Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Part Fibre Toxicol ; 19(1): 43, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35739584

ABSTRACT

BACKGROUND: Exposure to ambient fine particulate matter (PM2.5) is associated with vascular diseases. Polycyclic aromatic hydrocarbons (PAHs) in PM2.5 are highly hazardous; however, the contribution of PM2.5-bound PAHs to PM2.5-associated vascular diseases remains unclear. The ToxCast high-throughput in vitro screening database indicates that some PM2.5-bound PAHs activate the aryl hydrocarbon receptor (AhR). The present study investigated whether the AhR pathway is involved in the mechanism of PM2.5-induced vascular toxicity, identified the PAH in PM2.5 that was the major contributor of AhR activation, and identified a biomarker for vascular toxicity of PM2.5-bound PAHs. RESULTS: Treatment of vascular smooth muscle cells (VMSCs) with an AhR antagonist inhibited the PM2.5-induced increase in the cell migration ability; NF-κB activity; and expression of cytochrome P450 1A1 (CYP1A1), 1B1 (CYP1B1), interleukin-6 (IL-6), and osteopontin (OPN). Most PM2.5-bound PAHs were extracted into the organic fraction, which drastically enhanced VSMC migration and increased mRNA levels of CYP1A1, CYP1B1, IL-6, and OPN. However, the inorganic fraction of PM2.5 moderately enhanced VSMC migration and only increased IL-6 mRNA levels. PM2.5 increased IL-6 secretion through NF-κB activation; however, PM2.5 and its organic extract increased OPN secretion in a CYP1B1-dependent manner. Inhibiting CYP1B1 activity and silencing OPN expression prevented the increase in VSMC migration ability caused by PM2.5 and its organic extract. The AhR activation potencies of seven PM2.5-bound PAHs, reported in the ToxCast database, were strongly correlated with their capabilities of enhancing the migration ability of VSMCs. Benzo(k)fluoranthene (BkF) contributed the most to the AhR agonistic activity of ambient PM2.5-bound PAHs. The association between PM2.5-induced vascular toxicity, AhR activity, and OPN secretion was further verified in mice; PM2.5-induced intimal hyperplasia in pulmonary small arteries and OPN secretion were alleviated in mice with low AhR affinity. Finally, urinary concentrations of 1-hydroxypyrene, a major PAH metabolite, were positively correlated with plasma OPN levels in healthy humans. CONCLUSIONS: The present study offers in vitro, animal, and human evidences supporting the importance of AhR activation for PM2.5-induced vascular toxicities and that BkF was the major contributor of AhR activation. OPN is an AhR-dependent biomarker of PM2.5-induced vascular toxicity. The AhR activation potency may be applied in the risk assessment of vascular toxicity in PAH mixtures.


Subject(s)
Polycyclic Aromatic Hydrocarbons , Vascular Diseases , Animals , Biomarkers , Cytochrome P-450 CYP1A1/genetics , Interleukin-6 , Mice , NF-kappa B , Osteopontin/genetics , Particulate Matter/toxicity , Polycyclic Aromatic Hydrocarbons/toxicity , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism
2.
Antioxidants (Basel) ; 10(5)2021 May 14.
Article in English | MEDLINE | ID: mdl-34069133

ABSTRACT

Epidemiological studies have demonstrated an association between ambient particulate matter (PM) exposure and vascular diseases. Here, we observed that treatment with ambient PM increased cell migration ability in vascular smooth muscle cells (VSMCs) and pulmonary arterial SMCs (PASMCs). These results suggest that VSMCs and PASMCs transitioned from a differentiated to a synthetic phenotype after PM exposure. Furthermore, treatment with PM increased intracellular reactive oxygen species (ROS), activated the NF-κB signaling pathway, and increased the expression of proinflammatory cytokines in VSMCs. Using specific inhibitors, we demonstrated that PM increased the migration ability of VSMCs via the nicotinamide-adenine dinucleotide phosphate (NADPH) oxidase 1 (NOX1)/ROS-dependent NF-κB signaling pathway, which also partially involved in the induction of proinflammatory cytokines. Finally, we investigated whether nature polyphenolic compounds prevent PM-induced migration and proinflammatory cytokines secretion in VSMCs. Curcumin, resveratrol, and gallic acid prevented PM2.5-induced migration via the ROS-dependent NF-κB signaling pathway. However, honokiol did not prevent PM2.5-induced migration or activation of the ROS-dependent NF-κB signaling pathway. On the other hand, all polyphenols prevented PM2.5-induced cytokines secretion. These data indicated that polyphenols prevented PM-induced migration and cytokine secretion via blocking the ROS-dependent NF-κB signaling pathway in VSMCs. However, other mechanisms may also contribute to PM-induced cytokine secretion.

