Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 10(2): e1003951, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24586160

RESUMEN

The tremendous success of S. aureus as a human pathogen has been explained primarily by its array of virulence factors that enable the organism to evade host immunity. Perhaps equally important, but less well understood, is the importance of the intensity of the host response in determining the extent of pathology induced by S. aureus infection, particularly in the pathogenesis of pneumonia. We compared the pathogenesis of infection caused by two phylogenetically and epidemiologically distinct strains of S. aureus whose behavior in humans has been well characterized. Induction of the type I IFN cascade by strain 502A, due to a NOD2-IRF5 pathway, was the major factor in causing severe pneumonia and death in a murine model of pneumonia and was associated with autolysis and release of peptidogylcan. In contrast to USA300, 502A was readily eliminated from epithelial surfaces in vitro. Nonetheless, 502A caused significantly increased tissue damage due to the organisms that were able to invade systemically and trigger type I IFN responses, and this was ameliorated in Ifnar⁻/⁻ mice. The success of USA300 to cause invasive infection appears to depend upon its resistance to eradication from epithelial surfaces, but not production of specific toxins. Our studies illustrate the important and highly variable role of type I IFN signaling within a species and suggest that targeted immunomodulation of specific innate immune signaling cascades may be useful to prevent the excessive morbidity associated with S. aureus pneumonia.


Asunto(s)
Interferón Tipo I/inmunología , Transducción de Señal/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/patogenicidad , Animales , Modelos Animales de Enfermedad , Immunoblotting , Interferón Tipo I/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/inmunología , Virulencia
2.
J Infect Dis ; 205(10): 1571-9, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22457275

RESUMEN

The USA300 strains of Staphylococcus aureus are the major cause of skin and soft tissue infection in the United States. Invasive USA300 infection has been attributed to several virulence factors, including protein A and the α-hemolysin (Hla), which cause pathology by activating host signaling cascades. Here we show that S. aureus exploits the proinflammatory bias of human keratinocytes to activate pyroptosis, a caspase 1-dependent form of inflammatory cell death, which was required for staphylococci to penetrate across a keratinocyte barrier. Keratinocyte necrosis was mediated by calpains, Ca(2+)-dependent intracellular proteases whose endogenous inhibitor, calpastatin, is targeted by Hla-induced caspase 1. Neither Panton-Valentine leukocidin nor protein A expression was essential, but inhibition of either calpain or caspase 1 activity was sufficient to prevent staphylococcal invasion across the keratinocytes. These studies suggest that pharmacological interruption of specific keratinocyte signaling cascades as well as targeting the Hla might prevent invasive skin infection by staphylococci.


Asunto(s)
Calpaína/metabolismo , Caspasa 1/metabolismo , Queratinocitos/microbiología , Staphylococcus aureus Resistente a Meticilina/fisiología , Transducción de Señal/fisiología , Infecciones Estafilocócicas/microbiología , Apoptosis/fisiología , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/farmacología , Calpaína/antagonistas & inhibidores , Inhibidores de Caspasas , Inhibidores de Cisteína Proteinasa/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Dipéptidos/farmacología , Activación Enzimática , Humanos , Queratinocitos/enzimología , Queratinocitos/patología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Mutación , Infecciones de los Tejidos Blandos/microbiología , Infecciones de los Tejidos Blandos/prevención & control , Infecciones Estafilocócicas/prevención & control , Infecciones Cutáneas Estafilocócicas/microbiología , Factores de Virulencia
3.
J Biol Chem ; 286(41): 35891-35898, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-21878647

RESUMEN

Staphyococcus aureus and especially the epidemic methicillin-resistant S. aureus strains cause severe necrotizing pneumonia. The mechanisms whereby these organisms invade across the mucosal epithelial barrier to initiate invasive infection are not well understood. Protein A (SpA), a highly conserved and abundant surface protein of S. aureus, activates TNF receptor 1 and EGF receptor (EGFR) signaling cascades that can perturb the cytoskeleton. We demonstrate that wild-type S. aureus, but not spa mutants, invade across polarized airway epithelial cell monolayers via the paracellular junctions. SpA stimulated a RhoA/ROCK/MLC cascade, resulting in the contraction of the cytoskeleton. SpA(+) but not SpA(-) mutants stimulated activation of EGFR and along with subsequent calpain activity cleaved the membrane-spanning junctional proteins occludin and E-cadherin, facilitating staphylococcal transmigration through the cell-cell junctions. Treatment of polarized human airway epithelial monolayers with inhibitors of ROCK, EGFR, MAPKs, or calpain prevented staphylococcal penetration through the monolayers. In vivo, blocking calpain activity impeded bacterial invasion into the lung parenchyma. Thus, S. aureus exploits multiple receptors available on the airway mucosal surface to facilitate invasion across epithelial barriers.


