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










Publication year range
1.
ALTEX ; 32(1): 9-24, 2015.
Article in English | MEDLINE | ID: mdl-25372315

ABSTRACT

SEURAT-1 is a European public-private research consortium that is working towards animal-free testing of chemical compounds and the highest level of consumer protection. A research strategy was formulated based on the guiding principle to adopt a toxicological mode-of-action framework to describe how any substance may adversely affect human health.The proof of the initiative will be in demonstrating the applicability of the concepts on which SEURAT-1 is built on three levels:(i) Theoretical prototypes for adverse outcome pathways are formulated based on knowledge already available in the scientific literature on investigating the toxicological mode-of-actions leading to adverse outcomes (addressing mainly liver toxicity);(ii)adverse outcome pathway descriptions are used as a guide for the formulation of case studies to further elucidate the theoretical model and to develop integrated testing strategies for the prediction of certain toxicological effects (i.e., those related to the adverse outcome pathway descriptions);(iii) further case studies target the application of knowledge gained within SEURAT-1 in the context of safety assessment. The ultimate goal would be to perform ab initio predictions based on a complete understanding of toxicological mechanisms. In the near-term, it is more realistic that data from innovative testing methods will support read-across arguments. Both scenarios are addressed with case studies for improved safety assessment. A conceptual framework for a rational integrated assessment strategy emerged from designing the case studies and is discussed in the context of international developments focusing on alternative approaches for evaluating chemicals using the new 21st century tools for toxicity testing.


Subject(s)
Animal Testing Alternatives , Toxicity Tests/methods , Animals , Europe , Humans , Risk Assessment/methods
2.
Arch Toxicol ; 87(2): 337-45, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23274394

ABSTRACT

Cultivated hepatocytes represent a well-established in vitro system. However, the applicability of hepatocytes in toxicogenomics is still controversially discussed. Recently, an in vivo/in vitro discrepancy has been described, whereby the non-genotoxic rat liver carcinogen methapyrilene alters the expression of the metabolizing genes SULT1A1 and ABAT, as well as the DNA damage response gene GADD34 in vitro, but not in vivo. If the collagen sandwich cultures of hepatocytes really produce false-positive data, this would compromise its application in toxicogenomics. To revisit the putative in vivo/in vitro discrepancy, we first analyzed and modeled methapyrilene concentrations in the portal vein of rats. The relatively short half-life of 2.8 h implies a rapid decrease in orally administered methapyrilene in vivo below concentrations that can cause gene expression alterations. This corresponded to the time-dependent alteration levels of GADD34, ABAT and SULT1A1 RNA in the liver: RNA levels are altered 1, 6 and 12 h after methapyrilene administration, but return to control levels after 24 and 72 h. In contrast, methapyrilene concentrations in the culture medium supernatant of primary rat hepatocyte cultures decreased slowly. This explains why GADD34, ABAT and SULT1A1 were still deregulated after 24 h exposure in vitro, but not in vivo. It should also be considered that the earliest analyzed time point in the previous in vivo studies was 24 h after methapyrilene administration. In conclusion, previously observed in vitro/in vivo discrepancy can be explained by different pharmacokinetics present in vitro and in vivo. When the in vivo half-life is short, levels of some initially altered genes may have returned to control levels already 24 h after administration.


Subject(s)
Carcinogens/pharmacokinetics , Gene Expression Regulation, Neoplastic/drug effects , Hepatocytes/drug effects , Liver/drug effects , Methapyrilene/pharmacokinetics , 4-Aminobutyrate Transaminase/genetics , Animals , Antigens, Differentiation/genetics , Arylsulfotransferase/genetics , Carcinogens/toxicity , Cells, Cultured , Half-Life , Hepatocytes/metabolism , Liver/metabolism , Male , Methapyrilene/toxicity , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar
3.
Comput Biol Chem ; 34(3): 193-202, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20643583

