Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Toxicol In Vitro ; 80: 105317, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35065218

ABSTRACT

Chlorine (Cl2) is a common toxic industrial gas and human inhalation exposure causes tissue damage with symptoms ranging from wheezing to more severe symptoms such as lung injury or even death. Because the mechanism behind Cl2-induced cell death is not clearly understood, the present study aimed to study the cellular effects in vitro after Cl2 exposure of human A549 lung epithelial cells. In addition, the possible treatment effects of the anti-inflammatory antioxidant N-acetyl cysteine (NAC) were evaluated. Exposure of A549 cells to Cl2 (100-1000 ppm) in the cell medium induced cell damage and toxicity within 1 h in a dose-dependent manner. The results showed that 250 ppm Cl2 increased cell death and formation of apoptotic-like bodies, while 500 ppm Cl2 exposure resulted in predominantly necrotic death. Pre-treatment with NAC was efficient to prevent cell damage at lower Cl2 concentrations in part by averting the formation of apoptotic-like bodies and increasing the expression of the anti-apoptotic proteins clusterin and phosphorylated tumour protein p53(S46). Analysis showed that Cl2 induced cell death by a possibly caspase-independent mechanism, since no cleavage of caspase-3 could be detected after exposure to 250 ppm. Currently, these results justifies further research into new treatment strategies for Cl2-induced lung injury.


Subject(s)
Chlorine/toxicity , Lung/cytology , Oxidants/toxicity , A549 Cells , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Caspase 3 , Cell Physiological Phenomena/drug effects , Cytokines/metabolism , Humans
2.
J Appl Toxicol ; 42(2): 203-215, 2022 02.
Article in English | MEDLINE | ID: mdl-34050537

ABSTRACT

Metal oxide fumes form at high temperatures, for instance, during welding or firing ammunition. Inhalation exposure to high levels of airborne metal oxide particles can cause metal fume fever, cardiovascular effects, and lung damage in humans, but the associated underlying pathological mechanisms are still not fully understood. Using human alveolar epithelial cells, vascular endothelial cells, and whole blood model systems, we aimed to elucidate the short-term effects of well-characterized metal particles emitted while firing pistol ammunition. Human lung epithelial cells exposed to gunshot smoke particles (0.1-50 µg/ml) produced reactive oxygen species (ROS) and pro-inflammatory cytokines (interleukin 8 (IL-8), granulocyte-macrophage colony-stimulating factor (GM-CSF)) that activate and recruit immune cells. Particles comprising high copper (Cu) and zinc (Zn) content activated human endothelial cells via a non-ROS-mediated mechanism that triggered immune activation (IL-8, GM-CSF), leukocyte adhesion to the endothelium (soluble intercellular adhesion molecule 1 (sICAM-1)), and secretion of regulators of the acute-phase protein synthesis (interleukin 6 (IL-6)). In human whole blood, metal oxides in gunshot smoke demonstrated intrinsic properties that activated platelets (release of soluble cluster of differentiation 40 ligand (sCD40L), platelet-derived growth factor B-chain homodimer(PDGF-BB), and vascular endothelial growth factor A (VEGF-A)) and blood coagulation and induced concomitant release of pro-inflammatory cytokines from blood leukocytes that further orchestrate thrombogenesis. The model systems applied provide useful tools for health risk assessment of particle exposures, but more studies are needed to further elucidate the mechanisms of metal fume fever and to evaluate the potential risk of long-term cardiovascular diseases.


Subject(s)
Endothelial Cells/drug effects , Environmental Exposure/adverse effects , Metal Nanoparticles/toxicity , A549 Cells , Humans
3.
Toxicol Lett ; 322: 58-65, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-31962155

ABSTRACT

High-level concentrations of chlorine (Cl2) can cause life-threatening lung injuries and the objective in this study was to understand the pathogenesis of short-term sequelae of Cl2-induced lung injury and to evaluate whether pre-treatment with the antioxidant N-acetyl cysteine (NAC) could counteract these injuries using Cl2-exposed precision-cut lung slices (PCLS). The lungs of Sprague-Dawley rats were filled with agarose solution and cut into 250 µm-thick slices that were exposed to Cl2 (20-600 ppm) and incubated for 30 min. The tissue slices were pre-treated with NAC (5-25 mM) before exposure to Cl2. Toxicological responses were analyzed after 5 h by measurement of LDH, WST-1 and inflammatory mediators (IL-1ß, IL-6 and CINC-1) in medium or lung tissue homogenate. Exposure to Cl2 induced a concentration-dependent cytotoxicity (LDH/WST-1) and IL-1ß release in medium. Similar cytokine response was detected in tissue homogenate. Contraction of larger airways was measured using electric-field-stimulation method, 200 ppm and control slices had similar contraction level (39 ± 5%) but in the 400 ppm Cl2 group, the evoked contraction was smaller (7 ± 3%) possibly due to tissue damage. NAC-treatment improved cell viability and reduced tissue damage and the contraction was similar to control levels (50 ± 11%) in the NAC treated Cl2-exposed slices. In conclusion, Cl2 induced a concentration-dependent lung tissue damage that was effectively prevented with pre-treatment with NAC. There is a great need to improve the medical treatment of acute lung injury and this PCLS method offers a way to identify and to test new concepts of treatment of Cl2-induced lung injuries.


