Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Sci Rep ; 6: 26091, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27184933

ABSTRACT

As a sensor of polyaromatic chemicals the aryl hydrocarbon receptor (AhR) exerts an important role in immune regulation besides its requirement for xenobiotic metabolism. Transcriptional activation of AhR target genes is counterregulated by the AhR repressor (AhRR) but the exact function of the AhRR in vivo is currently unknown. We here show that the AhRR is predominantly expressed in immune cells of the skin and intestine, different from other AhR target genes. Whereas AhRR antagonizes the anti-inflammatory function of the AhR in the context of systemic endotoxin shock, AhR and AhRR act in concert to dampen intestinal inflammation. Specifically, AhRR contributes to the maintenance of colonic intraepithelial lymphocytes and prevents excessive IL-1ß production and Th17/Tc17 differentiation. In contrast, the AhRR enhances IFN-γ-production by effector T cells in the inflamed gut. Our findings highlight the physiologic importance of cell-type specific balancing of AhR/AhRR expression in response to microbial, nutritional and other environmental stimuli.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Enteritis/pathology , Gene Expression Regulation , Receptors, Aryl Hydrocarbon/biosynthesis , Repressor Proteins/metabolism , Animal Structures/enzymology , Animal Structures/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/genetics
2.
FEBS J ; 280(24): 6681-90, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24127753

ABSTRACT

The aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor that mediates the toxicity of dioxins, polycyclic aromatic hydrocarbons and related environmental pollutants. Besides drug metabolism, several studies have provided evidence that the AHR and its downstream targets trigger important developmental, physiological and pathophysiological processes. However, in contrast to the molecular mechanisms of AHR-dependent signaling pathways, the transcriptional regulation of the AHR gene itself is as yet only marginally understood. We found that the pleiotropic interleukin (IL)-6-type cytokine oncostatin M (OSM) is an inducer of AHR mRNA and protein expression in human HepG2 hepatocarcinoma cells. Analyses of the human AHR promoter revealed the existence of a putative signal transducer and activator of transcription (STAT)-binding element 5'-upstream of the transcription start site. By means of site-directed mutagenesis, inhibitor experiments and electrophoretic mobility shift assays, we demonstrated that this STAT motif is recognized by STAT3 to regulate basal and cytokine-inducible AHR expression in HepG2 cells. The identification of the AHR as a downstream target of IL-6-type cytokine-stimulated STAT3 signaling may contribute to a better understanding of the multiple facets of AHR during development, physiology and disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Interleukin-6/metabolism , Oncostatin M/metabolism , Promoter Regions, Genetic/genetics , Receptors, Aryl Hydrocarbon/genetics , STAT3 Transcription Factor/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Hep G2 Cells , Humans , Luciferases/metabolism , Mutagenesis, Site-Directed , Oncostatin M/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Aryl Hydrocarbon/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics
3.
PLoS One ; 8(9): e74917, 2013.
Article in English | MEDLINE | ID: mdl-24069365

ABSTRACT

Khellin and visnagin are two furanochromones that can be frequently found in ethnomedical formulations in Asia and the Middle East. Both compounds possess anti-inflammatory and analgesic properties, therefore modern medicine uses these compounds or structurally related derivatives for treatment of vitiligo, bronchial asthma and renal colics. Despite their frequent usage, the potential toxic properties of visnagin and khellin are not well characterized up-to-now. Many natural compounds modulate the expression and activity of cytochrome P450 1A1 (CYP1A1), which is well-known to bioactivate pro-carcinogens. The expression of this enzyme is controlled by the aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor and regulator of drug metabolism. Here, we investigated the influence of both furanochromones on AHR signaling in human HepG2 hepatocarcinoma cells and primary human hepatocytes. Both compounds transactivated xenobiotic response element (XRE)-driven reporter gene activity in a dose-dependent manner and induced CYP1A1 transcription in HepG2 cells and primary hepatocytes. The latter was abolished in presence of a specific AHR antagonist. CYP1A enzyme activity assays done in HepG2 cells and primary hepatocytes revealed an inhibition of enzyme activity by both furanochromones, which may become relevant regarding the metabolism of xenobiotics and co-administered therapeutic drugs. The observed induction of several other members of the AHR gene battery, whose gene products are involved in regulation of cell growth, differentiation and migration, indicates that a further toxicological characterization of visnagin and khelllin is urgently required in order to minimize potential drug-drug interactions and other toxic side-effects that may occur during therapeutic usage of these furanochromones.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Khellin/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/drug effects , Cell Line , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Enzyme Activation/drug effects , Gene Expression , Gene Expression Regulation/drug effects , Genes, Reporter , Humans
4.
Exp Dermatol ; 22(5): 349-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23614742