ABSTRACT

Exposing eukaryotic cells to a toxic compound and subsequent gene expression profiling may allow the prediction of selected toxic effects based on changes in gene expression. This objective is complicated by the observation that compounds with different modes of toxicity cause similar changes in gene expression and that a global stress response affects many genes. We developed an elastic network model of global stress response with nodes representing genes which are connected by edges of graded coexpression. The expression of only few genes have to be known to model the global stress response of all but a few atypical responder genes. Those required genes and the atypical response genes are shown to be good biomarker for tox predictions. In total, 138 experiments and 13 different compounds were used to train models for different toxicity classes. The deduced biomarkers were shown to be biologically plausible. A neural network was trained to predict the toxic effects of compounds from profiling experiments. On a validation data set of 189 experiments with 16 different compounds the accuracy of the predictions was assessed: 14 out of 16 compounds have been classified correctly. Derivation of model based biomarkers through the elastic network approach can naturally be extended to other areas beyond toxicology since subtle signals against a broad response background are common in biological studies.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/genetics , Gene Expression Profiling/methods , Gene Regulatory Networks , Neural Networks, Computer , Stress, Physiological/genetics , Biomarkers , Eukaryota , Gene Expression Regulation/drug effects
4.
Reprod Toxicol ; 30(1): 50-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20074635

ABSTRACT

Despite about two decades of research in the field of endocrine active compounds, still no validated human recombinant (hr) estrogen receptor-alpha (ERalpha) binding assay is available, although hr-ERalpha is available from several sources. In a joint effort, US EPA and Bayer Schering Pharma with funding from the EU-sponsored 6th framework project, ReProTect, developed a model protocol for such a binding assay. Important features of this assay are the use of a full length hr-ERalpha and performance in a 96-well plate format. A full length hr-ERalpha was chosen, as it was considered to provide the most accurate and human-relevant results, whereas truncated receptors could perform differently. Besides three reference compounds [17beta-estradiol, norethynodrel, dibutylphthalate] nine test compounds with different affinities for the ERalpha [diethylstilbestrol (DES), ethynylestradiol, meso-hexestrol, equol, genistein, o,p'-DDT, nonylphenol, n-butylparaben, and corticosterone] were used to explore the performance of the assay. Three independent experiments per compound were performed on different days, and dilutions of test compounds from deep-frozen stocks, solutions of radiolabeled ligand and receptor preparation were freshly prepared for each experiment. The ERalpha binding properties of reference and test compounds were well detected. As expected dibutylphthalate and corticosterone were non-binders in this assay. In terms of the relative ranking of binding affinities, there was good agreement with published data obtained from experiments using a human recombinant ERalpha ligand binding domain. Irrespective of the chemical nature of the compound, individual IC(50)-values for a given compound varied by not more than a factor of 2.5. Our data demonstrate that the assay was robust and reliably ranked compounds with strong, weak, and no affinity for the ERalpha with high accuracy. It avoids the manipulation and use of animals, i.e., the preparation of uterine cytosol as receptor source from ovariectomized rats, as a recombinant protein is used and thus contributes to the 3R concept (reduce, replace, and refine). Furthermore, in contrast to other assays, this assay could be adjusted to an intermediate/high throughput format. On the whole, this assay is a promising candidate for further validation.


Subject(s)
Animal Testing Alternatives , Biological Assay/methods , Endocrine Disruptors/pharmacology , Estrogen Receptor alpha/metabolism , Models, Biological , Recombinant Proteins/metabolism , Binding, Competitive , Biological Assay/standards , Dose-Response Relationship, Drug , Estrogen Receptor alpha/chemistry , Humans , Ligands , Protein Binding , Radioligand Assay , Recombinant Proteins/chemistry , Reproducibility of Results
5.
Eur J Cancer ; 46(1): 21-32, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19954965

ABSTRACT

Physiologically based modelling of pharmacodynamics/toxicodynamics requires an a priori knowledge on the underlying mechanisms causing toxicity or causing the disease. In the context of cancer, the objective of the expert meeting was to discuss the molecular understanding of the disease, modelling approaches used so far to describe the process, preclinical models of cancer treatment and to evaluate modelling approaches developed based on improved knowledge. Molecular events in cancerogenesis can be detected using 'omics' technology, a tool applied in experimental carcinogenesis, but also for diagnostics and prognosis. The molecular understanding forms the basis for new drugs, for example targeting protein kinases specifically expressed in cancer. At present, empirical preclinical models of tumour growth are in great use as the development of physiological models is cost and resource intensive. Although a major challenge in PKPD modelling in oncology patients is the complexity of the system, based in part on preclinical models, successful models have been constructed describing the mechanism of action and providing a tool to establish levels of biomarker associated with efficacy and assisting in defining biologically effective dose range selection for first dose in man. To follow the concentration in the tumour compartment enables to link kinetics and dynamics. In order to obtain a reliable model of tumour growth dynamics and drug effects, specific aspects of the modelling of the concentration-effect relationship in cancer treatment that need to be accounted for include: the physiological/circadian rhythms of the cell cycle; the treatment with combinations and the need to optimally choose appropriate combinations of the multiple agents to study; and the schedule dependence of the response in the clinical situation.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/genetics , Models, Biological , Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Transformation, Neoplastic/pathology , Circadian Rhythm/physiology , Drug Chronotherapy , Drug Screening Assays, Antitumor/methods , Humans , Neoplasms/diagnosis , Neoplasms/genetics
6.
Reprod Toxicol ; 30(1): 2-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19833195

