Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Immunol ; 10: 2169, 2019.
Article in English | MEDLINE | ID: mdl-31608051

ABSTRACT

Ozone exposure causes irritation, airway hyperreactivity (AHR), inflammation of the airways, and destruction of alveoli (emphysema), the gas exchange area of the lung in human and mice. This review focuses on the acute disruption of the respiratory epithelial barrier in mice. A single high dose ozone exposure (1 ppm for 1 h) causes first a break of the bronchiolar epithelium within 2 h with leak of serum proteins in the broncho-alveolar space, disruption of epithelial tight junctions and cell death, which is followed at 6 h by ROS activation, AHR, myeloid cell recruitment, and remodeling. High ROS levels activate a novel PGAM5 phosphatase dependent cell-death pathway, called oxeiptosis. Bronchiolar cell wall damage and inflammation upon a single ozone exposure are reversible. However, chronic ozone exposure leads to progressive and irreversible loss of alveolar epithelial cells and alveoli with reduced gas exchange space known as emphysema. It is further associated with chronic inflammation and fibrosis of the lung, resembling other environmental pollutants and cigarette smoke in pathogenesis of asthma, and chronic obstructive pulmonary disease (COPD). Here, we review recent data on the mechanisms of ozone induced injury on the different cell types and pathways with a focus on the role of the IL-1 family cytokines and the related IL-33. The relation of chronic ozone exposure induced lung disease with asthma and COPD and the fact that ozone exacerbates asthma and COPD is emphasized.


Subject(s)
Blood-Air Barrier/immunology , Ozone/toxicity , Respiratory Mucosa/immunology , Acute Disease , Animals , Asthma/chemically induced , Asthma/immunology , Asthma/pathology , Blood-Air Barrier/pathology , Cigarette Smoking/adverse effects , Cigarette Smoking/immunology , Humans , Mice , Phosphoprotein Phosphatases/immunology , Pneumonia/chemically induced , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Disease, Chronic Obstructive/chemically induced , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/immunology , Pulmonary Emphysema/pathology , Reactive Oxygen Species/immunology , Respiratory Mucosa/pathology , Tight Junctions/immunology , Tight Junctions/pathology
2.
Shock ; 51(2): 235-246, 2019 02.
Article in English | MEDLINE | ID: mdl-29438223

ABSTRACT

As activation of the coagulation system is both a consequence and contributor to acute lung injury (ALI), pulmonary coagulopathy has become a potential target for therapeutic intervention in ALI patients. We investigated the effects and possible mechanisms of endothelial cell (EC)-anchored tissue factor pathway inhibitor (TFPI) on lipopolysaccharide (LPS)-induced ALI in mice. To assess the effect of EC-anchored TFPI deletion on ALI indices, TFPI knockout (cKO) mice were generated. Mice were instilled by direct intratracheal injection LPS for the preparation of an ALI model. Evans blue dye (EBD) was injected intravenously 2 h prior to animal sacrifice (48 h post-LPS). Lungs were fixed for histopathology and the prepared tissue was homogenized or used to extract bronchoalveolar lavage fluid (BALF) or detect EBD concentration. TFPI knockdown mice with ALI were compared to wild-type (WT) mice with ALI to assess the effect of TFPI on endothelial barrier function and inflammation. TFPI deletion markedly exacerbated LPS histopathological changes in lung, and the LPS changes in protein, EBD extravasation, proinflammatory cytokines TNF-α, IL-1ß, and IL-6 in BALF in lung. The number and infiltration of white blood cells (WBCs) from BALF and lung tissue of TFPI cKO mice with LPS-challenged ALI was increased compared to WT mice with LPS-challenged ALI. We also found further increased toll-like receptor 4 and nuclear factor kappa-light-chain-enhancer of activated B cells activation and additional expression of vascular cell adhesion molecule 1 and reduction of angiotensin converting enzyme 2 expression in TFPI cKO+LPS mice compared with WT+LPS mice. Endothelial-specific TFPI deficiency promoted LPS-induced pulmonary inflammation and endothelial barrier permeability possibly via toll-like receptor 4-mediated nuclear factor kappa-light-chain-enhancer of activated B cells signaling pathway activation.