Asunto(s)
Neumonía Estafilocócica/metabolismo , Mucosa Respiratoria/metabolismo , Transducción de Señal , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Cadherinas/genética , Cadherinas/metabolismo , Calpaína/antagonistas & inhibidores , Calpaína/genética , Calpaína/metabolismo , Línea Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Humanos , Uniones Intercelulares/genética , Uniones Intercelulares/metabolismo , Ratones , Mutación , Neumonía Estafilocócica/tratamiento farmacológico , Neumonía Estafilocócica/genética , Inhibidores de Proteasas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Mucosa Respiratoria/microbiología , Proteína Estafilocócica A/genética , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Proteínas de Unión al GTP rho/genética , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/genética
4.
Infect Immun ; 79(5): 1898-904, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21402768

RESUMEN

Staphylococcus aureus causes especially severe pulmonary infection, associated with high morbidity and mortality. In addition to the effects of specific virulence factors, it appears that the intensity of the host proinflammatory response, particularly in the initial stages of infection, contributes substantially to pulmonary damage. We tested the hypothesis that the CD11c(+) leukocytes are important in the host response to pulmonary infection with methicillin-resistant S. aureus (MRSA) USA300. Clodronate-induced depletion of the alveolar macrophage population resulted in increased numbers of dendritic cells (DCs) and CD4(+) cells in bronchoalveolar lavage (BAL) fluid and was associated with significantly increased mortality by 18 h following S. aureus inoculation but had no effect on bacterial load or polymorphonuclear leukocyte (PMN) numbers in the lung. These clodronate-treated mice also had increased expression of interleukin-17A/F (IL-17A/F) and CXCL10 but not of gamma interferon (IFN-γ) or tumor necrosis factor (TNF). Depletion of the dendritic cell population in mice expressing a CD11c-enhanced green fluorescent protein (EGFP)-diphtheria toxin receptor (DTR) transgene was associated with an increased bacterial load in the lung but not increased mortality. Both DCs and airway epithelial cells produced CXCL9, -10, and -11 in response to S. aureus. Pretreatment of mice with an anti-CXCR3 antibody prior to inoculation with MRSA substantially reduced CD4(+) cells and decreased pulmonary inflammation at 18 h postinfection compared to pretreatment with an IgG control. The results of these experiments suggest that CD11c(+) cells, the induction of CXCR3 ligand expression, and subsequent CD4(+) cell recruitment have an important role in the pathogenesis of severe MRSA pulmonary infection.


Asunto(s)
Antígeno CD11c/inmunología , Linfocitos T CD4-Positivos/inmunología , Leucocitos/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , Infecciones del Sistema Respiratorio/inmunología , Infecciones Estafilocócicas/inmunología , Animales , Antígeno CD11c/metabolismo , Separación Celular , Citocinas/biosíntesis , Citocinas/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Leucocitos/metabolismo , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores CXCR3/inmunología , Receptores CXCR3/metabolismo , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones Estafilocócicas/microbiología
5.
Nat Med ; 10(8): 842-8, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15247912

RESUMEN

Staphylococcus aureus is a major human pathogen that is associated with diverse types of local and systemic infection characterized by inflammation dominated by polymorphonuclear leukocytes. Staphylococci frequently cause pneumonia, and these clinical isolates often have increased expression of protein A, suggesting that this protein may have a role in virulence. Here we show that TNFR1, a receptor for tumor-necrosis factor-alpha (TNF-alpha) that is widely distributed on the airway epithelium, is a receptor for protein A. We also show that the protein A-TNFR1 signaling pathway has a central role in the pathogenesis of staphylococcal pneumonia.