Subject(s)
Acetylcysteine/pharmacology , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Chlorine/toxicity , Inflammation Mediators/metabolism , Lung Injury/prevention & control , Lung/drug effects , Animals , Cell Survival/drug effects , Chemokine CXCL1/metabolism , Cytoprotection , Dose-Response Relationship, Drug , Female , In Vitro Techniques , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lung/metabolism , Lung/pathology , Lung/physiopathology , Lung Injury/chemically induced , Lung Injury/metabolism , Lung Injury/pathology , Rats, Sprague-Dawley
4.
J Appl Toxicol ; 39(8): 1143-1154, 2019 08.
Article in English | MEDLINE | ID: mdl-30957912

ABSTRACT

The use of electronic cigarettes (E-cigs) is rapidly increasing. The latest generation of E-cigs is highly customizable, allowing for high heating coil temperatures. The aim of this study was to assess the toxic potential of a fourth-generation E-cig. Aerosols generated from E-liquid with (24 mg/mL) and without nicotine, using a fourth-generation E-cig, were chemically analysed and compared with cigarette smoke (K3R4F). Human lung epithelial cell lines and distal lung tissue explants were exposed to E-cig vapour extract (EVE) and cigarette smoke extract for 24 hours and assessed for viability, inflammation, oxidative stress and genotoxicity. E-cig aerosols contained measurable levels of volatile organic compounds, aldehydes and polycyclic aromatic hydrocarbons, in general, to a much lesser extent than cigarette smoke. Higher levels of certain carbonyls, e.g. formaldehyde, were detected in the E-cig aerosols. EVEs decreased cell viability of BEAS-2B cells, whereas little effect was seen in A549 cells and distal lung tissue. The nicotine-containing EVE caused a greater decrease in cell viability and significant increase in DNA damage than the nicotine-free EVE. Increased cytotoxicity, reactive oxygen species production and genotoxicity were seen with cells and tissue exposed to cigarette smoke extract compared with EVEs. Although E-cig aerosols were less toxic than cigarette smoke, it was not benign. Moreover, the EVE containing nicotine was more toxic than the nicotine-free EVE. More research is needed on the short- and long-term health effects of vaping and the usage of newly emerging E-cig devices to evaluate better the potential negative effects of E-cigs on human health.


Subject(s)
DNA Damage , Electronic Nicotine Delivery Systems , Lung/drug effects , Nicotine/toxicity , Volatile Organic Compounds/toxicity , A549 Cells , Aerosols , Cell Cycle/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Cytokines/metabolism , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Lung/immunology , Lung/metabolism , Lung/pathology , Nicotine/analysis , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Smoke/adverse effects , Volatile Organic Compounds/analysis
5.
Nanomedicine ; 14(3): 735-744, 2018 04.
Article in English | MEDLINE | ID: mdl-29277639

ABSTRACT

Iron-oxide nanoparticles (NPs) generated by environmental events are likely to represent health problems. α-Fe2O3 NPs were synthesized, characterized and tested in a model for toxicity utilizing human whole blood without added anticoagulant. MALDI-TOF of the corona was performed and activation markers for plasma cascade systems (complement, contact and coagulation systems), platelet consumption and release of growth factors, MPO, and chemokine/cytokines from blood cells were analyzed. The coronas formed on the pristine α-Fe2O3 NPs contained contact system proteins and they induced massive activation of the contact (kinin/kallikrein) system, as well as thrombin generation, platelet activation, and release of two pro-angiogeneic growth factors: platelet-derived growth factor and vascular endothelial growth factor, whereas complement activation was unaffected. The α-Fe2O3 NPs exhibited a noticeable toxicity, with kinin/kallikrein activation, which may be associated with hypotension and long-term angiogenesis in vivo, with implications for cancer, arteriosclerosis and pulmonary disease.


Subject(s)
Blood Coagulation , Ferric Compounds/chemistry , Immunity, Innate/drug effects , Kallikrein-Kinin System , Metal Nanoparticles/administration & dosage , Humans , Metal Nanoparticles/chemistry , Platelet-Derived Growth Factor/metabolism , Protein Corona/metabolism , Vascular Endothelial Growth Factor A/metabolism
6.
J Toxicol Environ Health A ; 78(10): 645-61, 2015.
Article in English | MEDLINE | ID: mdl-26039682

ABSTRACT

Numerous studies demonstrated that the use of lead (Pb)-containing ammunition is associated with mainly chronic health problems and also is a burden on the environment and wildlife. Recently, a number of reports showed evidence of undesirable acute health effects related to the use of newly developed Pb-free small-caliber ammunition. In this study, particles from leaded and Pb-free ammunition were collected in liquid collection medium, in a highly controlled chamber, while firing a pistol (9 mm) or a rifle (7.62 × 51 mm). The emitted particles were typically smaller than 4 µm, with the great majority in even smaller size ranges, as shown by gravimetrical analysis and a multistage impactor. Chemical analysis revealed significant differences in content and concentration of several metals in the particles. After administration of the liquids to alveolar and bronchial in vitro cell systems, particles were taken up by the cells; the Pb-free particles displayed higher cytotoxicity (EC50 = 2 µg/cm(2)) than particles from Pb ammunition. High correlation factors (>0.9) were found between cell death and content of copper and zinc. Particles from both Pb-containing and Pb-free ammunition were able to induce oxidative stress and the proinflammatory marker interleukin (IL)-8 in both in vitro systems. These results support previous findings that indicate an association between gunshot emissions and metal fume fever. This study demonstrates the usefulness of combining chemical data with biological in vitro responses in assessing acute toxicological effects from emissions from firing both Pb and Pb-free ammunition.


Subject(s)
Bronchi/drug effects , Cytokines/metabolism , Epithelial Cells/drug effects , Firearms , Particulate Matter/toxicity , Reactive Oxygen Species/metabolism , Cell Line , Cells, Cultured , Humans , Particle Size , Pulmonary Alveoli
7.
Biomaterials ; 51: 58-68, 2015 May.
Article in English | MEDLINE | ID: mdl-25770998

ABSTRACT

There is a compelling need to understand and assess the toxicity of industrially produced nanoparticles (NPs). In order to appreciate the long-term effects of NPs, sensitive human-based screening tests that comprehensively map the NP properties are needed to detect possible toxic mechanisms. Animal models can only be used in a limited number of test applications and are subject to ethical concerns, and the interpretation of experiments in animals is also distorted by the species differences. Here, we present a novel easy-to-perform highly sensitive whole-blood model using fresh non-anticoagulated human blood, which most justly reflects complex biological cross talks in a human system. As a demonstrator of the tests versatility, we evaluated the toxicity of TiO2 NPs that are widely used in various applications and otherwise considered to have relatively low toxic properties. We show that TiO2 NPs at very low concentrations (50 ng/mL) induce strong activation of the contact system, which in this model elicits thromboinflammation. These data are in line with the finding of components of the contact system in the protein corona of the TiO2 NPs after exposure to blood. The contact system activation may lead to both thrombotic reactions and generation of bradykinin, thereby representing fuel for chronic inflammation in vivo and potentially long-term risk of autoimmunity, arteriosclerosis and cancer. These results support the notion that this novel whole-blood model represents an important contribution to testing of NP toxicity.


Subject(s)
Kallikreins/blood , Models, Biological , Nanoparticles/toxicity , Titanium/toxicity , Adsorption , Antithrombin III/metabolism , Blood Coagulation/drug effects , Blood Proteins/metabolism , Complement System Proteins/metabolism , Humans , Inflammation/pathology , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Peptide Hydrolases/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors , Titanium/chemistry
8.
J Inorg Biochem ; 138: 1-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24837331

ABSTRACT

Bacterial biofilms cause a range of problems in many areas and especially in health care. Biofilms are difficult to eradicate with traditional antibiotics and consequently there is a need for alternative ways to prevent and/or remove bacterial biofilms. Furthermore, the emergence of antibiotic resistance in bacteria creates a challenge to find new types of antibiotics with a lower evolutionary pressure for resistance development. One route to develop such drugs is to target the so called virulence factors, i.e. bacterial systems used when bacteria infect a host cell. This study investigates synergy effects between Ga(III) ions, previously reported to suppress biofilm formation and growth in bacteria, and salicylidene acylhydrazides (hydrazones) that have been proposed as antivirulence drugs targeting the type three secretion system used by several Gram-negative pathogens, including Pseudomonas aerugionosa, during bacterial infection of host cells. A library of hydrazones was screened for: Fe(III) binding, enhanced anti-biofilm effect with Ga(III) on P. aeruginosa, and low cytotoxicity to mammalian cells. The metal coordination for the most promising ligand, 2-Oxo-2-[N-(2,4,6-trihydroxy-benzylidene)-hydrazino]-acetamide (ME0163) with Ga(III) was investigated using extended X-ray absorption fine structure spectroscopy as well as density functional theory. The results showed that Ga(III) chelates the hydrazone with 5- and 6-membered chelating rings, and that the Ga(III)-ME0163 complex enhanced the antibiofilm effect of Ga(III) while suppressing the type three secretion system in P. aeruginosa. The latter effect was not observed for the hydrazone alone and was similar for Ga(III)-citrate and Ga(III)-ME0163 complexes, indicating that the inhibition of virulence was caused by Ga(III).


Subject(s)
Acetamides/pharmacology , Biofilms , Gallium/pharmacology , Hydrazines/pharmacology , Hydrazones/pharmacology , Leukocidins/antagonists & inhibitors , Organometallic Compounds/pharmacology , Pseudomonas aeruginosa/drug effects , HeLa Cells , Humans , Leukocidins/biosynthesis , Virulence/drug effects , Virulence Factors/biosynthesis
9.
Biophys J ; 105(2): 310-9, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23870252

ABSTRACT

It is a challenging task to characterize the biodistribution of nanoparticles in cells and tissue on a subcellular level. Conventional methods to study the interaction of nanoparticles with living cells rely on labeling techniques that either selectively stain the particles or selectively tag them with tracer molecules. In this work, Raman imaging, a label-free technique that requires no extensive sample preparation, was combined with multivariate classification to quantify the spatial distribution of oxide nanoparticles inside living lung epithelial cells (A549). Cells were exposed to TiO2 (titania) and/or α-FeO(OH) (goethite) nanoparticles at various incubation times (4 or 48 h). Using multivariate classification of hyperspectral Raman data with partial least-squares discriminant analysis, we show that a surprisingly large fraction of spectra, classified as belonging to the cell nucleus, show Raman bands associated with nanoparticles. Up to 40% of spectra from the cell nucleus show Raman bands associated with nanoparticles. Complementary transmission electron microscopy data for thin cell sections qualitatively support the conclusions.


Subject(s)
Cell Nucleus/metabolism , Epithelial Cells/metabolism , Ferric Compounds/metabolism , Metal Nanoparticles , Titanium/metabolism , Biological Transport , Cell Line, Tumor , Cell Nucleus/ultrastructure , Epithelial Cells/ultrastructure , Humans , Lung/cytology , Multivariate Analysis , Spectrum Analysis, Raman
10.
Toxicol In Vitro ; 27(2): 825-33, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23276820

ABSTRACT

In this study aerosol samples collected in an Asian mega-city (Kabul, Afghanistan) were compared to PM samples collected in a European location with traffic (Umeå, Sweden) and a reference urban dust material (SRM 1649b). The toxicity of each sample towards normal human bronchial epithelial (NHBE) cells and a human bronchial epithelial cell line (BEAS-2B) was tested along with their ability to induce reactive oxygen species (ROS) formation and inflammatory responses. The extracts' morphology and elemental composition was studied by SEM-EDXRF, and filter samples were analyzed for metals and organic compounds. The PM from Kabul contained a larger fraction of fine particles, 19 times more polyaromatic hydrocarbons (PAH) and 37 times more oxygenated PAH (oxy-PAH) compared to samples from Umeå. The PM-samples from Kabul and the reference material (SRM 1649b) induced significantly stronger oxidative stress responses than the samples from Umeå. Furthermore, samples collected in Kabul induced significantly higher secretion of the cytokines IL-6, IL-8 and GM-CSF while SRM1649b induced a cytokine pattern more similar to samples collected in Umeå. Several properties of the particles could potentially explain these differences, including differences in their size distribution and contents of PAH and oxy-PAH, possibly in combination with their relative transition metal contents.


Subject(s)
Air Pollutants/toxicity , Cytokines/metabolism , Epithelial Cells/drug effects , Oxidative Stress/drug effects , Aerosols , Afghanistan , Bronchi/cytology , Cell Line , Cell Survival/drug effects , Epithelial Cells/metabolism , Humans , Reactive Oxygen Species/metabolism
11.
Nanoscale ; 4(23): 7383-93, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23070150

ABSTRACT

Nanoparticles of iron oxide generated by wearing of vehicles have been modelled with a tailored solution of size-uniform engineered magnetite particles produced by the Bradley reaction, a solvothermal metal-organic approach rendering hydrophilic particles. The latter does not bear any pronounced surface charge in analogy with that originating from anthropogenic sources in the environment. Physicochemical properties of the nanoparticles were thoroughly characterized by a wide range of methods, including XPD, TEM, SEM, DLS and spectroscopic techniques. The magnetite nanoparticles were found to be sensitive for transformation into maghemite under ambient conditions. This process was clearly revealed by Raman spectroscopy for high surface energy magnetite particles containing minor impurities of the hydromaghemite phase and was followed by quantitative measurements with EXAFS spectroscopy. In order to assess the toxicological effects of the produced nanoparticles in humans, with and without surface modification with ATP (a model of bio-corona formed in alveolar liquid), a pathway of potential uptake and clearance was modelled with a sequence of in vitro studies using A549 lung epithelial cells, lymphocyte 221-B cells, and 293T embryonal kidney cells, respectively. Raman microscopy unambiguously showed that magnetite nanoparticles are internalized within the A549 cells after 24 h co-incubation, and that the ATP ligand is retained on the nanoparticles throughout the uptake process. The toxicity of the nanoparticles was estimated using confocal fluorescence microscopy and indicated no principal difference for unmodified and modified particles, but revealed considerably different biochemical responses. The IL-8 cytokine response was found to be significantly lower for the magnetite nanoparticles compared to TiO(2), while an enhancement of ROS was observed, which was further increased for the ATP-modified nanoparticles, implicating involvement of the ATP signalling pathway in the epithelium.


Subject(s)
Ferric Compounds/chemistry , Magnetite Nanoparticles/chemistry , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Cell Survival/drug effects , HEK293 Cells , Humans , Interleukin-8/metabolism , Magnetite Nanoparticles/toxicity , Microscopy, Confocal , Reactive Oxygen Species/metabolism , Spectrum Analysis, Raman , Titanium/chemistry
12.
Nanotoxicology ; 6(6): 623-34, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21781018

ABSTRACT

We have compared the cellular uptake and responses of five preparations of nanocrystalline titanium dioxide (TiO(2)) between normal human bronchial epithelial (NHBE) cells and epithelial cell lines (A549 and BEAS-2B). The P25 nanoparticles, containing both anatase and rutile modifications, induced reactive oxygen species (ROS) and secretion of the neutrophil chemoattractant IL-8 in all three cell types used. Pure anatase and rutile particles provoked differential IL-8 response in A549 and no response in BEAS-2B cells despite similar formation of ROS. The pure TiO(2) modifications also provoked release of the inflammatory mediators: IL-6, G-CSF and VEGF, in NHBE cells but not in the two cell lines. We conclude that the responsiveness of lung epithelial cells is strongly dependent on both the physicochemical properties of TiO(2) nanoparticles and the type of responder cells. The differential pro-inflammatory responsiveness of primary lung epithelial cells compared with immortalized cell lines should be considered in the assessment of adverse reactions to inhaled nanoparticles.


Subject(s)
Epithelial Cells/drug effects , Nanoparticles/chemistry , Titanium/pharmacology , Analysis of Variance , Cell Line, Transformed , Cell Line, Tumor , Cell Survival/drug effects , Epithelial Cells/metabolism , Humans , Interleukin-8/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Primary Cell Culture , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism
13.
Antimicrob Agents Chemother ; 55(12): 5568-80, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21947396

ABSTRACT

Gallium ions have previously been shown to exhibit antibacterial and antibiofilm properties. In this study, we report differential bactericidal activities of two gallium complexes, gallium desferrioxamine B (Ga-DFOB) and gallium citrate (Ga-Cit). Modeling of gallium speciation in growth medium showed that DFOB and citrate both can prevent precipitation of Ga(OH)(3), but some precipitation can occur above pH 7 with citrate. Despite this, Ga-Cit 90% inhibitory concentrations (IC(90)) were lower than those of Ga-DFOB for clinical isolates of Pseudomonas aeruginosa and several reference strains of other bacterial species. Treatment with Ga compounds mitigated damage inflicted on murine J774 macrophage-like cells infected with P. aeruginosa PAO1. Again, Ga-Cit showed more potent mitigation than did Ga-DFOB. Ga was also taken up more efficiently by P. aeruginosa in the form of Ga-Cit than in the form of Ga-DFOB. Neither Ga-Cit nor Ga-DFOB was toxic to several human cell lines tested, and no proinflammatory activity was detected in human lung epithelial cells after exposure in vitro. Metabolomic analysis was used to delineate the effects of Ga-Cit on the bacterial cell. Exposure to Ga resulted in lower concentrations of glutamate, a key metabolite for P. aeruginosa, and of many amino acids, indicating that Ga affects various biosynthesis pathways. An altered protein expression profile in the presence of Ga-Cit suggested that some compensatory mechanisms were activated in the bacterium. Furthermore, the antibacterial effect of Ga was shown to vary depending on the carbon source, which has importance in the context of medical applications of gallium.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Carbon/metabolism , Gallium Radioisotopes/pharmacology , Gallium/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/toxicity , Biofilms/growth & development , Cell Line , Citrates/metabolism , Citrates/pharmacology , Citrates/toxicity , Culture Media , Deferoxamine/metabolism , Deferoxamine/pharmacology , Deferoxamine/toxicity , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Gallium/metabolism , Gallium/toxicity , Gallium Radioisotopes/toxicity , Humans , Ligands , Lung/cytology , Lung/drug effects , Macrophages/drug effects , Macrophages/microbiology , Mice , Microbial Sensitivity Tests , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/growth & development
14.
Small ; 7(4): 514-23, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21265017

ABSTRACT

The cellular uptake and distribution of five types of well-characterized anatase and rutile TiO(2) nanoparticles (NPs) in A549 lung epithelial cells is reported. Static light scattering (SLS), in-vitro Raman microspectroscopy (µ-Raman) and transmission electron spectroscopy (TEM) reveal an intimate correlation between the intrinsic physicochemical properties of the NPs, particle agglomeration, and cellular NP uptake. It is shown that µ-Raman facilitates chemical-, polymorph-, and size-specific discrimination of endosomal-particle cell uptake and the retention of particles in the vicinity of organelles, including the cell nucleus, which quantitatively correlates with TEM and SLS data. Depth-profiling µ-Raman coupled with hyperspectral data analysis confirms the location of the NPs in the cells and shows that the NPs induce modifications of the biological matrix. NP uptake is found to be kinetically activated and strongly dependent on the hard agglomeration size-not the primary particle size-which quantitatively agrees with the measured intracellular oxidative stress. Pro-inflammatory responses are also found to be sensitive to primary particle size.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/metabolism , Lung/cytology , Nanoparticles/chemistry , Nanoparticles/toxicity , Titanium/metabolism , Titanium/toxicity , Cell Line , Chemokine CCL2/metabolism , Humans , Interleukin-8/metabolism , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Spectrum Analysis, Raman
15.
Toxicology ; 280(3): 88-97, 2011 Feb 27.
Article in English | MEDLINE | ID: mdl-21129433

ABSTRACT

Low-dose exposure of alkylating mustard gas causes long-term respiratory complications characterized by bronchitis and lung fibrosis. In this study, we utilized a mouse model for lung exposure of the nitrogen mustard melphalan, in order to define early and late events in the pathogenesis such as expression of pro-inflammatory cytokines, recruitment of inflammatory cells to airways and late-phase fibrosis. We investigated the roles of different T lymphocyte subsets on the inflammatory response by using knockout mice lacking either the genes expressing T cell receptor (TCR)αß or TCRγδ, and compared the responsiveness with that of wild type mice and double knockout mice completely deficient in T cells. Exposure to melphalan induced an early burst of the pro-inflammatory cytokines interleukin (IL)-1ß, IL-6 and IL-23 in airways, followed by extensive infiltration of neutrophils in the lung tissue and airways within 24h. The acute phase was followed by a sustained lymphocytic response that persisted for at least 14 days with resulting lung fibrosis. Engagement of T lymphocytes, particularly the γδ T cell subset, was crucial both for the acute cytokine and neutrophil response and for the late-phase lung fibrosis as indicated by the lack of response in γδ T cell deficient mice. Our data demonstrate that T lymphocytes play a prominent role in the pathogenesis of long-term lung injuries caused by strong alkylating agents.


Subject(s)
Bronchi/pathology , Connective Tissue/drug effects , Connective Tissue/growth & development , Inflammation Mediators/toxicity , Inhalation Exposure/adverse effects , Melphalan/toxicity , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Alkylating Agents/administration & dosage , Alkylating Agents/toxicity , Animals , Bronchi/drug effects , Bronchi/immunology , Connective Tissue/pathology , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/administration & dosage , Melphalan/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocytes/drug effects , Time Factors
16.
Inhal Toxicol ; 21(11): 958-64, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19572781

ABSTRACT

The pathogenesis of lung injury by exposure to highly toxic sulfur and nitrogen mustards involves alkylating damage of the respiratory epithelium followed by an acute inflammatory response and lung edema. The acute phase is followed by long-term respiratory complications characterized by bronchitis, lung fibrosis, and airway hyperreactivity. In this study, we utilized a mouse model for airway inflammation induced by inhalation exposure to the alkylating nitrogen mustard melphalan, in order to investigate possible beneficial treatment effects by the corticosteroid dexamethasone. In addition, we investigated therapeutic efficacy of liposome-encapsuled vitamin E, an antioxidant formulation previously shown to be efficient in counteracting inflammatory conditions. Influx of inflammatory cells to airways, edema formation, and expression of different cytokines were analyzed 6 and 18 hours after exposure to melphalan. In order to evaluate long-term lung effects, we also investigated collagen deposition and accumulation of lymphocytes at 2 and 4 weeks after exposure. A single intraperitoneal injection of dexamethasone (10 mg/kg body weight) 1 hour after melphalan exposure significantly reduced interleukin (IL)-1 and IL-6 in bronchoalveolar lavage fluid (BALF) and diminished the acute airway inflammation. Our results also indicate that early single-dose treatment with dexamethasone protects against long-term effects observed 2-4 weeks after melphalan exposure, as indicated by reduced lymphocytic response in airways and decreased collagen deposition. Furthermore, our results indicate that also vitamin E (50 mg/kg) reduces acute inflammatory cell influx, and suppresses collagen formation in lung tissue, indicating that this drug could be used in combination with corticosteroids for protection against chemical-induced lung injury.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Burns, Chemical/drug therapy , Dexamethasone/therapeutic use , Lung Injury/drug therapy , Vitamin E/therapeutic use , Acute-Phase Reaction/prevention & control , Animals , Antioxidants/administration & dosage , Bronchoalveolar Lavage Fluid/cytology , Collagen/metabolism , Cytokines/biosynthesis , Drug Carriers , Female , Liposomes , Lung Injury/pathology , Lymphocytes/drug effects , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/prevention & control , Vitamin E/administration & dosage
17.
COPD ; 6(2): 112-20, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19378224

ABSTRACT

The mechanisms behind airway inflammation in chronic obstructive pulmonary disease (COPD) are still not well understood. Here we investigated lymphocyte subtypes in bronchoalveolar lavage fluid, likely to be involved in the pathogenesis of COPD, as well as exploring the effect of smoking cessation. Differential cell counts and T cell subsets were determined in BAL fluid from nineteen individuals with stable COPD (seven smokers, twelve ex-smokers) compared to twelve age-matched never-smokers and thirteen smoking-matched smokers with normal lung function. COPD-patients had higher percentages of airway CD8(+) T cells compared to never-smokers. An increased population of CD4(+) T cells expressed high levels of CD25 in smokers and COPD patients compared to never-smokers, suggesting the presence of regulatory T cells. As the T cell populations in smokers with normal lung function and COPD-patients were similar, the impact of current smoking in COPD was addressed in a subgroup analysis. Activation of CD8(+) T cells was found regardless of smoking habits. In contrast, the enhanced expression of gamma/delta T cells, was mainly associated with current smoking, whilst the increase in T regulatory cells appeared related to both smoking and COPD. Regardless of smoking habits, CD8(+) T cell activation was found in COPD, supporting the contention that this T cell subset may play a role in the pathogenesis of COPD. As CD8(+) T cells coexist with immunoregulatory CD4(+) T cells in airways of COPD patients, it is likely that both cytotoxic T-cell responses and immunosuppressive mechanisms may be of importance in COPD pathogenesis.


Subject(s)
Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Smoking Cessation , Smoking/immunology , T-Lymphocyte Subsets/physiology , Aged , Antigens, Differentiation, T-Lymphocyte/metabolism , Bronchoalveolar Lavage Fluid/cytology , Case-Control Studies , Female , Forced Expiratory Volume , Humans , Lymphocyte Activation/physiology , Lymphocyte Count , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/metabolism , Smoking/metabolism , Smoking/pathology
18.
Biomaterials ; 30(8): 1524-31, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19108885

ABSTRACT

This study investigates the antibacterial and cytotoxic effect of surfaces with sulphonate brushes containing silver salts. By using the same type of samples for both cytotoxicity and antibacterial studies, these two parameters could be compared in a controlled way. The silver was incorporated into the brush in four different forms to enable release of silver ions at different concentrations and different rates. It was found that although the surfaces displayed very good antibacterial properties in buffer solutions, this effect disappeared in systems with high protein content. Similarly, the silver-containing surfaces displayed cytotoxic effects in the absence of serum proteins but this effect was reduced in the presence of serum. The speciation of silver in the different solutions is discussed. Cytotoxic and antibacterial effects are compared at the different silver concentrations released. The implications of a concentration range where silver could be used to kill bacterial without harmful effects on mammalian cells are also discussed and questioned.


Subject(s)
Epithelial Cells/cytology , Fibroblasts/cytology , Methacrylates/pharmacology , Polymers/pharmacology , Pseudomonas aeruginosa/cytology , Silver Compounds/pharmacology , Staphylococcus aureus/cytology , Animals , Anti-Bacterial Agents/pharmacology , Buffers , Cell Count , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Chlorides , Culture Media , Diffusion/drug effects , Epithelial Cells/drug effects , Fibroblasts/drug effects , Humans , Mice , Microbial Sensitivity Tests , Microbial Viability/drug effects , Proteins/metabolism , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development
19.
J Appl Toxicol ; 25(4): 328-37, 2005.
Article in English | MEDLINE | ID: mdl-16025434

ABSTRACT

To investigate how respiratory epithelial cells react to an alkylating agent, we exposed human bronchial (BEAS-2B) and alveolar (A549) cells to the nitrogen mustard derivative melphalan. The BEAS-2B cells were highly sensitive to melphalan, as shown by a reduced viability after a 10-min incubation with 300 microM melphalan. The A549 cells were less sensitive and required several hours of exposure to reduce significantly in viability. However, exposure to melphalan also induces activation of intracellular signal transduction pathways, as indicated by phosphorylation of extracellular signal-regulated kinase (ERK1/2) and p38 (proteins belonging to the family of stress-induced mitogen-activated phosphorylated kinases, MAPK) within 5 min, as well as translocation of the transcription factor nuclear factor (NF)-kappaB to the nucleus within 45 min. This early activation was followed by elevated levels of tumor necrosis factor (TNF)-alpha mRNA within 2 h. We also observed increased expression of intercellular adhesion molecule-1 (ICAM-1) on the surface of both cell lines 18 h after exposure to 25 microM melphalan and an increased adhesion of monocytes to the epithelial cells in vitro.In conclusion, we have demonstrated that alkylating compounds not only cause cell death of lung epithelial cells but also activate stress-associated MAPK signal transduction pathways and induce expression of mediators known to participate in the recruitment of inflammatory cells.


Subject(s)
Lung/pathology , Melphalan/pharmacology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/drug effects , Pneumonia/chemically induced , Pneumonia/pathology , Blotting, Western , Cell Adhesion/drug effects , Electrophoretic Mobility Shift Assay , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Flavonoids/pharmacology , Flow Cytometry , Intercellular Adhesion Molecule-1/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Monocytes/drug effects , Monocytes/metabolism , Oxazines , Protein Kinase C/antagonists & inhibitors , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/biosynthesis , Xanthenes
20.
Am J Respir Cell Mol Biol ; 28(2): 199-207, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12540487

ABSTRACT

We investigated the pharmacologic effects of the antioxidant Vitamin E (alpha-tocopherol [alpha-toc]) in airway inflammation induced by inhaled endotoxin. A preparation of alpha-toc incorporated in liposomes was administered intraperitoneally in mice 1 h after exposure of aerosolized endotoxin. Injection of 50 mg alpha-toc/kg significantly decreased the number of neutrophils in airspaces and prevented lung injury, monitored both as decreased lactate dehydrogenase activity in airways and reduced lung edema when compared with animals treated with plain liposomes. Immunofluorescence staining of lung tissue revealed that treatment with alpha-toc decreased the number of neutrophils in lung interstitium, whereas the number in lung blood vessels and peripheral blood did not differ between mice treated with alpha-toc and control mice. Our results indicate that alpha-toc downmodulates the migration of neutrophils across the endothelial barrier, but in contrast to strong anti-inflammatory drugs such as corticosteroids, without inhibition of transcription factors involved in the early inflammatory response (nuclear factor-kappaB/activator protein-1). Neither was the endotoxin-induced expression of proinflammatory cytokines in lung tissue downregulated. Treatment with a combination of alpha-toc and a suboptimal dose of 0.5 mg/kg dexamethasone enhanced the effect, suggesting that alpha-toc, in combination with low doses of corticosteroids, might be effective for therapeutic treatment of acute lung injury.


Subject(s)
Lung/cytology , Lung/drug effects , Neutrophils/drug effects , Vitamin E/pharmacology , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Movement/drug effects , Chemokines/genetics , Cytokines/genetics , Dexamethasone/administration & dosage , Edema/chemically induced , Edema/prevention & control , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Lipopolysaccharides/toxicity , Lung/metabolism , Lung Injury , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neutrophils/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Respiratory Burst/drug effects , Transcription Factor AP-1/metabolism , Vitamin E/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...