ABSTRACT

Despite more than a decade of research in the field of endocrine active compounds with affinity for the androgen receptor (AR), still no validated recombinant AR binding assay is available, although recombinant AR can be obtained from several sources. With funding from the European Union (EU)-sponsored 6th framework project, ReProTect, we developed a model protocol for such an assay based on a simple AR binding assay recently developed at our institution. Important features of the protocol were the use of a rat recombinant fusion protein to thioredoxin containing both the hinge region and ligand binding domain (LBD) of the rat AR (which is identical to the human AR-LBD) and performance in a 96-well plate format. Besides two reference compounds [dihydrotestosterone (DHT), androstenedione] ten test compounds with different affinities for the AR [levonorgestrel, progesterone, prochloraz, 17alpha-methyltestosterone, flutamide, norethynodrel, o,p'-DDT, dibutylphthalate, vinclozolin, linuron] were used to explore the performance of the assay. At least three independent experiments per compound were performed. The AR binding properties of reference and test compounds were well detected, in terms of the relative ranking of binding affinities, there was good agreement with published data obtained from experiments using recombinant AR preparations. Irrespective of the chemical nature of the compound, individual IC(50)-values for a given compound varied by not more than a factor of 2.6. Our data demonstrate that the assay reliably ranked compounds with strong, weak, and no/marginal affinity for the AR with high accuracy. It avoids the manipulation and use of animals, as a recombinant protein is used and thus contributes to the 3R concept. On the whole, this assay is a promising candidate for further validation.


Subject(s)
Androgens/pharmacology , Animal Testing Alternatives , Biological Assay/methods , Endocrine Disruptors/pharmacology , Receptors, Androgen , Recombinant Fusion Proteins , Androgen Receptor Antagonists , Androgens/chemistry , Animals , Binding, Competitive , Biological Assay/standards , Dose-Response Relationship, Drug , Endocrine Disruptors/chemistry , Humans , Inhibitory Concentration 50 , Ligands , Protein Binding , Rats , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/antagonists & inhibitors , Reproducibility of Results
7.
Reprod Toxicol ; 30(1): 36-43, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19897028

ABSTRACT

Short-term dynamic culture of rat testicular fragments was evaluated as a model to assess effects on steroidogenesis. A total of 11 compounds differentially affecting testosterone synthesis (aminoglutethimide, ketoconazole, danazol, flutamide, diethylstilbestrol, genistein, butylparaben, nonoxynol-9, dimethoate, RU 486, and cadmium chloride) were used to explore the performance of the assay. Testosterone secretion into the medium and testosterone retained in tissue fragments was determined as a measure of steroidogenesis. Three independent experiments per compound were performed. The known in vitro inhibitory properties of most compounds could be detected. Whenever significant inhibition of testosterone synthesis was observed, low effect concentrations for a given compound differed frequently only by a factor of

Subject(s)
Animal Testing Alternatives , Endocrine Disruptors/pharmacology , Models, Biological , Testis/drug effects , Testosterone/biosynthesis , Animals , Dose-Response Relationship, Drug , Male , Organ Culture Techniques , Radioimmunoassay , Rats , Rats, Wistar , Testis/cytology , Testis/metabolism , Time Factors
8.
Am J Pathol ; 175(4): 1686-98, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19717638

ABSTRACT

Kidneys are the second most frequent site for chemically induced cancers in rats. However, there is still limited information on direct effects of carcinogens on pathways involved in the development of kidney tumors. Since transformed tumor cells have different characteristics than their cell of origin, it was hypothesized that healthy tissue and progressing stages of preneoplastic lesions are differentially influenced by chemical carcinogens. To elucidate this question, TSC2(-/-) Eker rats were gavaged with genotoxic aristolochic acid or nongenotoxic ochratoxin A for 3 and 6 months, respectively. Histopathology and cell proliferation analysis demonstrated a compound- and sex-specific onset of preneoplastic lesions. In contrast, comparable gene expression profiles of laser-microdissected preneoplastic lesions from carcinogen-treated and control rats, including reduced expression of genes involved in carcinogen uptake and metabolism, point to a compound-independent lesion progression. Gene expression profiles and additional immunostaining suggested that clonal expansion of renal lesions appears primarily driven by disturbed mammalian target of rapamycin complex 1 and mammalian target of rapamycin complex 2 pathway regulation. Finally, prolonged carcinogen exposure resulted in only marginal gene expression changes in tubules with normal morphology, indicating that some tubules may have adapted to the treatment. Taken together, these findings indicate that the final outcome of in vivo carcinogenicity studies is primarily determined by time-restricted initial events, while lesion progression may be a compound-independent process, involving deregulated mTOR signaling in the Eker rat model.


Subject(s)
Kidney Neoplasms/pathology , Precancerous Conditions/pathology , Animals , Apoptosis/drug effects , Aristolochic Acids/toxicity , Bromodeoxyuridine/metabolism , Cell Proliferation/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm , Kidney Neoplasms/genetics , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Kidney Tubules/pathology , Models, Biological , Ochratoxins/toxicity , Precancerous Conditions/genetics , Rats , Staining and Labeling , Trans-Activators , Transcription Factors/metabolism
11.
Mutat Res ; 659(3): 202-10, 2008.
Article in English | MEDLINE | ID: mdl-18514569

ABSTRACT

Recent changes in the European legislation of chemical-related substances have forced the scientific community to speed up the search for alternative methods that could partly or fully replace animal experimentation. The Sixth Framework Program project carcinoGENOMICS was specifically raised to develop omics-based in vitro screens for testing the carcinogenic potential of chemical compounds in a pan-European context. This paper provides an in-depth analysis of the complexity of choosing suitable reference compounds used for creating and fine-tuning the in vitro carcinogenicity assays. First, a number of solid criteria for the selection of the model compounds are defined. Secondly, the strategy followed, including resources consulted, is described and the selected compounds are briefly illustrated. Finally, limitations and problems encountered during the selection procedure are discussed. Since selecting an appropriate set of chemicals is a frequent impediment in the early stages of similar research projects, the information provided in this paper might be extremely valuable.


Subject(s)
Carcinogenicity Tests/methods , Genomics/methods , Animal Testing Alternatives , European Union , Gene Expression Profiling , Hazardous Substances , International Cooperation , Toxicogenetics/trends
12.
Environ Health Perspect ; 114(3): 420-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16507466

ABSTRACT

This is the report of the first workshop "Validation of Toxicogenomics-Based Test Systems" held 11-12 December 2003 in Ispra, Italy. The workshop was hosted by the European Centre for the Validation of Alternative Methods (ECVAM) and organized jointly by ECVAM, the U.S. Interagency Coordinating Committee on the Validation of Alternative Methods (ICCVAM), and the National Toxicology Program (NTP) Interagency Center for the Evaluation of Alternative Toxicological Methods (NICEATM). The primary aim of the workshop was for participants to discuss and define principles applicable to the validation of toxicogenomics platforms as well as validation of specific toxicologic test methods that incorporate toxicogenomics technologies. The workshop was viewed as an opportunity for initiating a dialogue between technologic experts, regulators, and the principal validation bodies and for identifying those factors to which the validation process would be applicable. It was felt that to do so now, as the technology is evolving and associated challenges are identified, would be a basis for the future validation of the technology when it reaches the appropriate stage. Because of the complexity of the issue, different aspects of the validation of toxicogenomics-based test methods were covered. The three focus areas include a) biologic validation of toxicogenomics-based test methods for regulatory decision making, b) technical and bioinformatics aspects related to validation, and c) validation issues as they relate to regulatory acceptance and use of toxicogenomics-based test methods. In this report we summarize the discussions and describe in detail the recommendations for future direction and priorities.


Subject(s)
Toxicogenetics/legislation & jurisprudence , Animal Testing Alternatives/legislation & jurisprudence , Computational Biology , Government Regulation , Reproducibility of Results , Toxicity Tests/methods
13.
Toxicology ; 217(2-3): 169-75, 2006 Jan 16.
Article in English | MEDLINE | ID: mdl-16297522

ABSTRACT

N,N,N',N'-Tetramethylthiourea (TMTU) is a rat goitrogen inducing thyroid hyperplasia, hypertrophy, and tumor formation. Little is known about the exact underlying mechanism of action. As thyroid peroxidase (TPO) and type I iodothyronine deiodinase (ID-I) have been established as targets of goitrogenic thiourea derivatives, we investigated interactions of TMTU with target enzymes using a partially purified fraction from hog thyroids or solubilized hog thyroid microsomes and 10,000g supernatant from rat liver homogenate, respectively, as enzyme sources. For comparison, comprehensively characterized goitrogenic thiourea derivatives were studied as well. In contrast to propylthiouracil (PTU), and like ethylenethiourea (ETU), TMTU only marginally affected TPO-catalyzed oxidation of guaiacol. TMTU, like ETU, concentration-dependently suppressed TPO-catalyzed iodine formation with concomitant oxidative metabolism. Suppression ceased upon consumption of thiourea derivatives, the rate of the reappearing iodine formation was similar to that of controls. TMTU, like ETU, also suppressed non-enzymatic and TPO-catalyzed monoiodination of l-tyrosine with a stoichiometry of 2:1, i.e., one molecule of thiourea derivative suppressed two times monoiodination. TMTU and ETU were unable to irreversibly inhibit TPO. In contrast to PTU, TMTU did not inhibit ID-I. These findings provide evidence that TMTU interferes with thyroid hormone synthesis at the level of iodination and demonstrate a metabolic route for the oxidative detoxification of TMTU in the thyroid suggesting that low-level or intermittent exposure to TMTU would have only minimal effects on thyroid hormone synthesis. Finally, it can be concluded that meaningful toxicological studies on TPO inhibition can be performed without a need for highly purified TPO.


Subject(s)
Goiter/chemically induced , Thiourea/analogs & derivatives , Amitrole/pharmacology , Animals , Antithyroid Agents/metabolism , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Ethylenethiourea/administration & dosage , Ethylenethiourea/toxicity , Goiter/enzymology , Guaiacol/metabolism , Hydrogen Peroxide/pharmacology , Iodide Peroxidase/antagonists & inhibitors , Iodide Peroxidase/metabolism , Iodine/metabolism , Oxidation-Reduction/drug effects , Propylthiouracil/pharmacology , Rats , Swine , Thiourea/metabolism , Thiourea/toxicity , Time Factors , Tyrosine/metabolism
14.
J Photochem Photobiol B ; 79(1): 25-34, 2005 Apr 04.
Article in English | MEDLINE | ID: mdl-15792876

ABSTRACT

Phototoxic side effects of pharmaceutical and cosmetic products are of increasing concern for patients, dermatologists and the chemical industry. Moreover, the need of new chemicals and drugs puts pressure on pre-clinical test methods for side effects, especially interactive adverse-effects with UV-light. So, the predictive potential of different established test methods, which are used regularly in our departments in order to detect the phototoxic potential of chemicals, were analyzed. Namely the fibroblast 3T3 test, the photo hen's egg test, a guinea pig test for measuring acute photoreactions, and a modified Local Lymph Node Assay, the Integrated Model for the Differentiation of Skin Reactions. Various agents with different photoreactive potential were tested: quinolones like Bay y 3118, ciprofloxacin, enoxacin, lomefloxacin, moxifloxacin, ofloxacin, sparfloxacin, as well as promethazine, chlorpromazine, 8-methoxypsoralen and olaquindox serving as control. Special emphasis was taken to evaluate the capability of the employed test procedures to predict phototoxic side effects in patients. Following our results, both in vitro assays were useful tools to detect photoirritancy while the photoallergic potentials of tested compounds were exclusively detected by an in vivo assay. As long as no in vitro model for photoallergy is available, the UV-IMDS should be considered to evaluate photoallergic properties of a supposed photoreactive agent.


Subject(s)
Dermatitis, Photoallergic/pathology , Dermatitis, Phototoxic/pathology , Drug-Related Side Effects and Adverse Reactions , Animals , Cell Line , Chick Embryo , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Female , Guinea Pigs , Mice , Mice, Inbred BALB C , Pharmaceutical Preparations/administration & dosage , Ultraviolet Rays/adverse effects
15.
BMC Dev Biol ; 5: 1, 2005 Jan 26.
Article in English | MEDLINE | ID: mdl-15673475

ABSTRACT

BACKGROUND: Recently, tissue engineering has merged with stem cell technology with interest to develop new sources of transplantable material for injury or disease treatment. Eminently interesting, are bone and joint injuries/disorders because of the low self-regenerating capacity of the matrix secreting cells, particularly chondrocytes. ES cells have the unlimited capacity to self-renew and maintain their pluripotency in culture. Upon induction of various signals they will then differentiate into distinctive cell types such as neurons, cardiomyocytes and osteoblasts. RESULTS: We present here that BMP-2 can drive ES cells to the cartilage, osteoblast or adipogenic fate depending on supplementary co-factors. TGFbeta1, insulin and ascorbic acid were identified as signals that together with BMP-2 induce a chondrocytic phenotype that is characterized by increased expression of cartilage marker genes in a timely co-ordinated fashion. Expression of collagen type IIB and aggrecan, indicative of a fully mature state, continuously ascend until reaching a peak at day 32 of culture to approximately 80-fold over control values. Sox9 and scleraxis, cartilage specific transcription factors, are highly expressed at very early stages and show decreased expression over the time course of EB differentiation. Some smaller proteoglycans, such as decorin and biglycan, are expressed at earlier stages. Overall, proteoglycan biosynthesis is up-regulated 7-fold in response to the supplements added. BMP-2 induced chondrocytes undergo hypertrophy and begin to alter their expression profile towards osteoblasts. Supplying mineralization factors such as beta-glycerophosphate and vitamin D3 with the culture medium can facilitate this process. Moreover, gene expression studies show that adipocytes can also differentiate from BMP-2 treated ES cells. CONCLUSIONS: Ultimately, we have found that ES cells can be successfully triggered to differentiate into chondrocyte-like cells, which can further alter their fate to become hypertrophic, and adipocytes. Compared with previous reports using a brief BMP-2 supplementation early in differentiation, prolonged exposure increased chondrogenic output, while supplementation with insulin and ascorbic acid prevented dedifferentiation. These results provide a foundation for the use of ES cells as a potential therapy in joint injury and disease.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Chondrogenesis , Pluripotent Stem Cells/cytology , Transforming Growth Factor beta/pharmacology , Adipocytes/cytology , Adipogenesis , Aggrecans , Animals , Ascorbic Acid/pharmacology , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Bone Morphogenetic Protein 2 , Cartilage/cytology , Cell Differentiation , Cell Line , Chondrocytes/cytology , Collagen/biosynthesis , Collagen/genetics , Embryo, Mammalian/cytology , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Gene Expression/drug effects , Gene Expression/physiology , High Mobility Group Proteins/biosynthesis , High Mobility Group Proteins/genetics , Insulin/pharmacology , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Mice , Osteoblasts/cytology , Osteogenesis , Pluripotent Stem Cells/physiology , Proteins/pharmacology , Proteoglycans/biosynthesis , Proteoglycans/genetics , SOX9 Transcription Factor , Transcription Factors/biosynthesis , Transcription Factors/genetics
16.
Toxicology ; 195(2-3): 113-26, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14751668

ABSTRACT

Concerns have been raised whether natural and man-made chemicals might have the potential of interfering with the endocrine system. Especially interactions with sex hormone receptors are considered as a critical issue. Weak anti-androgenicity has been demonstrated for some environmental pollutants such as p,p'-DDE, and androgenic activity was found in feedlot and pulp mill effluents. In order to be able to screen for compounds with affinity for the androgen receptor (AR), we developed an AR binding assay using a recombinant AR as receptor source and the synthetic androgen methyltrienolone (R 1881) as ligand. Experiments were performed on 96-well microtitre plates. Following method optimization, compounds recently recommended for the validation of assays characterizing AR-mediated effects and those being used for the OECD validation of the Hershberger assay were employed amongst others to standardize the method. The assay readily detected and discriminated compounds with strong and weak affinity for the AR such as natural and synthetic androgens, anti-androgens in therapeutic use, and a variety of chemicals with weak anti-androgenic side effects, whereas in line with previous findings, AR binding properties of dibutylphthalate and its metabolites could not be demonstrated. Detergents interfered with receptor binding, but showed characteristic effects different from that of true AR binding compounds. The assay is simple and sensitive, avoids the use of animals as a receptor source, and should be of value when screening for endocrine-modulating compounds.


Subject(s)
Androgen Antagonists/pharmacology , Biological Assay/methods , Receptors, Androgen/drug effects , Animals , Biological Assay/standards , Dose-Response Relationship, Drug , Metribolone/pharmacology , Protein Binding , Rats , Receptors, Androgen/metabolism , Recombinant Fusion Proteins/pharmacology , Reference Standards , Thioredoxins/pharmacology
17.
Toxicol Sci ; 77(1): 19-34, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14600272

ABSTRACT

When applied in toxicological studies, the recently developed gene expression profiling techniques using microarrays, which brought forth the new field of toxicogenomics, facilitate the interpretation of a toxic compound's mechanism of action. In this study, we investigated whether genotoxic carcinogens at doses known to induce liver tumors in the 2-year rat bioassay deregulate a common set of genes in a short-term in vivo study and, if so, whether these deregulated genes represent defined biological pathways. Rats were dosed with the four genotoxic hepatocarcinogens dimethylnitrosamine (4 mg/kg/day), 2-nitrofluorene (44 mg/kg/day), aflatoxin B1 (0.24 mg/kg/day), and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK, 20 mg/kg/day). After treatment for up to 14 days, the expression profiles of the livers were analyzed on Affymetrix RG_U34A microarrays. Among the significantly upregulated genes were a set of target genes of the tumor suppressor protein p53, indicating a DNA damage response. Such a response was expected and, therefore, confirmed the validity of our approach. In addition, the gene expression changes suggest a specific detoxification response, the activation of proliferative and survival signaling pathways, and some cell structural changes. These responses were strong throughout the 14 day time course for 2-nitrofluorene and aflatoxin B1; in the case of dimethylnitrosamine and NNK, the effects were weakly detectable at day 1 and then increased with time. For dimethylnitrosamine and aflatoxin B1, which caused observable inflammation in vivo, we found a corresponding upregulation of inflammatory genes at the same time points. Thus, by the toxicogenomic analysis of short-term in vivo studies, we identified genes and pathways commonly deregulated by genotoxic carcinogens, which may be indicative for the early events in tumorigenesis and, thus, predictive of later tumor development.


Subject(s)
Carcinogens/toxicity , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Liver/drug effects , Toxicogenetics , Animals , Liver/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Wistar
18.
Toxicol Sci ; 73(2): 315-28, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12700408

ABSTRACT

Ochratoxin A (OTA) is a mycotoxin often found in cereals as a contaminant, and it is known to cause severe nephrotoxicity in animals and humans. There have been several investigations studying the mode of action of this toxicant, suggesting inhibition of protein synthesis, formation of DNA adducts, and provocation of DNA single-strand breaks as a result of oxidative stress, but little is known about the transcriptional alterations underlying OTA-derived nephrotoxicity so far. We carried out DNA microarray analyses to assess OTA-specific expression profiles in vivo and in vitro. Cultures of primary rat proximal tubular cells and male Wistar rats were treated with a low dose (5 microM and 1 mg/kg, respectively) or a high dose (12.5 microM and 10 mg/kg, respectively) of OTA for 24 or 72 h. Microarray experiments were carried out after dual fluorescent labeling of sample cDNA, and data analysis was performed utilizing different statistical methods. Validity of selected microarray data was confirmed by quantitative real-time PCR. We were able to demonstrate that microarray data derived from our proximal tubule cell (PTC) culture model were highly comparable to the in vivo situation. Marked treatment-specific transcriptional changes were detected for genes involved in DNA damage response and apoptosis (upregulation of GADD 153, GADD 45, annexin V), response to oxidative stress (differential expression of hypoxia-inducible factor 1 and catalase), and inflammatory reactions (upregulation of alpha 2 macroglobulin, ceruloplasmin, and cathepsin S). We conclude that our results provide a molecular basis for interpretation of OTA-induced nephrotoxicity.


Subject(s)
Carcinogens/toxicity , Gene Expression Profiling , Kidney Diseases/chemically induced , Mycotoxins/toxicity , Ochratoxins/toxicity , Oligonucleotide Array Sequence Analysis , Acute-Phase Reaction/genetics , Acute-Phase Reaction/metabolism , Administration, Oral , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carcinogens/administration & dosage , Cell Culture Techniques , DNA/analysis , DNA Damage/drug effects , DNA Damage/genetics , DNA Primers/chemistry , DNA Repair/drug effects , DNA Repair/genetics , Dose-Response Relationship, Drug , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mycotoxins/administration & dosage , Ochratoxins/administration & dosage , Oxidative Stress/drug effects , Oxidative Stress/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
19.
Neurotoxicology ; 24(1): 55-64, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12564382

ABSTRACT

Treatment of rats with fentrazamide for 2 years at 3000 ppm (males) and 4000 ppm (females) led to an increased incidence and degree of axonal degeneration in sciatic nerve as well as to effects on red blood cells. The mechanism underlying these effects was investigated in vitro using various cell cultures (permanent rodent cell lines from the nervous system, liver, kidney, skeletal and heart muscle and fibroblasts, primary cortical neurons and erythrocytes from the rat). Added to cultured rat cortical neurons for 1 week, fentrazamide considerably decreased glucose consumption, ATP levels and mitochondrial membrane potential and lowered the GSH level, however, it had little impact on viability and neurofilaments and did not induce oxidative stress (ROS) over the first 2 h. After recovery for 1 week, in addition some destruction of neurofilaments had occurred probably secondary to the disturbance of energy production. These effects were prevented by pyruvate. Further studies indicated that fentrazamide primarily inhibited glucose utilization, most likely by interfering with glycolysis. Similar effects were found in erythrocytes treated with fentrazamide over a period of 7 days. Primarily, the glucose consumption was reduced after 1-day treatment, followed by a marked reduction of the energy supply at days 3 and 7. Comparable to the neurons, the GSH level was significantly reduced. A marked hemolysis of the red blood cells was then observed after prolonged treatment. The extensive energy demand and exclusive dependency on glucose utilization of neurons and erythrocytes may explain the specific vulnerability of motor neurons and erythrocytes. When comparing the concentrations necessary for inducing effects in vitro on neuronal cells and erythrocytes to the very low plasma concentrations of fentrazamide in treated rats it is suggested that only a small impact of fentrazamide on the energy status at high doses will occur in vivo. Therefore, aging of the rat as another factor compromising mitochondrial energy production in motor neurons must be considered as additional contribution for the induction of axonal degeneration. It is concluded that this effect of fentrazamide in rats poses no specific risk under the exposure conditions relevant to humans.


Subject(s)
Erythrocytes/drug effects , Glucose/metabolism , Herbicides/pharmacology , Neurons/drug effects , Aging/drug effects , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Erythrocytes/cytology , Erythrocytes/metabolism , Female , Male , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar
20.
Arch Toxicol ; 76(7): 414-22, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12111006

ABSTRACT

After repeated-dose toxicity studies with the fungicide propineb, reversible effects on muscle functions were found. Therefore, mechanistic investigations should contribute to clarification of its mode of action in relation to disulfiram and diethyldithiocarbamate neurotoxicity or direct effects on muscle cells. In principle, besides the dithiocarbamate effects, two different mechanisms have been discussed for this fungicide. One mechanism is the degradation to carbon disulfide (CS(2)) and propylenthiourea (PTU) and the other are direct effects of zinc. Primary neuronal cell cultures of the rat are a well established model to identify neurotoxic compounds like n-hexane or acrylamide. In this cell culture model, endpoints such as viability, energy supply, glucose consumption and cytoskeleton elements were determined. Additionally, skeletal muscle cells were used for comparison. Propineb and its metabolite PTU were investigated in comparison to CS(2), disulfiram and diethyldithiocarbamate. The toxicity of zinc was tested using zinc chloride (ZnCl(2)). It was clearly shown that propineb exerted strong effects on the cytoskeleton of neuronal and non-neuronal cell cultures (astrocytes, muscle cells). This was similar to ZnCl(2,) but not to CS(2). With CS(2) and disulfiram effects on the energy supply were more prominent. In conclusion, the toxicity of propineb is not comparable to disulfiram, diethyldithiocarbamate or CS(2) neurotoxicity. In regard to these findings, a direct reversible effect of propineb on skeletal muscle cells seems to be more likely.


Subject(s)
Fungicides, Industrial/toxicity , Muscle, Skeletal/drug effects , Neurons/drug effects , Thiourea/analogs & derivatives , Zineb/analogs & derivatives , Zineb/toxicity , Animals , Carbon Disulfide/toxicity , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Chlorides/toxicity , Cytoskeleton/drug effects , Disulfiram/toxicity , Ditiocarb/toxicity , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Fungicides, Industrial/metabolism , Glucose/metabolism , Muscle, Skeletal/cytology , Neurons/cytology , Rats , Thiourea/toxicity , Zinc Compounds/toxicity , Zineb/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...