ABSTRACT

Findings from large epidemiologic studies indicate that there is a link between smoking and extrinsic skin ageing. We previously reported that matrix metalloproteinases (MMPs) mediate connective tissue damage in skin exposed to tobacco smoke extracts. Tobacco smoke contains more than 3800 constituents, including numerous water-insoluble polycyclic aromatic hydrocarbons (PAHs) that trigger aryl hydrocarbon receptor (AhR) signalling pathways. To analyse the molecular mechanisms involved in tobacco smoke-induced skin ageing, we exposed primary human fibroblasts and keratinocytes to tobacco smoke extracts. Hexane- and water-soluble tobacco smoke extracts significantly induced MMP-1 mRNA in both human cultured fibroblasts and keratinocytes in a dose-dependent manner. To clarify the involvement of the AhR pathway, we used a stable AhR-knockdown HaCaT cell line. AhR knockdown abolished the increased transcription of the AhR-dependent genes CYP1A1/CYP1B1 and MMP-1 induced by either of the tobacco smoke extracts. Furthermore, the tobacco smoke extracts induced 7-ethoxyresorufin-O-deethylase activity, which was almost completely abolished by AhR knockdown. Likewise, treating fibroblasts with AhR pathway inhibitors, that is, the flavonoids 3-methoxy-4-nitroflavone and α-naphthoflavone, blocked the expression of CYP1B1 and MMP-1. These findings suggest that the tobacco smoke extracts induce MMP-1 expression in human fibroblasts and keratinocytes via activation of the AhR pathway. Thus, the AhR pathway may be pathogenetically involved in extrinsic skin ageing.


Subject(s)
Keratinocytes/drug effects , Keratinocytes/physiology , Matrix Metalloproteinase 1/genetics , Receptors, Aryl Hydrocarbon/genetics , Skin Aging/physiology , Tobacco Smoke Pollution/adverse effects , Aryl Hydrocarbon Hydroxylases/genetics , Cell Line , Cells, Cultured , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1B1 , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , Hexanes/pharmacology , Humans , Keratinocytes/cytology , Matrix Metalloproteinase 1/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Skin Aging/drug effects , Solubility
5.
J Invest Dermatol ; 133(1): 87-96, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22951721

ABSTRACT

The skin reacts to environmental noxae by inducing cytochrome P450 (CYP)-catalyzed reactions via activation of the aryl hydrocarbon receptor (AhR). A drawback of this response is the generation of oxidative stress, which is especially dangerous for postreplicative cells such as dermal fibroblasts, in which damage may accumulate over time. Accordingly, in dermal fibroblasts, CYP1 expression is repressed and it has been proposed that this is due to the AhR repressor (AhRR), which is supposedly overexpressed in fibroblasts as compared with other skin cells. Here, we revisited this "AhRR hypothesis", which has been mainly based on ectopic overexpression studies and correlation analyses of high AhRR gene expression with CYP1A1 repression in certain cell types. In primary human skin fibroblasts (NHDFs) of 25 individuals, we found that (i) the AhRR was expressed only at moderate RNA copy numbers and that, against the common view, (ii) in some fibroblast strains, CYP1A1 mRNA expression could be induced by AhR activators. However, even the highest induction did not translate into measurable CYP1 enzyme activity, and neither basal expression nor mRNA inducibility correlated with AhRR expression. In addition, enhancement of CYP1A1 mRNA expression by trichostatin A, which inhibits AhRR-recruited histone deacetylases at the CYP1A1 promoter, failed to induce measurable CYP1 activity. Finally, AhRR-deficient ((-/-)) mouse embryonic fibroblasts were not induced to biologically relevant CYP1 enzyme activity despite impressive mRNA induction. These data clearly indicate that repressed CYP1 activity in NHDFs is not causally related to AhRR expression, which may serve a different, yet unknown, biological function.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cytochrome P-450 CYP1A1/metabolism , Fibroblasts/metabolism , Repressor Proteins/biosynthesis , Skin/metabolism , Adult , Animals , Benzo(a)pyrene/pharmacology , Cells, Cultured , Cytochrome P-450 CYP1A1/genetics , Female , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Methylcholanthrene/pharmacology , Mice , Middle Aged , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/agonists , Skin/drug effects , Young Adult
6.
Exp Dermatol ; 21(5): 364-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22509834

