Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Infect Immun ; 88(11)2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-32839185

RESUMEN

Mycoplasma pneumoniae is a cell wall-less bacterial pathogen of the conducting airways, causing bronchitis and atypical or "walking" pneumonia in humans. M. pneumoniae recognizes sialylated and sulfated oligosaccharide receptors to colonize the respiratory tract, but the contribution of the latter is particularly unclear. We used chamber slides coated with sulfatide (3-O-sulfogalactosylceramide) to provide a baseline for M. pneumoniae binding and gliding motility. As expected, M. pneumoniae bound to surfaces coated with sulfatide in a manner that was dependent on sulfatide concentration and incubation temperature and inhibited by competing dextran sulfate. However, mycoplasmas bound to sulfatide exhibited no gliding motility, regardless of receptor density. M. pneumoniae also bound lactose 3'-sulfate ligated to an inert polymer scaffold, and binding was inhibited by competing dextran sulfate. The major adhesin protein P1 mediates adherence to terminal sialic acids linked α-2,3, but P1-specific antibodies that blocked M. pneumoniae hemadsorption (HA) and binding to the sialylated glycoprotein laminin by 95% failed to inhibit mycoplasma binding to sulfatide, suggesting that P1 does not mediate binding to sulfated galactose. Consistent with this conclusion, the M. pneumoniae HA-negative mutant II-3 failed to bind to sialylated receptors but adhered to sulfatide in a temperature-dependent manner.


Asunto(s)
Adhesión Bacteriana/fisiología , Glicoproteínas/metabolismo , Mycoplasma pneumoniae/patogenicidad , Neumonía por Mycoplasma/microbiología , Proteínas Bacterianas/metabolismo , Humanos , Mycoplasma pneumoniae/metabolismo
2.
Mol Microbiol ; 109(6): 735-744, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29885004

RESUMEN

Mycoplasma pneumoniae is a common cause of human respiratory tract infections, including bronchitis and atypical pneumonia. M. pneumoniae binds glycoprotein receptors having terminal sialic acid residues via the P1 adhesin protein. Here, we explored the impact of sialic acid presentation on M. pneumoniae adherence and gliding on surfaces coated with sialylated glycoproteins, or chemically functionalized with α-2,3- and α-2,6-sialyllactose ligated individually or in combination to a polymer scaffold in precisely controlled densities. In both models, gliding required a higher receptor density threshold than adherence, and receptor density influenced gliding frequency but not gliding speed. However, very high densities of α-2,3-sialyllactose actually reduced gliding frequency over peak levels observed at lower densities. Both α-2,3- and α-2,6-sialyllactose supported M. pneumoniae adherence, but gliding was only observed on the former. Finally, gliding on α-2,3-sialyllactose was inhibited on surfaces also conjugated with α-2,6-sialyllactose, suggesting that both moieties bind P1 despite the inability of the latter to support gliding. Our results indicate that the nature and density of host receptor moieties profoundly influences M. pneumoniae gliding, which could affect pathogenesis and infection outcome. Furthermore, precise functionalization of polymer scaffolds shows great promise for further analysis of sialic acid presentation and M. pneumoniae adherence and gliding.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Lactosa/análogos & derivados , Movimiento/fisiología , Mycoplasma pneumoniae/metabolismo , Ácidos Siálicos/metabolismo , Glicoproteínas/metabolismo , Humanos , Lactosa/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Neumonía por Mycoplasma/microbiología , Neumonía por Mycoplasma/patología
3.
Mol Microbiol ; 108(3): 306-318, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29470845

RESUMEN

The Mycoplasma pneumoniae terminal organelle functions in adherence and gliding motility and is comprised of at least eleven substructures. We used electron cryotomography to correlate impaired gliding and adherence function with changes in architecture in diverse terminal organelle mutants. All eleven substructures were accounted for in the prkC, prpC and P200 mutants, and variably so for the HMW3 mutant. Conversely, no terminal organelle substructures were evident in HMW1 and HMW2 mutants. The P41 mutant exhibits a terminal organelle detachment phenotype and lacked the bowl element normally present at the terminal organelle base. Complementation restored this substructure, establishing P41 as either a component of the bowl element or required for its assembly or stability, and that this bowl element is essential to anchor the terminal organelle but not for leverage in gliding. Mutants II-3, III-4 and topJ exhibited a visibly lower density of protein knobs on the terminal organelle surface. Mutants II-3 and III-4 lack accessory proteins required for a functional adhesin complex, while the topJ mutant lacks a DnaJ-like co-chaperone essential for its assembly. Taken together, these observations expand our understanding of the roles of certain terminal organelle proteins in the architecture and function of this complex structure.


Asunto(s)
Mycoplasma pneumoniae/genética , Mycoplasma pneumoniae/fisiología , Orgánulos/genética , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana/genética , Proteínas Bacterianas/metabolismo , Tomografía con Microscopio Electrónico/métodos , Electrones , Orgánulos/metabolismo
4.
Cell Microbiol ; 20(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29155483

RESUMEN

Mycoplasma pneumoniae is a human respiratory tract pathogen causing acute and chronic airway disease states that can include long-term carriage and extrapulmonary spread. The mechanisms of persistence and migration beyond the conducting airways, however, remain poorly understood. We previously described an acute exposure model using normal human bronchial epithelium (NHBE) in air-liquid interface culture, showing that M. pneumoniae gliding motility is essential for initial colonisation and subsequent spread, including localisation to epithelial cell junctions. We extended those observations here, characterizing M. pneumoniae infection of NHBE for up to 4 weeks. Colonisation of the apical surface was followed by pericellular invasion of the basolateral compartment and migration across the underlying transwell membrane. Despite fluctuations in transepithelial electrical resistance and increased NHBE cell desquamation, barrier function remained largely intact. Desquamation was accompanied by epithelial remodelling that included cytoskeletal reorganisation and development of deep furrows in the epithelium. Finally, M. pneumoniae strains S1 and M129 differed with respect to invasion and histopathology, consistent with contrasting virulence in experimentally infected mice. In summary, this study reports pericellular invasion, NHBE cytoskeletal reorganisation, and tissue remodelling with persistent infection in a human airway epithelium model, providing clear insight into the likely route for extrapulmonary spread.


Asunto(s)
Mycoplasma pneumoniae/patogenicidad , Neumonía por Mycoplasma/metabolismo , Neumonía por Mycoplasma/microbiología , Sistema Respiratorio/microbiología , Bronquios/citología , Células Cultivadas , Células Epiteliales/microbiología , Humanos
5.
PLoS One ; 10(6): e0131831, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26121242

RESUMEN

Mycoplasma pneumoniae is a cell wall-less bacterial pathogen of the human respiratory tract that accounts for > 20% of all community-acquired pneumonia (CAP). At present the most effective means for detection and strain-typing is quantitative polymerase chain reaction (qPCR), which can exhibit excellent sensitivity and specificity but requires separate tests for detection and genotyping, lacks standardization between available tests and between labs, and has limited practicality for widespread, point-of-care use. We have developed and previously described a silver nanorod array-surface enhanced Raman Spectroscopy (NA-SERS) biosensing platform capable of detecting M. pneumoniae with statistically significant specificity and sensitivity in simulated and true clinical throat swab samples, and the ability to distinguish between reference strains of the two main genotypes of M. pneumoniae. Furthermore, we have established a qualitative lower endpoint of detection for NA-SERS of < 1 genome equivalent (cell/µl) and a quantitative multivariate detection limit of 5.3 ± 1 cells/µl. Here we demonstrate using partial least squares- discriminatory analysis (PLS-DA) of sample spectra that NA-SERS correctly identified M. pneumoniae clinical isolates from globally diverse origins and distinguished these from a panel of 12 other human commensal and pathogenic mycoplasma species with 100% cross-validated statistical accuracy. Furthermore, PLS-DA correctly classified by strain type all 30 clinical isolates with 96% cross-validated accuracy for type 1 strains, 98% cross-validated accuracy for type 2 strains, and 90% cross-validated accuracy for type 2V strains.


Asunto(s)
Mycoplasma pneumoniae/patogenicidad , Nanotubos , Neumonía por Mycoplasma/microbiología , Espectrometría Raman/métodos , Humanos , Límite de Detección , Mycoplasma pneumoniae/genética , Sensibilidad y Especificidad
6.
Analyst ; 139(24): 6426-34, 2014 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-25335653

RESUMEN

Mycoplasma pneumoniae is a cell wall-less bacterial pathogen of the human respiratory tract that accounts for up to 20% of community-acquired pneumonia. At present, the standard for detection and genotyping is quantitative polymerase chain reaction (qPCR), which can exhibit excellent sensitivity but lacks standardization and has limited practicality for widespread, point-of-care use. We previously described a Ag nanorod array-surface enhanced Raman spectroscopy (NA-SERS) biosensing platform capable of detecting M. pneumoniae in simulated and true clinical throat swab samples with statistically significant specificity and sensitivity. We report here that differences in sample preparation influence the integrity of mycoplasma cells for NA-SERS analysis, which in turn impacts the resulting spectra. We have established a multivariate detection limit (MDL) using NA-SERS for M. pneumoniae intact-cell sample preparations. Using an adaptation of International Union of Pure and Applied Chemistry (IUPAC)-recommended methods for analyzing multivariate data sets, we found that qPCR had roughly 10× better detection limits than NA-SERS when expressed in CFU ml(-1) and DNA concentration (fg). However, the NA-SERS MDL for intact M. pneumoniae was 5.3 ± 1.0 genome equivalents (cells per µl). By comparison, qPCR of a parallel set of samples yielded a limit of detection of 2.5 ± 0.25 cells per µl. Therefore, for certain standard metrics NA-SERS provides a multivariate detection limit for M. pneumoniae that is essentially identical to that determined via qPCR.


Asunto(s)
Mycoplasma pneumoniae/aislamiento & purificación , Nanotubos/química , Neumonía por Mycoplasma/diagnóstico , Espectrometría Raman/métodos , ADN Bacteriano/análisis , ADN Bacteriano/genética , Genotipo , Humanos , Límite de Detección , Mycoplasma pneumoniae/genética , Reacción en Cadena de la Polimerasa
7.
Analyst ; 139(17): 4287-95, 2014 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-25017005

RESUMEN

Mycoplasma pneumoniae is a major cause of respiratory disease in humans and accounts for as much as 20% of all community-acquired pneumonia. Existing mycoplasma diagnosis is primarily limited by the poor success rate at culturing the bacteria from clinical samples. There is a critical need to develop a new platform for mycoplasma detection that has high sensitivity, specificity, and expediency. Here we report the layer-by-layer (LBL) encapsulation of M. pneumoniae cells with Ag nanoparticles in a matrix of the polyelectrolytes poly(allylamine hydrochloride) (PAH) and poly(styrene sulfonate) (PSS). We evaluated nanoparticle encapsulated mycoplasma cells as a platform for the differentiation of M. pneumoniae strains using surface enhanced Raman scattering (SERS) combined with multivariate statistical analysis. Three separate M. pneumoniae strains (M129, FH and II-3) were studied. Scanning electron microscopy and fluorescence imaging showed that the Ag nanoparticles were incorporated between the oppositely charged polyelectrolyte layers. SERS spectra showed that LBL encapsulation provides excellent spectral reproducibility. Multivariate statistical analysis of the Raman spectra differentiated the three M. pneumoniae strains with 97-100% specificity and sensitivity, and low (0.1-0.4) root mean square error. These results indicated that nanoparticle and polyelectrolyte encapsulation of M. pneumoniae is a potentially powerful platform for rapid and sensitive SERS-based bacterial identification.


Asunto(s)
Mycoplasma pneumoniae/aislamiento & purificación , Neumonía por Mycoplasma/microbiología , Poliaminas/química , Poliestirenos/química , Espectrometría Raman/métodos , Células Inmovilizadas/química , Humanos , Nanopartículas del Metal/química , Mycoplasma pneumoniae/química , Mycoplasma pneumoniae/clasificación , Neumonía por Mycoplasma/diagnóstico , Reproducibilidad de los Resultados , Plata/química
8.
J Bacteriol ; 194(11): 3000-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22544269

RESUMEN

The cell wall-less prokaryote Mycoplasma pneumoniae is a major cause of community-acquired bronchitis and pneumonia in humans. Colonization is mediated largely by a differentiated terminal organelle, which is also the leading end in gliding motility. Cytadherence-associated proteins P30 and P65 appear to traffic concurrently to the distal end of developing terminal organelles. Here, truncation of P65 due to transposon insertion in the corresponding gene resulted in lower gliding velocity, reduced cytadherence, and decreased steady-state levels of several terminal organelle proteins, including P30. Utilizing fluorescent protein fusions, we followed terminal organelle development over time. New P30 foci appeared at nascent terminal organelles in P65 mutants, as in the wild type. However, with forward cell motility, P30 in the P65 mutants appeared to drag toward the trailing cell pole, where it was released, yielding a fluorescent trail to which truncated P65 colocalized. In contrast, P30 was only rarely observed at the trailing end of gliding wild-type cells. Complementation with the recombinant wild-type P65 allele by transposon delivery restored P65 levels and stabilized P30 localization to the terminal organelle.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Mycoplasma pneumoniae/citología , Mycoplasma pneumoniae/metabolismo , Eliminación de Secuencia , Adhesinas Bacterianas/genética , Secuencias de Aminoácidos , Proteínas Bacterianas/genética , Mutagénesis Insercional , Mycoplasma pneumoniae/química , Mycoplasma pneumoniae/genética , Orgánulos/metabolismo , Transporte de Proteínas
9.
J Bacteriol ; 193(20): 5841-6, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21821772

RESUMEN

The cell wall-less prokaryote Mycoplasma pneumoniae causes bronchitis and atypical pneumonia in humans. Mycoplasma attachment to the host respiratory epithelium is required for colonization and mediated largely by a differentiated terminal organelle. P30 is an integral membrane protein located at the distal end of the terminal organelle. The P30 null mutant II-3 is unable to attach to host cells and nonmotile and has a branched cellular morphology compared to the wild type, indicating an important role for P30 in M. pneumoniae biology. P30 is predicted to have an N-terminal signal sequence, but the presence of such a motif has not been confirmed experimentally. In the current study we analyzed P30 derivatives having epitope tags engineered at various locations to demonstrate that posttranslational processing occurred in P30. Several potential cleavage sites predicted in silico were examined, and a processing-defective mutant was created to explore P30 maturation further. Our results suggested that signal peptide cleavage occurs between residues 52 and 53 to yield mature P30. The processing-defective mutant exhibited reduced gliding velocity and cytadherence, indicating that processing is required for fully functional maturation of P30. We speculate that P30 processing may trigger a conformational change in the extracellular domain or expose a binding site on the cytoplasmic domain to allow interaction with a binding partner as a part of functional maturation.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Mycoplasma pneumoniae/fisiología , Neumonía por Mycoplasma/microbiología , Procesamiento Proteico-Postraduccional , Adhesinas Bacterianas/química , Adhesinas Bacterianas/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Línea Celular , Humanos , Datos de Secuencia Molecular , Mycoplasma pneumoniae/química , Mycoplasma pneumoniae/genética , Señales de Clasificación de Proteína
10.
J Bacteriol ; 188(17): 6335-45, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16923901

RESUMEN

The wall-less prokaryote Mycoplasma pneumoniae, a common cause of chronic respiratory tract infections in humans, is considered to be among the smallest and simplest known cells capable of self-replication, yet it has a complex architecture with a novel cytoskeleton and a differentiated terminal organelle that function in adherence, cell division, and gliding motility. Recent findings have begun to elucidate the hierarchy of protein interactions required for terminal organelle assembly, but the engineering of its gliding machinery is largely unknown. In the current study, we assessed gliding in cytadherence mutants lacking terminal organelle proteins B, C, P1, and HMW1. Furthermore, we screened over 3,500 M. pneumoniae transposon mutants individually to identify genes associated with gliding but dispensable for cytadherence. Forty-seven transformants having motility defects were characterized further, with transposon insertions mapping to 32 different open reading frames widely distributed throughout the M. pneumoniae genome; 30 of these were dispensable for cytadherence. We confirmed the clonality of selected transformants by Southern blot hybridization and PCR analysis and characterized satellite growth and gliding by microcinematography. For some mutants, satellite growth was absent or developed more slowly than that of the wild type. Others produced lawn-like growth largely devoid of typical microcolonies, while still others had a dull, asymmetrical leading edge or a filamentous appearance of colony spreading. All mutants exhibited substantially reduced gliding velocities and/or frequencies. These findings significantly expand our understanding of the complexity of M. pneumoniae gliding and the identity of possible elements of the gliding machinery, providing a foundation for a detailed analysis of the engineering and regulation of motility in this unusual prokaryote.


Asunto(s)
Genes Bacterianos , Locomoción/genética , Mycoplasma pneumoniae/fisiología , Proteínas Bacterianas/genética , Adhesión Celular/genética , Medios de Cultivo , Elementos Transponibles de ADN/genética , Gelatina , Vidrio , Mutagénesis , Mycoplasma pneumoniae/genética , Mycoplasma pneumoniae/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...