinical outcomes, from a mild cold to severe bronchiolitis or even acute interstitial pneumonia. Among the known factors influencing this clinical diversity, genetic background has often been mentioned. In parallel, recent evidence has also pointed out that an early infectious experience affects heterologous infections severity. Here, we analyzed the importance of these two host-related factors in shaping the immune response in pneumoviral disease. We show that a prior gammaherpesvirus infection improves, in a genetic background-dependent manner, the immune system response against a subsequent lethal dose of pneumovirus primary infection notably by inducing a systematic expansion of the CD8+ bystander cell pool and by modifying the resident alveolar macrophages (AMs) phenotype to induce immediate cyto/chemokinic responses upon pneumovirus exposure, thereby drastically attenuating the host inflammatory response without affecting viral replication. Moreover, we show that these AMs present similar rapid and increased production of neutrophil chemokines both in front of pneumoviral or bacterial challenge, confirming recent studies attributing a critical antibacterial role of primed AMs. These results corroborate other recent studies suggesting that the innate immunity cells are themselves capable of memory, a capacity hitherto reserved for acquired immunity.


Subject(s)
Genetic Background , Herpesviridae Infections/immunology , Macrophages, Alveolar/immunology , Pneumovirus Infections/immunology , Pneumovirus/immunology , Rhadinovirus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Female , Herpesviridae Infections/genetics , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Immunity, Innate , Inflammation/immunology , Lung/immunology , Lung/pathology , Lung/virology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/immunology , Pneumococcal Infections/immunology , Pneumovirus/physiology , Pneumovirus Infections/genetics , Pneumovirus Infections/pathology , Pneumovirus Infections/virology , Rhadinovirus/physiology
2.
Viruses ; 12(12)2020 12 02.
Article in English | MEDLINE | ID: mdl-33276587

ABSTRACT

The paramyxo- and pneumovirus family includes a wide range of viruses that can cause respiratory and/or systemic infections in humans and animals. The significant disease burden of these viruses is further exacerbated by the limited therapeutics that are currently available. Host cellular proteins that can antagonize or limit virus replication are therefore a promising area of research to identify candidate molecules with the potential for host-targeted therapies. Host proteins known as host cell restriction factors are constitutively expressed and/or induced in response to virus infection and include proteins from interferon-stimulated genes (ISGs). Many ISG proteins have been identified but relatively few have been characterized in detail and most studies have focused on studying their antiviral activities against particular viruses, such as influenza A viruses and human immunodeficiency virus (HIV)-1. This review summarizes current literature regarding host cell restriction factors against paramyxo- and pneumoviruses, on which there is more limited data. Alongside discussion of known restriction factors, this review also considers viral countermeasures in overcoming host restriction, the strengths and limitations in different experimental approaches in studies reported to date, and the challenges in reconciling differences between in vitro and in vivo data. Furthermore, this review provides an outlook regarding the landscape of emerging technologies and tools available to study host cell restriction factors, as well as the suitability of these proteins as targets for broad-spectrum antiviral therapeutics.


Subject(s)
Host-Pathogen Interactions , Paramyxoviridae Infections/virology , Paramyxovirinae/physiology , Pneumovirus Infections/virology , Pneumovirus/physiology , Animals , Biomarkers , Gene Expression Regulation, Viral , Host Specificity , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Paramyxoviridae Infections/genetics , Paramyxoviridae Infections/metabolism , Pneumovirus Infections/genetics , Pneumovirus Infections/metabolism , Viral Tropism , Virus Replication
3.
J Leukoc Biol ; 102(3): 905-913, 2017 09.
Article in English | MEDLINE | ID: mdl-28619948