ABSTRACT

The 7th Amendment to the EU Cosmetics Directive prohibits the use of animals in cosmetic testing for certain endpoints, such as genotoxicity. Therefore, skin in vitro models have to replace chemical testing in vivo. However, the metabolic competence neither of human skin nor of alternative in vitro models has so far been fully characterized, although skin is the first-pass organ for accidentally or purposely (cosmetics and pharmaceuticals) applied chemicals. Thus, there is an urgent need to understand the xenobiotic-metabolizing capacities of human skin and to compare these activities to models developed to replace animal testing. We have measured the activity of the phase II enzymes glutathione S-transferase, UDP-glucuronosyltransferase and N-acetyltransferase in ex vivo human skin, the 3D epidermal model EpiDerm 200 (EPI-200), immortalized keratinocyte-based cell lines (HaCaT and NCTC 2544) and primary normal human epidermal keratinocytes. We show that all three phase II enzymes are present and highly active in skin as compared to phase I. Human skin, therefore, represents a more detoxifying than activating organ. This work systematically compares the activities of three important phase II enzymes in four different in vitro models directly to human skin. We conclude from our studies that 3D epidermal models, like the EPI-200 employed here, are superior over monolayer cultures in mimicking human skin xenobiotic metabolism and thus better suited for dermatotoxicity testing.


Subject(s)
Acetyltransferases/metabolism , Epidermis/metabolism , Glucuronosyltransferase/metabolism , Glutathione Transferase/metabolism , Keratinocytes/metabolism , Models, Biological , Skin/metabolism , Xenobiotics/metabolism , Animal Testing Alternatives , Cell Line , Cells, Cultured , Cosmetics , Dermotoxins , Epidermal Cells , Humans , In Vitro Techniques , Keratinocytes/cytology , Skin/cytology , Skin/drug effects , Toxicology
7.
J Invest Dermatol ; 132(1): 7-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22158605

ABSTRACT

The ligand-activated transcription factor AhR mediates the cutaneous stress response toward a variety of environmental noxae and is therefore currently of interest for modern preventive medicine. In this issue, Tsuji et al. identify the antifungal agent ketoconazole as an inducer of AhR signaling and the Nrf2 antioxidant response in human keratinocytes. Ketoconazole-stimulated nuclear translocation of Nrf2 and its cytoprotective effects against oxidative stress strongly depend on a functional AhR. This newly identified AhR-Nrf2 pathway opens up new opportunities to prevent and treat inflammatory skin diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , Ketoconazole/pharmacology , NF-E2-Related Factor 2/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Humans
8.
Toxicol Lett ; 203(3): 258-64, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21458546

ABSTRACT

Benzo[a]pyrene (B[a]P) and related procarcinogens found in cigarette smoke and roasted foodstuff require metabolic activation to build mutagenic DNA adducts that may cause tumor diseases like colorectal cancer. The major B[a]P-activating enzymes belong to the cytochrome-P450 (CYP)-1 family and are regulated by the aryl hydrocarbon receptor (AhR). Previous studies have indicated that an inhibition of AhR is accompanied with a reduced metabolic activation of B[a]P and therefore may act protective against carcinogenesis. We investigated if the green tea flavonoid (-)-epigallocatechin-3-gallate (EGCG), a known AhR inhibitor, is able to influence B[a]P-metabolizing and B[a]P-transporting enzymes in human Caco-2 colon carcinoma cells. Strikingly, treatment with EGCG did neither affect constitutive and B[a]P-inducible expression of CYP1A1 and UDP-glucuronosyltransferase (UGT)-1A1 nor overall CYP1 and UGT enzyme activities, indicating that EGCG does not antagonize the AhR in Caco-2 cells. Since flavonoids were also identified to enhance the activity of B[a]P-carrying transporter, we analyzed if EGCG exposure alters cellular excretion of B[a]P conjugates. In contrast to the positive control fisetin, EGCG did not affect cellular excretion of B[a]P metabolites. Our data provide evidence that EGCG does not alter the metabolism and transport of B[a]P in Caco-2 cells, and thus may not protect against procarcinogenic food contaminants.


Subject(s)
Benzo(a)pyrene/metabolism , Catechin/analogs & derivatives , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Biotransformation , Caco-2 Cells , Catechin/pharmacology , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Flavonoids/pharmacology , Flavonols , Glucuronosyltransferase/genetics , Humans , RNA, Messenger/analysis
9.
Biol Chem ; 391(11): 1235-48, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20868221

ABSTRACT

Depending on their chemical structure and properties, environmental chemicals and other xenobiotics that enter the cell can affect cellular function by either nonselective binding to cellular macromolecules or by interference with cellular receptors, which would initiate a more defined cell biological response. One of these intracellular chemosensor molecules is the aryl hydrocarbon receptor (AhR), a transcription factor of the bHLH/PAS family that is known to mediate the biochemical and toxic effects of dioxins, polyaromatic hydrocarbons and related compounds. Numerous investigations have revealed that the AhR is not only a master regulator of drug metabolism activated by anthropogenic chemicals, but is also triggered by natural and endogenous ligands and can influence cell biological endpoints such as growth and differentiation. Cutting-edge research has identified new intriguing functions of the AhR, such as during proteasomal degradation of steroid hormone receptors, the cellular UVB stress response and the differentiation of certain T-cell subsets. In this review we provide both a survey of the fundamental basics of AhR biology and an insight into new functional aspects of AhR signaling to further stimulate research on this intriguing transcription factor at the interface between toxicology, cell biology and immunology.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins , Receptors, Aryl Hydrocarbon/physiology , Signal Transduction , Xenobiotics , Alkaloids/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caenorhabditis elegans , Cell Differentiation/drug effects , Cytochrome P-450 Enzyme System/metabolism , Dioxins/pharmacology , Drosophila melanogaster , Environmental Pollutants/toxicity , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Humans , Hydrocarbons, Aromatic/pharmacology , Hydrocarbons, Halogenated/pharmacology , Indoles/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Rats , Receptors, Aryl Hydrocarbon/chemistry , Signal Transduction/drug effects , Signal Transduction/radiation effects , Structure-Activity Relationship , Transcription Factors/metabolism , Ubiquitination , Ultraviolet Rays , Xenobiotics/metabolism , Xenobiotics/pharmacology
10.
Free Radic Biol Med ; 49(4): 597-605, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20570722

ABSTRACT

Cell-membrane-dependent proliferative signal transduction activated by ultrafine carbon particles in lung epithelial cells involves the specific induction of Akt and ERK1/2 phosphorylation. Particle-induced generation of reactive oxygen species (ROS) and oxidative stress are regarded as initial molecular mechanisms leading to the induction of diverse cellular responses. Therefore, we aimed to analyze the ROS dependence of the induced activation of the Akt/ERK1/2 signaling pathway upon exposure to ultrafine particulate matter (UPM). For this, ultrafine carbon black (ufCB) and ferric sulfate (FS) were used as a model representing the carbonaceous core and a nonparticulate Fenton-reactive transition metal salt often found in combustion-derived UPM. Cell-free as well as intracellular particle-induced ROS generation was assessed and related to the induced Akt and ERK1/2 phosphorylation by inhibiting oxidative stress with catalase, superoxide dismutase, and N-acetylcysteine. We show here that the activation of this signal transduction pathway was mainly due to intracellular, rather than extracellular, ROS production induced by both ufCB and FS. Further inhibitor studies on the role of cell membrane receptors pointed to the epidermal growth factor receptor as a common mediator for particle- as well as transition metal-induced signaling, whereas integrin-dependent Akt and ERK1/2 activation seems to be particle-specific.


Subject(s)
Carbon/chemistry , Cell Membrane/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Animals , Cells, Cultured , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Ferric Compounds/chemistry , Oxidative Stress , Particle Size , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats
11.
Environ Health Perspect ; 118(11): 1571-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20570779

ABSTRACT

BACKGROUND: Because of their lipophilicity, persistent organic pollutants (POPs) cross the human placenta, possibly affecting central nervous system development. Most POPs are known aryl hydrocarbon receptor (AhR) ligands and activators of AhR signaling. Therefore, AhR activation has been suggested to cause developmental neurotoxicity (DNT). OBJECTIVE: We studied the effects of AhR ligands on basic processes of brain development in two comparative in vitro systems to determine whether AhR-activation is the underlying mechanism for reported DNT of POPs in humans. METHODS: We employed neurosphere cultures based on human neural progenitor cells (hNPCs) and wild-type and AhR-deficient mouse NPCs (mNPCs) and studied the effects of different AhR agonists [3-methylcholanthrene (3-MC), benzo(a)pyrene [B(a)P], and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)] and an antagonist [3'-methoxy-4'-nitroflavone (MNF)] on neurosphere development. Moreover, we analyzed expression of AhR and genes involved in AhR signaling. RESULTS: In contrast to wild-type mNPCs, hNPCs and AhR-deficient mNPCs were insensitive to AhR agonism or antagonism. Although AhR modulation attenuated wild-type mNPC proliferation and migration, hNPCs and AhR-deficient mNPCs remained unaffected. Results also suggest that species-specific differences resulted from nonfunctional AhR signaling in hNPCs. CONCLUSION: Our findings suggest that in contrast to wild-type mNPCs, hNPCs were protected against polycyclic aromatic hydrocarbon-induced DNT because of an absence of AhR This difference may contribute to species-specific differences in sensitivity to POPs.


Subject(s)
Environmental Pollutants/toxicity , Neurons/drug effects , Polycyclic Aromatic Hydrocarbons/toxicity , Receptors, Aryl Hydrocarbon/metabolism , Stem Cells/drug effects , Animals , Benzo(a)pyrene/toxicity , Cells, Cultured , Gene Expression/drug effects , Humans , Methylcholanthrene/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System/drug effects , Nervous System/growth & development , Nervous System/metabolism , Neurons/metabolism , Polychlorinated Dibenzodioxins/toxicity , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction/drug effects , Species Specificity , Stem Cells/metabolism
12.
Arch Toxicol ; 84(8): 619-29, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20449727

ABSTRACT

The dioxin receptor, also known as arylhydrocarbon receptor (AhR), is a ligand-activated transcription factor that mediates the toxicity of dioxins and related environmental contaminants. In addition, there is a growing list of natural compounds, mainly plant polyphenols that can modulate AhR function and downstream signaling with quite unknown consequences for cellular function. We investigate the potential of four different beta-carboline alkaloids to stimulate AhR signaling in human hepatoma cells and keratinocytes. Three test substances, namely rutaecarpine, annomontine and xestomanzamine A, increase AhR-driven reporter gene activity as well as expression of two AhR target genes in a dose-dependent and time-dependent manner. Additionally, the three test alkaloids stimulate cytochrome P450 (CYP) 1 enzyme activity without showing any antagonistic effects regarding benzo(a)pyrene-stimulated CYP1 activation. The AhR-activating property of the beta-carbolines is completely abrogated in AhR-deficient cells providing evidence that rutaecarpine, annomontine and xestomanzamine A are natural stimulators of the human AhR. The toxicological relevance of beta-carboline-mediated AhR activation is discussed.


Subject(s)
Alkaloids/toxicity , Carbolines/toxicity , Gene Expression Regulation/drug effects , Receptors, Aryl Hydrocarbon/metabolism , Alkaloids/chemistry , Carbolines/chemistry , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Gene Expression/drug effects , Hep G2 Cells , Humans , Indole Alkaloids/toxicity , Pyrimidines/toxicity , Quinazolines/toxicity
13.
Arch Biochem Biophys ; 498(2): 111-8, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20403331

ABSTRACT

We investigated the effect of luteolin, a plant-derived flavonoid, on benzo(a)pyrene (B(a)P)-stimulated drug metabolism and transport in human colon carcinoma cells. While luteolin treatment inhibited B(a)P-induced expression and activity of arylhydrocarbon receptor-dependent cytochrome P450 enzymes, the overall activity of UDP-glucuronosyltransferases and sulfotransferases was not affected by luteolin, indicating that luteolin affects phase-I but not phase-II function. Luteolin exposure decreased apical transport of B(a)P metabolites due to its interaction with the transporter breast cancer resistance protein. Inhibitor studies provide a first clue to the mechanism of luteolin-mediated inhibition of this transporter. The inhibition of both phase-I metabolism as well as phase-III transport by luteolin resulted in a 3-fold intracellular accumulation of radioactively labeled B(a)P. Our data reveal that luteolin is able to interfere with crucial steps of drug metabolism and thereby enhances the bioavailability of B(a)P. These findings are of special importance regarding future benefit-risk evaluations of preventive flavonoid usage.


Subject(s)
Benzo(a)pyrene/pharmacokinetics , Colonic Neoplasms/metabolism , Luteolin/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , Benzo(a)pyrene/pharmacology , Biological Availability , Biological Transport/drug effects , Caco-2 Cells , Cytochrome P-450 Enzyme System/biosynthesis , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glucuronosyltransferase/biosynthesis , Humans , Neoplasm Proteins/biosynthesis , Receptors, Aryl Hydrocarbon/metabolism , Sulfotransferases/biosynthesis
14.
Environ Health Perspect ; 118(4): 572-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20368126

ABSTRACT

BACKGROUND: Polybrominated diphenyl ethers (PBDEs) are persistent and bioaccumulative flame retardants, which are found in rising concentrations in human tissues. They are of concern for human health because animal studies have shown that they possess the potential to be developmentally neurotoxic. OBJECTIVE: Because there is little knowledge of the effects of PBDEs on human brain cells, we investigated their toxic potential for human neural development in vitro. Moreover, we studied the involvement of thyroid hormone (TH) disruption in the effects caused by PBDEs. METHODS: We used the two PBDE congeners BDE-47 and BDE-99 (0.1-10 microM), which are most prominent in human tissues. As a model of neural development, we employed primary fetal human neural progenitor cells (hNPCs), which are cultured as neurospheres and mimic basic processes of brain development in vitro: proliferation, migration, and differentiation. RESULTS: PBDEs do not disturb hNPC proliferation but decrease migration distance of hNPCs. Moreover, they cause a reduction of differentiation into neurons and oligodendrocytes. Simultaneous exposure with the TH receptor (THR) agonist triiodothyronine rescues these effects on migration and differentiation, whereas the THR antagonist NH-3 does not exert an additive effect. CONCLUSION: PBDEs disturb development of hNPCs in vitro via endocrine disruption of cellular TH signaling at concentrations that might be of relevance for human exposure.


Subject(s)
Brain/cytology , Brain/drug effects , Halogenated Diphenyl Ethers/toxicity , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Flame Retardants/toxicity , Humans , Immunohistochemistry , Microscopy, Fluorescence , Neurons/drug effects
15.
Environ Health Perspect ; 117(7): 1131-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19654924

ABSTRACT

BACKGROUND: Developmental neurotoxicity (DNT) of environmental chemicals is a serious threat to human health. Current DNT testing guidelines propose investigations in rodents, which require large numbers of animals. With regard to the "3 Rs" (reduction, replacement, and refinement) of animal testing and the European regulation of chemicals [Registration, Evaluation, and Authorisation of Chemicals (REACH)], alternative testing strategies are needed in order to refine and reduce animal experiments and allow faster and less expensive screening. OBJECTIVES: The goal of this study was to establish a three-dimensional test system for DNT screening based on human fetal brain cells. METHODS: We established assays suitable for detecting disturbances in basic processes of brain development by employing human neural progenitor cells (hNPCs), which grow as neurospheres. Furthermore, we assessed effects of mercury and oxidative stress on these cells. RESULTS: We found that human neurospheres imitate proliferation, differentiation, and migration in vitro. Exposure to the proapoptotic agent staurosporine further suggests that human neurospheres possess functioning apoptosis machinery. The developmental neurotoxicants methylmercury chloride and mercury chloride decreased migration distance and number of neuronal-like cells in differentiated hNPCs. Furthermore, hNPCs undergo caspase-independent apoptosis when exposed toward high amounts of oxidative stress. CONCLUSIONS: Human neurospheres are likely to imitate basic processes of brain development, and these processes can be modulated by developmental neurotoxicants. Thus, this three-dimensional cell system is a promising approach for DNT testing.


Subject(s)
Neurogenesis/drug effects , Apoptosis/drug effects , Brain/cytology , Brain/embryology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Fetus/cytology , Fetus/embryology , Flow Cytometry , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mercury/toxicity , Methylmercury Compounds/toxicity
16.
Am J Respir Crit Care Med ; 180(1): 29-35, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19324973

ABSTRACT

RATIONALE: Inflammatory reactions of the airways induced by nanoparticles of occupational and environmental origin contribute to organ-specific and systemic human diseases. Because this kind of exposure in modern societies is often unavoidable, a strategy of molecular prevention on an individual level could help to prevent inflammation-derived secondary diseases. OBJECTIVES: To test whether the compatible solute ectoine [(S)-2-methyl-1,4,5,6-tetrahydropyrimidine-4-carboxylic acid], which is known to reduce cell stress effects on a molecular level, prevents nanoparticle-induced lung inflammation. METHODS: Inflammatory parameters were studied in Fischer 344 rats treated with model carbon nanoparticles. The molecular effects of ectoin on proinflammatory signal transduction were demonstrated in the rat and in the human system using cultured lung epithelial cells. MEASUREMENTS AND MAIN RESULTS: Ectoine, given with or before the nanoparticles, dose-dependently reduced neutrophil inflammation in the lung. This preventive effect was not observed when lung inflammation was induced by bacterial lipopolysaccharide. Analyses of the underlying mode of action revealed that ectoine acted on lung epithelial cells. Ectoine administration inhibited nanoparticle-induced signaling, which is known to be responsible for proinflammatory reactions in rat lung epithelial cells in vitro as well as in vivo. These findings were corroborated and extended in experiments with cultured human bronchial epithelial cells in which ectoine inhibited nanoparticle-triggered cell signaling and IL-8 induction. CONCLUSIONS: Because compatible solutes are compliant natural products without known toxic potential, we propose that this group of substances may be used for the prevention of particle-induced airway inflammation in humans.


Subject(s)
Acute Lung Injury/prevention & control , Air Pollutants/adverse effects , Amino Acids, Diamino/administration & dosage , Epithelial Cells/drug effects , Mitogen-Activated Protein Kinase Kinases/drug effects , Respiratory System Agents/administration & dosage , Acute Lung Injury/etiology , Acute Lung Injury/immunology , Animals , Bronchi/cytology , Cells, Cultured , Disease Models, Animal , Epithelial Cells/immunology , Female , Humans , Interleukin-8/drug effects , Nanoparticles/adverse effects , Neutrophils/immunology , Rats , Rats, Inbred F344 , Signal Transduction/drug effects , Vehicle Emissions
17.
Biochem Pharmacol ; 77(4): 508-20, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-18848820

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a widespread environmental pollutant, which causes a variety of severe health effects, e.g. immunosuppression, hepatotoxicity, and carcinogenesis. The main mediator of TCDD toxicity is the arylhydrocarbon receptor (AhR), which, upon activation, translocates into the nucleus and enforces gene expression. Since most of the pleiotropic effects caused by TCDD are associated with alterations in cell growth and differentiation, the analysis of the interference of the AhR with factors controlling these cellular functions seems to be a promising target regarding the prevention and treatment of chemical-provoked diseases. Cell growth and differentiation are regulated by numerous growth factors and cytokines. These multifunctional peptides promote or inhibit cell growth and regulate differentiation and other cellular processes, depending on cell-type and developmental stage. They are involved in the regulation of a broad range of physiological processes, including immune response, hematopoiesis, neurogenesis, and tissue remodeling. The complex network of growth factors and cytokines is accurately regulated and disturbances of this system are associated with adverse health effects. The molecular mechanisms by which the AhR interferes with this signaling network are multifaceted and the physiological consequences of this cross-talk are quite enigmatic. The investigation of this complex interaction is an exciting task, especially with respect to the recently described non-genomic and/or ligand-independent activities of AhR. Therefore, we summarize the current knowledge about the interaction of the AhR with three cytokine-/growth factor-related signal transducers -- the epidermal growth factor (EGF) family, tumor necrosis factor-alpha (TNF-alpha), and transforming growth factor-beta (TGF-beta) -- with regard to pathophysiological findings.


Subject(s)
Cytokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Cytokine/metabolism , Receptors, Growth Factor/metabolism , Signal Transduction/physiology , Animals , Humans , Ligands , Receptor Cross-Talk , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction/drug effects
18.
J Toxicol Environ Health A ; 71(13-14): 887-97, 2008.
Article in English | MEDLINE | ID: mdl-18569591

ABSTRACT

Tobacco smoking, alcohol drinking, and occupational exposures to polycyclic aromatic hydrocarbons are the major proven risk factors for human head and neck squamous-cell cancer (HNSCC). Major research focus on gene-environment interactions concerning HNSCC has been on genes encoding enzymes of metabolism for tobacco smoke constituents and repair enzymes. To investigate the role of genetically determined individual predispositions in enzymes of xenobiotic metabolism and in repair enzymes under the exogenous risk factor tobacco smoke in the carcinogenesis of HNSCC, we conducted a case-control study on 312 cases and 300 noncancer controls. We focused on the impact of 22 sequence variations in CYP1A1, CYP1B1, CYP2E1, ERCC2/XPD, GSTM1, GSTP1, GSTT1, NAT2, NQO1, and XRCC1. To assess relevant main and interactive effects of polymorphic genes on the susceptibility to HNSCC we used statistical models such as logic regression and a Bayesian version of logic regression. In subgroup analysis of nonsmokers, main effects in ERCC2 (Lys751Gln) C/C genotype and combined ERCC2 (Arg156Arg) C/A and A/A genotypes were predominant. When stratifying for smokers, the data revealed main effects on combined CYP1B1 (Leu432Val) C/G and G/G genotypes, followed by CYP1B1 (Leu432Val) G/G genotype and CYP2E1 (-70G>T) G/T genotype. When fitting logistic regression models including relevant main effects and interactions in smokers, we found relevant associations of CYP1B1 (Leu432Val) C/G genotype and CYP2E1 (-70G>T) G/T genotype (OR, 10.84; 95% CI, 1.64-71.53) as well as CYP1B1 (Leu432Val) G/G genotype and GSTM1 null/null genotype (OR, 11.79; 95% CI, 2.18-63.77) with HNSCC. The findings underline the relevance of genotypes of polymorphic CYP1B1 combined with exposures to tobacco smoke.


Subject(s)
Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , Polymorphism, Genetic/genetics , Xenobiotics/metabolism , Age Distribution , Base Sequence , Bayes Theorem , Case-Control Studies , DNA Repair/genetics , Female , Genetic Predisposition to Disease , Genotype , Humans , Logistic Models , Male , Middle Aged , Sex Characteristics , Smoking , Toxicogenetics
19.
J Toxicol Environ Health A ; 71(11-12): 803-15, 2008.
Article in English | MEDLINE | ID: mdl-18569579

ABSTRACT

Tobacco smoke and occupational exposures to chemicals such as polycyclic aromatic hydrocarbons (PAHs) are, aside from alcohol, the major risk factors for development of head and neck squamous-cell cancer (HNSCC). In this study, new statistical methods were applied. We employ new statistical methods to detect genetic interactions perhaps of higher order, that might play a role in developing HNSCC. The underlying study comprises 312 HNSCC cases and 300 controls. Single-nucleotide polymorphisms (SNPs) of PAH metabolizing and repair enzymes, somatic p53 mutations, and tobacco smoke were examined. Key statistical tools for our analysis are methods of unsupervised and supervised learning. In unsupervised learning, one performs cluster analyses based on well-known and new distance measures to find differences in the SNP patterns of cases and controls, and to understand the role of p53. Our main goal in supervised learning was to identify SNPs and SNP interactions that are likely to alter the susceptibility to HNSCC. Logic regression, a classification method well suited for SNPs, was employed as well as a Bayesian generalization that allows for incorporating additional expert knowledge. These methods detected several important interactions, such as an association between CYP1B1, tobacco smokes and p53 mutations and some interactions between CYP1B1 and glutathione S-transferases in smokers, which included a three-way interaction between CYP1B1, CYP2E1-70G>T, and GSTP1 (exon 5).


Subject(s)
Carcinoma, Squamous Cell/genetics , Genetic Predisposition to Disease , Head and Neck Neoplasms/genetics , Models, Statistical , Aryl Hydrocarbon Hydroxylases/genetics , Bayes Theorem , Carcinoma, Squamous Cell/blood , Case-Control Studies , Cluster Analysis , Cytochrome P-450 CYP1B1 , DNA, Neoplasm/blood , Data Interpretation, Statistical , Female , Genes, p53/genetics , Glutathione Transferase/genetics , Head and Neck Neoplasms/blood , Humans , Logistic Models , Male , Middle Aged , Polymorphism, Single Nucleotide , Smoking , Tobacco Use Disorder/genetics
20.
Am J Physiol Lung Cell Mol Physiol ; 294(2): L358-67, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18083769

ABSTRACT

Treatment of lung epithelial cells with different kinds of nano-sized particles leads to cell proliferation. Because bigger particles fail to induce this reaction, it is suggested that the special surface properties, due to the extremely small size of these kinds of materials, is the common principle responsible for this specific cell reaction. Here the activation of the protein kinase B (Akt) signaling cascade by carbon nanoparticles was investigated with regard to its relevance for proliferation. Kinetics and dose-response experiments demonstrated that Akt is specifically activated by nanoparticulate carbon particles in rat alveolar type II epithelial cells as well as in human bronchial epithelial cells. This pathway appeared to be dependent on epidermal growth factor receptor and beta(1)-integrins. The activation of Akt by these receptors is known to be a feature of adhesion-dependent signaling. However, intracellular proteins described in this context (focal adhesion kinase pp125(FAK) and integrin-linked kinase) were not activated, indicating a specific signaling mechanism. Inhibitor studies demonstrate that nanoparticle-induced proliferation is mediated by phosphoinositide 3-kinases and Akt. Moreover, overexpression of mutant Akt, as well as pretreatment with an Akt inhibitor, reduced nanoparticle-specific ERK1/2 phosphorylation, which is decisive for nanoparticle-induced proliferation. With this report, we describe the activation of a pathway by carbon nanoparticles that was so far known to be triggered by ligand receptor binding or on cell adhesion to extracellular matrix proteins.


Subject(s)
Carbon/pharmacology , Epithelial Cells/cytology , Lung/cytology , Lung/enzymology , Nanoparticles , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cell Surface/metabolism , Animals , Bronchi/cytology , Bronchi/drug effects , Bronchi/enzymology , Cell Line , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/metabolism , Humans , Lung/drug effects , Models, Biological , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Rats , Signal Transduction/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...