Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Mater ; 20(4): 518-524, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33398117

ABSTRACT

It is commonly assumed that charge-carrier transport in doped π-conjugated polymers is dominated by one type of charge carrier, either holes or electrons, as determined by the chemistry of the dopant. Here, through Seebeck coefficient and Hall effect measurements, we show that mobile electrons contribute substantially to charge-carrier transport in π-conjugated polymers that are heavily p-doped with strong electron acceptors. Specifically, the Seebeck coefficient of several p-doped polymers changes sign from positive to negative as the concentration of the oxidizing agents FeCl3 or NOBF4 increase, and Hall effect measurements for the same p-doped polymers reveal that electrons become the dominant delocalized charge carriers. Ultraviolet and inverse photoelectron spectroscopy measurements show that doping with oxidizing agents results in elimination of the transport gap at high doping concentrations. This approach of heavy p-type doping is demonstrated to provide a promising route to high-performance n-type organic thermoelectric materials.

3.
Toxicol Appl Pharmacol ; 264(2): 153-60, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22884995

ABSTRACT

Cadmium has been widely used in industry and is known to be carcinogenic to humans. Although it is widely accepted that chronic exposure to cadmium increases the incidence of cancer, the mechanisms underlying cadmium-induced carcinogenesis are unclear. The main aim of this study was to investigate the role of reactive oxygen species (ROS) in cadmium-induced carcinogenesis and the signal transduction pathways involved. Chronic exposure of human bronchial epithelial BEAS-2B cells to cadmium induced cell transformation, as evidenced by anchorage-independent growth in soft agar and clonogenic assays. Chronic cadmium treatment also increased the potential of these cells to invade and migrate. Injection of cadmium-stimulated cells into nude mice resulted in the formation of tumors. In contrast, the cadmium-mediated increases in colony formation, cell invasion and migration were prevented by transfection with catalase, superoxide dismutase-1 (SOD1), or SOD2. In particular, chronic cadmium exposure led to activation of signaling cascades involving PI3K, AKT, GSK-3ß, and ß-catenin and transfection with each of the above antioxidant enzymes markedly inhibited cadmium-mediated activation of these signaling proteins. Inhibitors specific for AKT or ß-catenin almost completely suppressed the cadmium-mediated increase in total and active ß-catenin proteins and colony formation. Moreover, there was a marked induction of AKT, GSK-3ß, ß-catenin, and carcinogenic markers in tumor tissues formed in mice after injection with cadmium-stimulated cells. Collectively, our findings suggest a direct involvement of ROS in cadmium-induced carcinogenesis and implicate a role of AKT/GSK-3ß/ß-catenin signaling in this process.


Subject(s)
Cadmium/toxicity , Carcinogens , Glycogen Synthase Kinase 3/physiology , Oncogene Protein v-akt/physiology , Phosphatidylinositol 3-Kinases/physiology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , beta Catenin/physiology , Animals , Blotting, Western , Bronchi/cytology , Bronchi/drug effects , Cell Line , Cell Movement/drug effects , Cell Transformation, Neoplastic/drug effects , Glycogen Synthase Kinase 3 beta , Humans , Mice , Mice, Nude , Neoplasm Invasiveness/pathology , Plasmids/genetics , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Transfection , Xenograft Model Antitumor Assays
4.
Toxicol Sci ; 123(2): 399-410, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21742780