ABSTRACT

Administration of immunobiotic Lactobacillus plantarum (Lp) directly to the respiratory mucosa promotes cross-protection against lethal pneumovirus infection via B-cell-independent mechanisms. In this study, we examined Lp-mediated cross protection in Rag1-/- mice which cannot clear virus from lung tissue. Although Lp was initially protective, Rag1-/- mice ultimately succumbed to a delayed lethal outcome associated with local production of the proinflammatory cytokines CCL1, -2, and -7, granulocyte recruitment, and ongoing virus replication. By contrast, CD8null mice, which are fully capable of clearing virus, are protected by Lp with no delayed lethal outcome, granulocyte recruitment to the airways, or induction of CCL7. Repeated administration of Lp to virus-infected Rag1-/- mice had no impact on delayed mortality. Moreover, administration of Lp to the respiratory mucosa resulted in no induction of IFN-α or -ß in Rag1-/- or wild-type mice, and IFN-abR gene deletion had no impact on Lp-mediated protection. Overall, our findings indicate that although Lp administered to the respiratory tract has substantial impact on lethal virus-induced inflammation in situ, endogenous virus clearance mechanisms are needed to promote sustained protection. Our results suggest that a larger understanding of the mechanisms and mediators that limit acute virus-induced inflammation may yield new and useful therapeutic modalities.


Subject(s)
Homeodomain Proteins/genetics , Lactobacillus plantarum , Pneumovirus Infections/immunology , Pneumovirus Infections/therapy , Pneumovirus/immunology , Animals , Chemokines, CC/genetics , Chemokines, CC/immunology , Mice , Mice, Knockout , Pneumovirus Infections/genetics
4.
Sci Rep ; 7(1): 2353, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28539639

ABSTRACT

Respiratory syncytial virus (RSV)-bronchiolitis is a major cause of infant morbidity and mortality and a risk factor for subsequent asthma. We showed previously that toll-like receptor (TLR)7 in plasmacytoid dendritic cells (pDCs) is critical for protection against bronchiolitis and asthma in mice infected with pneumonia virus of mice (PVM), the mouse homolog of RSV. This lack of redundancy was unexpected as interferon-ß promotor stimulator-1 (IPS-1) signalling, downstream of RIG-I-like receptor (RLR) and not TLR7 activation, contributes to host defence in hRSV-inoculated adult mice. To further clarify the role of IPS-1 signalling, we inoculated IPS-1-/- and WT mice with PVM in early-life, and again in later-life, to model the association between bronchiolitis and asthma. IPS-1 deficiency predisposed to severe PVM bronchiolitis, characterised by neutrophilic inflammation and necroptotic airway epithelial cell death, high mobility group box 1 (HMGB1) and IL-33 release, and downstream type-2 inflammation. Secondary infection induced an eosinophilic asthma-like pathophysiology in IPS-1-/- but not WT mice. Mechanistically, we identified that IPS-1 is necessary for pDC recruitment, IFN-α production and viral control. Our findings suggest that TLR7 and RLR signalling work collaboratively to optimally control the host response to pneumovirus infection thereby protecting against viral bronchiolitis and subsequent asthma.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Asthma/metabolism , Bronchiolitis/metabolism , Murine pneumonia virus/physiology , Pneumovirus Infections/virology , Adaptor Proteins, Signal Transducing/genetics , Animals , Asthma/genetics , Bronchiolitis/genetics , DEAD Box Protein 58/metabolism , Dendritic Cells/metabolism , Host-Pathogen Interactions , Interferon-alpha/metabolism , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Pneumovirus Infections/genetics , Pneumovirus Infections/metabolism , Signal Transduction , Toll-Like Receptor 7/metabolism
5.
Sci Rep ; 6: 38139, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27905537

