Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Appl Environ Microbiol ; 87(11)2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33741628

RESUMEN

MalF has been shown to be required for virulence in the important avian pathogen Mycoplasma gallisepticum To characterize the function of MalF, predicted to be part of a putative ABC transporter, we compared metabolite profiles of a mutant with a transposon inserted in malF (MalF-deficient ST mutant 04-1; ΔmalF) with those of wild-type bacteria using gas chromatography-mass spectrometry and liquid chromatography-mass spectrometry. Of the substrates likely to be transported by an ABC transport system, glycerol was detected at significantly lower abundance in the ΔmalF mutant, compared to the wild type. Stable isotope labeling using [U-13C]glycerol and reverse transcription-quantitative PCR analysis indicated that MalF was responsible for the import of glycerol into M. gallisepticum and that, in the absence of MalF, the transcription of gtsA, which encodes a second transporter, GtsA, was upregulated, potentially to increase the import of glycerol-3-phosphate into the cell to compensate for the loss of MalF. The loss of MalF appeared to have a global effect on glycerol metabolism, suggesting that it may also play a regulatory role, and cellular morphology was also affected, indicating that the change to glycerol metabolism may have a broader effect on cellular organization. Overall, this study suggests that the reduced virulence of the ΔmalF mutant is due to perturbed glycerol uptake and metabolism and that the operon including malF should be reannotated as golABC to reflect its function in glycerol transport.IMPORTANCE Many mycoplasmas are pathogenic and cause disease in humans and animals. M. gallisepticum causes chronic respiratory disease in chickens and infectious sinusitis in turkeys, resulting in economic losses in poultry industries throughout the world. Expanding our knowledge about the pathogenesis of mycoplasma infections requires better understanding of the specific gene functions of these bacteria. In this study, we have characterized the metabolic function of a protein involved in the pathogenicity of M. gallisepticum, as well as its effect on expression of selected genes, cell phenotype, and H2O2 production. This study is a key step forward in elucidating why this protein plays a key role in virulence in chickens. This study also emphasizes the importance of functional characterization of mycoplasma proteins, using tools such as metabolomics, since prediction of function based on homology to other bacterial proteins is not always accurate.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Proteínas Bacterianas/genética , Elementos Transponibles de ADN , Peróxido de Hidrógeno/metabolismo , Mycoplasma gallisepticum/genética , Mycoplasma gallisepticum/patogenicidad , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Bacterianas/metabolismo , Cromatografía Líquida de Alta Presión , Cromatografía de Gases y Espectrometría de Masas , Glicerol/metabolismo , Espectrometría de Masas , Mycoplasma gallisepticum/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virulencia/genética
2.
Microb Pathog ; 138: 103848, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31704462

RESUMEN

Mycoplasma gallisepticum (Mg) causes chronic respiratory disease (CRD) in chickens. However, the effect of Mg infection on energy metabolism in chicken lungs is still unknown. The present study was aimed to investigate the effect of Mg infection on energy metabolism in chicken lungs. Four-weeks-old white leghorn chickens were randomly divided into control group (L1) and Mg infection group (L2). Histopathology, transmission electron microscopy, qRT-PCR and Western blot were used to determine the hallmarks of ultrastructural analysis, inflammation and energy metabolism. Results revealed that Mg infection induced oxidative stress in the chicken lungs and serum cytokine activities were enhanced at the three time points. Chickens infected with Mg revealed abnormal morphology and cellular damage including increased inflammatory cells infiltrate, cellular debris and exudate, mitochondrial and DNA damage in the lungs. The mRNA and protein expression level of inflammation-related genes were significantly increased in L2 group, showing that Mg induced inflammation in chicken lungs. In addition, ATPase activities were reduced in L2 group compared to L1 group. Meanwhile, the expression of energy metabolism related genes were decreased at both mRNA and protein level at all assessed time points, which showed that Mg infection weakened energy metabolism in chicken lungs. In summary, the data suggested that Mg infection induced oxidative stress, inflammation and energy metabolism dysfunction in the chicken lungs, exploring new therapeutic targets and providing a reference for comparative veterinary medicine.