ABSTRACT

Hexavalent chromium [Cr(VI)] is a well-known human carcinogen associated with the incidence of lung cancer. Although overproduction of reactive oxygen species (ROS) has been suggested to play a major role in its carcinogenicity, the mechanisms of Cr(VI)-induced ROS production remain unclear. In this study, we investigated the role of NADPH oxidase (NOX), one of the major sources of cellular ROS, in Cr(VI)-induced oxidative stress and carcinogenesis. We found that short-term exposure to Cr(VI) (2µM) resulted in a rapid increase in ROS generation in Beas-2B cells, and concomitantly increased NOX activity and expression of NOX members (NOX1-3 and NOX5) and subunits (p22(phox), p47(phox), p40(phox), and p67(phox)). Cr(VI) also induced phosphorylation of p47(phox) and membrane translocation of p47(phox) and p67(phox), further confirming NOX activation. Knockdown of p47(phox) with a short hairpin RNA attenuated the ROS production induced by Cr(VI). Chronic exposure (up to 3 months) to low doses of Cr(VI) (0.125, 0.25, and 0.5µM) also promoted ROS generation and the expression of NOX subunits, such as p47(phox) and p67(phox), but inhibited the expression of main antioxidant enzymes, such as superoxidase dismutase (SOD) and glutathione peroxidase (GPx). Chronic Cr(VI) exposure resulted in transformation of Beas-2B cells, increasing cell proliferation, anchorage independent growth in soft agar, and forming aggressive tumors in nude mice. Stable knockdown of p47(phox) or overexpression of SOD1, SOD2, or catalase (CAT) eliminated Cr(VI)-induced malignant transformation. Our results suggest that NOX plays an important role in Cr(VI)-induced ROS generation and carcinogenesis.


Subject(s)
Carcinogens/toxicity , Chromates/toxicity , Chromium/toxicity , NADPH Oxidases/biosynthesis , Reactive Oxygen Species/metabolism , Animals , Bronchi/drug effects , Bronchi/metabolism , Bronchi/pathology , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Cell Transformation, Neoplastic/chemically induced , Enzyme Activation , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Silencing , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/pathology , Oxidative Stress/drug effects , Oxidoreductases/metabolism , Phosphorylation/drug effects , RNA, Small Interfering/genetics , Transfection , Xenograft Model Antitumor Assays
5.
Toxicol Appl Pharmacol ; 245(2): 226-35, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20298709

ABSTRACT

Cr(VI) compounds are known to cause serious toxic and carcinogenic effects. Cr(VI) exposure can lead to a severe damage to the skin, but the mechanisms involved in the Cr(VI)-mediated toxicity in the skin are unclear. The present study examined whether Cr(VI) induces cell death by apoptosis or necrosis using mouse skin epidermal cell line, JB6 Cl41 cells. We also investigated the cellular mechanisms of Cr(VI)-induced cell death. This study showed that Cr(VI) induced apoptotic cell death in a dose-dependent manner, as demonstrated by the appearance of cell shrinkage, the migration of cells into the sub-G1 phase, the increase of Annexin V positively stained cells, and the formation of nuclear DNA ladders. Cr(VI) treatment resulted in the increases of mitochondrial membrane depolarization and caspases activation. Electron spin resonance (ESR) and fluorescence analysis revealed that Cr(VI) increased intracellular levels of reactive oxygen species (ROS) such as hydrogen peroxide and superoxide anion radical in dose-dependent manner. Blockage of p53 by si-RNA transfection suppressed mitochondrial changes of Bcl-2 family composition, mitochondrial membrane depolarization, caspase activation and PARP cleavage, leading to the inhibition of Cr(VI)-induced apoptosis. Further, catalase treatment prevented p53 phosphorylation stimulated by Cr(VI) with the concomitant inhibition of caspase activation. These results suggest that Cr(VI) induced a mitochondrial-mediated and caspase-dependent apoptosis in skin epidermal cells through activation of p53, which are mainly mediated by reactive oxidants generated by the chemical.


Subject(s)
Apoptosis , Carcinogens, Environmental/toxicity , Caspases/biosynthesis , Chromium/toxicity , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/physiology , Animals , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Mice , Mitochondria/physiology
6.
Article in English | MEDLINE | ID: mdl-20176134

ABSTRACT

Although Lepomis species are abundant in a wide variety of habitats throughout North America and could serve as potentially valuable biomonitoring tools, few studies have examined the induction of pollutant biomarkers in this genus. We hypothesized that the induction of cytochrome P-450 1A (CYP1A), a sensitive and widely used indicator of response to aquatic contaminants, would serve as an effective biomarker of organic pollutant exposure in Lepomis species. We examined the response of CYP1A and two of the major pollutant-responsive phase II enzymes, glutathione S-transferase (GST), and uridine diphosphate glucuronyltransferase (UDPGT), in Lepomis exposed to organic pollutants under laboratory and field conditions. Two Lepomis species (longear sunfish, Lepomis megalottis and bluegill, Lepomis macrochirus) were exposed in the laboratory via intraperitoneal injection to corn oil (vehicle), benzo(a)pyrene (BaP) (10 and 50mg/kg), a polynuclear aromatic hydrocarbon (PAH) or 3,4,3',4'-tetrachlorobiphenyl (PCB 77) (0.1 and 1.0mg/kg), a dioxin-like planar halogenated aromatic hydrocarbon (HAH), and sacrificed 2 (BaP) or 7 (corn oil, PCB77) days later. Lepomis hepatic CYP1A exhibited differential sensitivity to these two classes of environmental contaminants. CYP1A activity was weakly induced in bluegill exposed to 1.0mg/kg PCB 77 (3 fold induction over controls) but strongly induced in both bluegill and longear sunfish exposed to 50mg/kg BaP (37 and 15 fold induction over controls, respectively). In contrast, hepatic GST activity in both species remained unchanged following the treatment with either compound and hepatic UDPGT activity, which was assessed only in BaP-treated longear sunfish, was unaffected by that chemical, indicating these phase II enzymes may not be sensitive pollutant biomarkers in this genus. Further, longear sunfish collected from a PCB contaminated site displayed relatively low levels of CYP1A activity despite PCB body burdens associated with strong induction of CYP1A activity in other fish species. The strong induction of CYP1A by BaP with much weaker CYP1A response to PCB indicates that CYP1A in Lepomis sp. could be an excellent biomarker for PAH pollution, but may not be a reliable indicator of site contamination by halogenated hydrocarbons. We conclude that Lepomis species provide a useful model for examining the regulation and potential consequences of differential pollutant sensitivity, but that CYP1A in these species should be used with caution as an indicator of halogenated contaminants.


Subject(s)
Benzo(a)pyrene/toxicity , Cytochrome P-450 CYP1A1/metabolism , Environmental Monitoring/standards , Perciformes/metabolism , Polychlorinated Biphenyls/toxicity , Water Pollutants, Chemical/toxicity , Animals , Dose-Response Relationship, Drug , Environmental Monitoring/methods
7.
Toxicol Sci ; 113(1): 127-37, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19887573

ABSTRACT

Cadmium is a toxic heavy metal and has been widely used in industry. The skin is an important target for this metal. The mechanisms by which cadmium leads to damage to the skin are unclear at present. The aims of this study were to examine whether cadmium induces apoptosis in mouse skin epidermal cell line, JB6 Cl41 cells, and to investigate the cellular mechanisms by which cadmium causes cytotoxicity in the cells. The present study showed that cadmium induced cell death by apoptosis in a dose-dependent manner, as proven by the appearance of cell shrinkage, the increase of Annexin V positively stained cells, and the formation of nuclear DNA ladders. Cadmium-induced apoptosis involved a mitochondria-mediated mechanism but not caspase-dependent pathway in that the critical apoptotic events induced by cadmium, such as the decrease of Bcl-2/Bcl-xL, the increase of GADD45alpha, and the nuclear translocation of apoptosis inducing factor, were not affected by the inhibition of executive caspases. In contrast, blockage of p53 and JNK by pharmacological inhibitors or small interference RNA transfection suppressed the cadmium-induced apoptosis with the concomitant inhibition of antiapoptotic Bcl-2 family proteins and GADD45alpha, respectively. Furthermore, the activation of p53 and JNK and their downstream proteins in cadmium-exposed cells were inhibited by individual treatment with catalase and Bapta-acetoxymethyl. These results suggest that cadmium induces apoptosis via the activation of JNK- and p53-mediated signaling, where calcium ion and hydrogen peroxide act as the pivotal mediators of the apoptotic signaling.


Subject(s)
Apoptosis/drug effects , Cadmium Chloride/toxicity , Calcium/metabolism , Epidermis/drug effects , Hydrogen Peroxide/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antioxidants/pharmacology , Apoptosis Inducing Factor/metabolism , Caspases/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Dose-Response Relationship, Drug , Epidermis/enzymology , Epidermis/pathology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/pathology , Nuclear Proteins/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Signal Transduction/drug effects , Transfection , Tumor Suppressor Protein p53/genetics , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...