3.
Sci Total Environ ; 719: 137243, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32147111

ABSTRACT

Exposure to ambient fine particulate matter (PM2.5) has been associated with vascular diseases in epidemiological studies. We have demonstrated previously that exposure to ambient PM2.5 caused pulmonary vascular remodeling in mice and increased vascular smooth muscle cells (VSMCs) viability. Here, we further demonstrated that exposure of mice to ambient PM2.5 increased urinary 8­hydroxy­2'­deoxyguanosine (8-OHdG) and cytokines concentrations in the broncheoalveolar lavage. The objective of the present study was to identify the PM2.5 components related to vascular dysfunction. Exposure to PM2.5 collected from various areas and seasons in Taiwan significantly increased viability, oxidative stress, and inflammatory cytokines secretion in VSMCs. The mass concentrations of benz[a]anthracene (BaA), benzo[e]pyrene (BeP), perylene, dibenzo[a,e]pyrene, molybdenum, zinc (Zn), vanadium (V), and nickel in the PM2.5 were significantly associated with increased viability of VSMCs. These components, except BaA and BeP, also were significantly associated with chemokine (CC motif) ligand 5 (CCL5) concentrations in the VSMCs. The effects of V and Zn on cell viability and CCL5 expression, respectively, were verified. In addition, the mass concentrations of sulfate and manganese (Mn) in PM2.5 were significantly correlated with increased oxidative stress; this correlation was also confirmed. After extraction, the inorganic fraction of PM2.5 increased cell viability and oxidative stress, but the organic fraction of PM2.5 increased only cell viability, which was inhibited by an aryl hydrocarbon receptor antagonist. These data suggest that controlling the emission of Zn, V, Mn, sulfate, and PAHs may prevent the occurrence of PM2.5-induced vascular diseases.


Subject(s)
Particulate Matter/toxicity , Air Pollutants , Animals , Mice , Muscle, Smooth, Vascular , Oxidative Stress , Taiwan
4.
Environ Pollut ; 245: 975-985, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30682754

ABSTRACT

Ambient particulate matter (PM) exposure is associated with pulmonary and cardiovascular diseases; however, there is scant research linking data on animal and human cells. The objective of this study was to investigate these associations. Vascular remodeling plays a crucial role in both pulmonary and cardiovascular diseases. Therefore, we conducted a transcriptomic analysis using vascular smooth muscle cells (VSMCs) to identify potential regulators or markers of PM exposure. We demonstrated that fine and coarse PM increased VSMC proliferation in mice. We conducted a genome-wide cDNA microarray analysis, followed by a pathway analysis of VSMCs treated with coarse PM for durations of 24, 48, and 72 h. Sixteen genes were discovered to be time-dependently upregulated and involved in VSMC proliferation. Osteopontin (OPN) is indicated as one of the regulators of these upregulated genes. Both fine and coarse PM from industrial and urban areas significantly increased OPN expression in VSMCs and macrophages. Moreover, oropharyngeal instillation of fine and coarse PM for 8 weeks increased the VSMCs in the pulmonary arteries of mice. OPN level was consistently increased in the lung tissues, bronchoalveolar lavage fluid, and serum of mice. Moreover, we analyzed the plasma OPN levels of 72 healthy participants recruited from the studied metropolitan area. Each participant wore a personal PM2.5 sampler to assess their PM2.5 exposure over a 24 h period. Our results indicate that personal exposure to fine PM is positively correlated with plasma OPN level in young adults. The data obtained in this study suggest that exposure to fine and coarse PM may cause pulmonary vascular lesions in humans and that OPN level may be a biomarker of PM exposure in humans.