Asunto(s)
Pulmón/patología , Neumonía Estafilocócica/fisiopatología , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/fisiología , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/metabolismo , Animales , Western Blotting , Línea Celular , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunohistoquímica , Interleucina-8/inmunología , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Neutrófilos/inmunología , Neumonía Estafilocócica/metabolismo , Mucosa Respiratoria/metabolismo , Staphylococcus aureus/patogenicidad
6.
Optom Vis Sci ; 87(12): E948-57, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21076353

RESUMEN

PURPOSE: To investigate evidence-based visual field size criteria for referral of low-vision (LV) patients for mobility rehabilitation. METHODS: One hundred and nine participants with LV and 41 age-matched participants with normal sight (NS) were recruited. The LV group was heterogeneous with diverse causes of visual impairment. We measured binocular kinetic visual fields with the Humphrey Field Analyzer and mobility performance on an obstacle-rich, indoor course. Mobility was assessed as percent preferred walking speed (PPWS) and number of obstacle-contact errors. The weighted kappa coefficient of association (κr) was used to discriminate LV participants with both unsafe and inefficient mobility from those with adequate mobility on the basis of their visual field size for the full sample and for subgroups according to type of visual field loss and whether or not the participants had previously received orientation and mobility training. RESULTS: LV participants with both PPWS <38% and errors >6 on our course were classified as having inadequate (inefficient and unsafe) mobility compared with NS participants. Mobility appeared to be first compromised when the visual field was less than about 1.2 steradians (sr; solid angle of a circular visual field of about 70° diameter). Visual fields <0.23 and 0.63 sr (31 to 52° diameter) discriminated patients with at-risk mobility for the full sample and across the two subgroups. A visual field of 0.05 sr (15° diameter) discriminated those with critical mobility. CONCLUSIONS: Our study suggests that: practitioners should be alert to potential mobility difficulties when the visual field is less than about 1.2 sr (70° diameter); assessment for mobility rehabilitation may be warranted when the visual field is constricted to about 0.23 to 0.63 sr (31 to 52° diameter) depending on the nature of their visual field loss and previous history (at risk); and mobility rehabilitation should be conducted before the visual field is constricted to 0.05 sr (15° diameter; critical).


Asunto(s)
Derivación y Consulta , Baja Visión/fisiopatología , Baja Visión/rehabilitación , Campos Visuales , Caminata , Adulto , Anciano , Anciano de 80 o más Años , Medicina Basada en la Evidencia/métodos , Humanos , Persona de Mediana Edad , Orientación , Prioridad del Paciente , Valor Predictivo de las Pruebas , Seguridad , Factores de Tiempo , Resultado del Tratamiento , Baja Visión/psicología
7.
Infect Immun ; 77(9): 3722-30, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19564377

RESUMEN

Streptococcus pneumoniae remains a major cause of bacteremia, pneumonia, and otitis media despite vaccines and effective antibiotics. The neuraminidase of S. pneumoniae, which catalyzes the release of terminal sialic acid residues from glycoconjugates, is involved in host colonization in animal models of infection and may provide a novel target for preventing pneumococcal infection. We demonstrate that the S. pneumoniae neuraminidase (NanA) cleaves sialic acid and show that it is involved in biofilm formation, suggesting an additional role in pathogenesis, and that it shares this property with the neuraminidase of Pseudomonas aeruginosa even though we show that the two enzymes are phylogenetically divergent. Using an in vitro model of biofilm formation incorporating human airway epithelial cells, we demonstrate that small-molecule inhibitors of NanA block biofilm formation and may provide a novel target for preventative therapy. This work highlights the role played by the neuraminidase in pathogenesis and represents an important step in drug development for prevention of colonization of the respiratory tract by this important pathogen.


Asunto(s)
Proteínas Bacterianas/fisiología , Biopelículas/crecimiento & desarrollo , Neuraminidasa/fisiología , Streptococcus pneumoniae/fisiología , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Humanos , Ácido N-Acetilneuramínico/farmacología , Neuraminidasa/antagonistas & inhibidores , Filogenia , Streptococcus pneumoniae/enzimología
8.
J Clin Invest ; 116(8): 2297-2305, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16862214