Asunto(s)
Pulmón/microbiología , Infecciones por Mycoplasma/veterinaria , Mycoplasma gallisepticum/patogenicidad , Enfermedades de las Aves de Corral/microbiología , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Animales , Pollos/microbiología , Citocinas/sangre , Metabolismo Energético/genética , Expresión Génica , Inflamación/microbiología , Pulmón/patología , Infecciones por Mycoplasma/inmunología , Infecciones por Mycoplasma/metabolismo , Estrés Oxidativo/genética
3.
Avian Pathol ; 49(4): 342-354, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32270701

RESUMEN

Leukocyte differentials are a useful tool for assessing systemic immunological changes during pathogen infections, particularly for non-model species. To date, no study has explored how experimental infection with a common bacterial pathogen, Mycoplasma gallisepticum (MG), influences the course and strength of haematological changes in the natural songbird host, house finches. Here we experimentally inoculated house finches with MG isolates known to vary in virulence, and quantified the proportions of circulating leukocytes over the entirety of infection. First, we found significant temporal effects of MG infection on the proportions of most cell types, with strong increases in heterophil and monocyte proportions during infection. Marked decreases in lymphocyte proportions also occurred during infection, though these proportional changes may simply be driven by correlated increases in other leukocytes. Second, we found significant effects of isolate virulence, with the strongest changes in cell proportions occurring in birds inoculated with the higher virulence isolates, and almost no detectable changes relative to sham treatment groups in birds inoculated with the lowest virulence isolate. Finally, we found that variation in infection severity positively predicted the proportion of circulating heterophils and lymphocytes, but the strength of these correlations was dependent on isolate. Taken together, these results indicate strong haematological changes in house finches during MG infection, with markedly different responses to MG isolates of varying virulence. These results are consistent with the possibility that evolved virulence in house finch MG results in higher degrees of immune stimulation and associated immunopathology, with potential direct benefits for MG transmission. RESEARCH HIGHLIGHTS House finches show a marked pro-inflammatory response to M. gallisepticum infection. Virulent pathogen isolates produce stronger finch white blood cell responses. Among birds, stronger white blood cell responses are associated with higher infection severity.


Asunto(s)
Enfermedades de las Aves/prevención & control , Pinzones/microbiología , Infecciones por Mycoplasma/veterinaria , Mycoplasma gallisepticum/patogenicidad , Animales , Enfermedades de las Aves/microbiología , Femenino , Leucocitos/inmunología , Masculino , Infecciones por Mycoplasma/microbiología , Infecciones por Mycoplasma/prevención & control , Mycoplasma gallisepticum/inmunología , Virulencia
4.
Infect Immun ; 87(9)2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31235640

RESUMEN

Mycoplasma gallisepticum is an avian respiratory and reproductive tract pathogen that has a significant economic impact on the poultry industry worldwide. Although membrane proteins of Mycoplasma spp. are thought to play crucial roles in host interactions, very few have had their biochemical function defined. In this study, we found that the GroEL protein (heat shock protein 60) of Mycoplasma gallisepticum could induce apoptosis in peripheral blood mononuclear cells, and the underlying molecular mechanism was further determined. The GroEL gene from Mycoplasma gallisepticum was cloned and expressed in Escherichia coli to facilitate the functional analysis of recombinant protein. The purified GroEL protein was shown to adhere to peripheral blood mononuclear cells (PBMCs) and DF-1 cells and cause apoptosis in PBMCs. A protein pulldown assay coupled with mass spectrometry identified that annexin A2 possibly interacted with GroEL protein. Coimmunoprecipitation assays confirmed that GroEL proteins could bind to annexin A2, and confocal analysis further demonstrated that GroEL colocolized with annexin A2 in HEK293T cells and PBMCs. Moreover, annexin A2 expression was significantly induced by a recombinant GroEL protein in PBMCs, and knocking down annexin A2 expression resulted in significantly reduced apoptosis. Taken together, these data suggest that GroEL induces apoptosis in host cells by interacting with annexin A2, a novel virulence mechanism in Mycoplasma gallisepticum Our findings lead to a better understanding of molecular pathogenesis in Mycoplasma gallisepticum.


