Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Respir Res ; 19(1): 42, 2018 03 10.
Article in English | MEDLINE | ID: mdl-29524964

ABSTRACT

BACKGROUND: Smoking is a leading cause of respiratory infections worldwide. Tobacco particulate matter disrupts iron homeostasis in the lungs and increases the iron content in the airways of smokers. The airway epithelia secrete lactoferrin to quench iron required for bacteria to proliferate and cause lung infections. We hypothesized that smokers would have increased bacterial growth and biofilm formation via iron lactoferrin imbalance. METHODS: We collected bronchoalveolar lavage (BAL) samples from non-smokers and smokers. We challenged these samples using a standard inoculum of Staphylococcus aureus and Pseudomonas aeruginosa and quantified bacterial growth and biofilm formation. We measured both iron and lactoferrin in the samples. We investigated the effect of supplementing non-smoker BAL with cigarette smoke extract (CSE) or ferric chloride and the effect of supplementing smoker BAL with lactoferrin on bacterial growth and biofilm formation. RESULTS: BAL from smokers had increased bacterial growth and biofilm formation compared to non-smokers after both S. aureus and P. aeruginosa challenge. In addition, we found that samples from smokers had a higher iron to lactoferrin ratio. Supplementing the BAL of non-smokers with cigarette smoke extract and ferric chloride increased bacterial growth. Conversely, supplementing the BAL of smokers with lactoferrin had a concentration-dependent decrease in bacterial growth and biofilm formation. CONCLUSION: Cigarette smoking produces factors which increase bacterial growth and biofilm formation in the BAL. We propose that smoking disrupts the iron-to-lactoferrin in the airways. This finding offers a new avenue for potential therapeutic interventions to prevent respiratory infections in smokers.


Subject(s)
Biofilms/growth & development , Iron/metabolism , Lactoferrin/metabolism , Pseudomonas aeruginosa/growth & development , Smoking/metabolism , Staphylococcus aureus/growth & development , Adolescent , Adult , Biofilms/drug effects , Bronchoalveolar Lavage Fluid/chemistry , Female , Humans , Lactoferrin/pharmacology , Male , Middle Aged , Pseudomonas aeruginosa/drug effects , Smokers , Staphylococcus aureus/drug effects , Young Adult
2.
Environ Health Perspect ; 125(7): 077003, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28696208

ABSTRACT

BACKGROUND: Sustained exposure to ambient particulate matter (PM) is a global cause of mortality. Coal fly ash (CFA) is a byproduct of coal combustion and is a source of anthropogenic PM with worldwide health relevance. The airway epithelia are lined with fluid called airway surface liquid (ASL), which contains antimicrobial proteins and peptides (AMPs). Cationic AMPs bind negatively charged bacteria to exert their antimicrobial activity. PM arriving in the airways could potentially interact with AMPs in the ASL to affect their antimicrobial activity. OBJECTIVES: We hypothesized that PM can interact with ASL AMPs to impair their antimicrobial activity. METHODS: We exposed pig and human airway explants, pig and human ASL, and the human cationic AMPs ß-defensin-3, LL-37, and lysozyme to CFA or control. Thereafter, we assessed the antimicrobial activity of exposed airway samples using both bioluminescence and standard colony-forming unit assays. We investigated PM-AMP electrostatic interaction by attenuated total reflection Fourier-transform infrared spectroscopy and measuring the zeta potential. We also studied the adsorption of AMPs on PM. RESULTS: We found increased bacterial survival in CFA-exposed airway explants, ASL, and AMPs. In addition, we report that PM with a negative surface charge can adsorb cationic AMPs and form negative particle-protein complexes. CONCLUSION: We propose that when CFA arrives at the airway, it rapidly adsorbs AMPs and creates negative complexes, thereby decreasing the functional amount of AMPs capable of killing pathogens. These results provide a novel translational insight into an early mechanism for how ambient PM increases the susceptibility of the airways to bacterial infection. https://doi.org/10.1289/EHP876.


Subject(s)
Air Pollutants/toxicity , Antimicrobial Cationic Peptides/genetics , Coal Ash/toxicity , Particulate Matter/toxicity , Respiratory Mucosa/drug effects , Animals , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/metabolism , Humans , Respiratory System/drug effects , Sus scrofa
3.
Am J Respir Cell Mol Biol ; 55(6): 869-877, 2016 12.
Article in English | MEDLINE | ID: mdl-27486964

ABSTRACT

Protein kinase Cζ (PKCζ) is highly expressed in the lung, where it plays several regulating roles in the pathogenesis of acute lung injury (ALI). Proliferation and differentiation of integrin ß4+ distal lung epithelial progenitor cells seem to play a key role in proper lung regeneration. We investigated the effects of a myristoylated PKCζ inhibitor (PKCζi) in a murine model of bleomycin-induced ALI. After intratracheal injury, we treated mice three times a week with PKCζi or its vehicle, DMSO. We found that mice injured with bleomycin and then treated with PKCζi for one week showed decreased activation of PKCζ, improved lung compliance, and decreased lung protein permeability compared to injured mice treated with DMSO. Mice treated continuously with PKCζi for 6 weeks showed reduced evidence of lung fibrosis by computed tomographic images, decreased lung collagen deposition, and decreased active transforming growth factor-ß in the bronchoalveolar lavage fluid. In addition, we found an increased number of lung ß4+ cells compared to DMSO at Week 6. Therefore, we grew isolated integrin ß4+ lung progenitor cells in the presence of PKCζi or DMSO and found that ß4+ cells treated with PKCζi proliferated more in vitro compared to DMSO. We conclude that the use of a PKCζi promotes resolution of lung fibrosis in a bleomycin ALI model and increases the number of ß4+ progenitor cells with regenerative potential in the lung.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/enzymology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Acute Lung Injury/pathology , Animals , Bleomycin , Bronchoalveolar Lavage Fluid/cytology , Cell Death/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Separation , Compliance , Disease Models, Animal , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Mice, Inbred C57BL , Models, Biological , Neutrophils/cytology , Neutrophils/drug effects , Permeability , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology , Reactive Oxygen Species/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism
4.
Environ Sci Nano ; 1(2): 123-132, 2014 Apr.
Article in English | MEDLINE | ID: mdl-25221673

ABSTRACT

Given the increased use of iron-containing nanoparticles in a number of applications, it is important to understand any effects that iron-containing nanoparticles can have on the environment and human health. Since iron concentrations are extremely low in body fluids, there is potential that iron-containing nanoparticles may influence the ability of bacteria to scavenge iron for growth, affect virulence and inhibit antimicrobial peptide (AMP) function. In this study, Pseudomonas aeruginosa (PA01) and AMPs were exposed to iron oxide nanoparticles, hematite (α-Fe2O3), of different sizes ranging from 2 to 540 nm (2 ± 1, 43 ± 6, 85 ± 25 and 540 ± 90 nm) in diameter. Here we show that the greatest effect on bacterial growth, biofilm formation, and AMP function impairment is found when exposed to the smallest particles. These results are attributed in large part to enhanced dissolution observed for the smallest particles and an increase in the amount of bioavailable iron. Furthermore, AMP function can be additionally impaired by adsorption onto nanoparticle surfaces. In particular, lysozyme readily adsorbs onto the nanoparticle surface which can lead to loss of peptide activity. Thus, this current study shows that co-exposure of nanoparticles and known pathogens can impact host innate immunity. Therefore, it is important that future studies be designed to further understand these types of impacts.

5.
PLoS One ; 9(1): e84434, 2014.
Article in English | MEDLINE | ID: mdl-24400090

ABSTRACT

BACKGROUND: Paraoxonase 1 (PON1) is a protein found associated with high density lipoprotein (HDL), thought to prevent oxidative modification of low-density lipoprotein (LDL). This enzyme has been implicated in lowering the risk of cardiovascular disease. Anoxia-reoxygenation and oxidative stress are important elements in cardiovascular and cerebrovascular disease. However, the role of PON1 in anoxia-reoxygenation or anoxic injury is unclear. We hypothesize that PON1 prevents anoxia-reoxygenation injury. We set out to determine whether PON1 expression in Drosophila melanogaster protects against anoxia-reoxygenation (A-R) induced injury. METHODS: Wild type (WT) and transgenic PON1 flies were exposed to anoxia (100% Nitrogen) for different time intervals (from 1 to 24 hours). After the anoxic period, flies were placed in room air for reoxygenation. Activity and survival of flies was then recorded. RESULTS: Within 5 minutes of anoxia, all flies fell into a stupor state. After reoxygenation, survivor flies resumed activity with some delay. Interestingly, transgenic flies recovered from stupor later than WT. PON1 transgenic flies had a significant survival advantage after A-R stress compared with WT. The protection conferred by PON1 expression was present regardless of the age or dietary restriction. Furthermore, PON1 expression exclusively in CNS conferred protection. CONCLUSION: Our results support the hypothesis that PON1 has a protective role in anoxia-reoxygenation injury, and its expression in the CNS is sufficient and necessary to provide a 100% survival protection.


Subject(s)
Aryldialkylphosphatase/genetics , Hypoxia/genetics , Oxidative Stress/genetics , Animals , Animals, Genetically Modified , Aryldialkylphosphatase/metabolism , Central Nervous System/metabolism , Diet , Disease Models, Animal , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression , Hypoxia/metabolism , Hypoxia/mortality , Male , Polymorphism, Genetic , Reactive Oxygen Species/metabolism
6.
PLoS One ; 8(2): e57673, 2013.
Article in English | MEDLINE | ID: mdl-23469047

ABSTRACT

Air pollution is a risk factor for respiratory infections, and one of its main components is particulate matter (PM), which is comprised of a number of particles that contain iron, such as coal fly ash (CFA). Since free iron concentrations are extremely low in airway surface liquid (ASL), we hypothesize that CFA impairs antimicrobial peptides (AMP) function and can be a source of iron to bacteria. We tested this hypothesis in vivo by instilling mice with Pseudomonas aeruginosa (PA01) and CFA and determine the percentage of bacterial clearance. In addition, we tested bacterial clearance in cell culture by exposing primary human airway epithelial cells to PA01 and CFA and determining the AMP activity and bacterial growth in vitro. We report that CFA is a bioavailable source of iron for bacteria. We show that CFA interferes with bacterial clearance in vivo and in primary human airway epithelial cultures. Also, we demonstrate that CFA inhibits AMP activity in vitro, which we propose as a mechanism of our cell culture and in vivo results. Furthermore, PA01 uses CFA as an iron source with a direct correlation between CFA iron dissolution and bacterial growth. CFA concentrations used are very relevant to human daily exposures, thus posing a potential public health risk for susceptible subjects. Although CFA provides a source of bioavailable iron for bacteria, not all CFA particles have the same biological effects, and their propensity for iron dissolution is an important factor. CFA impairs lung innate immune mechanisms of bacterial clearance, specifically AMP activity. We expect that identifying the PM mechanisms of respiratory infections will translate into public health policies aimed at controlling, not only concentration of PM exposure, but physicochemical characteristics that will potentially cause respiratory infections in susceptible individuals and populations.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Coal Ash/pharmacology , Coal , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Respiratory System/drug effects , Respiratory System/microbiology , Animals , Biological Availability , Coal Ash/chemistry , Epithelium/drug effects , Epithelium/metabolism , Epithelium/microbiology , Humans , Iron/analysis , Iron/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Pseudomonas aeruginosa/metabolism , Respiratory System/metabolism
7.
Environ Health Perspect ; 121(6): 691-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23478268

ABSTRACT

BACKGROUND: On 20 March 2010, the Icelandic volcano Eyjafjallajökull erupted for the first time in 190 years. Despite many epidemiological reports showing effects of volcanic ash on the respiratory system, there are limited data evaluating cellular mechanisms involved in the response to ash. Epidemiological studies have observed an increase in respiratory infections in subjects and populations exposed to volcanic eruptions. METHODS: We physicochemically characterized volcanic ash, finding various sizes of particles, as well as the presence of several transition metals, including iron. We examined the effect of Eyjafjallajökull ash on primary rat alveolar epithelial cells and human airway epithelial cells (20-100 µg/cm(2)), primary rat and human alveolar macrophages (5-20 µg/cm(2)), and Pseudomonas aeruginosa (PAO1) growth (3 µg/104 bacteria). RESULTS: Volcanic ash had minimal effect on alveolar and airway epithelial cell integrity. In alveolar macrophages, volcanic ash disrupted pathogen-killing and inflammatory responses. In in vitro bacterial growth models, volcanic ash increased bacterial replication and decreased bacterial killing by antimicrobial peptides. CONCLUSIONS: These results provide potential biological plausibility for epidemiological data that show an association between air pollution exposure and the development of respiratory infections. These data suggest that volcanic ash exposure, while not seriously compromising lung cell function, may be able to impair innate immunity responses in exposed individuals.


Subject(s)
Bacteria/growth & development , Immunity, Innate , Volcanic Eruptions/adverse effects , Adult , Animals , Autophagy , Bacteria/drug effects , Cells, Cultured , Female , Humans , Iceland , MAP Kinase Signaling System , Macrophages/immunology , Male , Microbial Viability/drug effects , Rats , Rats, Sprague-Dawley , beta-Defensins/pharmacology
8.
Am J Respir Cell Mol Biol ; 48(3): 306-13, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23221045

ABSTRACT

Recently, we reported that diesel exhaust particles (DEPs) disrupt tight junctions (TJs) in alveolar epithelial cells (AECs) via an increase in reactive oxygen species (ROS). In this study, we investigated the role of protein kinase C (PKC)-ζ activation in DEP-induced lung injury. C57/bl6 mice were instilled intratracheally with 50 µl of saline containing 100 µg of DEPs or titanium dioxide (TiO2). Twenty-four hours later, bronchoalveolar lavage was performed to assess neutrophil counts and protein concentrations. In addition, in vitro experiments were performed in primary rat and human AECs exposed to DEPs (50 µg/cm(2)) for 3 hours. Transepithelial electrical conductance was measured, and TJ protein association was analyzed by immunoprecipitation. To determine whether the overexpression of antioxidants prevented DEP-induced lung injury, AECs and mice were infected with adenoviruses containing catalase and manganese superoxide dismutase (MnSOD) plasmids. In vivo, the overexpression of catalase and MnSOD prevented DEP-induced neutrophil recruitment. The inhibition of PKC-ζ activation also prevented DEP-induced neutrophil recruitment in vivo. In vitro, DEPs activated PKC-ζ in AECs, but not in alveolar macrophages. Using a specific myristolated PKC-ζ pseudosubstrate pepetide (PKC-ζ ps), we showed that PKC-ζ mediated the DEP-induced dissociation of occludin and zonula occludin-1 (ZO1) in rat and human AECs. In addition, the overexpression of constitutively active PKC-ζ induced the dissociation of occludin and ZO1 in AECs. DEP-induced TJ disruption occurs via PKC-ζ. TJ disruption seems to be in part responsible for DEP-induced lung injury.


Subject(s)
Lung Injury/chemically induced , Lung Injury/enzymology , Protein Kinase C/metabolism , Vehicle Emissions/toxicity , Animals , Bronchoalveolar Lavage , Catalase/metabolism , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Lung Injury/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Occludin/metabolism , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Titanium/toxicity , Zonula Occludens-1 Protein/metabolism
9.
Am J Physiol Lung Cell Mol Physiol ; 300(4): L569-78, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21257729

ABSTRACT

During pulmonary edema, the alveolar space is exposed to a hypoxic environment. The integrity of the alveolar epithelial barrier is required for the reabsorption of alveolar fluid. Tight junctions (TJ) maintain the integrity of this barrier. We set out to determine whether hypoxia creates a dysfunctional alveolar epithelial barrier, evidenced by an increase in transepithelial electrical conductance (G(t)), due to a decrease in the abundance of TJ proteins at the plasma membrane. Alveolar epithelial cells (AEC) exposed to mild hypoxia (Po(2) = 50 mmHg) for 30 and 60 min decreased occludin abundance at the plasma membrane and significantly increased G(t). Other cell adhesion molecules such as E-cadherin and claudins were not affected by hypoxia. AEC exposed to hypoxia increased superoxide, but not hydrogen peroxide (H(2)O(2)). Overexpression of superoxide dismutase 1 (SOD1) but not SOD2 prevented the hypoxia-induced G(t) increase and occludin reduction in AEC. Also, overexpression of catalase had a similar effect as SOD1, despite not detecting any increase in H(2)O(2) during hypoxia. Blocking PKC-ζ and protein phosphatase 2A (PP2A) prevented the hypoxia-induced occludin reduction at the plasma membrane and increase in G(t). In summary, we show that superoxide, PKC-ζ, and PP2A are involved in the hypoxia-induced increase in G(t) and occludin reduction at the plasma membrane in AEC.


Subject(s)
Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/enzymology , Cell Membrane/metabolism , Electric Conductivity , Membrane Proteins/metabolism , Protein Kinase C/metabolism , Protein Phosphatase 2/metabolism , Animals , Catalase/metabolism , Cell Hypoxia , Hydrogen-Ion Concentration , Lanthanum/metabolism , Male , Occludin , Partial Pressure , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Signal Transduction , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...