Subject(s)
Acute Lung Injury/immunology , Blood-Air Barrier/immunology , Endothelial Cells/immunology , Lipopolysaccharides/toxicity , Lipoproteins/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Blood-Air Barrier/pathology , Cytokines/genetics , Cytokines/immunology , Endothelial Cells/pathology , Gene Knockout Techniques , Lipoproteins/genetics , Mice , Mice, Transgenic , NF-kappa B/genetics , Signal Transduction/genetics
3.
Sud Med Ekspert ; 60(4): 9-11, 2017.
Article in Russian | MEDLINE | ID: mdl-28766520

ABSTRACT

The acute respiratory viral infections (ARVI) are ranked among the most widespread diseases affecting the children in the early infancy. They account for 60 to 85.4% of all infections recorded in the young children. AIM: The objective of the present study was to elucidate the peculiar features of the accumulation of the effector cells of the local immunity system and intercellular interplay in the broncho-vascular barrier of the breast-fed infants presenting with various ARVIs. MATERIAL AND METHODS: We undertook the analysis of 32 cases of infections caused by influenza A and B viruses and of the adenovirus infection verified by the immunofluorescence assay. The group of comparison was comprised of 10 children presenting with congenial heart disease in the absence of the signs of inflammatory processes in the lungs. The tissue samples were harvested from the upper lobe of the lung at the level of the lobe bronchus and a terminal bronchiola. The materials for the histological study were prepared using the Romanovsky method. The effector cells were examined in the lamina propria of bronchial mucosa and the submucous layer. The morphometric analysis included direct counting the number of lymphocytes, mast cells, macrophages, plasmocytes, eosinophils, and neutrophils with the subsequent recalculation of the data thus obtained per unit volume of the connective tissue. The results of the morphometric analysis were subjected to the statistical treatment. RESULTS: The study has demonstrated that the young children suffering from a viral infection, regardless of the type of the causative factor, experience a change in the total number and the ratio of the effector cells at all the levels of the broncho-vascular barrier.


Subject(s)
Blood-Air Barrier , Heart Defects, Congenital , Immune System/pathology , Respiratory Tract Infections , Virus Diseases , Acute Disease , Blood-Air Barrier/immunology , Blood-Air Barrier/pathology , Cell Communication/immunology , Female , Fluorescent Antibody Technique/methods , Forensic Medicine/methods , Heart Defects, Congenital/immunology , Heart Defects, Congenital/pathology , Humans , Infant , Male , Respiratory Tract Infections/immunology , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Virus Diseases/immunology , Virus Diseases/pathology
6.
J Immunol ; 193(5): 2469-82, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25063875

ABSTRACT

A disintegrin and a metalloproteinase domain (ADAM) 9 is known to be expressed by monocytes and macrophages. In this study, we report that ADAM9 is also a product of human and murine polymorphonuclear neutrophils (PMNs). ADAM9 is not synthesized de novo by circulating PMNs. Rather, ADAM9 protein is stored in the gelatinase and specific granules and the secretory vesicles of human PMNs. Unstimulated PMNs express minimal quantities of surface ADAM9, but activation of PMNs with degranulating agonists rapidly (within 15 min) increases PMN surface ADAM9 levels. Human PMNs produce small quantities of soluble forms of ADAM9. Surprisingly, ADAM9 degrades several extracellular matrix (ECM) proteins, including fibronectin, entactin, laminin, and insoluble elastin, as potently as matrix metalloproteinase-9. However, ADAM9 does not degrade types I, III, or IV collagen or denatured collagens in vitro. To determine whether Adam9 regulates PMN recruitment or ECM protein turnover during inflammatory responses, we compared wild-type and Adam9(-/-) mice in bacterial LPS- and bleomycin-mediated acute lung injury (ALI). Adam9 lung levels increase 10-fold during LPS-mediated ALI in wild-type mice (due to increases in leukocyte-derived Adam9), but Adam9 does not regulate lung PMN (or macrophage) counts during ALI. Adam9 increases mortality, promotes lung injury, reduces lung compliance, and increases degradation of lung elastin during LPS- and/or bleomycin-mediated ALI. Adam9 does not regulate collagen accumulation in the bleomycin-treated lung. Thus, ADAM9 is expressed in an inducible fashion on PMN surfaces where it degrades some ECM proteins, and it promotes alveolar-capillary barrier injury during ALI in mice.


Subject(s)
ADAM Proteins/immunology , Acute Lung Injury/immunology , Extracellular Matrix/immunology , Membrane Proteins/immunology , Neutrophils/immunology , Proteolysis , ADAM Proteins/genetics , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/pharmacology , Bleomycin/adverse effects , Bleomycin/pharmacology , Blood-Air Barrier/immunology , Blood-Air Barrier/pathology , Collagen/genetics , Collagen/immunology , Elastin/genetics , Elastin/immunology , Extracellular Matrix/genetics , Humans , Lipopolysaccharides/toxicity , Membrane Proteins/genetics , Mice , Mice, Knockout , Neutrophils/pathology
7.
Am J Respir Cell Mol Biol ; 51(4): 526-35, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24773184

ABSTRACT

In inhalation therapy, drugs are deposited as aerosols onto the air-facing lung epithelium. The currently used in vitro cell assays for drug testing, however, typically dissolve drugs in the medium, completely covering the cells, which represents an unphysiological drug application scenario. Although physiologically realistic in vitro cell culture models of the pulmonary air-blood barrier are available, reliable, easy-to-handle, and efficient technologies for direct aerosol-to-cell delivery are lacking. Here, we introduce the Air-Liquid Interface (ALI) Cell Exposure-Cloud (ALICE-CLOUD) technology, which uses principles of cloud motion for fast and quantitative delivery of aerosolized liquid drugs to pulmonary cells cultured under realistic ALI conditions. Aerosol-to-cell delivery proved to be highly efficient, reproducible, and rapid when using aerosolized fluorescein as surrogate drug. As a proof-of-concept study for the ALICE-CLOUD, we performed functional efficacy studies with the U.S. Food and Drug Administration-approved proteasome inhibitor, Bortezomib, a novel candidate drug for inhalation therapy. Aerosolized Bortezomib had a pronounced anti-inflammatory effect on human epithelial lung cells (A549), as indicated by a significant reduction of (TNFα-induced) IL-8 promoter activation. Importantly, cell-based therapeutic efficacy of aerosolized Bortezomib under ALI conditions was similar to that under dissolved and nonaerosolized submerged conditions, but with faster uptake kinetics. Our data indicate that the ALICE-CLOUD is a reliable tool for aerosolized drug screening with cells cultured under ALI conditions, which combines ease of handling with rapid, efficient, and dosimetrically accurate drug-to-cell delivery. This may pave the way for screening of inhalable drugs under physiologically more relevant and, hence, potentially more predictive conditions than the currently used submerged cell culture systems.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Blood-Air Barrier/drug effects , Boronic Acids/administration & dosage , Epithelial Cells/drug effects , Proteasome Inhibitors/administration & dosage , Pyrazines/administration & dosage , Respiratory Mucosa/drug effects , Administration, Inhalation , Aerosols , Anti-Inflammatory Agents/metabolism , Blood-Air Barrier/immunology , Blood-Air Barrier/metabolism , Boronic Acids/metabolism , Bortezomib , Cell Culture Techniques , Cell Line, Tumor , Dose-Response Relationship, Drug , Epithelial Cells/immunology , Epithelial Cells/metabolism , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Kinetics , Promoter Regions, Genetic , Proteasome Inhibitors/metabolism , Pyrazines/metabolism , Reproducibility of Results , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
8.
Eur J Immunol ; 43(12): 3125-37, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24165907

ABSTRACT

Chronic asthma is an inflammatory disease of the airway wall that leads to bronchial smooth muscle hyperreactivity and airway obstruction, caused by inflammation, goblet cell metaplasia, and airway wall remodeling. In response to allergen presentation by airway DCs, T-helper lymphocytes of the adaptive immune system control many aspects of the disease through secretion of IL-4, IL-5, IL-13, IL-17, and IL-22, and these are counterbalanced by cytokines produced by Treg cells. Many cells of the innate immune system such as mast cells, basophils, neutrophils, eosinophils, and innate lymphoid cells also play an important role in disease pathogenesis. Barrier epithelial cells are being ever more implicated in disease pathogenesis than previously thought, as these cells have in recent years been shown to sense exposure to allergens via pattern recognition receptors and to activate conventional and inflammatory-type DCs and other innate immune cells through the secretion of thymic stromal lymphopoietin, granulocyte-macrophage colony stimulating factor, IL-1, IL-33, and IL-25. Understanding this cytokine crosstalk between barrier epithelial cells, DCs, and immune cells provides important insights into the mechanisms of allergic sensitization and asthma progression as discussed in this review.


Subject(s)
Allergens/immunology , Antigen Presentation , Asthma/immunology , Blood-Air Barrier/immunology , Dendritic Cells/immunology , Goblet Cells/immunology , Animals , Asthma/pathology , Blood-Air Barrier/pathology , Cytokines/immunology , Dendritic Cells/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Goblet Cells/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Mast Cells/immunology , Mast Cells/pathology , Metaplasia , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
9.
J Immunol ; 189(10): 4960-9, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23045615

ABSTRACT

A feature shared by many inflammatory lung diseases is excessive neutrophilic infiltration. Neutrophil homing to airspaces involve multiple factors produced by several distinct cell types. Hepoxilin A(3) is a neutrophil chemoattractant produced by pathogen-infected epithelial cells that is hypothesized to facilitate neutrophil breach of mucosal barriers. Using a Transwell model of lung epithelial barriers infected with Pseudomonas aeruginosa, we explored the role of hepoxilin A(3) in neutrophil transepithelial migration. Pharmacological inhibitors of the enzymatic pathways necessary to generate hepoxilin A(3), including phospholipase A(2) and 12-lipoxygenase, potently interfere with P. aeruginosa-induced neutrophil transepithelial migration. Both transformed and primary human lung epithelial cells infected with P. aeruginosa generate hepoxilin A(3) precursor arachidonic acid. All four known lipoxygenase enzymes capable of synthesizing hepoxilin A(3) are expressed in lung epithelial cell lines, primary small airway epithelial cells, and human bronchial epithelial cells. Lung epithelial cells produce increased hepoxilin A(3) and lipid-derived neutrophil chemotactic activity in response to P. aeruginosa infection. Lipid-derived chemotactic activity is soluble epoxide hydrolase sensitive, consistent with hepoxilin A(3) serving a chemotactic role. Stable inhibitory structural analogs of hepoxilin A(3) are capable of impeding P. aeruginosa-induced neutrophil transepithelial migration. Finally, intranasal infection of mice with P. aeruginosa promotes enhanced cellular infiltrate into the airspace, as well as increased concentration of the 12-lipoxygenase metabolites hepoxilin A(3) and 12-hydroxyeicosa-5Z,8Z,10E,14Z-tetraenoic acid. Data generated from multiple models in this study provide further evidence that hepoxilin A(3) is produced in response to lung pathogenic bacteria and functions to drive neutrophils across epithelial barriers.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Arachidonate 12-Lipoxygenase/immunology , Blood-Air Barrier/immunology , Neutrophils/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Transendothelial and Transepithelial Migration/immunology , 8,11,14-Eicosatrienoic Acid/immunology , 8,11,14-Eicosatrienoic Acid/metabolism , Animals , Arachidonate 12-Lipoxygenase/metabolism , Blood-Air Barrier/metabolism , Blood-Air Barrier/microbiology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Humans , Male , Mice , Neutrophils/metabolism , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/metabolism , Pseudomonas Infections/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology
10.
Toxicol Appl Pharmacol ; 245(3): 361-9, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20399800

ABSTRACT

Cadmium (Cd(2+)) is a widespread environmental pollutant, which is associated with a wide variety of cytotoxic and metabolic effects. Recent studies showed that intoxication with the heavy metal most importantly targets the integrity of the epithelial barrier. In our study, the lung epithelial cell line, NCI H441, was cultured with the endothelial cell line, ISO-HAS-1, as a bilayer on a 24-well HTS-Transwell filter plate. This coculture model was exposed to various concentrations of CdCl(2). The transepithelial electrical resistance decreased on the apical side only after treatment with high Cd(2+) concentrations after 48 h. By contrast, a breakdown of TER to less than 5% of baseline could be observed much earlier (after 24 h) when Cd(2+) was administered from the basal side. Observations of cell layer fragmentation and widening of intercellular spaces confirmed the barrier breakdown only for the basolaterally treated samples. Furthermore, the cytotoxicity and release of proinflammatory markers was enhanced if samples were exposed to Cd(2+) from the basal side compared to treatment from the apical side. Moreover, we could demonstrate that a high concentration of Ca(2+) could prevent the barrier-disrupting effect of Cd(2+). In conclusion, the exposure of Cd(2+) to cocultures of lung cells caused a decrease in TER, major morphological changes, a reduction of cell viability and an increase of cytokine release, but the effects markedly differed between the two modes of exposure. Therefore, our results suggest that intact epithelial TJs may play a major role in protecting the air-blood barrier from inhaled Cd(2+).


Subject(s)
Blood-Air Barrier/drug effects , Cadmium Chloride/toxicity , Cell Polarity , Endothelial Cells/drug effects , Epithelial Cells/drug effects , Adherens Junctions/drug effects , Adherens Junctions/pathology , Blood-Air Barrier/immunology , Blood-Air Barrier/pathology , Calcium/metabolism , Cell Line, Tumor , Cell Shape/drug effects , Cell Survival/drug effects , Coculture Techniques , Cytokines/metabolism , Cytoprotection , Dose-Response Relationship, Drug , Electric Impedance , Endothelial Cells/immunology , Endothelial Cells/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Humans , Inflammation Mediators/metabolism , Tight Junctions/drug effects , Tight Junctions/pathology , Time Factors
11.
Exp Lung Res ; 33(6): 321-35, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17694441

ABSTRACT

In vitro models of the alveolo-pulmonary barrier consist of microvascular endothelial cells and alveolar epithelial cells cultured on opposing sides of synthetic porous membranes. However, these simple models do not reflect the physiological microenvironment of pulmonary cells, wherein cells are exposed to a complex milieu of mechanical and soluble stimuli. In this report, we studied alveolar epithelial (A549) and microvascular endothelial (HMEC-1) cells within varying microfluidic environments as a first step towards building a microfluidic analog of the gas-exchange interface. We fabricated polydimethylsiloxane (PDMS) microdevices for parallel studies of cell growth under multiple flow rates. Cells adhered and proliferated in the microculture chambers for shear stresses up to approximately 2 x 10(-3) dynes/cm(2), corresponding to media turnover rates of approximately 53 seconds. Proliferation of these cells into confluent monolayers and expression of cell-specific markers (SP-A and CD-31) demonstrated successful pulmonary cell culture in microscale devices, a first for alveolar epithelial cells. These results represent the initial steps towards the development of microfluidic analogs of the alveolo-pulmonary barrier and tissue engineering of the lung.


Subject(s)
Blood-Air Barrier/pathology , Cell Culture Techniques/instrumentation , Cell Proliferation , Endothelial Cells/cytology , Epithelial Cells/pathology , Lung/blood supply , Microfluidic Analytical Techniques/instrumentation , Pulmonary Alveoli/pathology , Blood-Air Barrier/chemistry , Blood-Air Barrier/immunology , Cell Adhesion , Cell Culture Techniques/methods , Cell Line, Tumor , Dimethylpolysiloxanes , Endothelial Cells/immunology , Epithelial Cells/chemistry , Equipment Design , Humans , Microcirculation/cytology , Perfusion , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Pulmonary Alveoli/chemistry , Pulmonary Surfactant-Associated Protein A/analysis , Silicones , Stress, Mechanical , Time Factors
12.
Virchows Arch ; 442(1): 56-65, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12536315

ABSTRACT

We tested the hypothesis whether allergic airway inflammation in ovalbumin sensitized and challenged Brown Norway rats is associated with intrinsic surfactant alteration and dysfunction. The determination of intra-alveolar surfactant subtypes and alveolar edema within their original microenvironment is only possible using an ultrastructural stereological approach. Therefore both lungs of control and asthmatic rats were fixed by vascular perfusion. The volume fractions of surfactant subtypes and the epithelial surface fraction covered with alveolar edema were determined by point and intersection counting. Furthermore, lung resistance was measured by means of whole-body plethysmography. The surface activity of surfactant from bronchoalveolar lavage was determined as minimum surface tension at minimal bubble size with a pulsating bubble surfactometer. Compared with controls, in asthmatics (1) the fraction of inactive unilamellar forms was significantly increased from 56% to 66%, (2) the fraction of alveolar epithelium covered with alveolar edema visible by light microscopy was significantly increased from 0.7% to 5.0%, (3) the fraction of alveolar epithelium covered with fluid seen by electron microscopy expanded significantly from 5% to 21%, (4) lung resistance was significantly elevated from 14% to 86% and (5) surface tension was enhanced from 6 mN/m to 12 mN/m. Thus, the inflammatory process after allergen challenge of sensitized Brown Norway rats causes intra-alveolar surfactant alterations. These surfactant alterations might contribute to small airway dysfunction.


Subject(s)
Asthma/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Surfactants/metabolism , Administration, Inhalation , Aerosols , Animals , Asthma/immunology , Asthma/pathology , Blood-Air Barrier/immunology , Disease Models, Animal , Edema/immunology , Edema/metabolism , Edema/pathology , Immunization , Injections, Subcutaneous , Lung/immunology , Lung/pathology , Lung/physiopathology , Male , Ovalbumin/administration & dosage , Ovalbumin/immunology , Pulmonary Alveoli/ultrastructure , Rats , Rats, Inbred BN , Respiratory Function Tests
13.
Infect Immun ; 70(7): 3865-73, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12065530

ABSTRACT

Fibronectin-binding proteins mediate Staphylococcus aureus internalization into nonphagocytic cells in vitro. We have investigated whether fibronectin-binding proteins are virulence factors in the pathogenesis of pneumonia by using S. aureus strain 8325-4 and isogenic mutants in which fibronectin-binding proteins were either deleted (DU5883) or overexpressed [DU5883(pFnBPA4)]. We first demonstrated that fibronectin-binding proteins mediate S. aureus internalization into alveolar epithelial cells in vitro and that S. aureus internalization into alveolar epithelial cells requires actin rearrangement and protein kinase activity. Second, we established a rat model of S. aureus-induced pneumonia and measured lung injury and bacterial survival at 24 and 96 h postinoculation. S. aureus growth and the extent of lung injury were both increased in rats inoculated with the deletion mutant (DU5883) in comparison with rats inoculated with the wild-type (8325-4) and the fibronectin-binding protein-overexpressing strain DU5883(pFnBPA4) at 24 h postinfection. Morphological evaluation of infected lungs at the light and electron microscopic levels demonstrated that S. aureus was present within neutrophils from both 8325-4- and DU5883-inoculated lungs. Our data suggest that fibronectin-binding protein-mediated internalization into alveolar epithelial cells is not a virulence mechanism in a rat model of pneumonia. Instead, our data suggest that fibronectin-binding proteins decrease the virulence of S. aureus in pneumonia.


Subject(s)
Adhesins, Bacterial , Bacterial Proteins/immunology , Carrier Proteins/immunology , Fibronectins/immunology , Pneumonia, Bacterial/microbiology , Pulmonary Alveoli/microbiology , Staphylococcus aureus/pathogenicity , Animals , Antigens, Polyomavirus Transforming/genetics , Bacterial Proteins/genetics , Blood-Air Barrier/immunology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , Carrier Proteins/genetics , Cell Line, Transformed , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Leukocytes/cytology , Leukocytes/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Male , Mutagenesis , Neutrophil Infiltration/immunology , Phagocytosis/immunology , Pneumonia, Bacterial/blood , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/pathology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/immunology , Rats , Staphylococcal Infections/blood , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...