RESUMEN

Many respiratory pathogens, including Hemophilus influenzae, Streptococcus pneumoniae, and Pseudomonas aeruginosa, express neuraminidases that can cleave alpha2,3-linked sialic acids from glycoconjugates. As mucosal surfaces are heavily sialylated, neuraminidases have been thought to modify epithelial cells by exposing potential bacterial receptors. However, in contrast to neuraminidase produced by the influenza virus, a role for bacterial neuraminidase in pathogenesis has not yet been clearly established. We constructed a mutant of P. aeruginosa PAO1 by deleting the PA2794 neuraminidase locus (Delta2794) and tested its virulence and immunostimulatory capabilities in a mouse model of infection. Although fully virulent when introduced i.p., the Delta2794 mutant was unable to establish respiratory infection by i.n. inoculation. The inability to colonize the respiratory tract correlated with diminished production of biofilm, as assessed by scanning electron microscopy and in vitro assays. The importance of neuraminidase in biofilm production was further demonstrated by showing that viral neuraminidase inhibitors in clinical use blocked P. aeruginosa biofilm production in vitro as well. The P. aeruginosa neuraminidase has a key role in the initial stages of pulmonary infection by targeting bacterial glycoconjugates and contributing to the formation of biofilm. Inhibiting bacterial neuraminidases could provide a novel mechanism to prevent bacterial pneumonia.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Haemophilus influenzae/enzimología , Mucosa Intestinal/microbiología , Neuraminidasa/farmacología , Pseudomonas aeruginosa/enzimología , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/farmacología , Biopelículas/efectos de los fármacos , Modelos Animales de Enfermedad , Eliminación de Gen , Haemophilus influenzae/genética , Mucosa Intestinal/crecimiento & desarrollo , Ratones , Datos de Secuencia Molecular , Neuraminidasa/metabolismo , Fagocitosis , Neumonía Bacteriana/patología
9.
J Bacteriol ; 190(8): 2814-21, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18165298

RESUMEN

The type III secreted toxins of Pseudomonas aeruginosa are important virulence factors associated with clinically important infection. However, their effects on bacterial invasion across mucosal surfaces have not been well characterized. One of the most commonly expressed toxins, ExoS, has two domains that are predicted to affect cytoskeletal integrity, including a GTPase-activating protein (GAP) domain, which targets Rho, a major regulator of actin polymerization; and an ADP-ribosylating domain that affects the ERM proteins, which link the plasma membrane to the actin cytoskeleton. The activities of these toxins, and ExoS specifically, on the permeability properties of polarized airway epithelial cells with intact tight junctions were examined. Strains expressing type III toxins altered the distribution of the tight junction proteins ZO-1 and occludin and were able to transmigrate across polarized airway epithelial monolayers, in contrast to DeltaSTY mutants. These effects on epithelial permeability were associated with the ADP-ribosylating domain of ExoS, as bacteria expressing plasmids lacking expression of the ExoS GAP activity nonetheless increased the permeation of fluorescent dextrans, as well as bacteria, across polarized airway epithelial cells. Treatment of epithelial cells with cytochalasin D depolymerized actin filaments and increased permeation across the monolayers but did not eliminate the differential effects of wild-type and toxin-negative mutants on the epithelial cells, suggesting that additional epithelial targets are involved. Confocal imaging studies demonstrated that ZO-1, occludin, and ezrin undergo substantial redistribution in human airway cells intoxicated by ExoS, -T, and -Y. These studies support the hypothesis that type III toxins enhance P. aeruginosa's invasive capabilities by interacting with multiple eukaryotic cytoskeletal components.


Asunto(s)
ADP Ribosa Transferasas/toxicidad , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/toxicidad , Células Epiteliales/microbiología , Proteínas Activadoras de GTPasa/toxicidad , Glucosiltransferasas/toxicidad , Pseudomonas aeruginosa/patogenicidad , Uniones Estrechas/fisiología , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/genética , Proteínas Bacterianas/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Línea Celular , Proteínas del Citoesqueleto/análisis , Células Epiteliales/química , Células Epiteliales/efectos de los fármacos , Eliminación de Gen , Humanos , Proteínas de la Membrana/análisis , Ocludina , Permeabilidad , Fosfoproteínas/análisis , Estructura Terciaria de Proteína , Pseudomonas aeruginosa/crecimiento & desarrollo , Proteína de la Zonula Occludens-1
10.
Retina ; 28(8): 1111-6, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18779718

RESUMEN

PURPOSE: To examine if vision in subjects with macular heterotopia (MH) secondary to retinopathy of prematurity (ROP) is related to anatomical macular structure. METHODS: Six subjects with MH who were between 18 years and 65 years of age and three age-matched subjects with normal vision were recruited for the study. Vision and macular structure of the better eye of the subjects with MH and the dominant eye of age-matched subjects with normal vision were assessed. High contrast visual acuity and contrast sensitivity were measured using Early Treatment of Diabetic Retinopathy Study and Pelli-Robson charts, respectively. The Micro Perimeter (Nidek Technologies MP-1) was used to assess macular sensitivity and fixation stability. Using optical coherence tomography, macular thickness and relative retinal thickness at fixation were measured. RESULTS: Subjects with MH had significantly reduced visual acuity and macular sensitivity compared with age-matched subjects with normal vision. In comparison with their age-matched counterparts, subjects with MH had significantly increased macular thickness and increased relative retinal thickness at fixation. A normal foveal architecture was absent in three subjects with MH (50%). CONCLUSION: Patients with MH secondary to ROP have increased macular thickness and reduced vision.


Asunto(s)
Coristoma/complicaciones , Coristoma/patología , Mácula Lútea/patología , Enfermedades de la Retina/complicaciones , Enfermedades de la Retina/patología , Retinopatía de la Prematuridad/complicaciones , Baja Visión/etiología , Adolescente , Adulto , Anciano , Coristoma/etiología , Coristoma/fisiopatología , Fóvea Central/patología , Humanos , Recién Nacido , Mácula Lútea/fisiopatología , Persona de Mediana Edad , Enfermedades de la Retina/etiología , Enfermedades de la Retina/fisiopatología , Baja Visión/fisiopatología , Agudeza Visual , Adulto Joven
11.
J Clin Invest ; 113(10): 1482-9, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15146246

RESUMEN

Toll-like receptors (TLRs) mediate host responses to bacterial gene products. As the airway epithelium is potentially exposed to many diverse inhaled bacteria, TLRs involved in defense of the airways must be broadly responsive, available at the exposed apical surface of the cells, and highly regulated to prevent activation following trivial encounters with bacteria. We demonstrate that TLR2 is enriched in caveolin-1-associated lipid raft microdomains presented on the apical surface of airway epithelial cells after bacterial infection. These receptor complexes include myeloid differentiation protein (MyD88), interleukin-1 receptor-activated kinase-1, and TNF receptor-associated factor 6. The signaling capabilities of TLR2 are amplified through its association with the asialoganglioside gangliotetraosylceramide (Gal beta 1,2GalNAc beta 1,4Gal beta 1,4Glc beta 1,1Cer), which has receptor function itself for many pulmonary pathogens. Ligation of either TLR2 or asialoGM1 by ligands with specificity for either receptor, by Pseudomonas aeruginosa, or by Staphylococcus aureus stimulates IL-8 production through activation of NF-kappa B, as mediated by TLR2 and MyD88. Thus, TLR2 in association with asialo-glycolipids presented within the context of lipid rafts provides a broadly responsive signaling complex at the apical surfaces of airway cells to initiate the host response to potential bacterial infection.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Microdominios de Membrana/inmunología , Receptores de Superficie Celular/metabolismo , Sistema Respiratorio/inmunología , Proteínas Adaptadoras Transductoras de Señales , Antígenos de Diferenciación/metabolismo , Secuencia de Carbohidratos , Línea Celular , Células Epiteliales/inmunología , Gangliósidos , Glicoesfingolípidos/química , Glicoesfingolípidos/metabolismo , Humanos , Interleucina-8/biosíntesis , Ligandos , Microdominios de Membrana/metabolismo , Datos de Secuencia Molecular , Factor 88 de Diferenciación Mieloide , Pseudomonas aeruginosa/patogenicidad , Receptores Inmunológicos/metabolismo , Sistema Respiratorio/citología , Transducción de Señal , Staphylococcus aureus/patogenicidad , Receptor Toll-Like 2 , Receptores Toll-Like
12.
Clin Exp Optom ; 90(2): 115-23, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17311573

RESUMEN

BACKGROUND: As vision plays a significant role in mobility performance, it is usual to refer low vision patients, particularly those who complain of mobility difficulties, for orientation and mobility (O&M) training to help them maintain safe independent travel. Our study aimed to determine whether there was a relationship between measures of vision and self-reported mobility, and the applicability of a patient-based mobility assessment in patients with heterogeneous causes of visual loss. METHOD: We assessed the high and low contrast visual acuity, visual field and scanning ability of 30 patients with low vision. A validated mobility questionnaire was used to assess the participants' perceived visual ability for independent mobility. RESULTS: Vision was significantly correlated with self-reported mobility performance, however, visual field was a significant predictor accounting for 56.5 per cent of the variance. The instrument was well constructed with valid content and high reliability scores. CONCLUSIONS: Self-reported mobility performance together with measures of vision could be used as a guide to refer patients for O&M training. The patient-based assessment instrument was valid to measure perceived visual ability for independent mobility in patients with heterogeneous causes of visual loss.