Subject(s)
Air Pollutants/analysis , Environmental Exposure/analysis , Macrophages/drug effects , Myocytes, Smooth Muscle/drug effects , Osteopontin/metabolism , Particulate Matter/analysis , Adult , Air Pollutants/toxicity , Animals , Biomarkers/metabolism , Bronchoalveolar Lavage Fluid/cytology , Cell Proliferation/drug effects , Female , Humans , Lung/drug effects , Lung/metabolism , Macrophages/metabolism , Male , Mice , Myocytes, Smooth Muscle/metabolism , Osteopontin/genetics , Particle Size , Particulate Matter/toxicity , RAW 264.7 Cells , Young Adult
5.
Toxicol Appl Pharmacol ; 324: 1-11, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28351761

ABSTRACT

Exposure to environmental aryl hydrocarbon receptor (AhR) agonists, such as halogenated aromatic hydrocarbons and polycyclic aromatic hydrocarbons (PAHs), has great impacts on the development of various lung diseases. As emerging molecular targets for AhR agonists, cytokines may contribute to the inflammatory or immunotoxic effects of environmental AhR agonists. However, general cytokine expression may not specifically indicate environmental AhR agonist exposure. By comparing cytokine and chemokine expression profiles in human lung adenocarcinoma cell line CL5 treated with AhR agonists and the non-AhR agonist polychlorinated biphenyl (PCB) 39, we identified a target cytokine of environmental AhR agonist exposure of in the lungs. Thirteen cytokine and chemokine genes were altered in the AhR agonists-treated cells, but none were altered in the PCB39-treated cells. Interleukin (IL)-24 was the most highly induced gene among AhR-modulated cytokines. Cotreatment with AhR antagonist completely prevented IL-24 induction by AhR agonists in the CL5 cells. Knockdown AhR expression with short-hairpin RNA (shRNA) significantly reduced benzo[a]pyrene (BaP)-induced IL-24 mRNA levels. We further confirmed that gene transcription, but not mRNA stability, was involved in IL-24 upregulation by BaP. Particulate matter (PM) in the ambient air contains some PAHs and is reported to activate AhR. Oropharyngeal aspiration of PM significantly increased IL-24 levels in lung epithelia and in bronchoalveolar lavage fluid of mice 4weeks after treatment. Thus, our data suggests that IL-24 is a pulmonary exposure target cytokine of environmental AhR agonists.


Subject(s)
Cytokines/biosynthesis , Hydrocarbons, Halogenated/toxicity , Lung/metabolism , Receptors, Aryl Hydrocarbon/agonists , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Cytokines/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Lung/drug effects , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Random Allocation
6.
Nanotoxicology ; 11(3): 360-370, 2017 04.
Article in English | MEDLINE | ID: mdl-28285566

ABSTRACT

Zinc oxide nanoparticles (ZnONPs) are widely used in our daily life, such as in sunscreens and electronic nanodevices. However, pulmonary exposure to ZnONPs causes acute pulmonary inflammation, which is considered as an initial event for various respiratory diseases. Thus, elucidation of the underlying cellular mechanisms of ZnONPs can help us in predicting their potential effects in respiratory diseases. In this study, we observed that ZnONPs increased proinflammatory cytokines, accompanied with an increased expression of aryl hydrocarbon receptor (AhR) and its downstream target cytochrome P450 1A1 (CYP1A1) in macrophages in vitro and in mouse lung epithelia in vivo. Moreover, zinc nitrate, but not silica or titanium dioxide nanoparticles (NPs), had similar effects on macrophages, indicating that the zinc element or ion released from ZnONPs is likely responsible for the activation of the AhR pathway. Cotreatment with an AhR antagonist or AhR knockout reduced ZnONPs-induced cytokine secretion in macrophages or mice, respectively. Furthermore, kynurenine (KYN), an endogenous AhR agonist and a tryptophan metabolite catalyzed by indoleamine 2,3-dioxygenase (IDO), was increased in the serums of mice that aspirated ZnONPs. Consistently, ZnONPs increased IDO1 expression in lung cells in vitro and in vivo. Finally, AhR knockout reduced ZnONPs-induced pulmonary inflammation, cytokine secretion and KYN production in mice, suggesting that AhR activation is involved in ZnONPs-induced cytokine secretion and pulmonary inflammation. In summary, we demonstrated that the pulmonary exposure of ZnONPs stimulated the cytokine-IDO1-AhR loop in the lungs, which has been implied to play roles in immune dysfunctions.


Subject(s)
Cytokines/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Nanoparticles/toxicity , Pneumonia/chemically induced , Receptors, Aryl Hydrocarbon/physiology , Zinc Oxide/toxicity , Animals , Cells, Cultured , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
7.
Nano Lett ; 15(10): 6446-53, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26339979

ABSTRACT

In this study, we sought to control the assembly of an endotoxin known as the biologically supramolecular lipopolysaccharide (LPS, which consists of three portions: an O antigen, a core carbohydrate, and a lipid A molecule) in order to modulate immunological responses in a manner that has the potential for utilization in vaccine development. Changing the structures of LPS aggregates from lamellas to specific nonlamellas (i.e., cubosomes and hexosomes) can dramatically enhance the strength of LPS in causing inflammatory responses, leading to highly active responses. In order to control the formation of cubosome-free and hexosome-free nonlamellas, we designed a simple strategy based on the use of hydrophilic gold nanodots (AuNDs) to control LPS assembly to facilitate the formation of stable endotoxin nanovesicles, which are stable precursors of cubosomes and hexosomes with specific immunological effects. Structurally, the wall thicknesses of these nanovesicles are exactly twice the lengths of a single LPS molecule, indicating that the LPS molecules adopt a tail-to-tail arrangement (with the lipid A portions acting as the tail domain). The involvement of the hydrophilic AuNDs to laterally link polar domains of LPS can result in the progressive extension of an endotoxically active zone of lipid A assembly, leading to the eventual formation of large-size nanovesicles. Our results showed that endotoxin nanovesicles with such dense lipid A units can elicit the stronger inflammatory gene expressions, including interleukin 6 (IL-6), IL-1A, TNF-α, C-X-C chemokine ligand (CXCL) 1, 2, and 11, which have characteristics of T-helper 1 adjuvants. These findings provide evidence that the concept of manipulating the surface hydrophilicity of AuNDs to control LPS assembly in order to avoid the formation of highly active cubosomes and hexosomes, and thereby modulate immunological responses appropriately, could prove useful in vaccine development.


Subject(s)
Chemokines/metabolism , Cytokines/metabolism , Gold/chemistry , Lipopolysaccharides/chemistry , Metal Nanoparticles , Humans , Hydrophobic and Hydrophilic Interactions , Microscopy, Electron, Transmission
8.
Nanotoxicology ; 9(1): 43-53, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24559390

ABSTRACT

Inhalation of zinc oxide nanoparticles (ZnONP) has potential health impact. Because zinc ion may involve in the toxicity of ZnONP, we compared adverse effects of inhaled aerosolized ZnONP and zinc nitrate in mice. Aerosolized ZnONP and zinc nitrate were well-dispersed in the inhalation chamber. Inhalation of 0.86 mg/m(3) ZnONP or 1.98 mg/m(3) zinc nitrate for 5 h caused acute inflammation mainly at bronchioloalveolar junctions of lungs at 24-h post-exposure. Inhalation of ZnONP or zinc nitrate increased metallothionein expression in the epithelial cells of brochioloalveolar junction. While the effects on cytokines secretion in bronchoalveolar lavage were similar between ZnONP and zinc nitrate, only ZnONP increased lactate dehydrogenase activity. However, repeated exposure to 0.86 mg/m(3) ZnONP 5 h/day for 5 days failed to cause a similar adverse effect. Either single or repeated exposure to 0.86 mg/m(3) ZnONP increased activities of glutamate oxaloacetate transaminase, glutamate pyruvate transaminase and creatine phosphokinase in blood. In contrast, exposure to zinc nitrate had no similar systemic effects. In human bronchial epithelial cells, ZnONP-induced interleukin-8 secretion was partially prevented by co-treatment with the Toll-like receptor 4 (TLR4) inhibitor. Furthermore, ZnONP-induced pulmonary inflammation was greater in wild-type mice than in TLR4-deficent mice. It appears that ZnONP-induced acute pulmonary inflammation partially depended on TLR4. In summary, we demonstrated the dose-responsive effects for inhalation of ZnONP and zinc nitrate in mice. The threshold of cytokines induction for inhalation of ZnONP for 5 h was 0.43 mg/m(3). The particulate characters of ZnONP might contribute to the systemic adverse effects and shall be evaluated for assessing its health impact in humans.


Subject(s)
Lung/drug effects , Metal Nanoparticles/toxicity , Pneumonia/chemically induced , Zinc Oxide/toxicity , Administration, Inhalation , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/analysis , Dose-Response Relationship, Drug , Female , Humans , Lung/cytology , Lung/pathology , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Mice , Mice, Inbred BALB C , Nitrates/administration & dosage , Nitrates/toxicity , Particle Size , Pneumonia/metabolism , Zinc Compounds/administration & dosage , Zinc Compounds/toxicity , Zinc Oxide/administration & dosage , Zinc Oxide/chemistry
9.
Exp Toxicol Pathol ; 65(6): 887-96, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23352990