Asunto(s)
Anexina A2/fisiología , Apoptosis/fisiología , Chaperonina 60/fisiología , Infecciones por Mycoplasma/microbiología , Mycoplasma gallisepticum/patogenicidad , Animales , Leucocitos Mononucleares/metabolismo , Enfermedades de las Aves de Corral/microbiología
5.
Infect Immun ; 87(3)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30559221

RESUMEN

Mycoplasmas are small bacterial commensals or pathogens that commonly colonize host mucosal tissues and avoid rapid clearance, in part by stimulating inflammatory, immunopathogenic responses. We previously characterized a wide array of transcriptomic perturbations in avian host tracheal mucosae infected with virulent, immunopathologic Mycoplasma gallisepticum; however, mechanisms delineating these from protective responses, such as those induced upon vaccination, have not been thoroughly explored. In this study, host transcriptomic responses to two experimental M. gallisepticum vaccines were assessed during the first 2 days of infection. Relative to virulent infection, host metabolic and immune gene responses to both vaccines were greatly decreased, including early innate immune responses critical to disease development and subsequent adaptive immunity. These data specify host genes and potential mechanisms contributing to maladaptive versus beneficial host responses-information critical for design of vaccines efficacious in both limiting inflammation and enabling pathogen clearance.


Asunto(s)
Vacunas Bacterianas/inmunología , Pollos/inmunología , Infecciones por Mycoplasma/veterinaria , Mycoplasma gallisepticum/patogenicidad , Enfermedades de las Aves de Corral/microbiología , Inmunidad Adaptativa , Animales , Femenino , Regulación de la Expresión Génica/inmunología , Infecciones por Mycoplasma/inmunología , Enfermedades de las Aves de Corral/inmunología , Organismos Libres de Patógenos Específicos , Vacunas Atenuadas , Virulencia
6.
Microb Pathog ; 117: 225-231, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29471139

RESUMEN

Mycoplasma gallisepticum (M. gallisepticum) is one of the most important pathogens that cause chronic respiratory disease in chickens. M. gallisepticum-derived lipid-associated membrane proteins (LAMPs) are thought to be one of the major factors in mycoplasma pathogenesis and are potent inducers of the host innate immune response. However, the interaction of pathogenic M. gallisepticum-derived LAMPs with Toll-like receptors (TLRs) and the signaling pathways responsible for activating inflammation and NF-κB have not been fully elucidated. In this study, we found that IL-1ß expression was induced in DF-1 cells stimulated with M. gallisepticum LAMPs. Subcellular localization experiments using immunofluorescence assays (IFAs) showed p65 translocation from the cytoplasm to the nucleus in DF-1 cells following stimulation with M. gallisepticum LAMPs. Phosphorylation of p65 was detected in LAMP-stimulated DF-1 cells. Treatment with an NF-κB-specific inhibitor showed that NF-κB is required for M. gallisepticum LAMP-induced IL-1ß expression. In addition, the results indicated that TLR2 and myeloid differentiation primary-response protein 88 (MyD88)-dependent signaling pathways were involved in the activation of NF-κB by M. gallisepticum LAMPs. Together, these results provide evidence that M. gallisepticum LAMPs activate IL-1ß production through the NF-κB pathway via TLR2 and MyD88.


Asunto(s)
Interleucina-1beta/biosíntesis , Infecciones por Mycoplasma/inmunología , Mycoplasma gallisepticum/metabolismo , Factor 88 de Diferenciación Mieloide/biosíntesis , Transducción de Señal , Receptor Toll-Like 2/biosíntesis , Animales , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/farmacología , Línea Celular , Pollos , Regulación de la Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Inflamación/inmunología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Mycoplasma gallisepticum/inmunología , Mycoplasma gallisepticum/patogenicidad , Factor 88 de Diferenciación Mieloide/genética , Receptor Toll-Like 2/genética
7.
J Evol Biol ; 31(11): 1704-1714, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30107064

RESUMEN

Emergent infectious diseases can have a devastating impact on host populations. The high selective pressures on both the hosts and the pathogens frequently lead to rapid adaptations not only in pathogen virulence but also host resistance following an initial outbreak. However, it is often unclear whether hosts will evolve to avoid infection-associated fitness costs by preventing the establishment of infection (here referred to as qualitative resistance) or by limiting its deleterious effects through immune functioning (here referred to as quantitative resistance). Equally, the evolutionary repercussions these different resistance mechanisms have for the pathogen are often unknown. Here, we investigate the co-evolutionary dynamics of pathogen virulence and host resistance following the epizootic outbreak of the highly pathogenic bacterium Mycoplasma gallisepticum in North American house finches (Haemorhous mexicanus). Using an evolutionary modelling approach and with a specific emphasis on the evolved resistance trait, we demonstrate that the rapid increase in the frequency of resistant birds following the outbreak is indicative of strong selection pressure to reduce infection-associated mortality. This, in turn, created the ecological conditions that selected for increased bacterial virulence. Our results thus suggest that quantitative host resistance was the key factor underlying the evolutionary interactions in this natural host-pathogen system.


Asunto(s)
Enfermedades de las Aves/microbiología , Pinzones , Infecciones por Mycoplasma/veterinaria , Mycoplasma gallisepticum/patogenicidad , Animales , Evolución Biológica , Modelos Biológicos , Infecciones por Mycoplasma/microbiología , Mycoplasma gallisepticum/genética , Virulencia/genética
8.
Int J Mol Sci ; 19(4)2018 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-29652844

RESUMEN

Mycoplasma gallisepticum (MG) is the most economically significant mycoplasma pathogen of poultry that causes chronic respiratory disease (CRD) in chickens. Although miRNAs have been identified as a major regulator effect on inflammatory response, it is largely unclear how they regulate MG-induced inflammation. The aim of this study was to investigate the functional roles of gga-miR-451 and identify downstream targets regulated by gga-miR-451 in MG infection of chicken. We found that the expression of gga-miR-451 was significantly up-regulated during MG infection of chicken embryo fibroblast cells (DF-1) and chicken embryonic lungs. Overexpression of gga-miR-451 decreased the MG-induced inflammatory cytokine production, including tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6), whereas inhibition of gga-miR-451 had the opposite effect. Gene expression data combined with luciferase reporter assays demonstrated that tyrosine3-monooxygenase/tryptophan5-monooxygenase activation protein zeta (YWHAZ) was identified as a direct target of gga-miR-451 in the context of MG infection. Furthermore, upregulation of gga-miR-451 significantly inhibited the MG-infected DF-1 cells proliferation, induced cell-cycle arrest, and promoted apoptosis. Collectively, our results demonstrate that gga-miR-451 negatively regulates the MG-induced production of inflammatory cytokines via targeting YWHAZ, inhibits the cell cycle progression and cell proliferation, and promotes cell apoptosis. This study provides a better understanding of the molecular mechanisms of MG infection.


Asunto(s)
Proteínas 14-3-3/genética , Infecciones por Mycoplasma/genética , Mycoplasma gallisepticum/patogenicidad , Enfermedades de las Aves de Corral/microbiología , Infecciones del Sistema Respiratorio/veterinaria , Animales , Apoptosis , Línea Celular , Embrión de Pollo , Pollos , Citocinas/genética , Fibroblastos/química , Fibroblastos/citología , Fibroblastos/microbiología , Pulmón/química , Pulmón/microbiología , Infecciones por Mycoplasma/microbiología , Infecciones por Mycoplasma/veterinaria , Enfermedades de las Aves de Corral/genética , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/microbiología , Regulación hacia Arriba
9.
Int J Mol Sci ; 19(8)2018 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-30044397

RESUMEN

Mycoplasma gallisepticum (MG) is the pathogen of chronic respiratory disease (CRD), hallmarked by vigorous inflammation in chickens, causing the poultry industry enormous losses. miRNAs have emerged as important regulators of animal diseases. Previous miRNA sequencing data has demonstrated that miR-130b-3p is up-regulated in MG-infected chicken embryo lungs. Therefore, we aimed to investigate the function of miR-130b-3p in MG infection of chickens. RT-qPCR results confirmed that miR-130b-3p was up-regulated both in MG-infected chicken embryo lungs and chicken embryonic fibroblast cells (DF-1 cells). Furthermore, functional studies showed that overexpression of miR-130b-3p promoted MG-infected DF-1 cell proliferation and cell cycle, whereas inhibition of miR-130b-3p weakened these cellular processes. Luciferase reporter assay combined with gene expression data supported that phosphatase and tensin homolog deleted on chromosome ten (PTEN) was a direct target of miR-130b-3p. Additionally, overexpression of miR-130b-3p resulted in up-regulations of phosphatidylinositol-3 kinase (PI3K), serine/threonine kinase (AKT), and nuclear factor-κB (NF-κB), whereas inhibition of miR-130b-3p led to the opposite results. Altogether, upon MG infection, up-regulation of miR-130b-3p activates the PI3K/AKT/NF-κB pathway, facilitates cell proliferation and cell cycle via down-regulating PTEN. This study helps to understand the mechanism of host response to MG infection.


Asunto(s)
Pollos/microbiología , MicroARNs/metabolismo , Infecciones por Mycoplasma/veterinaria , Mycoplasma gallisepticum/patogenicidad , Enfermedades de las Aves de Corral/microbiología , Animales , Ciclo Celular , Línea Celular , Proliferación Celular , Embrión de Pollo , Fibroblastos/microbiología , Humanos , Pulmón/microbiología , MicroARNs/genética , Infecciones por Mycoplasma/microbiología , FN-kappa B/genética , FN-kappa B/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Enfermedades de las Aves de Corral/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Regulación hacia Arriba
10.
Infect Immun ; 85(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28396323

RESUMEN

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.


Asunto(s)
Pollos/microbiología , Pinzones/microbiología , Infecciones por Mycoplasma/epidemiología , Mycoplasma gallisepticum/aislamiento & purificación , Mycoplasma gallisepticum/patogenicidad , Animales , Femenino , Infecciones por Mycoplasma/microbiología , Infecciones por Mycoplasma/veterinaria , Fenotipo , Filogenia , Virginia , Virulencia
11.
Infect Immun ; 85(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28348054

RESUMEN

Relatively few virulence genes have been identified in pathogenic mycoplasmas, so we used signature-tagged mutagenesis to identify mutants of the avian pathogen Mycoplasma gallisepticum with a reduced capacity to persist in vivo and compared the levels of virulence of selected mutants in experimentally infected chickens. Four mutants had insertions in one of the two incomplete oppABCDF operons, and a further three had insertions in distinct hypothetical genes, two containing peptidase motifs and one containing a member of a gene family. The three hypothetical gene mutants and the two with insertions in oppD1 were used to infect chickens, and all five were shown to have a reduced capacity to induce respiratory tract lesions. One oppD1 mutant and the MGA_1102 and MGA_1079 mutants had a greatly reduced capacity to persist in the respiratory tract and to induce systemic antibody responses against M. gallisepticum The other oppD1 mutant and the MGA_0588 mutant had less capacity than the wild type to persist in the respiratory tract but did elicit systemic antibody responses. Although M. gallisepticum carries two incomplete opp operons, one of which has been acquired by horizontal gene transfer, our results suggest that one of the copies of oppD may be required for full expression of virulence. We have also shown that three hypothetical genes, two of which encode putative peptidases, may be required for full expression of virulence in M. gallisepticum. None of these genes has previously been shown to influence virulence in pathogenic mycoplasmas.


Asunto(s)
Proteínas Bacterianas/metabolismo , Pollos/microbiología , Mutagénesis Insercional/genética , Mycoplasma gallisepticum/genética , Mycoplasma gallisepticum/patogenicidad , Enfermedades de las Aves de Corral/microbiología , Animales , Proteínas Bacterianas/genética , Infecciones por Mycoplasma/genética , Infecciones por Mycoplasma/microbiología , Enfermedades de las Aves de Corral/genética , Virulencia/genética , Factores de Virulencia/genética
12.
Infect Immun ; 85(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28739827

RESUMEN

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.


Asunto(s)
Pollos/microbiología , Redes y Vías Metabólicas/genética , Infecciones por Mycoplasma/veterinaria , Mycoplasma gallisepticum/inmunología , Mycoplasma gallisepticum/patogenicidad , Enfermedades de las Aves de Corral/inmunología , Tráquea/microbiología , Animales , Quimiocinas/genética , Quimiocinas/inmunología , Pollos/inmunología , Citocinas/genética , Citocinas/inmunología , Perfilación de la Expresión Génica/métodos , Infecciones por Mycoplasma/inmunología , Infecciones por Mycoplasma/microbiología , Mycoplasma gallisepticum/genética , Enfermedades de las Aves de Corral/microbiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Análisis de Secuencia de ARN , Transducción de Señal , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Tráquea/inmunología
13.
Int J Med Microbiol ; 306(3): 174-86, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27079856

RESUMEN

Multiple respiratory infections have a significant impact on health and economy. Pathogenesis of co-infecting viruses and bacteria and their interaction with mucosal surfaces are poorly characterized. In this study we established a co-infection model based on pre-incubation of tracheal organ cultures (TOC) with Mycoplasma (M.) gallisepticum and a subsequent infection with avian influenza virus (AIV). Mycoplasma gallisepticum modified the pathogenesis of AIV as demonstrated in TOC of two different avian species (chickens and turkeys). Co-infection promoted bacterial growth in tracheal epithelium. Depending on the interaction time of M. gallisepticum with the host cells, AIV replication was either promoted or suppressed. M. gallisepticum inhibited the antiviral gene expression and affected AIV attachment to the host cell by desialylation of α-2,3 linked sialic acids. Ultrastructural analysis of co-infected TOC suggests that both pathogens may attach to and possibly infect the same epithelial cell. The obtained results contribute to better understanding of the interaction dynamics between M. gallisepticum and AIV. They highlight the importance of the time interval between infections as well as the biological properties of the involved pathogens as influencing factors in the outcome of respiratory infections.


Asunto(s)
Coinfección/veterinaria , Interacciones Huésped-Patógeno , Virus de la Influenza A/patogenicidad , Gripe Aviar/patología , Infecciones por Mycoplasma/patología , Mycoplasma gallisepticum/patogenicidad , Enfermedades de las Aves de Corral/patología , Animales , Apoptosis , Pollos/microbiología , Pollos/virología , Coinfección/microbiología , Coinfección/virología , Células Epiteliales/metabolismo , Epitelio/microbiología , Epitelio/virología , Gripe Aviar/microbiología , Gripe Aviar/virología , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Infecciones por Mycoplasma/microbiología , Infecciones por Mycoplasma/veterinaria , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/virología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ácidos Siálicos/metabolismo , Tráquea/microbiología , Tráquea/virología , Pavos/microbiología , Pavos/virología
14.
PLoS Biol ; 11(5): e1001570, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23723736

RESUMEN

The evolution of higher virulence during disease emergence has been predicted by theoretical models, but empirical studies of short-term virulence evolution following pathogen emergence remain rare. Here we examine patterns of short-term virulence evolution using archived isolates of the bacterium Mycoplasma gallisepticum collected during sequential emergence events in two geographically distinct populations of the host, the North American house finch (Haemorhous [formerly Carpodacus] mexicanus). We present results from two complementary experiments, one that examines the trend in pathogen virulence in eastern North American isolates over the course of the eastern epidemic (1994-2008), and the other a parallel experiment on Pacific coast isolates of the pathogen collected after M. gallisepticum established itself in western North American house finch populations (2006-2010). Consistent with theoretical expectations regarding short-term or dynamic evolution of virulence, we show rapid increases in pathogen virulence on both coasts following the pathogen's establishment in each host population. We also find evidence for positive genetic covariation between virulence and pathogen load, a proxy for transmission potential, among isolates of M. gallisepticum. As predicted by theory, indirect selection for increased transmission likely drove the evolutionary increase in virulence in both geographic locations. Our results provide one of the first empirical examples of rapid changes in virulence following pathogen emergence, and both the detected pattern and mechanism of positive genetic covariation between virulence and pathogen load are consistent with theoretical expectations. Our study provides unique empirical insight into the dynamics of short-term virulence evolution that are likely to operate in other emerging pathogens of wildlife and humans.


Asunto(s)
Pinzones/microbiología , Variación Genética , Mycoplasma gallisepticum/patogenicidad , Animales , Evolución Biológica , Pinzones/genética , Interacciones Huésped-Patógeno/genética , Virulencia
15.
Am Nat ; 185(3): 332-42, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25674688

RESUMEN

We explore pathogen virulence evolution during the spatial expansion of an infectious disease epidemic in the presence of a novel host movement trade-off, using a simple, spatially explicit mathematical model. This work is motivated by empirical observations of the Mycoplasma gallisepticum invasion into North American house finch (Haemorhous mexicanus) populations; however, our results likely have important applications to other emerging infectious diseases in mobile hosts. We assume that infection reduces host movement and survival and that across pathogen strains the severity of these reductions increases with pathogen infectiousness. Assuming these trade-offs between pathogen virulence (host mortality), pathogen transmission, and host movement, we find that pathogen virulence levels near the epidemic front (that maximize wave speed) are lower than those that have a short-term growth rate advantage or that ultimately prevail (i.e., are evolutionarily stable) near the epicenter and where infection becomes endemic (i.e., that maximize the pathogen basic reproductive ratio). We predict that, under these trade-offs, less virulent pathogen strains will dominate the periphery of an epidemic and that more virulent strains will increase in frequency after invasion where disease is endemic. These results have important implications for observing and interpreting spatiotemporal epidemic data and may help explain transient virulence dynamics of emerging infectious diseases.


Asunto(s)
Enfermedades de las Aves/transmisión , Mycoplasma gallisepticum/patogenicidad , Passeriformes/microbiología , Distribución Animal , Animales , Evolución Biológica , Interacciones Huésped-Patógeno , Modelos Teóricos , Infecciones por Mycoplasma/veterinaria , Mycoplasma gallisepticum/genética , Virulencia
16.
Avian Pathol ; 44(4): 296-304, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25925422

RESUMEN

Live Mycoplasma gallisepticum vaccines are used for the control of respiratory disease, egg production losses and egg transmission associated with M. gallisepticum infection in long-lived poultry. The first field case of apparent increased virulence and vertical transmission of ts-11, a live M. gallisepticum vaccine, has been reported. In that study a M. gallisepticum isolate from the broiler progeny of ts-11-vaccinated breeders was genotyped as ts-11 by sequence analysis of four different genetic targets and Random Amplified Polymorphic DNA and found to be significantly more virulent than ts-11 vaccine. The objective of the current study was to evaluate the rate of egg transmission and pathogenicity of ts-11 vaccine and isolates recovered from ts-11-vaccinated breeders (K6222B) and their broiler progeny (K6216D) which had been genotyped as ts-11. Groups of 28-week-old specific pathogen-free chickens at 87% average weekly egg production were inoculated with sterile broth media (negative controls), ts-11 vaccine, K6222B, K6216D or R strain (positive controls) by eye-drop and aerosol. K6216D transmitted via the egg at an average rate of 4.0% in the third and fourth weeks post-infection, while egg transmission of K6222B and ts-11 vaccine was not detected. M. gallisepticum was isolated from the air sacs, ovaries and oviducts of hens infected with K6216D and K6222B, but not from those infected with ts-11 vaccine. K6216D and K6222B both induced respiratory signs and significantly more tracheal colonization and more severe tracheal and air sac lesions than ts-11 vaccine (P ≤ 0.05). There were no substantial differences in the egg production of ts-11, K6216D and K6222B infected groups. These results provide the first conclusive evidence of transovarian transmission of an isolate genotyped as ts-11 and indicate that isolates genotyed as ts-11 vary in their virulence and ability to transmit via the egg.


Asunto(s)
Vacunas Bacterianas/inmunología , Pollos/microbiología , Infecciones por Mycoplasma/veterinaria , Mycoplasma gallisepticum/inmunología , Enfermedades de las Aves de Corral/transmisión , Sacos Aéreos/microbiología , Animales , Femenino , Genotipo , Masculino , Infecciones por Mycoplasma/prevención & control , Infecciones por Mycoplasma/transmisión , Mycoplasma gallisepticum/patogenicidad , Óvulo/microbiología , Enfermedades de las Aves de Corral/prevención & control , Organismos Libres de Patógenos Específicos , Tráquea/microbiología , Vacunas Atenuadas/inmunología , Virulencia
17.
PLoS Genet ; 8(2): e1002511, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22346765

RESUMEN

Measureable rates of genome evolution are well documented in human pathogens but are less well understood in bacterial pathogens in the wild, particularly during and after host switches. Mycoplasma gallisepticum (MG) is a pathogenic bacterium that has evolved predominantly in poultry and recently jumped to wild house finches (Carpodacus mexicanus), a common North American songbird. For the first time we characterize the genome and measure rates of genome evolution in House Finch isolates of MG, as well as in poultry outgroups. Using whole-genome sequences of 12 House Finch isolates across a 13-year serial sample and an additional four newly sequenced poultry strains, we estimate a nucleotide diversity in House Finch isolates of only ∼2% of ancestral poultry strains and a nucleotide substitution rate of 0.8-1.2×10(-5) per site per year both in poultry and in House Finches, an exceptionally fast rate rivaling some of the highest estimates reported thus far for bacteria. We also found high diversity and complete turnover of CRISPR arrays in poultry MG strains prior to the switch to the House Finch host, but after the invasion of House Finches there is progressive loss of CRISPR repeat diversity, and recruitment of novel CRISPR repeats ceases. Recent (2007) House Finch MG strains retain only ∼50% of the CRISPR repertoire founding (1994-95) strains and have lost the CRISPR-associated genes required for CRISPR function. Our results suggest that genome evolution in bacterial pathogens of wild birds can be extremely rapid and in this case is accompanied by apparent functional loss of CRISPRs.


Asunto(s)
Evolución Molecular , Pinzones/microbiología , Secuencias Invertidas Repetidas/genética , Tasa de Mutación , Mycoplasma gallisepticum/genética , Mycoplasma gallisepticum/patogenicidad , Enfermedades de las Aves de Corral/microbiología , Animales , Evolución Biológica , Pollos/microbiología , Pinzones/genética , Genoma Bacteriano , Especificidad del Huésped/genética , Filogenia , Polimorfismo de Nucleótido Simple , Enfermedades de las Aves de Corral/genética , Análisis de Secuencia de ADN , Pavos/microbiología
18.
Infect Immun ; 82(12): 4915-20, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25156740

RESUMEN

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.


Asunto(s)
Glicerol/metabolismo , Peróxido de Hidrógeno/metabolismo , Infecciones por Mycoplasma/veterinaria , Mycoplasma gallisepticum/patogenicidad , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/patología , Tráquea/patología , Animales , Pollos , Elementos Transponibles de ADN , Mutagénesis Insercional , Infecciones por Mycoplasma/microbiología , Índice de Severidad de la Enfermedad , Tráquea/microbiología , Virulencia
19.
J Evol Biol ; 27(6): 1271-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24750277

RESUMEN

In the mid-1990s, the common poultry pathogen Mycoplasma gallisepticum (MG) made a successful species jump to the eastern North American house finch Haemorhous mexicanus (HM). Subsequent strain diversification allows us to directly quantify, in an experimental setting, the transmission dynamics of three sequentially emergent geographic isolates of MG, which differ in the levels of pathogen load they induce. We find significant among-strain variation in rates of transmission as well as recovery. Pathogen strains also differ in their induction of host morbidity, measured as the severity of eye lesions due to infection. Relationships between pathogen traits are also investigated, with transmission and recovery rates being significantly negatively correlated, whereas transmission and virulence, measured as average eye lesion score over the course of infection, are positively correlated. By quantifying these disease-relevant parameters and their relationships, we provide the first analysis of the trade-offs that shape the evolution of this important emerging pathogen.


Asunto(s)
Enfermedades de las Aves/transmisión , Pinzones/microbiología , Mycoplasma gallisepticum/patogenicidad , Animales , Enfermedades Transmisibles Emergentes/microbiología , Enfermedades Transmisibles Emergentes/transmisión , Enfermedades Transmisibles Emergentes/veterinaria , Mycoplasma gallisepticum/aislamiento & purificación
20.
Vet Microbiol ; 295: 110126, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38896939

RESUMEN

The co-infection of Newcastle disease virus (NDV) and Mycoplasma gallisepticum (MG) has a detrimental effect on chicken production performance, exerts a deleterious impact on poultry production performance, resulting in substantial economic losses. However, the exact impact and underlying mechanisms remain ambiguous. In this study, co-infection models were established both in vivo and in vitro. Through these models, it was found that the co-infection facilitated the replication of MG and NDV, as well as MG induced pathogenesis. The administration of lentogenic NDV resulted in the suppression of the innate immune response in vivo. At cellular level, co-infection promoted MG induced apoptosis through caspase-dependent mitochondrial endogenous pathway and suppressed the inflammatory secretion. This research contributes novel insights in co-infection.


Asunto(s)
Pollos , Coinfección , Infecciones por Mycoplasma , Mycoplasma gallisepticum , Enfermedad de Newcastle , Virus de la Enfermedad de Newcastle , Enfermedades de las Aves de Corral , Mycoplasma gallisepticum/patogenicidad , Animales , Virus de la Enfermedad de Newcastle/patogenicidad , Virus de la Enfermedad de Newcastle/fisiología , Coinfección/microbiología , Coinfección/veterinaria , Coinfección/virología , Infecciones por Mycoplasma/veterinaria , Infecciones por Mycoplasma/microbiología , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/virología , Enfermedad de Newcastle/virología , Apoptosis , Inmunidad Innata , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA