Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Vet Microbiol ; 251: 108891, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33120088

ABSTRACT

Mycoplasma gallisepticum, a significant poultry pathogen, has evolved rapidly in its new passerine host since its first reported isolation from house finches in the US in 1994. In poultry, M. gallisepticum infects the upper respiratory tract, causing tracheal mucosal thickening and inflammation, in addition to inflammation of the reproductive tract. However, in house finches M. gallisepticum primarily causes inflammation of the conjunctiva. Given that different tissues are primarily affected by the same pathogen in different hosts, we have compared the early changes in gene expression of the phase-variable lipoproteins (vlhA) gene family of M. gallisepticum collected directly from target tissues in both hosts. Previous data have demonstrated that vlhA genes may be related to virulence, exhibiting changes in expression in a non-stochastic, temporal progression and we hypothesize that this may be influenced by differences in the target host tissue. If this is true, we would expect M. gallisepticum to display a different vlhA gene expression pattern in the chicken trachea compared to its expression pattern in house finch conjunctiva. Here we report significant differences in vlhA gene expression patterns between M. gallisepticum collected from chicken tracheas compared to those collected from house finch conjunctiva. While many of the predominant vlhA genes expressed in the input population showed an increase in expression in the chicken trachea at day one postinfection, those same vlhA genes decreased in expression in the house finch. These data suggest that discrete suites of vlhA genes may be involved in M. gallisepticum pathogenesis and tropism for unique tissues in two disparate avian hosts.


Subject(s)
Bacterial Proteins/genetics , Gene Expression , Host Microbial Interactions/genetics , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/genetics , Poultry Diseases/microbiology , Animals , Chickens/microbiology , Conjunctiva/microbiology , Female , Finches/microbiology , Poultry Diseases/pathology , Sequence Analysis, RNA , Specific Pathogen-Free Organisms , Trachea/microbiology , Virulence
2.
Infect Immun ; 88(1)2019 12 17.
Article in English | MEDLINE | ID: mdl-31591166

ABSTRACT

The avian pathogen Mycoplasma gallisepticum, the etiological agent of chronic respiratory disease in chickens, exhibits enhanced pathogenesis in the presence of a copathogen such as low-pathogenic avian influenza virus (LPAIV). To further investigate the intricacies of this copathogenesis, chickens were monoinfected or coinfected with either virulent M. gallisepticum strain Rlow or LPAIV H3N8 (A/duck/Ukraine/1963), with assessment of tracheal histopathology, pathogen load, and transcriptomic host responses to infection by RNA sequencing. Chickens coinfected with M. gallisepticum Rlow followed by LPAIV H3N8 exhibited significantly more severe tracheal lesions and mucosal thickening than chickens infected with LPAIV H3N8 alone and greater viral loads than chickens infected first with H3N8 and subsequently with M. gallisepticum Rlow Recovery of live M. gallisepticum was significantly higher in chickens infected first with LPAIV H3N8 and then with M. gallisepticum Rlow, compared to chickens given a mock infection followed by M. gallisepticum Rlow The transcriptional responses to monoinfection and coinfection with M. gallisepticum and LPAIV highlighted the involvement of differential expression of genes such as Toll-like receptor 15, Toll-like receptor 21, and matrix metallopeptidase 1. Pathway and gene ontology analyses of these differentially expressed genes suggest that coinfection with virulent M. gallisepticum and LPAIV induces decreases in the expression of genes related to ciliary activity in vivo and alters multiple immune-related signaling cascades. These data aid in the understanding of the relationship between M. gallisepticum and LPAIV during copathogenesis in the natural host and may contribute to further understanding of copathogen infections of humans and other animals.


Subject(s)
Coinfection/pathology , Influenza in Birds/pathology , Mycoplasma Infections/pathology , Poultry Diseases/pathology , Trachea/pathology , Animals , Bacterial Load , Chickens , Gene Expression Profiling , Gene Expression Regulation , Histocytochemistry , Host-Pathogen Interactions , Influenza A virus/growth & development , Influenza in Birds/complications , Mycoplasma Infections/complications , Mycoplasma gallisepticum/growth & development , Viral Load
3.
Infect Immun ; 86(11)2018 11.
Article in English | MEDLINE | ID: mdl-30181349

ABSTRACT

Mycoplasma gallisepticum, the primary etiologic agent of chronic respiratory disease, is a significant poultry pathogen, causing severe inflammation and leading to economic losses worldwide. Immunodominant proteins encoded by the variable lipoprotein and hemagglutinin (vlhA) gene family are thought to be important for M. gallisepticum-host interaction, pathogenesis, and immune evasion, but their exact role remains unknown. Previous work has demonstrated that vlhA phase variation is dynamic throughout the earliest stages of infection, with vlhA 3.03 being the predominant vlhA expressed during the initial infection, and that the pattern of dominant vlhA expression may be nonrandom and regulated by previously unrecognized mechanisms. To further investigate this gene family, we assessed the vlhA profile of two well-characterized vaccine strains, GT5 and Mg7, a vlhA 3.03 mutant strain, and an M. gallisepticum population expressing an alternative immunodominant vlhA Here, we report that two M. gallisepticum vaccine strains show different vlhA profiles over the first 2 days of infection compared to that of wild-type Rlow, while the population expressing an alternative immunodominant vlhA gene reverted to a profile indistinguishable from that of wild-type Rlow Additionally, we observed a slight shift in the vlhA gene expression profile but no reduction in virulence in a vlhA 3.03 mutant. Taken together, these data further support the hypothesis that M. gallisepticum vlhA genes change in a nonstochastic temporal progression of expression and that vlhA 3.03, while preferred, is not required for virulence. Collectively, these data may be important in elucidating mechanisms of colonization and overall pathogenesis of M. gallisepticum.


Subject(s)
Bacterial Proteins/biosynthesis , Gene Expression Regulation, Bacterial , Hemagglutinins/biosynthesis , Lipoproteins/biosynthesis , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/genetics , Poultry Diseases/microbiology , Animals , Antigenic Variation , Bacterial Proteins/genetics , Chickens , Gene Expression Profiling , Hemagglutinins/genetics , Lipoproteins/genetics , Multigene Family , Mycoplasma Infections/microbiology , Mycoplasma gallisepticum/metabolism , Poultry Diseases/pathology
4.
Infect Immun ; 85(10)2017 10.
Article in English | MEDLINE | ID: mdl-28739827

ABSTRACT

Mycoplasma gallisepticum, the primary etiologic agent of chronic respiratory disease (CRD) in poultry, leads to prolonged recruitment and activation of inflammatory cells in the respiratory mucosa. This is consistent with the current model of immune dysregulation that ostensibly allows the organism to evade clearance mechanisms and establish chronic infection. To date, studies using quantitative reverse transcription-PCR (qRT-PCR) and microarrays have shown a significant transient upregulation of cytokines and chemokines from tracheal epithelial cells (TECs) in vitro and tracheal tissue ex vivo in response to virulent strain Rlow that contributes to the infiltration of inflammatory cells into the tracheal mucosa. To expand upon these experiments, RNA was isolated from tracheas of 20 chickens infected with M. gallisepticum Rlow and 20 mock-infected animals at days 1, 3, 5, and 7 postinoculation, and samples were analyzed for differential gene expression using Illumina RNA sequencing. A rapid host response was observed 24 h postinfection, with over 2,500 significantly differentially expressed genes on day 3, the peak of infection. Many of these genes have immune-related functions involved in signaling pathways, including Toll-like receptor (TLR), mitogen-activated protein kinase, Jak-STAT, and the nucleotide oligomerization domain-like receptor pathways. Of interest was the increased expression of numerous cell surface receptors, including TLR4 and TLR15, which may contribute to the production of cytokines. Metabolic pathways were also activated on days 1 and 3 postinfection, ostensibly due to epithelial cell distress that occurs upon infection. Early perturbations in tissue-wide gene expression, as observed here, may underpin a profound immune dysregulation, setting the stage for disease manifestations characteristic of M. gallisepticum infection.


Subject(s)
Chickens/microbiology , Metabolic Networks and Pathways/genetics , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/immunology , Mycoplasma gallisepticum/pathogenicity , Poultry Diseases/immunology , Trachea/microbiology , Animals , Chemokines/genetics , Chemokines/immunology , Chickens/immunology , Cytokines/genetics , Cytokines/immunology , Gene Expression Profiling/methods , Mycoplasma Infections/immunology , Mycoplasma Infections/microbiology , Mycoplasma gallisepticum/genetics , Poultry Diseases/microbiology , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Sequence Analysis, RNA , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Trachea/immunology
5.
Infect Immun ; 85(6)2017 06.
Article in English | MEDLINE | ID: mdl-28396323

ABSTRACT

Mycoplasma gallisepticum, known primarily as a respiratory pathogen of domestic poultry, has emerged since 1994 as a significant pathogen of the house finch (Haemorhousmexicanus) causing severe conjunctivitis and mortality. House finch-associated M. gallisepticum (HFMG) spread rapidly and increased in virulence for the finch host in the eastern United States. In the current study, we assessed virulence in domestic poultry with two temporally distant, and yet geographically consistent, HFMG isolates which differ in virulence for house finches-Virginia 1994 (VA1994), the index isolate of the epidemic, and Virginia 2013 (VA2013), a recent isolate of increased house finch virulence. Here we report a significant difference between VA1994 and VA2013 in their levels of virulence for chickens; notably, this difference correlated inversely to the difference in their levels of virulence for house finches. VA1994, while moderately virulent in house finches, displayed significant virulence in the chicken respiratory tract. VA2013, while highly virulent in the house finch, was significantly attenuated in chickens relative to VA1994, displaying less-severe pathological lesions in, and reduced bacterial recovery from, the respiratory tract. Overall, these data indicate that a recent isolate of HFMG is greatly attenuated in the chicken host relative to the index isolate, notably demonstrating a virulence phenotype in chickens inversely related to that in the finch host.


Subject(s)
Chickens/microbiology , Finches/microbiology , Mycoplasma Infections/epidemiology , Mycoplasma gallisepticum/isolation & purification , Mycoplasma gallisepticum/pathogenicity , Animals , Female , Mycoplasma Infections/microbiology , Mycoplasma Infections/veterinary , Phenotype , Phylogeny , Virginia , Virulence
6.
Infect Immun ; 84(1): 351-5, 2016 01.
Article in English | MEDLINE | ID: mdl-26553465

ABSTRACT

Mycoplasma gallisepticum is the primary etiologic agent of chronic respiratory disease in poultry, a disease largely affecting the respiratory tract and causing significant economic losses worldwide. Immunodominant proteins encoded by members of the variable lipoprotein and hemagglutinin (vlhA) gene family are thought to be important for mechanisms of M. gallisepticum-host interaction, pathogenesis, and immune evasion, but their exact role and the overall nature of their phase variation are unknown. To better understand these mechanisms, we assessed global transcriptomic vlhA gene expression directly from M. gallisepticum populations present on tracheal mucosae during a 7-day experimental infection in the natural chicken host. Here we report differences in both dominant and minor vlhA gene expression levels throughout the first week of infection and starting as early as day 1 postinfection, consistent with a functional role not dependent on adaptive immunity for driving phase variation. Notably, data indicated that, at given time points, specific vlhA genes were similarly dominant in multiple independent hosts, suggesting a nonstochastic temporal progression of dominant vlhA gene expression in the colonizing bacterial population. The dominant expression of a given vlhA gene was not dependent on the presence of 12-copy GAA trinucleotide repeats in the promoter region and did not revert to the predominate vlhA gene when no longer faced with host pressures. Overall, these data indicate that vlhA phase variation is dynamic throughout the earliest stages of infection and that the pattern of dominant vlhA expression may be nonrandom and regulated by previously unrecognized mechanisms.


Subject(s)
Bacterial Proteins/biosynthesis , Hemagglutinins/biosynthesis , Lectins/biosynthesis , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Base Sequence , Chickens , Female , Hemagglutinins/genetics , Lectins/genetics , Lipoproteins/biosynthesis , Lipoproteins/genetics , Mycoplasma Infections/immunology , Mycoplasma Infections/microbiology , Mycoplasma gallisepticum/immunology , Poultry Diseases/immunology , Poultry Diseases/microbiology , Promoter Regions, Genetic , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/microbiology , Sequence Analysis, DNA
7.
Infect Immun ; 82(12): 4915-20, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25156740

ABSTRACT

Hydrogen peroxide (H2O2) is a by-product of glycerol metabolism in mycoplasmas and has been shown to cause cytotoxicity for cocultured eukaryotic cells. There appears to be selective pressure for mycoplasmas to retain the genes needed for glycerol metabolism. This has generated interest and speculation as to their function during infection. However, the actual effects of glycerol metabolism and H2O2 production on virulence in vivo have never been assessed in any Mycoplasma species. To this end, we determined that the wild-type (WT) R(low) strain of the avian pathogen Mycoplasma gallisepticum is capable of producing H2O2 when grown in glycerol and is cytotoxic to eukaryotic cells in culture. Transposon mutants with mutations in the genes present in the glycerol transport and utilization pathway, namely, glpO, glpK, and glpF, were identified. All mutants assessed were incapable of producing H2O2 and were not cytotoxic when grown in glycerol. We also determined that vaccine strains ts-11 and 6/85 produce little to no H2O2 when grown in glycerol, while the naturally attenuated F strain does produce H2O2. Chickens were infected with one of two glpO mutants, a glpK mutant, R(low), or growth medium, and tracheal mucosal thickness and lesion scores were assessed. Interestingly, all glp mutants were reproducibly virulent in the respiratory tracts of the chickens. Thus, there appears to be no link between glycerol metabolism/H2O2 production/cytotoxicity and virulence for this Mycoplasma species in its natural host. However, it is possible that glycerol metabolism is required by M. gallisepticum in a niche that we have yet to study.


Subject(s)
Glycerol/metabolism , Hydrogen Peroxide/metabolism , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/pathogenicity , Poultry Diseases/microbiology , Poultry Diseases/pathology , Trachea/pathology , Animals , Chickens , DNA Transposable Elements , Mutagenesis, Insertional , Mycoplasma Infections/microbiology , Severity of Illness Index , Trachea/microbiology , Virulence
8.
Microbiology (Reading) ; 158(Pt 8): 2073-2088, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22628486

ABSTRACT

Mycoplasma gallisepticum, a significant respiratory and reproductive pathogen of domestic poultry, has since 1994 been recognized as an emergent pathogen of the American house finch (Carpodacus mexicanus). Epizootic spread and pathognomonic characteristics of house finch-associated Mycoplasma gallisepticum (HFMG) have been studied as a model of an emergent to endemic pathogen in a novel host. Here we present comparative analysis of eight HFMG genomes, including one from an index isolate and seven isolates separated spatially and temporally (1994-2008) across the epizootic, and notably having differences in virulence. HFMG represented a monophyletic clade relative to sequenced poultry isolates, with genomic changes indicating a novel M. gallisepticum lineage and including unique deletions of coding sequence. Though most of the HFMG genome was highly conserved among isolates, genetic distances correlated with temporal-spatial distance from the index. The most dramatic genomic differences among HFMG involved phase-variable and immunodominant VlhA lipoprotein genes, including those variable in presence and genomic location. Other genomic differences included tandem copy number variation of a 5 kbp repeat, changes in and adjacent to the clustered regularly interspaced short palindromic repeats, and small-scale changes affecting coding potential and association of genes with virulence. Divergence of monophyletic isolates from similar time/space in the epizootic indicated local diversification of distinct HFMG sublineages. Overall, these data identify candidate virulence genes and reveal the importance of phase-variable lipoproteins during the evolution of M. gallisepticum during its emergence and dissemination in a novel host in nature, likely mediating an important role at the interface between pathogen virulence and host immunity.


Subject(s)
Bacterial Proteins/genetics , Bird Diseases/microbiology , Evolution, Molecular , Genetic Variation , Lipoproteins/genetics , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/genetics , Passeriformes/microbiology , Animals , Bacterial Proteins/metabolism , Base Sequence , Genome, Bacterial , Genomics , Lipoproteins/metabolism , Molecular Sequence Data , Mycoplasma Infections/microbiology , Mycoplasma gallisepticum/classification , Mycoplasma gallisepticum/isolation & purification , Mycoplasma gallisepticum/pathogenicity , Phylogeny , Virulence , Zoonoses/microbiology
9.
Infect Immun ; 80(3): 1007-14, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22252865

ABSTRACT

Mycoplasma pneumoniae is a significant human respiratory pathogen that causes high morbidity worldwide. No vaccine to prevent M. pneumoniae infection currently exists, since the mechanisms of pathogenesis are poorly understood. To this end, we constructed a P30 cytadhesin mutant (P-130) with a drastically reduced capacity for binding to erythrocytes and an inability to glide on glass substrates. This mutant was determined to be avirulent and cannot survive in the lungs of BALB/c mice. We also ascertained that the previously identified P30 gliding motility mutant II-3R is avirulent and also cannot be recovered from the lungs of mice after infection. Mutant P130 was then assessed for its efficacy as a live attenuated vaccine candidate in mice after challenge with wild-type M. pneumoniae. After vaccination with the P-130 P30 mutant, mice showed evidence of exacerbated disease upon subsequent challenge with the wild-type strain PI1428, which appears to be driven by a Th17 response and corresponding eosinophilia. Our results are in accordance with other reports of vaccine-induced disease exacerbation in rodents and emphasize the need to better understand the basic mechanisms of M. pneumoniae pathogenesis.


Subject(s)
Adhesins, Bacterial/genetics , Bacterial Vaccines/adverse effects , Bacterial Vaccines/immunology , Disease Progression , Gene Knockout Techniques , Mycoplasma pneumoniae/immunology , Pneumonia, Mycoplasma/prevention & control , Animals , Bacterial Adhesion , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Eosinophilia , Erythrocytes/microbiology , Female , Lung/microbiology , Mice , Mice, Inbred BALB C , Microbial Viability , Mycoplasma pneumoniae/genetics , Pneumonia, Mycoplasma/immunology , Pneumonia, Mycoplasma/microbiology , Th17 Cells/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Virulence
10.
Infect Immun ; 78(8): 3475-83, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20515935

ABSTRACT

Many lipoproteins are expressed on the surfaces of mycoplasmas, and some have been implicated as playing roles in pathogenesis. Family 2 lipoproteins of Mycoplasma pneumoniae have a conserved "mycoplasma lipoprotein X" central domain and a "mycoplasma lipoprotein 10" C-terminal domain and are differentially expressed in response to environmental conditions. Homologues of family 2 lipoproteins are Mycoplasma specific and include the lipoprotein of Mycoplasma gallisepticum, encoded by the MGA0674 gene. Comparative transcriptomic analysis of the M. gallisepticum live attenuated vaccine strain F and the virulent strain R(low), reported in this study, indicated that MGA0674 is one of several differentially expressed genes. The MGA0674-encoded lipoprotein is a proteolytically processed, immunogenic, TX-114 detergent-phase protein which appears to have antigenic divergence between field strains R(low) and S6. We examined the virulence of an R(low) Delta MGA0674 mutant (P1H9) in vivo and observed reduced recovery and attenuated virulence in the tracheas of experimentally infected chickens. The virulence of two additional R(low) Delta MGA0674 mutants, 2162 and 2204, was assessed in a second in vivo virulence experiment. These mutants exhibited partial to complete attenuation in vivo, but recovery was observed more frequently. Since only Mycoplasma species harbor homologues of MGA0674, the gene product has been renamed "Mycoplasma-specific lipoprotein A" (MslA). Collectively, these data indicate that MslA is an immunogenic lipoprotein exhibiting reduced expression in an attenuated strain and plays a role in M. gallisepticum virulence.


Subject(s)
Bacterial Proteins/physiology , Lipoproteins/physiology , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/pathogenicity , Poultry Diseases/microbiology , Virulence Factors/physiology , Animals , Bacterial Proteins/genetics , Chickens , Female , Gene Deletion , Gene Expression Profiling , Lipoproteins/deficiency , Mycoplasma Infections/microbiology , Mycoplasma Infections/pathology , Poultry Diseases/pathology , Trachea/microbiology , Trachea/pathology , Virulence , Virulence Factors/deficiency
11.
Infect Immun ; 78(4): 1760-71, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20123709

ABSTRACT

Mycoplasma gallisepticum is a significant respiratory and reproductive pathogen of domestic poultry. While the complete genomic sequence of the virulent, low-passage M. gallisepticum strain R (R(low)) has been reported, genomic determinants responsible for differences in virulence and host range remain to be completely identified. Here, we utilize genome sequencing and microarray-based comparative genomic data to identify these genomic determinants of virulence and to elucidate genomic variability among strains of M. gallisepticum. Analysis of the high-passage, attenuated derivative of R(low), R(high), indicated that relatively few total genomic changes (64 loci) occurred, yet they are potentially responsible for the observed attenuation of this strain. In addition to previously characterized mutations in cytadherence-related proteins, changes included those in coding sequences of genes involved in sugar metabolism. Analyses of the genome of the M. gallisepticum vaccine strain F revealed numerous differences relative to strain R, including a highly divergent complement of vlhA surface lipoprotein genes, and at least 16 genes absent or significantly fragmented relative to strain R. Notably, an R(low) isogenic mutant in one of these genes (MGA_1107) caused significantly fewer severe tracheal lesions in the natural host compared to virulent M. gallisepticum R(low). Comparative genomic hybridizations indicated few genetic loci commonly affected in F and vaccine strains ts-11 and 6/85, which would correlate with proteins affecting strain R virulence. Together, these data provide novel insights into inter- and intrastrain M. gallisepticum genomic variability and the genetic basis of M. gallisepticum virulence.


Subject(s)
DNA, Bacterial/genetics , Genome, Bacterial , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/genetics , Mycoplasma gallisepticum/pathogenicity , Poultry Diseases/microbiology , Virulence Factors/genetics , Animals , Chickens , Comparative Genomic Hybridization , DNA, Bacterial/chemistry , Female , Molecular Sequence Data , Sequence Analysis, DNA
12.
Vaccine ; 26(16): 2010-9, 2008 Apr 07.
Article in English | MEDLINE | ID: mdl-18342996

ABSTRACT

In a previous study, signature sequence mutagenesis (SSM) was used to identify a mutant with a disruption of the gene encoding the metabolic factor, dihydrolipoamide dehydrogenase, and that mutant was designated Mg 7. The current study assessed the safety, immunogenicity and efficacy of Mg 7 in comparison to two commercially available vaccines (ts-11 and F) as well as a laboratory vaccine strain, GT5. Intratracheal vaccination of chickens with all four attenuated mutants induced varying levels of protection against intratracheal challenge with virulent Mycoplasma gallisepticum strain R(low). Mg 7 vaccinated chickens rapidly cleared the challenge strain, had lower histopathologic tracheal lesion scores when compared to unvaccinated chickens, and mounted a strong humoral anti-M. gallisepticum-specific IgG response. The IgG levels increased 2- to 3-fold upon R(low) challenge. Mg 7 induced a greater level of protection against intratracheal R(low) challenge than that observed with the other three attenuated strains, as evidenced by a lower recovery of R(low) from tracheas and lower histopathologic lesion scores in tracheas and air sacs. Based on these findings, Mg 7 appears to have good potential as a safe and effective vaccine for the prevention of avian mycoplasmosis.


Subject(s)
Bacterial Vaccines/administration & dosage , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/immunology , Poultry Diseases/prevention & control , Respiratory Tract Infections/veterinary , Vaccination , Air Sacs/pathology , Animals , Antibodies, Bacterial/blood , Chickens , Dihydrolipoamide Dehydrogenase/genetics , Female , Mutation , Mycoplasma Infections/pathology , Mycoplasma Infections/prevention & control , Mycoplasma gallisepticum/enzymology , Mycoplasma gallisepticum/genetics , Poultry Diseases/pathology , Respiratory Mucosa/pathology , Respiratory Tract Infections/pathology , Respiratory Tract Infections/prevention & control , Trachea/pathology , Vaccines, Attenuated/administration & dosage
13.
J Clin Microbiol ; 46(2): 438-42, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18032617

ABSTRACT

Sheeppox virus (SPPV) is a member of the Capripoxvirus (CaPV) genus of the Poxviridae family. Members of this genus, which also include goatpox and lumpy skin disease viruses, cause economically significant disease in sheep, goats, and cattle. A rapid diagnostic assay for CaPV would be useful for disease surveillance as well as for detection of CaPV in clinical samples and for outbreak management. Here we describe a fluorogenic probe hydrolysis (TaqMan) PCR assay designed for rapid detection of CaPV and tested on sheep experimentally infected with a virulent strain of SPPV. This assay can detect SPPV in buffy coats, nasal swabs, oral swabs, scabs, and skin lesions as well as in lung and lymph nodes collected at necropsy. This single-tube diagnostic assay can be performed in 2 h or less and can detect viral DNA in preclinical, clinical, and postmortem samples.


Subject(s)
Capripoxvirus/isolation & purification , Polymerase Chain Reaction/methods , Poxviridae Infections/veterinary , Sheep Diseases/diagnosis , Virology/methods , Animals , Capripoxvirus/genetics , Disease Outbreaks/prevention & control , Fluorescence , Fluorescent Dyes/metabolism , Poxviridae Infections/diagnosis , Poxviridae Infections/virology , Sheep , Sheep Diseases/virology , Time Factors
14.
J Virol ; 81(20): 11392-401, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17686843

ABSTRACT

Sheeppox virus (SPPV), a member of the Capripoxvirus genus of the Poxviridae, is the etiologic agent of a significant disease of sheep in the developing world. Genomic analysis of pathogenic and vaccine capripoxviruses identified genes with potential roles in virulence and host range, including three genes with similarity to kelch-like genes of other poxviruses and eukaryotes. Here, a mutant SPPV with a deletion in the SPPV-019 kelch-like gene, DeltaKLP, was derived from the pathogenic strain SPPV-SA. DeltaKLP exhibited in vitro growth characteristics similar to those of SPPV-SA and revertant virus (RvKLP). DeltaKLP-infected cells exhibited a reduction in Ca(2+)-independent cell adhesion, suggesting that SPPV-019 may modulate cellular adhesion. When inoculated in sheep by the intranasal or intradermal routes, DeltaKLP was markedly attenuated, since all DeltaKLP-infected lambs survived infection. In contrast, SPPV-SA and RvKLP induced mortality approaching 100%. Lambs inoculated with DeltaKLP exhibited marked reduction or delay in fever response, gross lesions, viremia, and virus shedding compared to parental and revertant viruses. Together, these findings indicate that SPPV-019 is a significant SPPV virulence determinant in sheep.


Subject(s)
Capripoxvirus/genetics , Capripoxvirus/pathogenicity , Viral Proteins/physiology , Animals , Genes, Viral/physiology , Mutation , Sheep , Virulence/genetics
15.
Infect Immun ; 74(2): 931-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16428737

ABSTRACT

To effectively analyze Mycoplasma gallisepticum for virulence-associated determinants, the ability to create stable genetic mutations is essential. Global M. gallisepticum mutagenesis is currently limited to the use of transposons. Using the gram-positive transposon Tn4001mod, a mutant library of 110 transformants was constructed and all insertion sites were mapped. To identify transposon insertion points, a unique primer directed outward from the end of Tn4001mod was used to sequence flanking genomic regions. By comparing sequences obtained in this manner to the annotated M. gallisepticum genome, the precise locations of transposon insertions were discerned. After determining the transposon insertion site for each mutant, unique reverse primers were synthesized based on the specific sequences, and PCR was performed. The resultant amplicons were used as unique Tn4001mod mutant identifiers. This procedure is referred to as signature sequence mutagenesis (SSM). SSM permits the comprehensive screening of the M. gallisepticum genome for the identification of novel virulence-associated determinants from a mixed mutant population. To this end, chickens were challenged with a pool of 27 unique Tn4001mod mutants. Two weeks postinfection, the birds were sacrificed, and organisms were recovered from respiratory tract tissues and screened for the presence or absence of various mutants. SSM is a negative-selection screening technique whereby those mutants possessing transposon insertions in genes essential for in vivo survival are not recovered from the host. We have identified a virulence-associated gene encoding dihydrolipoamide dehydrogenase (lpd). A transposon insertion in the middle of the coding sequence resulted in diminished biologic function and reduced virulence of the mutant designated Mg 7.


Subject(s)
DNA Transposable Elements , Dihydrolipoamide Dehydrogenase/genetics , Mutagenesis, Insertional , Mycoplasma gallisepticum/pathogenicity , Poultry Diseases/microbiology , Poultry Diseases/pathology , Animals , Bacterial Proteins , Chickens/microbiology , Dihydrolipoamide Dehydrogenase/metabolism , Mycoplasma Infections/microbiology , Mycoplasma Infections/pathology , Mycoplasma Infections/veterinary , Mycoplasma gallisepticum/genetics , Mycoplasma gallisepticum/metabolism , Sequence Analysis, DNA , Specific Pathogen-Free Organisms , Virulence
16.
Infect Immun ; 70(12): 6839-45, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12438360

ABSTRACT

It was previously demonstrated that avirulent Mycoplasma gallisepticum strain R(high) (passage 164) is lacking three proteins that are expressed in its virulent progenitor, strain R(low) (passage 15). These proteins were identified as the cytadhesin molecule GapA, the putative cytadhesin-related molecule CrmA, and a component of a high-affinity transporter system, HatA. Complementation of R(high) with wild-type gapA restored expression in the transformant (GT5) but did not restore the cytadherence phenotype and maintained avirulence in chickens. These results suggested that CrmA might play an essential role in the M. gallisepticum cytadherence process. CrmA is encoded by the second gene in the gapA operon and shares significant sequence homology to the ORF6 gene of Mycoplasma pneumoniae, which has been shown to play an accessory role in the cytadherence process. Complementation of R(high) with wild-type crmA resulted in the transformant (SDCA) that lacked the cytadherence and virulence phenotype comparable to that found in R(high) and GT5. In contrast, complementation of R(high) with the entire wild-type gapA operon resulted in the transformant (GCA1) that restored cytadherence to the level found in wild-type R(low). In vivo pathogenesis trials revealed that GCA1 had regained virulence, causing airsacculitis in chickens. These results demonstrate that both GapA and CrmA are required for M. gallisepticum cytadherence and pathogenesis.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion , Bacterial Proteins/metabolism , Mycoplasma Infections/veterinary , Mycoplasma/pathogenicity , Poultry Diseases/microbiology , Animals , Bacterial Proteins/genetics , Chickens/microbiology , Mycoplasma/physiology , Mycoplasma Infections/microbiology , Mycoplasma Infections/physiopathology , Poultry Diseases/physiopathology , Sequence Analysis, DNA , Transformation, Bacterial , Virulence
17.
Vaccine ; 20(31-32): 3709-19, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12399199

ABSTRACT

The aim of this study was to assess the efficacy of a modified live Mycoplasma gallisepticum vaccine (GT5) for the protection of chickens against infection and respiratory disease. GT5 was constructed by the reconstitution of the avirulent high passage R (R(high)) strain with the gene encoding the major cytadhesin GapA. GT5 expressed GapA on its surface yet retained the phenotypic characteristics of the parental R(high) strain. Birds vaccinated with GT5 were protected upon challenge with the virulent low passage R (R(low)) strain as evidenced by a complete absence of tracheal lesions 2 and 4 weeks post-challenge, in contrast to sham immunized/challenged control birds. Modest amounts of IgG, and little, if any secretory IgA or IgM anti-M. gallisepticum were found in tracheal washings following vaccination. However, copious amounts of specific IgA were found following challenge, especially in sham immunized birds. This suggests that the tracheal IgG elicited by GT5 vaccination may have been responsible for blocking the initial colonization of R(low), thereby resulting in protection.


Subject(s)
Bacterial Vaccines/therapeutic use , Mycoplasma Infections/prevention & control , Mycoplasma Infections/veterinary , Mycoplasma/immunology , Poultry Diseases/microbiology , Poultry Diseases/prevention & control , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/veterinary , Vaccination/veterinary , Animals , Bacterial Vaccines/administration & dosage , Chickens , Drug Administration Routes/veterinary , Immunity, Mucosal , Immunoglobulin A, Secretory/biosynthesis , Immunoglobulin A, Secretory/blood , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin Isotypes/biosynthesis , Immunoglobulin Isotypes/blood , Immunoglobulin M/biosynthesis , Immunoglobulin M/blood , Mycoplasma/isolation & purification , Respiratory Tract Infections/microbiology , Trachea/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/therapeutic use
18.
Infect Immun ; 68(12): 6643-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11083776

ABSTRACT

Comparison of the phenotypic expression of Mycoplasma gallisepticum strain R low (passage 15) to that of strain R high (passage 164) revealed that three proteins, i.e., the cytadhesin molecule GapA, a 116-kDa protein (p116), and a 45-kDa protein (p45), are missing in strain R high. Sequence analysis confirmed that the insertion of an adenine 105 bp downstream of the gapA translational start codon resulted in premature termination of translation in R high. A second adenine insertion had also occurred at position 907. Restoration of expression of wild-type gapA in R high (clone designated GT5) allowed us to evaluate the extent to which the diminished cytadherence capacity could be attributed to GapA alone. The results indicated that GT5 attached to the same limited extent as the parental R high, from which it was derived. The cytadherence capability of the parental R high was not restored solely by gapA complementation alone, indicating that either p116 or p45 or both may play a role in the overall cytadherence process. The gene encoding p116 was found to be immediately downstream of gapA in the same operon and was designated crmA. This gene exhibited striking homology to genes encoding molecules with cytadhesin-related functions in both Mycoplasma pneumoniae and Mycoplasma genitalium. Transcriptional analysis revealed that crmA is not transcribed in R high. We are currently constructing a shuttle vector containing both the wild-type gapA and crmA for transformation into R high to assess the role of CrmA in the cytadherence process.


Subject(s)
Adhesins, Bacterial/physiology , Bacterial Adhesion , Mycoplasma/physiology , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/genetics , Amino Acid Sequence , Cells, Cultured , Humans , Molecular Sequence Data , Mycoplasma/genetics , Open Reading Frames
19.
Microbiology (Reading) ; 144 ( Pt 11): 2971-2978, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9846732

ABSTRACT

The identification of a gene (gapA) from Mycoplasma gallisepticum with homology to the P1 cytadherence gene of Mycoplasma pneumoniae is reported. The gapA gene is a 2895 bp ORF encoding a protein with a molecular mass of 105 kDa. Nucleotide sequence analysis of the gapA gene revealed 45% homology to the M. pneumoniae P1 gene, 46% homology to the Mycoplasma genitalium MgPa gene and 47% homology to the Mycoplasma pirum P1-like protein gene. It has a 64 mol % A+T content compared to 46, 60 and 72 mol % respectively for the P1, MgPa and the P1-like protein genes. As with the P1 and MgPa genes, gapA is a central gene in a multi-gene operon, but unlike the P1 and MgPa genes, there is only a single copy of gapA in the genome. GapA is a trypsin-sensitive surface-exposed protein. Chicken tracheal-ring inhibition-of-attachment assays, using anti-GapA Fab fragments, resulted in 64% inhibition of attachment. These results indicated that GapA plays a role in cytadherence of M. gallisepticum to host cells.


Subject(s)
Adhesins, Bacterial/genetics , Mycoplasma/physiology , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/metabolism , Animals , Bacterial Adhesion , Blotting, Southern , Genes, Bacterial , Immunoblotting , Molecular Sequence Data , Mycoplasma/genetics , Nucleic Acid Hybridization , Open Reading Frames/genetics , Physical Chromosome Mapping , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
20.
J Wildl Dis ; 34(2): 403-6, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9577796

ABSTRACT

Northern mockingbirds (Mimus polyglottos) and blue jays (Cyanocitta cristata) in a Florida (USA) wildlife care facility developed clinical signs and gross lesions suggestive of the ongoing outbreak of Mycoplasma gallisepticum (MG) conjunctivitis in house finches (Carpodacus mexicanus) and American goldfinches (Carduelis tristis). Mycoplasmal organisms were cultured from conjunctival/corneal swabs of birds with sinusitis, conjunctivitis, and/or epiphora. All of the isolates tested were identified as Mycoplasma sturni by indirect immunofluorescence. Mycoplasma sturni as well as MG should be considered in the differential diagnosis of songbirds with conjunctivitis.


Subject(s)
Bird Diseases/microbiology , Conjunctivitis, Bacterial/veterinary , Disease Outbreaks/veterinary , Mycoplasma Infections/veterinary , Animals , Bird Diseases/epidemiology , Birds , Conjunctivitis, Bacterial/epidemiology , Conjunctivitis, Bacterial/microbiology , Florida/epidemiology , Fluorescent Antibody Technique, Indirect/veterinary , Mycoplasma Infections/epidemiology , Mycoplasma Infections/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...