ABSTRACT

Zinc oxide nanoparticles (ZnONP) have great potential for medical applications. However, ZnONP is reported to induce acute lung inflammation, which limits its application in humans. We designed in vivo and in vitro studies to clarify ZnONP inflammation and its associated molecular signals. ZnONP with a single dose of 80 µg/30 µl was instilled into the tracheas of mice sacrificed at days 2, 7, 14, and 28 after instillation. Bronchoalveolar lavage fluid showed increased neutrophils and macrophages after treatment. Lung pathology showed a mixed inflammatory infiltrate of neutrophils, lymphocytes, and macrophages primarily in the bronchioles and peribronchiolar areas. Proinflammatory gene expression of TNF-α, IL-6, CXCL1, and MCP-1 was increased at day 2 and decreased after 7 days. The lung pathology resolved at day 28, without fibrosis. It remains unclear whether this acute lung inflammation was caused by ZnONP themselves or Zn(2+) iron released from the nanoparticles. In vitro studies confirming the results of in vivo studies showed increased expression of proinflammatory genes in both MLE12 cells (mouse lung epithelial cells) and RAW264.7 cells (mouse macrophages) with either ZnONP or Zn(NO3)2 treatment; notably, increased levels of proinflammatory genes were obviously higher in cells treated with ZnONP than in cells treated with Zn(NO3)2 at the same molarity dose. TNF-α and MCP-1 were induced only in MLE12 cells. MyD88, an adaptor protein for most Toll-like receptors (TLR) signaling pathways, initiated the ZnONP or Zn(NO3)2-induced lung inflammation. Silencing MyD88 expression with siRNA significantly reduced ZnONP or Zn(NO3)2-induced proinflammatory gene expression in MLE12 and RAW264.7 cells. Single-dose exposure to ZnONP produced the short-term lung inflammation via a MyD88-dependent TLR pathway. These data suggest that although both ZnONP and zinc ion might participate in the inflammatory reactions, ZnONP more effectively induced MyD88-dependent proinflammatory cytokines than zinc ion in lung epithelial cells.


Subject(s)
Myeloid Differentiation Factor 88/metabolism , Nanoparticles/toxicity , Pneumonia/chemically induced , Zinc Oxide/toxicity , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Cell Survival/drug effects , Cytokines/immunology , Cytokines/metabolism , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred ICR , Myeloid Differentiation Factor 88/genetics , Particle Size , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , RNA, Small Interfering/genetics , Respiratory Function Tests
10.
Nanotoxicology ; 7(1): 105-15, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22107365

ABSTRACT

Some quantum dots (QDs) have been applied for drug delivery and imaging in biological systems. Drug delivery via the lung and lung imaging are potential applications of QDs. QD705 is cadmium based. The aims of the study were to evaluate the biological effects of QD705 in the lungs and the protective effects of polyethylene glycol (PEG) coating against QD705-induced biological responses. Intratracheal instillation of QD705-COOH persistently induced acute neutrophil infiltration, followed by interstitial lymphocyte infiltration and a granulomatous reaction on days 17 and 90. QD705-COOH also induced gene expression of cytokines, chemokines and metalloproteinase 12 in lung tissues. Furthermore, QD705-COOH transiently reduced pulmonary function on day 17. Treatment with QD705-PEG induced similar inflammatory responses and reduced pulmonary function on day 17, but the granulomatous reaction disappeared by day 90 These data indicated that administration of QD705 via the lung caused adverse responses and PEG coating failed to prevent these effects.


Subject(s)
Cadmium/chemistry , Granuloma/pathology , Nanoparticles , Pneumonia/pathology , Quantum Dots , Animals , Cell Line , Mice , Mice, Inbred ICR
11.
Mol Pharmacol ; 83(3): 605-12, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23229511

ABSTRACT

We have demonstrated that aryl hydrocarbon receptor (AhR) is overexpressed in lung adenocarcinoma (AD). AhR is usually associated with heat shock protein 90 (Hsp90) in the cytoplasm. 17-Allylamino-17-demethoxygeldanamycin (17-AAG), an Hsp90 inhibitor, is currently under evaluation for its anticancer activity in clinical trials. Here we investigated whether AhR plays a role in 17-AAG-mediated anticancer activity by functioning as a downstream target or by modulating its anticancer efficacy. AhR expression in lung AD cells was modulated by siRNA interference or overexpression. Tumor growth was determined with colony formation in vitro or in vivo. Anticancer activity of 17-AAG was determined by measuring cell viability, cell cycle distribution, and expression of cell cycle regulators. Proteins and mRNA levels were examined by immunoblotting and the real-time reverse transcription-polymerase chain reaction, respectively. In this study, AhR overexpression positively modulated growth of lung AD cells, at least partially, via RelA-dependent mechanisms. Although treatment with 17-AAG reduced AhR levels and AhR-regulated gene expression in lung AD cells, AhR expression increased anticancer activity of 17-AAG. In addition, 17-AAG treatment reduced cell viability, CDK2, CDK4, cyclin E, cyclin D1, and phosphorylated Rb levels in AhR-expressing lung AD cells. NAD(P)H:quinone oxidoreductase (NQO1), which is regulated by AhR, was shown to increase anticancer activity of 17-AAG in cells. Knockdown of NQO1 expression attenuated the reduction of cell cycle regulators by 17-AAG treatment in AhR overexpressed cells. We demonstrated that AhR protein not only functions as a downstream target of 17-AAG, but also enhances anticancer activity of 17-AAG in lung AD cells.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , Lactams, Macrocyclic/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Animals , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Humans , Lung Neoplasms/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , RNA, Messenger/genetics , Receptors, Aryl Hydrocarbon/genetics , Xenograft Model Antitumor Assays
12.
Nanotechnology ; 23(8): 085102, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22293282

ABSTRACT

Although zinc oxide nanoparticles (ZnONPs) have been applied in nanotechnology, their kinetics and tissue distribution in vivo are unknown. Here we compared the kinetics and tissue distribution of 10 nm (65)ZnONPs, 71 nm (65)ZnONPs and (65)Zn(NO(3))(2) in mice after intravenous injection. The areas under the curves and the half-lives in the second compartment of (65)Zn(NO(3))(2) were greater than those of (65)ZnONPs; the kinetic parameters were similar for both (65)ZnONPs. However, the tissue distributions for the three forms were different. ZnONPs preferentially accumulated in the liver and spleen at 24 h. At day 28, (65)Zn concentration was highest in bone and the proportion of recovered (65)Zn radioactivity was highest in the carcass; these had the same ranking, 10 nm (65)ZnONPs > 71 nm (65)ZnONPs>  (65)Zn(NO(3))(2). Although more than 80% of the 10 nm (65)ZnONPs had been excreted by day 28, greater amounts of the 10 nm (65)ZnONPs than the 71 nm (65)ZnONPs or (65)Zn(NO(3))(2) had accumulated in other organs (brain, lung, heart and kidneys). Zn ions seem to have a longer half-life in the plasma, but ZnONPs show greater tissue accumulation. Although the size of the ZnONPs had no obvious effect on the kinetics, nevertheless the smaller ZnONPs tended to accumulate preferentially in some organs.


Subject(s)
Nanoparticles/chemistry , Nitrates/pharmacokinetics , Zinc Compounds/pharmacokinetics , Zinc Oxide/pharmacokinetics , Animals , Kinetics , Male , Materials Testing , Metabolic Clearance Rate , Mice , Mice, Inbred ICR , Nanoparticles/radiation effects , Nanoparticles/ultrastructure , Neutrons , Nitrates/chemistry , Nitrates/radiation effects , Particle Size , Tissue Distribution , Zinc Compounds/chemistry , Zinc Compounds/radiation effects , Zinc Oxide/chemistry , Zinc Oxide/radiation effects
13.
Nanotoxicology ; 5(1): 91-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21155710

ABSTRACT

Cadmium (Cd) is a component in quantum dot 705 (QD705). Whether QD705 behaves similar to Cd in vivo is of great concern. We compared the distributional kinetics of cadmium chloride (CdCl(2)) and QD705 in mice after intravenous injection. QD705 showed a longer plasma and body retention than CdCl(2) and could be detected in the brain during early exposure. While both the liver and spleen demonstrated a constant Cd concentration for 28 days after QD705 injection, it is likely that this represents intact QD705 stored in mononuclear phagocytes. The kidneys showed a time-dependent accumulation of Cd in the QD705-exposed animals. By day 28, Cd in the kidneys from QD705 was 3-fold that of CdCl(2). QD705 and CdCl(2) have very different kinetics in distribution and metabolism. The long body retention of QD705 in the kidneys may mean that QD705 has even more renal toxicity than CdCl(2).


Subject(s)
Cadmium Chloride/pharmacokinetics , Hazardous Substances/toxicity , Metal Nanoparticles/toxicity , Quantum Dots , Animals , Cadmium Chloride/toxicity , Diagnostic Imaging/methods , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Metallothionein/metabolism , Mice , Mice, Inbred ICR , Tissue Distribution
14.
Food Chem Toxicol ; 47(7): 1703-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19406192

ABSTRACT

To investigate whether diabetes-induced alterations of CYP2E1 and oxidative stress can be modulated by dietary taurine supplementation, male Wistar rats were divided into non-diabetic, diabetic, and diabetic taurine-supplemented groups (administered at 2% in the drinking water). Increased levels of CYP2E1-catalyzed p-nitrophenol hydroxylation were found in liver and kidney microsomes of rats with STZ-induced diabetes compared to those of non-diabetic control rats. Immunoblot and RT-PCR analyses of CYP2E1 protein and mRNA levels in the liver and kidneys showed the same trend as with enzyme activities. Taurine supplementation significantly decreased the enzyme activity and expression (protein and mRNA) of CYP2E1 in diabetic rat kidneys. Plasma beta-hydroxybutyrate concentration was significantly reduced in taurine-treated diabetic rats. The induction of heme oxygenase-1 mRNA was suppressed by taurine treatment in diabetic rat kidneys. An increase in reduced glutathione (GSH) and a higher ratio of reduced to oxidized glutathione (GSH/GSSG) together with lower values of thiobarbituric acid-reactive substances (TBARS) were observed in the kidneys of taurine-treated diabetic rats. However, taurine supplementation caused only a slight or insignificant effect on these alternations in the liver of diabetic rats. Our results show dietary taurine may reduce CYP2E1 expression and activity, and oxidative stress in kidneys of diabetic rats.


Subject(s)
Cytochrome P-450 CYP2E1/metabolism , Diabetes Mellitus, Experimental/metabolism , Kidney/metabolism , Liver/metabolism , Oxidative Stress/drug effects , Taurine/pharmacology , Animals , Antioxidants/metabolism , Blood Glucose/metabolism , Blotting, Western , Body Weight/drug effects , Cholesterol/metabolism , Diabetes Mellitus, Experimental/enzymology , Dietary Supplements , Drinking/drug effects , Heme Oxygenase-1/metabolism , Kidney/drug effects , Kidney/enzymology , Lipid Peroxidation/drug effects , Lipids/blood , Liver/drug effects , Liver/enzymology , Male , Organ Size/drug effects , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/metabolism
15.
Toxicol Sci ; 107(2): 385-93, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19033395

ABSTRACT

Epidemiological evidence indicated that there was a synergistic interaction between arsenic and cigarette smoke on enhancement of lung cancer risk. Benzo[a]pyrene (B[a]P), a component in cigarette smoke, is one of the most carcinogenic compounds known. Animal studies have demonstrated that there were increased benzo[a]pyrene-7,8-diol-9,10-epoxide (BPDE) adduct formation and lung tumorigenesis in animals when they were coexposed to B[a]P and arsenic. Since BPDE adduct is a by-product of B[a]P metabolism, elevation of B[a]P metabolism by arsenic is suspected. However, the effects of arsenic on cytochrome P450 1A1 (CYP1A1) status (expression and activity), which is essential for B[a]P metabolism, either in lung cells or in lung tissues, are never demonstrated. We hypothesized that arsenic would enhance aryl hydrocarbon receptor (AhR) activation leading to CYP1A1 expression and activity in lung cells. Indeed, our present study successfully demonstrated the elevation of CYP1A1 messenger RNA expression in H1355 cells, a human lung adenocarcinoma cell line, as well as CYP1A1 expression and activity in lung tissues of arsenic-exposed mice. We further demonstrated that this elevation of CYP1A1 expression could be effectively blocked with AhR antagonist, 3',4'-dimethoxyflavone, indicating that the arsenic-induced CYP1A1 expression and activity were via AhR activation. Furthermore, we found that arsenic-induced AhR activation and -enhanced CYP1A1 expression can be further increased by a prooxidant, buthionine-(S,R)-sulfoximine, and suppressed by antioxidants, such as N-acetylcysteine and catalase. Our findings provided clear evidence that arsenic can enhance CYP1A1 expression and activity via AhR activation, and the arsenic-induced AhR activation is probably triggered by oxidative stress.


Subject(s)
Arsenic/toxicity , Cytochrome P-450 CYP1A1/biosynthesis , Lung/metabolism , Oxidative Stress/physiology , Receptors, Aryl Hydrocarbon/physiology , Animals , Arsenic/analysis , Arsenic/pharmacokinetics , Benzo(a)pyrene/metabolism , Cell Line , Cell Survival/drug effects , Genes, Reporter/genetics , In Vitro Techniques , Luciferases/genetics , Lung/chemistry , Lung/cytology , Male , Mice , Mice, Inbred ICR , Microsomes/drug effects , Microsomes/metabolism , Polychlorinated Dibenzodioxins/toxicity , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Aryl Hydrocarbon/drug effects , Receptors, Aryl Hydrocarbon/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
16.
Biochem Biophys Res Commun ; 376(3): 519-24, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18796293

ABSTRACT

Trans, trans-2,4-decadienal (tt-DDE), a specific type of dienaldehyde, is abundant in heated oils or cooking oil fumes. Ingestion of heated oils and exposure to cooking oil fumes has been suggested to have a great health impact in a variety of organs, including the lungs. Previous studies have demonstrated that acute exposures to high doses of tt-DDE have induced oxidative stress, genotoxicity, and cytotoxicity in human lung cells. The objective in utilizing proteomic techniques of this study was to identify protein biomarkers associated with tt-DDE-induced oxidative stress and cytotoxicity in human bronchial epithelial cells BEAS-2B. Experimental results suggested that DJ-1 and cofilin proteins were protein biomarkers for tt-DDE-induced cytotoxicity and oxidative stress in lung cells. DJ-1 was especially an early biomarker for tt-DDE exposure.


Subject(s)
Actin Depolymerizing Factors/metabolism , Aldehydes/toxicity , Bronchi/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Oncogene Proteins/metabolism , Oxidative Stress , Proteomics , Acetylcysteine/pharmacology , Actin Depolymerizing Factors/analysis , Biomarkers/analysis , Biomarkers/metabolism , Bronchi/cytology , Bronchi/metabolism , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Intracellular Signaling Peptides and Proteins/analysis , Oncogene Proteins/analysis , Protein Deglycase DJ-1
17.
Toxicol Sci ; 104(2): 294-302, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18480072

ABSTRACT

Some epidemiological studies suggest women may be at greater risk for lung cancer than men. Hydroxyestradiols (OHE(2)) are genotoxic and considered as carcinogenic metabolites of estrogens. In this study, we demonstrate that treatment with 0.1 or 1 nM 2/4 OHE(2) significantly increased intracellular oxidative stress, nuclear factor kappa B (NF-kappaB) activity, and cyclooxygenase-2 (COX-2) expression within 24 h in human bronchial epithelial cells BEAS-2B. Cotreatment with the NF-kappaB inhibitor, Bay 117085, prevented OHE(2)-induced COX-2 mRNA accumulation, suggesting that OHE(2) induced COX-2 expression via the NF-kappaB dependent pathway. Furthermore, cotreatment with 10nM 17-beta estradiol (E(2)) significantly enhanced OHE(2)-increased intracellular oxidative stress and significantly increased not only NF-kappaB activity but also COX-2 levels. As COX-2 participates in biosynthesis of prostaglandin E2 (PGE2), PGE2 secretion was enhanced by the cotreatment of 1 nM OHE(2) and 10nM E(2). To understand the enhancement mechanism between OHE(2) and E(2), cells were cotreated with an antioxidant, N-acetylcysteine (NAC), or NF-kB inhibitor, Bay 117085. Both NAC and Bay 117085 prevented the enhancement in COX-2 expression and PGE2 secretion by the cotreatment of E(2) and OHE(2) in BEAS-2B cells. Similarly, Bay 117085 prevented PGE2 secretion induced by the cotreatment of E(2) and OHE(2) in rat lung slice cultures. These results suggest that E(2) enhanced OHE(2)-increased intracellular oxidative stress which increased NF-kappaB activity, COX-2 expression, and PGE2 secretion. Elevated COX-2 expression and PGE2 secretion has been shown to increase the risk of cancer development. Our present data suggest a pathway that contributes an epigenetic mechanism to the overall mechanism of carcinogenesis.


Subject(s)
Bronchi/drug effects , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , NF-kappa B/metabolism , Acetylcysteine/pharmacology , Animals , Bronchi/metabolism , Cyclooxygenase 2/genetics , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Epigenesis, Genetic/drug effects , Estrogens/pharmacology , Estrogens, Catechol/pharmacology , Female , Humans , Lung/drug effects , Lung/metabolism , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Sulfones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...