ABSTRACT

Pneumonia Virus of Mice (PVM) is the only virus that shares the Pneumovirus genus of the Paramyxoviridae family with Respiratory Syncytial Virus (RSV). A deadly mouse pathogen, PVM has the potential to serve as a robust animal model of RSV infection, since human RSV does not fully replicate the human pathology in mice. Like RSV, PVM also encodes two nonstructural proteins that have been implicated to suppress the IFN pathway, but surprisingly, they exhibit no sequence similarity with their RSV equivalents. The molecular mechanism of PVM NS function, therefore, remains unknown. Here, we show that recombinant PVM NS proteins degrade the mouse counterparts of the IFN pathway components. Proteasomal degradation appears to be mediated by ubiquitination promoted by PVM NS proteins. Interestingly, NS proteins of PVM lowered the levels of several ISG (IFN-stimulated gene) proteins as well. These results provide a molecular foundation for the mechanisms by which PVM efficiently subverts the IFN response of the murine cell. They also reveal that in spite of their high sequence dissimilarity, the two pneumoviral NS proteins are functionally and mechanistically similar.


Subject(s)
Interferons/metabolism , Murine pneumonia virus/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interferons/genetics , Metabolic Networks and Pathways/immunology , Mice , Murine pneumonia virus/genetics , Murine pneumonia virus/pathogenicity , Pneumovirus Infections/genetics , Pneumovirus Infections/immunology , Pneumovirus Infections/virology , Proteolysis , Respiratory Syncytial Virus Infections/etiology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/metabolism , Respiratory Syncytial Viruses/pathogenicity , Viral Nonstructural Proteins/genetics
6.
Immunobiology ; 221(3): 494-502, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26621546

ABSTRACT

The pneumonia virus of mice (PVM) model is used to study respiratory syncytial virus (RSV) pathogenesis. The outcome of PVM infection varies in different inbred mouse strains, BALB/c being highly susceptible and C57BL/6 more resistant. As the disease symptoms induced by RSV infection can become more severe as people age, we examined the primary and secondary immune responses to infection with PVM in aged BALB/c and C57BL/6 mice. Based on clinical parameters, aged C57BL/6 mice displayed less severe disease than young adult mice when infected with 3000pfu of PVM-15, while BALB/c mice were equally susceptible at both ages showing significant weight loss and high levels of virus replication. Furthermore, after primary infection the CD4(+) T cell numbers in the lungs were higher in young adult mice, while the CD8(+) T cell numbers were comparable in both age groups and strains. When either C57BL/6 or BALB/c mice were infected with PVM as young adults and then re-infected as aged mice, they were protected from clinical disease, while virus replication was reduced. In contrast to mice with a primary PVM-infection, re-infected mice did not have infiltration of neutrophils or inflammatory mediators in the lung. BALB/c mice had higher virus neutralizing antibody levels in the serum and lung than C57BL/6 mice upon re-infection. Re-infection with PVM led to significant influx of effector CD4(+) T cells into the lungs when compared to aged mice with a primary infection, while this cell population was decreased in the lung draining lymph nodes in both mouse strains. After re-infection the effector CD8(+) T cell population was also decreased in the lung draining lymph nodes in both mouse strain when compared to aged mice after primary infection. However, the central memory CD4(+) and CD8(+) T cells were significantly enhanced in numbers in the lungs and draining lymph nodes of both mouse strains after re-infection, and these numbers were higher for C57BL/6 mice.


Subject(s)
Genetic Background , Genetic Predisposition to Disease , Murine pneumonia virus/physiology , Pneumovirus Infections/genetics , Pneumovirus Infections/virology , Adaptive Immunity , Age Factors , Animals , Cell Line , Cytokines/metabolism , Immunologic Memory , Inflammation Mediators/metabolism , Lung/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pneumovirus Infections/immunology , Pneumovirus Infections/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
7.
J Immunol ; 192(11): 5265-72, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24748495

ABSTRACT

We have shown previously that priming of respiratory mucosa with live Lactobacillus species promotes robust and prolonged survival from an otherwise lethal infection with pneumonia virus of mice, a property known as heterologous immunity. Lactobacillus priming results in a moderate reduction in virus recovery and a dramatic reduction in virus-induced proinflammatory cytokine production; the precise mechanisms underlying these findings remain to be elucidated. Because B cells have been shown to promote heterologous immunity against respiratory virus pathogens under similar conditions, in this study we explore the role of B cells in Lactobacillus-mediated protection against acute pneumovirus infection. We found that Lactobacillus-primed mice feature elevated levels of airway Igs IgG, IgA, and IgM and lung tissues with dense, B cell (B220(+))-enriched peribronchial and perivascular infiltrates with germinal centers consistent with descriptions of BALT. No B cells were detected in lung tissue of Lactobacillus-primed B cell deficient µMT mice or Jh mice, and Lactobacillus-primed µMT mice had no characteristic infiltrates or airway Igs. Nonetheless, we observed diminished virus recovery and profound suppression of virus-induced proinflammatory cytokines CCL2, IFN-γ, and CXCL10 in both wild-type and Lactobacillus-primed µMT mice. Furthermore, Lactobacillus plantarum-primed, B cell-deficient µMT and Jh mice were fully protected from an otherwise lethal pneumonia virus of mice infection, as were their respective wild-types. We conclude that B cells are dispensable for Lactobacillus-mediated heterologous immunity and were not crucial for promoting survival in response to an otherwise lethal pneumovirus infection.


Subject(s)
B-Lymphocytes/immunology , Lactobacillus/immunology , Lung/immunology , Pneumovirus Infections/immunology , Pneumovirus/immunology , Respiratory Mucosa/immunology , Animals , Antibodies, Bacterial/genetics , Antibodies, Bacterial/immunology , Cytokines/genetics , Cytokines/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Pneumovirus/genetics , Pneumovirus Infections/genetics , Pneumovirus Infections/pathology , Respiratory Mucosa/pathology , Respiratory Mucosa/virology
8.
Blood ; 114(13): 2649-56, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19652202

ABSTRACT

Eosinophils are recruited to the lung in response to infection with pneumovirus pathogens and have been associated with both the pathophysiologic sequelae of infection and, more recently, with accelerated virus clearance. Here, we demonstrate that the pneumovirus pathogens, respiratory syncytial virus (RSV) and pneumonia virus of mice (PVM), can infect human and mouse eosinophils, respectively, and that virus infection of eosinophils elicits the release of disease-related proinflammatory mediators from eosinophils. RSV replication in human eosinophils results in the release of infectious virions and in the release of the proinflammatory mediator, interleukin-6 (IL-6). PVM replication in cultured bone marrow eosinophils (bmEos) likewise results in release of infectious virions and the proinflammatory mediators IL-6, IP-10, CCL2, and CCL3. In contrast to the findings reported in lung tissue of RSV-challenged mice, PVM replication is accelerated in MyD88 gene-deleted bmEos, whereas release of cytokines is diminished. Interestingly, exogenous IL-6 suppresses virus replication in MyD88 gene-deleted bmEos, suggesting a role for a MyD88-dependent cytokine-mediated feedback circuit in modulating this response. Taken together, our findings suggest that eosinophils are targets of virus infection and may have varied and complex contributions to the pathogenesis and resolution of pneumovirus disease.


Subject(s)
Chemokines/metabolism , Eosinophils/metabolism , Interleukin-6/metabolism , Myeloid Differentiation Factor 88/physiology , Pneumovirus Infections/immunology , Pneumovirus/physiology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/virology , Chemotactic Factors/metabolism , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/virology , Interleukin-6/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Pneumovirus Infections/genetics , Pneumovirus Infections/metabolism , Virus Replication/drug effects , Virus Replication/genetics , Virus Replication/physiology , Virus Shedding/drug effects , Virus Shedding/genetics , Virus Shedding/physiology
9.
BMC Immunol ; 10: 14, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-19298652

ABSTRACT

BACKGROUND: We have shown previously that acute infection with the respiratory pathogen, pneumonia virus of mice (PVM), results in local production of the proinflammatory chemokine, CCL3, and that neutrophil recruitment in response to PVM infection is reduced dramatically in CCL3 -/- mice. RESULTS: In this work, we demonstrate that CCL3-mediated neutrophil recruitment is coordinated by interferon-gamma (IFNgamma). Neutrophil recruitment in response to PVM infection was diminished five-fold in IFNgamma receptor gene-deleted mice, although neutrophils from IFNgammaR -/- mice expressed transcripts for the CCL3 receptor, CCR1 and responded functionally to CCL3 ex vivo. Similarly, in the absence of PVM infection, CCL3 overexpression alone could not elicit neutrophil recruitment in the absence of IFNgamma. Interestingly, although supplemental IFNgamma restored neutrophil recruitment and resulted in a sustained weight loss among CCL3-overexpressing IFNgamma -/- mice, CCL3-mediated neutrophil recruitment alone did not result in the pulmonary edema or respiratory failure characteristic of severe viral infection, suggesting that CCL3 and IFN-gamma together are sufficient to promote neutrophil recruitment but not pathologic activation. CONCLUSION: Our findings reveal a heretofore unrecognized hierarchical interaction between the IFNgamma and CCL3, which demonstrate that IFNgamma is crucial for CCL3-mediated neutrophil recruitment in vivo.


Subject(s)
Chemokine CCL3/metabolism , Interferon-gamma/metabolism , Lung/metabolism , Murine pneumonia virus/immunology , Neutrophils/metabolism , Pneumovirus Infections/immunology , Animals , Cell Movement/immunology , Chemokine CCL3/genetics , Chemokine CCL3/immunology , Gene Expression Profiling , Interferon-gamma/genetics , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lung/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Murine pneumonia virus/pathogenicity , Neutrophils/immunology , Neutrophils/pathology , Pneumovirus Infections/genetics , Pneumovirus Infections/physiopathology , Pulmonary Edema , Receptors, CCR1/genetics , Receptors, CCR1/immunology , Receptors, CCR1/metabolism , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Receptors, Interferon/metabolism , Respiratory Insufficiency , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Interferon gamma Receptor
10.
Am J Physiol Lung Cell Mol Physiol ; 291(3): L426-35, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16556725

ABSTRACT

Respiratory syncytial virus (RSV) is a prominent cause of airway morbidity in children under 1 yr of age. It is assumed that host factors influence the severity of the disease presentation and thus the need for hospitalization. As a first step toward the identification of the underlying genes involved, this study was undertaken to establish whether inbred mouse strains differ in susceptibility to pneumonia virus of mice (PVM), the murine counterpart of RSV, which has been shown to accurately mimic the RSV disease of children. With this purpose in mind, double-chamber plethysmography and carbon monoxide uptake data were collected daily for 7 days after inoculation of PVM in six inbred strains of mice. In parallel, histological examinations and lung viral titration were carried out from day 5 to day 7 after inoculation. Pulmonary structure/function values reflected the success of viral replication in the lungs and revealed a pattern of continuous variation, with resistant, intermediate, and susceptible strains. The results suggest that SJL (resistant) and 129/Sv (susceptible) strains should be used in crossing experiments aimed at identifying genes controlling pneumovirus replication by the positional cloning approach. Similarly, crossing experiments using BALB/c or C57BL/6 (resistant) and DBA/2 or 129/Sv (susceptible) will allow the identification of the genes involved in the control of pulmonary inflammation during pneumovirus infection.


Subject(s)
Genetic Predisposition to Disease , Immunity, Innate/genetics , Mice, Inbred Strains/genetics , Murine pneumonia virus , Pneumovirus Infections/genetics , Animals , Female , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred Strains/virology , Pneumovirus Infections/immunology , Pneumovirus Infections/pathology , Species Specificity , Time Factors , Viral Load , Virus Replication
11.
Poult Sci ; 82(6): 885-92, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12817442

ABSTRACT

Respiratory tract diseases are the single most important cause of economic loss due to infections among poultry populations worldwide. However, the molecular mechanisms of the host response to infections remain unknown. Here, we review the literature and describe the adoption of a conceptually simple approach to understand the genetic and biochemical responses of host cells during infection with respiratory pathogens, such as avian pneumovirus (APV). The strategy that we have adopted integrates the powerful techniques of cDNA subtraction hybridization and microarray analysis for global transcriptional profiling. The results of our investigations identify the specific transcriptional alterations in host-cell gene expression that result from an attempt by the host to combat and limit the spread of the pathogen or by the pathogen to enhance its own survival and ability to reproduce. Our studies suggest that a molecular description of host-pathogen interactions in terms of differential gene expression will provide key insights on the molecular basis of disease pathogenesis, pathogen virulence, and host immunity. In addition, the results suggest that the identification of genes and pathways with a role in host response to infection has considerable practical implications for the future design and development of effective immunomodulators and vaccines.


Subject(s)
Gene Expression Regulation , Oligonucleotide Array Sequence Analysis , Poultry Diseases/genetics , Respiratory Tract Diseases/veterinary , Transcription, Genetic , Animals , DNA, Complementary , In Situ Hybridization , Pneumovirus/pathogenicity , Pneumovirus Infections/genetics , Pneumovirus Infections/veterinary , Poultry , Respiratory Tract Diseases/genetics , Respiratory Tract Diseases/virology , Virulence
12.
Respir Res ; 2(4): 225-33, 2001.
Article in English | MEDLINE | ID: mdl-11686888

ABSTRACT

Respiratory syncytial virus (RSV) and pneumonia virus of mice (PVM) are viruses of the family Paramyxoviridae, subfamily pneumovirus, which cause clinically important respiratory infections in humans and rodents, respectively. The respiratory epithelial target cells respond to viral infection with specific alterations in gene expression, including production of chemoattractant cytokines, adhesion molecules, elements that are related to the apoptosis response, and others that remain incompletely understood. Here we review our current understanding of these mucosal responses and discuss several genomic approaches, including differential display reverse transcription-polymerase chain reaction (PCR) and gene array strategies, that will permit us to unravel the nature of these responses in a more complete and systematic manner.


Subject(s)
Gene Expression , Murine pneumonia virus , Pneumovirus Infections/genetics , Respiratory Syncytial Virus Infections/genetics , Animals , Apoptosis , Chemokines/biosynthesis , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Oligonucleotide Array Sequence Analysis
13.
J Vet Diagn Invest ; 13(3): 201-5, 2001 May.
Article in English | MEDLINE | ID: mdl-11482596

ABSTRACT

Avian pneumovirus (APV) or turkey rhinotracheitis virus (TRTV) is an important respiratory pathogen of domesticated poultry in many countries in Europe, Africa, and Asia. Until recently, the United States was considered free of APV. In late 1996, an atypical upper respiratory tract infection appeared in turkey flocks in Colorado and shortly thereafter in turkey flocks in Minnesota. An avian pneumovirus (APV-US) that was serologically distinct from the previously described TRTV was isolated as the primary cause of the new syndrome. The nucleotide sequence of a fragment of the APV-US fusion gene was determined and used to develop a polymerase chain reaction-based assay that specifically detects APV-US viral nucleic acid sequences in RNA extracts of tracheal swabs and turbinate homogenates. The assay is highly sensitive in that it can detect <0.01 TCID50 of APV. The availability of this assay enables the rapid and accurate determination of APV-US in infected poultry flocks.


Subject(s)
Disease Outbreaks/veterinary , Pneumovirus Infections/veterinary , Pneumovirus/pathogenicity , Poultry Diseases/virology , Animals , Base Sequence , DNA, Viral/analysis , DNA, Viral/genetics , Diagnosis, Differential , Molecular Sequence Data , Pneumovirus/genetics , Pneumovirus/isolation & purification , Pneumovirus Infections/genetics , Pneumovirus Infections/transmission , Polymerase Chain Reaction/veterinary , Poultry Diseases/genetics , Sensitivity and Specificity , Serologic Tests , Turkeys
SELECTION OF CITATIONS
SEARCH DETAIL
...