Asunto(s)
Limitación de la Movilidad , Trastornos de la Visión/fisiopatología , Visión Ocular , Adulto , Anciano , Anciano de 80 o más Años , Sensibilidad de Contraste , Humanos , Persona de Mediana Edad , Encuestas y Cuestionarios , Agudeza Visual , Campos Visuales
13.
Cell Rep ; 18(11): 2742-2751, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28297676

RESUMEN

Human skin is commonly colonized and infected by Staphylococcus aureus. Exactly how these organisms are sensed by keratinocytes has not been clearly delineated. Using a combination of metabolic and transcriptomic methodologies, we found that S. aureus infection is sensed as a metabolic stress by the hypoxic keratinocytes. This induces HIF1α signaling, which promotes IL-1ß production and stimulates aerobic glycolysis to meet the metabolic requirements of infection. We demonstrate that staphylococci capable of glycolysis, including WT and agr mutants, readily induce HIF1α responses. In contrast, Δpyk glycolytic mutants fail to compete with keratinocytes for their metabolic needs. Suppression of glycolysis using 2-DG blocked keratinocyte production of IL-1ß in vitro and significantly exacerbated the S. aureus cutaneous infection in a murine model. Our data suggest that S. aureus impose a metabolic stress on keratinocytes that initiates signaling necessary to promote both glycolysis and the proinflammatory response to infection.


Asunto(s)
Queratinocitos/inmunología , Queratinocitos/microbiología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Estrés Fisiológico , Animales , Línea Celular , Citocinas/metabolismo , Glucólisis , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Queratinocitos/patología , Ratones Endogámicos C57BL , Consumo de Oxígeno , Transducción de Señal , Piel/microbiología , Piel/patología
14.
Cell Rep ; 16(8): 2219-2230, 2016 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-27524612

RESUMEN

Staphylococcus aureus triggers inflammation through inflammasome activation and recruitment of neutrophils, responses that are critical for pathogen clearance but are associated with substantial tissue damage. We postulated that necroptosis, cell death mediated by the RIPK1/RIPK3/MLKL pathway, would function to limit pathological inflammation. In models of skin infection or sepsis, Mlkl-/- mice had high bacterial loads, an inability to limit interleukin-1b (IL-1b) production, and excessive inflammation. Similarly, mice treated with RIPK1 or RIPK3 inhibitors had increased bacterial loads in a model of sepsis. Ripk3-/- mice exhibited increased staphylococcal clearance and decreased inflammation in skin and systemic infection, due to direct effects of RIPK3 on IL-1b activation and apoptosis. In contrast to Casp1/4-/- mice with defective S. aureus killing, the poor outcomes of Mlkl-/- mice could not be attributed to impaired phagocytic function. We conclude that necroptotic cell death limits the pathological inflammation induced by S. aureus.


Asunto(s)
Apoptosis/inmunología , Necrosis/inmunología , Proteínas Quinasas/inmunología , Sepsis/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Carga Bacteriana , Caspasa 1/genética , Caspasa 1/inmunología , Caspasas/genética , Caspasas/inmunología , Caspasas Iniciadoras , Línea Celular , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inflamasomas/genética , Inflamasomas/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Queratinocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas/deficiencia , Proteínas Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Sepsis/genética , Sepsis/mortalidad , Sepsis/patología , Transducción de Señal , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/patología , Infecciones Cutáneas Estafilocócicas/genética , Infecciones Cutáneas Estafilocócicas/patología , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/patogenicidad , Análisis de Supervivencia
15.
Chest ; 128(2): 912-9, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16100186

RESUMEN

There is considerable interest in the use of azithromycin for the treatment of lung disease in patients with cystic fibrosis (CF). Although its mechanism of action as an inhibitor of bacterial protein synthesis has been well-established, it is less clear how azithromycin ameliorates the lung disease associated with Pseudomonas aeruginosa, which is considered to be resistant to the drug. We tested the effects of azithromycin on clinical isolates (CIs) from CF patients and compared them with laboratory reference strains to establish how this drug might interfere with the production of bacterial virulence factors that are relevant to the pathogenesis of airway disease in CF patients. Azithromycin inhibited P aeruginosa PAO1 protein synthesis by 80%, inhibiting bacterial growth and the expression of immunostimulatory exoproducts such as pyocyanin, as well as the gene products necessary for biofilm formation. In contrast, the effects of azithromycin on CIs of P aeruginosa were much more variable, due in large part to their slow growth and limited exoproduct expression. Culture supernatants for two of three clinical strains induced appreciable CXCL8 expression from cultured epithelial cells. Azithromycin treatment of the organisms inhibited 65 to 70% of this induction; azithromycin had no direct effect on the ability of either normal cells or CF epithelial cells to produce CXCL8. Azithromycin does decrease the P aeruginosa synthesis of immunostimulatory exoproducts and is likely to be most effective against planktonic, actively growing bacteria. This effect is less predictable against CIs than the prototypic strain PAO1.


Asunto(s)
Antibacterianos/farmacología , Azitromicina/farmacología , Fibrosis Quística/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/crecimiento & desarrollo , Humanos , Pseudomonas aeruginosa/aislamiento & purificación
16.
mBio ; 6(2)2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25900653

RESUMEN

UNLABELLED: Skin is the most common site of Staphylococcus aureus infection. While most of these infections are self-limited, recurrent infections are common. Keratinocytes and recruited immune cells participate in skin defense against infection. We postulated that S. aureus is able to adapt to the milieu within human keratinocytes to avoid keratinocyte-mediated clearance. From a collection of S. aureus isolated from chronically infected patients with atopic dermatitis, we noted 22% had an agr mutant-like phenotype. Using several models of human skin infection, we demonstrate that toxin-deficient, agr mutants of methicillin-resistant S. aureus (MRSA) USA300 are able to persist within keratinocytes by stimulating autophagy and evading caspase-1 and inflammasome activation. MRSA infection induced keratinocyte autophagy, as evidenced by galectin-8 and LC3 accumulation. Autophagy promoted the degradation of inflammasome components and facilitated staphylococcal survival. The recovery of more than 58% agr or RNAIII mutants (P < 0.0001) of an inoculum of wild-type (WT) MRSA from within wortmannin-treated keratinocytes compared to control keratinocytes reflected the survival advantage for mutants no longer expressing agr-dependent toxins. Our results illustrate the dynamic interplay between S. aureus and keratinocytes that can result in the selection of mutants that have adapted specifically to evade keratinocyte-mediated clearance mechanisms. IMPORTANCE: Human skin is a major site of staphylococcal infection, and keratinocytes actively participate in eradication of these pathogens. We demonstrate that methicillin-resistant Staphylococcus aureus (MRSA) is ingested by keratinocytes and activates caspase-1-mediated clearance through pyroptosis. Toxin-deficient MRSA mutants are selected within keratinocytes that fail to induce caspase-1 activity and keratinocyte-mediated clearance. These intracellular staphylococci induce autophagy that enhances their intracellular survival by diminishing inflammasome components. These findings suggest that S. aureus mutants, by exploiting autophagy, can persist within human keratinocytes.


Asunto(s)
Queratinocitos/microbiología , Staphylococcus aureus Resistente a Meticilina/fisiología , Adaptación Biológica , Animales , Autofagia , Proteínas Bacterianas , Toxinas Bacterianas/metabolismo , Humanos , Evasión Inmune , Ratones , Transactivadores/deficiencia
17.
Vision Res ; 44(27): 3193-202, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15482805

RESUMEN

Foveal detection of a Gabor patch (target) is facilitated by collinear, displaced high-contrast flankers. Polat and Sagi reported that the same phenomenon occurred in the periphery, but no data were presented [Proc. Natl. Acad. Sci. 91 (1994) 1206]. Others have found no facilitation in a limited number of conditions tested. To resolve this apparent conflict, we measured lateral facilitation in the near-periphery using a range of stimulus parameters. We found facilitation for a range of target-flanker distances for peripheral eccentricities up to 6 degrees , but the magnitude of the effect was less than found in central vision. Facilitation varied across subjects and with spatial frequency. Flanker contrast had no effect over the range evaluated (10-80%). Equal facilitation was found for two global arrangements of the stimulus pattern. Facilitation was found using a temporal, but not a spatial two-alternative forced-choice paradigm, accounting for the different results among previous studies. This finding supports previous indications of the role of attention in altering such facilitation. The value of facilitation from lateral interactions for persons with central vision impairment, who have to shift their attention to a peripheral locus constantly, needs to be examined.


Asunto(s)
Sensibilidad de Contraste/fisiología , Campos Visuales/fisiología , Adolescente , Adulto , Atención/fisiología , Femenino , Humanos , Masculino , Orientación/fisiología , Reconocimiento Visual de Modelos/fisiología , Estimulación Luminosa/métodos , Psicofísica , Umbral Sensorial
18.
mBio ; 2(3): e00016-11, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21586648

RESUMEN

UNLABELLED: The mucosal epithelium is the initial target for respiratory pathogens of all types. While type I interferon (IFN) signaling is traditionally associated with antiviral immunity, we demonstrate that the extracellular bacterial pathogen Streptococcus pneumoniae activates the type I IFN cascade in airway epithelial and dendritic cells. This response is dependent upon the pore-forming toxin pneumolysin. Pneumococcal DNA activates IFN-ß expression through a DAI/STING/TBK1/IRF3 cascade. Tlr4(-/-), Myd88(-/-), Trif(-/-), and Nod2(-/-) mutant mice had no impairment of type I IFN signaling. Induction of type I IFN signaling contributes to the eradication of pneumococcal carriage, as IFN-α/ß receptor null mice had significantly increased nasal colonization with S. pneumoniae compared with that of wild-type mice. These studies suggest that the type I IFN cascade is a central component of the mucosal response to airway bacterial pathogens and is responsive to bacterial pathogen-associated molecular patterns that are capable of accessing intracellular receptors. IMPORTANCE: The bacterium Streptococcus pneumoniae is a leading cause of bacterial pneumonia, leading to upwards of one million deaths a year worldwide and significant economic burden. Although it is known that antibody is critical for efficient phagocytosis, it is not known how this pathogen is sensed by the mucosal epithelium. We demonstrate that this extracellular pathogen activates mucosal signaling typically activated by viral pathogens via the pneumolysin pore to activate intracellular receptors and the type I interferon (IFN) cascade. Mice lacking the receptor to type I IFNs have a reduced ability to clear S. pneumoniae, suggesting that the type I IFN cascade is central to the mucosal clearance of this important pathogen.


Asunto(s)
ADN Bacteriano/inmunología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Sistema Respiratorio/inmunología , Transducción de Señal , Streptococcus pneumoniae/inmunología , Animales , Portador Sano/inmunología , Portador Sano/microbiología , Células Cultivadas , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucosa Nasal/microbiología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Sistema Respiratorio/microbiología , Streptococcus pneumoniae/genética
19.
J Clin Invest ; 119(7): 1931-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19603548

RESUMEN

The activation of type I IFN signaling is a major component of host defense against viral infection, but it is not typically associated with immune responses to extracellular bacterial pathogens. Using mouse and human airway epithelial cells, we have demonstrated that Staphylococcus aureus activates type I IFN signaling, which contributes to its virulence as a respiratory pathogen. This response was dependent on the expression of protein A and, more specifically, the Xr domain, a short sequence-repeat region encoded by DNA that consists of repeated 24-bp sequences that are the basis of an internationally used epidemiological typing scheme. Protein A was endocytosed by airway epithelial cells and subsequently induced IFN-beta expression, JAK-STAT signaling, and IL-6 production. Mice lacking IFN-alpha/beta receptor 1 (IFNAR-deficient mice), which are incapable of responding to type I IFNs, were substantially protected against lethal S. aureus pneumonia compared with wild-type control mice. The profound immunological consequences of IFN-beta signaling, particularly in the lung, may help to explain the conservation of multiple copies of the Xr domain of protein A in S. aureus strains and the importance of protein A as a virulence factor in the pathogenesis of staphylococcal pneumonia.


Asunto(s)
Interferón Tipo I/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Transducción de Señal , Proteína Estafilocócica A/inmunología , Animales , Humanos , Interferón Tipo I/metabolismo , Ratones , Ratones Endogámicos C57BL , Estructura Terciaria de Proteína , Receptor de Interferón alfa y beta/inmunología , Secuencias Repetitivas de Aminoácido , Factor de Transcripción STAT3/metabolismo , Proteína Estafilocócica A/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA