Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
N Z Vet J ; 72(4): 191-200, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38650102

ABSTRACT

AIMS: To isolate canine respiratory coronavirus (CRCoV) and canine pneumovirus (CnPnV) in cell culture and to compare partial genomic sequences of CRCoV and CnPnV from New Zealand with those from other countries. METHODS: Oropharyngeal swab samples from dogs affected by canine infectious respiratory disease syndrome that were positive for CnPnV (n = 15) or CRCoV (n = 1) by virus-specific reverse transcriptase quantitative PCR (RT-qPCR) in a previous study comprised the starting material. Virus isolation was performed in HRT-18 cells for CRCoV and RAW 264.7 and Vero cells for CnPnV. The entire sequence of CnPnV G protein (1,266 nucleotides) and most (8,063/9,707 nucleotides) of the 3' region of CRCoV that codes for 10 structural and accessory proteins were amplified and sequenced. The sequences were analysed and compared with other sequences available in GenBank using standard molecular tools including phylogenetic analysis. RESULTS: Virus isolation was unsuccessful for both CRCoV and CnPnV. Pneumovirus G protein was amplified from 3/15 (20%) samples that were positive for CnPnV RNA by RT-qPCR. Two of these (NZ-048 and NZ-049) were 100% identical to each other, and 90.9% identical to the third one (NZ-007). Based on phylogenetic analysis of the G protein gene, CnPnV NZ-048 and NZ-049 clustered with sequences from the USA, Thailand and Italy in group A, and CnPnV NZ-007 clustered with sequences from the USA in group B. The characteristics of the predicted genes (length, position) and their putative protein products (size, predicted structure, presence of N- and O-glycosylation sites) of the New Zealand CRCoV sequence were consistent with those reported previously, except for the region located between open reading frame (ORF)3 (coding for S protein) and ORF6 (coding for E protein). The New Zealand virus was predicted to encode 5.9 kDa, 27 kDa and 12.7 kDa proteins, which differed from the putative coding capacity of this region reported for CRCoV from other countries. CONCLUSIONS: This report represents the first characterisation of partial genomic sequences of CRCoV and CnPnV from New Zealand. Our results suggest that the population of CnPnV circulating in New Zealand is not homogeneous, and that the viruses from two clades described overseas are also present here. Limited conclusions can be made based on only one CRCoV sequence, but the putative differences in the coding capacity of New Zealand CRCoV support the previously reported variability of this region. The reasons for such variability and its biological implications need to be further elucidated.


Subject(s)
Coronavirus, Canine , Dog Diseases , Genome, Viral , Phylogeny , Pneumovirus , Animals , Dogs , New Zealand/epidemiology , Coronavirus, Canine/genetics , Coronavirus, Canine/classification , Coronavirus, Canine/isolation & purification , Dog Diseases/virology , Dog Diseases/epidemiology , Pneumovirus/genetics , Pneumovirus/classification , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/epidemiology , Vero Cells , Chlorocebus aethiops
2.
Emerg Microbes Infect ; 12(2): 2239938, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37470510

ABSTRACT

Respiratory disease is a significant economic issue in pig farming, with a complex aetiology that includes swine influenza A viruses (swIAV), which are common in European domestic pig populations. The most recent human influenza pandemic in 2009 showed swIAV's zoonotic potential. Monitoring pathogens and disease control are critical from a preventive standpoint, and are based on quick, sensitive, and specific diagnostic assays capable of detecting and distinguishing currently circulating swIAV in clinical samples. For passive surveillance, a set of multiplex quantitative reverse transcription real-time PCRs (mRT-qPCR) and MinION-directed sequencing was updated and deployed. Several lineages and genotypes of swIAV were shown to be dynamically developing, including novel reassortants between human pandemic H1N1 and the avian-derived H1 lineage of swIAV. Despite this, nearly 70% (842/1216) of individual samples from pigs with respiratory symptoms were swIAV-negative, hinting to different aetiologies. The complex and synergistic interactions of swIAV infections with other viral and bacterial infectious agents contribute to the aggravation of pig respiratory diseases. Using a newly developed mRT-qPCR for the combined detection of swIAV and the recently described porcine respirovirus 1 (PRV1) and swine orthopneumovirus (SOV) widespread co-circulation of PRV1 (19.6%, 238/1216 samples) and SOV (14.2%, 173/1216 samples) was evident. Because of the high incidence of PRV1 and SOV infections in pigs with respiratory disease, these viruses may emerge as new allies in the porcine respiratory disease syndrome.


Subject(s)
Orthomyxoviridae Infections , Pneumovirus Infections , Respiratory Tract Diseases , Respirovirus Infections , Swine Diseases , Germany/epidemiology , Swine Diseases/epidemiology , Swine Diseases/virology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/veterinary , Influenza A virus/genetics , Respirovirus/genetics , Respirovirus Infections/epidemiology , Respirovirus Infections/veterinary , Respiratory Tract Diseases/veterinary , Respiratory Tract Diseases/virology , Pneumovirus Infections/epidemiology , Pneumovirus Infections/veterinary , Pneumovirus/genetics , Reverse Transcriptase Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction , Phylogeny
3.
mBio ; 12(6): e0262121, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34724816

ABSTRACT

Multiple enveloped RNA viruses of the family Paramyxoviridae and Pneumoviridae, like measles virus (MeV), Nipah virus (NiV), canine distemper virus (CDV), or respiratory syncytial virus (RSV), are of high clinical relevance. Each year a huge number of lives are lost as a result of these viral infections. Worldwide, MeV infection alone is responsible for over a hundred thousand deaths each year despite available vaccine. Therefore, there is an urgent need for treatment options to counteract these viral infections. The development of antiviral drugs in general stands as a huge challenge due to the rapid emergence of viral escape mutants. Here, we disclose the discovery of a small-molecule antiviral, compound 1 (ZHAWOC9045), active against several pneumo-/paramyxoviruses, including MeV, NiV, CDV, RSV, and parainfluenza virus type 5 (PIV-5). A series of mechanistic characterizations revealed that compound 1 targets a host factor which is indispensable for viral genome replication. Drug resistance profiling against a paramyxovirus model (CDV) demonstrated no detectable adaptation despite prolonged time of investigation, thereby mitigating the rapid emergence of escape variants. Furthermore, a thorough structure-activity relationship analysis of compound 1 led to the invention of 100-times-more potent-derivatives, e.g., compound 2 (ZHAWOC21026). Collectively, we present in this study an attractive host-directed pneumoviral/paramyxoviral replication inhibitor with potential therapeutic application. IMPORTANCE Measles virus, respiratory syncytial virus, canine distemper virus, and Nipah virus are some of the clinically significant RNA viruses that threaten substantial number of lives each year. Limited to no availability of treatment options for these viral infections makes it arduous to handle the outbreaks. This highlights the major importance of developing antivirals to fight not only ongoing infections but also potential future epidemics. Most of the discovered antivirals, in clinical trials currently, are virus targeted, which consequently poses the challenge of rapid emergence of escape variants. Here, we present compound 1 (ZHAWOC9045), discovered to target viral replication in a host-dependent manner, thereby exhibiting broad-spectrum activity against several members of the family Pneumo-/Paramyxoviridae. The inability of viruses to mutate against the inhibitor mitigated the critical issue of generation of escape variants. Importantly, compound 1 was successfully optimized to a highly potent variant, compound 2 (ZHAWOC21026), with a promising profile for pharmacological intervention.


Subject(s)
Antiviral Agents/pharmacology , Paramyxoviridae/physiology , Pneumovirus/physiology , Virus Replication/drug effects , Antiviral Agents/chemistry , Drug Discovery , Humans , Paramyxoviridae/genetics , Paramyxoviridae Infections/drug therapy , Paramyxoviridae Infections/virology , Pneumovirus/genetics , Pneumovirus Infections/drug therapy , Pneumovirus Infections/virology
4.
Int J Mol Sci ; 22(4)2021 Feb 03.
Article in English | MEDLINE | ID: mdl-33546457

ABSTRACT

Mononegavirales phosphoproteins (P) are essential co-factors of the viral polymerase by serving as a linchpin between the catalytic subunit and the ribonucleoprotein template. They have highly diverged, but their overall architecture is conserved. They are multidomain proteins, which all possess an oligomerization domain that separates N- and C-terminal domains. Large intrinsically disordered regions constitute their hallmark. Here, we exemplify their structural features and interaction potential, based on the Pneumoviridae P proteins. These P proteins are rather small, and their oligomerization domain is the only part with a defined 3D structure, owing to a quaternary arrangement. All other parts are either flexible or form short-lived secondary structure elements that transiently associate with the rest of the protein. Pneumoviridae P proteins interact with several viral and cellular proteins that are essential for viral transcription and replication. The combination of intrinsic disorder and tetrameric organization enables them to structurally adapt to different partners and to act as adaptor-like platforms to bring the latter close in space. Transient structures are stabilized in complex with protein partners. This class of proteins gives an insight into the structural versatility of non-globular intrinsically disordered protein domains.


Subject(s)
Models, Molecular , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Pneumovirus/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Gene Expression Regulation, Viral , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Mononegavirales , Phosphoproteins/genetics , Pneumovirus/genetics , Protein Binding , Protein Folding , Respiratory Syncytial Virus, Human , Structure-Activity Relationship , Viral Proteins/genetics
5.
BMC Vet Res ; 15(1): 300, 2019 Aug 19.
Article in English | MEDLINE | ID: mdl-31426794

ABSTRACT

BACKGROUND: Canine pneumovirus (CPV) is a pathogen that causes respiratory disease in dogs, and recent outbreaks in shelters in America and Europe have been reported. However, based on published data and documents, the identification of CPV and its variant in clinically symptomatic individual dogs in Thailand through Asia is limited. Therefore, the aims of this study were to determine the emergence of CPV and to consequently establish the genetic characterization and phylogenetic analysis of the CPV strains from 209 dogs showing respiratory distress in Thailand. RESULTS: This study identified and described the full-length CPV genome from three strains, designated herein as CPV_CP13 TH/2015, CPV_CP82 TH/2016 and CPV_SR1 TH/2016, that were isolated from six dogs out of 209 dogs (2.9%) with respiratory illness in Thailand. Phylogenetic analysis suggested that these three Thai CPV strains (CPV TH strains) belong to the CPV subgroup A and form a novel lineage; proposed as the Asian prototype. Specific mutations in the deduced amino acids of these CPV TH strains were found in the G/glycoprotein sequence, suggesting potential substitution sites for subtype classification. Results of intragenic recombination analysis revealed that CPV_CP82 TH/2016 is a recombinant strain, where the recombination event occurred in the L gene with the Italian prototype CPV Bari/100-12 as the putative major parent. Selective pressure analysis demonstrated that the majority of the nucleotides in the G/glycoprotein were under purifying selection with evidence of positive selection sites. CONCLUSIONS: This collective information on the CPV TH strains is the first evidence of CPV emergence with genetic characterization in Thailand and as first report in Asia, where homologous recombination acts as a potential force driving the genetic diversity and shaping the evolution of canine pneumovirus.


Subject(s)
Dog Diseases/virology , Phylogeny , Pneumovirus Infections/veterinary , Pneumovirus/classification , Reassortant Viruses/genetics , Respiratory Tract Infections/veterinary , Amino Acid Sequence , Animals , Dog Diseases/epidemiology , Dogs , Genome, Viral , Mutation , Pneumovirus/genetics , Pneumovirus Infections/epidemiology , Pneumovirus Infections/virology , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/virology , Thailand/epidemiology , Viral Proteins/genetics , Viral Proteins/metabolism
6.
Virus Res ; 265: 68-73, 2019 05.
Article in English | MEDLINE | ID: mdl-30844414

ABSTRACT

Pneumoviruses represent a major public health burden across the world. Respiratory syncytial virus (RSV) and human metapneumovirus (HMPV), two of the most recognizable pediatric infectious agents, belong to this family. These viruses are enveloped with a non-segmented negative-sense RNA genome, and their replication occurs in specialized cytosolic organelles named inclusion bodies (IB). The critical role of IBs in replication of pneumoviruses has begun to be elucidated, and our current understanding suggests they are highly dynamic structures. From IBs, newly synthesized nucleocapsids are transported to assembly sites, potentially via the actin cytoskeleton, to be incorporated into nascent virions. Released virions, which generally contain one genome, can then diffuse in the extracellular environment to target new cells and reinitiate the process of infection. This is a challenging business for virions, which must face several risks including the extracellular immune responses. In addition, several recent studies suggest that successful infection may be achieved more rapidly by multiple, rather than single, genomic copies being deposited into a target cell. Interestingly, recent data indicate that pneumoviruses have several mechanisms that permit their transmission en bloc, i.e. transmission of multiple genomes at the same time. These mechanisms include the well-studied syncytia formation as well as the newly described formation of long actin-based intercellular extensions. These not only permit en bloc viral transmission, but also bypass assembly of complete virions. In this review we describe several aspects of en bloc viral transmission and how these mechanisms are reshaping our understanding of pneumovirus replication, assembly and spread.


Subject(s)
Paramyxoviridae Infections/transmission , Pneumovirus/physiology , Virus Assembly , Animals , Cell Line , Humans , Metapneumovirus/genetics , Metapneumovirus/physiology , Mice , Pneumovirus/genetics , RNA, Viral , Virion/genetics , Virion/physiology , Virus Replication
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.
PLoS One ; 9(1): e85220, 2014.
Article in English | MEDLINE | ID: mdl-24400129

ABSTRACT

An outbreak of canine infectious respiratory disease (CIRD) associated to canine pneumovirus (CnPnV) infection is reported. The outbreak occurred in a shelter of the Apulia region and involved 37 out of 350 dogs that displayed cough and/or nasal discharge with no evidence of fever. The full-genomic characterisation showed that the causative agent (strain Bari/100-12) was closely related to CnPnVs that have been recently isolated in the USA, as well as to murine pneumovirus, which is responsible for respiratory disease in mice. The present study represents a useful contribution to the knowledge of the pathogenic potential of CnPnV and its association with CIRD in dogs. Further studies will elucidate the pathogenicity and epidemiology of this novel pneumovirus, thus addressing the eventual need for specific vaccines.


Subject(s)
Dog Diseases/epidemiology , Dog Diseases/virology , Genome, Viral , Pneumovirus Infections/veterinary , Pneumovirus/genetics , Animals , Base Sequence , Disease Outbreaks , Dogs , Female , Gene Order , Italy/epidemiology , Male , Molecular Sequence Data , Phylogeny , Pneumovirus/classification , Pneumovirus/isolation & purification , Sequence Alignment
9.
Virology ; 443(2): 257-64, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23763766

ABSTRACT

A previous report of a novel pneumovirus (PnV) isolated from the respiratory tract of a dog described its significant homology to the rodent pathogen, pneumonia virus of mice (PVM). The original PnV-Ane4 pathogen replicated in and could be re-isolated in infectious state from mouse lung but elicited minimal mortality compared to PVM strain J3666. Here we assess phylogeny and physiologic responses to 10 new PnV isolates. The G/glycoprotein sequences of all PnVs include elongated amino-termini when compared to the characterized PVMs, and suggest division into groups A and B. While we observed significant differences in cytokine production and neutrophil recruitment to the lungs of BALB/c mice in response to survival doses (50 TCID50 units) of representative group A (114378-10-29-KY-F) and group B (7968-11-OK) PnVs, we observed no evidence for positive selection (dN > dS) among the PnV/PnV, PVM/PnV or PVM/PVM G/glycoprotein or F/fusion protein sequence pairs.


Subject(s)
Evolution, Molecular , Inflammation/pathology , Pneumovirus Infections/pathology , Pneumovirus/classification , Pneumovirus/pathogenicity , Amino Acid Sequence , Animals , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/metabolism , Inflammation/immunology , Inflammation/virology , Lung/immunology , Lung/pathology , Membrane Glycoproteins , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Murine pneumonia virus/genetics , Murine pneumonia virus/pathogenicity , Phylogeny , Pneumovirus/genetics , Pneumovirus Infections/virology , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
10.
Viruses ; 5(1): 406-22, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23344499

ABSTRACT

Pneumovirus infections cause a wide spectrum of respiratory disease in humans and animals. The airway epithelium is the major site of pneumovirus replication. Apoptosis or regulated cell death, may contribute to the host anti-viral response by limiting viral replication. However, apoptosis of lung epithelial cells may also exacerbate lung injury, depending on the extent, the timing and specific location in the lungs. Differential apoptotic responses of epithelial cells versus innate immune cells (e.g., neutrophils, macrophages) during pneumovirus infection can further contribute to the complex and delicate balance between host defense and disease pathogenesis. The purpose of this manuscript is to give an overview of the role of apoptosis in pneumovirus infection. We will examine clinical and experimental data concerning the various pro-apoptotic stimuli and the roles of apoptotic epithelial and innate immune cells during pneumovirus disease. Finally, we will discuss potential therapeutic interventions targeting apoptosis in the lungs.


Subject(s)
Apoptosis , Pneumovirus Infections/physiopathology , Pneumovirus/physiology , Animals , Antiviral Agents/pharmacology , Humans , Pneumovirus/drug effects , Pneumovirus/genetics , Pneumovirus Infections/drug therapy , Pneumovirus Infections/immunology , Pneumovirus Infections/virology
11.
J Interferon Cytokine Res ; 32(7): 332-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22385204

ABSTRACT

The respiratory syncytial virus (RSV) is a major pathogen of the human species. This pneumovirus is a prominent cause of airway morbidity in children and maintains an excessive hospitalization rate despite decades of research. As involvement of a genetic vulnerability is a possibility supported by recent data, we addressed the question of whether the Mx gene products, the typical target of which consists in single-stranded negative-polarity RNA viruses, could alter the course of pneumovirus-associated disease in vivo. Wild-type and Bos taurus Mx1-expressing transgenic FVB/J mice were inoculated with the mouse counterpart and closest phylogenetic relative of RSV, pneumonia virus of mice. Survival data and follow-up of body weight, histological scores, lung virus spread, and lung viral load unequivocally showed that the viral infection was severely repressed in Mx-transgenic mice, thus suggesting that pneumoviruses belong to the antiviral spectrum of mammalian Mx GTPases. Elucidating the underlying mechanisms at the molecular level could reveal critical information for the development of new anti-RSV molecules.


Subject(s)
GTP-Binding Proteins/immunology , Immunity, Innate , Pneumovirus Infections/immunology , Pneumovirus/immunology , Administration, Intranasal , Animals , Animals, Genetically Modified , Cattle , Female , GTP-Binding Proteins/genetics , Gene Expression Regulation , Humans , Mice , Myxovirus Resistance Proteins , Pneumovirus/genetics , Pneumovirus Infections/mortality , Pneumovirus Infections/pathology , Survival Analysis
12.
Curr Med Chem ; 19(10): 1424-31, 2012.
Article in English | MEDLINE | ID: mdl-22360479

ABSTRACT

Respiratory syncytial virus (RSV; Family Paramyxoviridae, Genus Pneumovirus) is a major respiratory pathogen of infants and children and an emerging pathogen of the elderly. Current management of RSV disease includes monoclonal antibody prophylaxis for infants identified as high risk and supportive care for those with active infection; there is no vaccine, although several are under study. In this manuscript, we review published findings from human autopsy studies, as well as experiments that focus on human clinical samples and mouse models of acute pneumovirus infection that elucidate basic principles of disease pathogenesis. Consideration of these data suggests that the inflammatory responses to RSV and related pneumoviral pathogens can be strong, persistent, and beyond the control of conventional antiviral and anti-inflammatory therapies, and can have profound negative consequences to the host. From this perspective, we consider the case for specific immunomodulatory strategies that may have the potential to alleviate some of the more serious sequelae of this disease.


Subject(s)
Immunologic Factors/therapeutic use , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus, Human/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antiviral Agents/therapeutic use , Cytokines/antagonists & inhibitors , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Oligodeoxyribonucleotides, Antisense/therapeutic use , Pneumovirus/genetics , Pneumovirus/immunology , Pneumovirus Infections/drug therapy , Pneumovirus Infections/immunology , Pneumovirus Infections/pathology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus, Human/genetics
13.
Virology ; 416(1-2): 26-31, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21600624

ABSTRACT

Canine pneumovirus (CnPnV) was recently isolated from the respiratory tracts of shelter dogs and shares sequence similarity with the rodent pathogen, pneumonia virus of mice (PVM). We show here that CnPnV replicates in and can elicit local proinflammatory cytokine production and neutrophil recruitment to lung tissue and the airways. In contrast to PVM J3666 infection, fatal CnPnV infections are observed only in response to high titer intranasal inocula (>67 TCID(50) units). Sera from mice that recover from CnPnV infection contain antibodies that cross-react with PVM antigens; these mice are protected against lethal PVM infection. Given these findings, it will be intriguing to determine the relative role(s) of CnPnV and PVM in eliciting respiratory symptoms in susceptible canine species.


Subject(s)
Inflammation/virology , Lung/virology , Pneumovirus Infections/virology , Pneumovirus/classification , Pneumovirus/physiology , Virus Replication/physiology , Animals , Dogs , Inflammation/pathology , Mice , Mice, Inbred BALB C , Phylogeny , Pneumovirus/genetics , Pneumovirus Infections/pathology
14.
Vet Microbiol ; 150(1-2): 88-95, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21324612

ABSTRACT

A previously unrecognized virus belonging to the subfamily Pneumovirinae and most closely related to murine pneumovirus (MPV) was identified in domestic dogs in 2 related animal shelters. Additional diagnostic testing yielded 3 new viral isolates and identified 6 additional PCR positive dogs from other USA locations indicating that its distribution is not geographically limited. Nucleotide sequences encompassing 9 of the 10 genes were compared to the only 2 available MPV strains, 15 and J3666. Several features distinguished the canine pneumovirus (CnPnV) from the murine strains. Two regions of diversity were identified in the amino-proximal region of P and the overlapping P2 ORF was only 54 amino acids (aa) compared to 137aa in MPV. The G protein had an amino-terminal cytoplasmic tail 18aa longer than in the MPV strains. The CnPnV SH protein showed the highest divergence with only 90.2% aa identity when compared to MPV strain 15. Like strain 15, the CnPnV SH ORF coded for a protein of 92aa while J3666 has a 114aa variant. Comparison of CnPnV isolates at culture passages 4 and 17 revealed 7nt differences within the 8598nt sequenced. Of note was a substitution at nt 364 in G resulting in a termination codon that would produce a truncated G protein of 122aa. Analysis of early passage and ex vivo samples showed the termination codon in G to be predominant after 6 days in culture indicating rapid selection of the mutation in A72 cells.


Subject(s)
Dog Diseases/virology , Dogs/virology , Genome, Viral , Pneumovirus Infections/veterinary , Pneumovirus/genetics , Animals , Cell Line , Genomics , Open Reading Frames , Phylogeny , Pneumovirus/classification , Pneumovirus/isolation & purification , Pneumovirus Infections/virology , RNA, Viral/genetics , Sequence Analysis, RNA , Sequence Homology, Amino Acid , United States , Viral Proteins/genetics
15.
Emerg Infect Dis ; 16(6): 993-5, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20507755

ABSTRACT

To determine which respiratory viruses circulate among confined dogs, we analyzed nasal and pharyngeal swab specimens from shelter dogs with acute respiratory disease. An unknown virus was isolated. Monoclonal antibody testing indicated that it was probably a pneumovirus. PCR and sequence analysis indicated that it was closely related to murine pneumovirus.


Subject(s)
Dog Diseases , Pneumovirus Infections/veterinary , Pneumovirus/isolation & purification , Respiratory Tract Infections/veterinary , Acute Disease , Animals , Cell Line , DNA, Viral/analysis , DNA, Viral/genetics , Disease Outbreaks , Dog Diseases/epidemiology , Dog Diseases/virology , Dogs , Fluorescent Antibody Technique , Genes, Viral , Nose/virology , Pharynx/virology , Pneumovirus/genetics , Pneumovirus Infections/complications , Pneumovirus Infections/epidemiology , Pneumovirus Infections/virology , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/etiology , Respiratory Tract Infections/pathology , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid
16.
Ann Clin Lab Sci ; 38(1): 41-6, 2008.
Article in English | MEDLINE | ID: mdl-18316781

ABSTRACT

This study evaluated the clinical usefulness of a newly introduced multiplex reverse transcription PCR assay (Seeplex RV; Seegene, Inc., Seoul, Korea) in patients with respiratory symptoms. Fifty clinical respiratory specimens (45 from children, 5 from adults) were tested for 8 viruses (influenza virus type A and B, parainfluenza virus type 1, 2, 3, respiratory syncytial virus type A and B, and adenovirus) by Seeplex RV (S-RV) and R-mix viral culture with immunofluorescence (VC-IF). Forty (80%) of the 50 samples showed concordant results between S-RV and VC-IF; 24 of these showed the same positive and 16 showed the same negative results. Among the 10 discrepant samples, 9 were S-RV-positive and VC-IF-negative. Six were obtained in patients with lower respiratory tract infection. Only 1 sample was VC-IF-positive and S-RV-negative. This patient had pneumonia. In 3 cases, more than 1 virus was identified by S-RV. The total running time of S-RV was 6 hr, which shortens the detection time for the viral presence by 2 workdays compared to VC-IF. In conclusion, S-RV is reliable, rapid, relatively easy to perform, and able to detect more than 1 virus simultaneously. Therefore, implementation of the S-RV assay in clinical laboratories will aid rapid diagnosis and treatment of major viral infections of the respiratory tract.


Subject(s)
Fluorescent Antibody Technique/methods , Pneumovirus/genetics , Pneumovirus/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction/methods , Adolescent , Adult , Aged , Child , Child, Preschool , DNA, Viral/analysis , DNA, Viral/genetics , Female , Humans , Infant , Male , Middle Aged , Pneumovirus/growth & development , Pneumovirus/immunology
17.
Mol Immunol ; 44(5): 993-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16650896

ABSTRACT

Internalisation of the plant toxin ricin occurs by retrograde transport which delivers the toxin to the ER where it intersects with the MHC class I system for peptide antigen display. Here, we describe the generation of an inactivated, non-toxic, ricin molecule fused to a peptide which elicits a CD8+ T-cell response in mice directed against pneumonia virus of mice, a pneumovirus related to human respiratory syncytial virus. The ricin fusion elicited a significant T-cell response when delivered by intraperitoneal inoculation in the absence of adjuvent. Challenge experiments showed that the T-cell response resulting from inoculation with the ricin-peptide fusion molecule delayed the onset of virus-induced disease.


Subject(s)
Pneumovirus Infections/immunology , Recombinant Fusion Proteins/administration & dosage , Ricin/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , Viral Proteins/administration & dosage , Animals , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Glycosides/pharmacology , Mice , Mice, Inbred BALB C , Peptides/administration & dosage , Peptides/genetics , Peptides/immunology , Pneumovirus/genetics , Pneumovirus/immunology , Triterpenes/pharmacology , Viral Proteins/genetics , Viral Proteins/immunology
18.
J Immunol ; 175(10): 6597-604, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16272314

ABSTRACT

Pneumonia virus of mice (PVM) is a natural pathogen of mice and has been proposed as a tractable model for the replication of a pneumovirus in its natural host, which mimics human infection with human respiratory syncytial virus (RSV). PVM infection in mice is highly productive in terms of virus production compared with the situation seen with RSV in mice. Because RSV suppresses CD8 T cell effector function in the lungs of infected mice, we have investigated the nature of PVM-induced CD8 T cell responses to study pneumovirus-induced T cell responses in a natural virus-host setting. PVM infection was associated with a massive influx of activated CD8 T cells into the lungs. After identification of three PVM-specific CD8 T cell epitopes, pulmonary CD8 T cell responses were enumerated. The combined frequency of cytokine-secreting CD8 T cells specific for the three epitopes was much smaller than the total number of activated CD8 T cells. Furthermore, quantitation of the CD8 T cell response against one of these epitopes (residues 261-270 from the phosphoprotein) by MHC class I pentamer staining and by in vitro stimulation followed by intracellular IFN-gamma and TNF-alpha staining indicated that the majority of pulmonary CD8 specific for the P261 epitope were deficient in cytokine production. This deficient phenotype was retained up to 96 days postinfection, similar to the situation in the lungs of human RSV-infected mice. The data suggest that PVM suppresses T cell effector functions in the lungs.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Pneumovirus Infections/immunology , Pneumovirus/immunology , Amino Acid Sequence , Animals , Antigens, Viral/genetics , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Epitopes/genetics , Female , Humans , Immunologic Memory , In Vitro Techniques , Interferon-gamma/biosynthesis , Lung/immunology , Lung/pathology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Pneumovirus/genetics , Pneumovirus Infections/pathology , Pneumovirus Infections/virology , Spleen/immunology , Spleen/pathology , Tumor Necrosis Factor-alpha/biosynthesis
19.
Avian Dis ; 47(2): 481-4, 2003.
Article in English | MEDLINE | ID: mdl-12887210

ABSTRACT

Four avian pneumovirus (APV) isolates from chickens clinically diagnosed with swollen head syndrome were genetically characterized as to the subtypes of the virus in Japan. The results of reverse transcriptase-polymerase chain reactions based on subtype-specific primers and direct sequence analysis of G genes indicated subtypes A and B but not C or D of APV were present in Japan. Several routes or sources are conceivable for APV to invade into Japan.


Subject(s)
Chickens/virology , Pneumovirus Infections/epidemiology , Pneumovirus Infections/veterinary , Pneumovirus/classification , Pneumovirus/isolation & purification , Poultry Diseases/epidemiology , Poultry Diseases/virology , Animals , Genes, Viral/genetics , Japan/epidemiology , Phylogeny , Pneumovirus/genetics , Pneumovirus Infections/virology
20.
Virus Res ; 93(1): 91-7, 2003 May.
Article in English | MEDLINE | ID: mdl-12727346

ABSTRACT

We report here the nucleotide and deduced amino acid (aa) sequences of the small hydrophobic (SH) gene of the avian pneumovirus strain Colorado (APV/CO). The SH gene of APV/CO is 628 nucleotides in length from gene-start to gene-end. The longest ORF of the SH gene encoded a protein of 177 aas in length. Comparison of the deduced aa sequence of the SH protein of APV/CO with the corresponding published sequences of other members of genera metapneumovirus showed 28% identity with the newly discovered human metapneumovirus (hMPV), but no discernable identity with the APV subgroup A or B. Collectively, this data supports the hypothesis that: (i) APV/CO is distinct from European APV subgroups and belongs to the novel subgroup APV/C (APV/US); (ii) APV/CO is more closely related to hMPV, a mammalian metapneumovirus, than to either APV subgroup A or B. The SH gene of APV/CO was cloned using a genomic walk strategy which initiated cDNA synthesis from genomic RNA that traversed the genes in the order 3'-M-F-M2-SH-G-5', thus confirming that gene-order of APV/CO conforms in the genus Metapneumovirus. We also provide the sequences of transcription-signals and the M-F, F-M2, M2-SH and SH-G intergenic regions of APV/CO.


Subject(s)
Metapneumovirus/classification , Metapneumovirus/genetics , Pneumovirus/classification , Pneumovirus/genetics , Viral Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Birds/virology , Humans , Molecular Sequence Data , Sequence Alignment , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Species Specificity , United States , Viral Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...