Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Toxicol Lett ; 384: 96-104, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37451652

ABSTRACT

The evaluation of chemical and pharmaceutical safety for humans is moving from animal studies to New Approach Methodologies (NAM), reducing animal use and focusing on mechanism of action, whilst enhancing human relevance. In developmental toxicology, the mechanistic approach is facilitated by the assessment of predictive biomarkers, which allow mechanistic pathways perturbation monitoring at the basis of human hazard assessment. In our search for biomarkers of maldevelopment, we focused on chemically-induced perturbation of the retinoic acid signaling pathway (RA-SP), a major pathway implicated in a plethora of developmental processes. A genome-wide expression screening was performed on zebrafish embryos treated with two teratogens, all-trans retinoic acid (ATRA) and valproic acid (VPA), and a non-teratogen reference compound, folic acid (FA). Each compound was found to have a specific mRNA expression profile with 248 genes commonly dysregulated by both teratogenic compounds but not by FA. These genes were implicated in several developmental processes (e.g., the circulatory and nervous system). Given the prominent response of neurodevelopmental gene sets, and the crucial need to better understand developmental neurotoxicity, our study then focused on nervous system development. We found 62 genes that are potential early neurodevelopmental toxicity biomarker candidates. These results advance NAM-based safety assessment evaluation by highlighting the usefulness of the RA-SP in providing early toxicity biomarker candidates.


Subject(s)
Tretinoin , Zebrafish , Animals , Humans , Tretinoin/toxicity , Zebrafish/genetics , Zebrafish/metabolism , Valproic Acid/toxicity , Gene Expression Regulation , Teratogens/toxicity , Biomarkers , Nervous System/metabolism , Gene Expression Regulation, Developmental , Embryo, Nonmammalian
2.
Reprod Toxicol ; 119: 108404, 2023 08.
Article in English | MEDLINE | ID: mdl-37207909

ABSTRACT

The zebrafish embryo (ZE) model provides a developmental model well conserved throughout vertebrate embryogenesis, with relevance for early human embryo development. It was employed to search for gene expression biomarkers of compound-induced disruption of mesodermal development. We were particularly interested in the expression of genes related to the retinoic acid signaling pathway (RA-SP), as a major morphogenetic regulating mechanism. We exposed ZE to teratogenic concentrations of valproic acid (VPA) and all-trans retinoic acid (ATRA), using folic acid (FA) as a non-teratogenic control compound shortly after fertilization for 4 h, and performed gene expression analysis by RNA sequencing. We identified 248 genes specifically regulated by both teratogens but not by FA. Further analysis of this gene set revealed 54 GO-terms related to the development of mesodermal tissues, distributed along the paraxial, intermediate, and lateral plate sections of the mesoderm. Gene expression regulation was specific to tissues and was observed for somites, striated muscle, bone, kidney, circulatory system, and blood. Stitch analysis revealed 47 regulated genes related to the RA-SP, which were differentially expressed in the various mesodermal tissues. These genes provide potential molecular biomarkers of mesodermal tissue and organ (mal)formation in the early vertebrate embryo.


Subject(s)
Tretinoin , Zebrafish , Animals , Humans , Zebrafish/genetics , Zebrafish/metabolism , Tretinoin/metabolism , Transcriptome , Mesoderm/metabolism , Signal Transduction , Biomarkers/metabolism , Gene Expression Profiling , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental
3.
ALTEX ; 40(1): 61-82, 2023.
Article in English | MEDLINE | ID: mdl-35536601

ABSTRACT

Thyroid hormones (THs) are crucial regulators of human metabolism and early development. During the safety assessment of plant protection products, the human relevance of chemically induced TH perturbations observed in test animals remains uncertain. European regulatory authorities request follow-up in vitro studies to elucidate human-relevant interferences on thyroid gland function or TH catabolism through hepatic enzyme induction. However, human in vitro assays based on single molecular initiating events poorly reflect the complex TH biology and related liver-thyroid axis. To address this complexity, we present human three-dimensional thyroid and liver organoids with key functions of TH metabolism. The thyroid model resembles in vivo-like follicular architecture and a TSH-dependent triiodothyronine synthesis over 21 days, which is inhibited by methimazole. The HepaRG-based liver model, secreting the critical TH-binding proteins albumin and thyroxine-binding globulin, emulates an active TH catabolism via the formation of glucuronidated and sulfated thyroxine (gT4/sT4). Activation of the nuclear receptors PXR and AHR was demonstrated via the induction of specific CYP isoenzymes by rifampicin, pregnenolone-16α-carbonitrile, and ß-naphthoflavone. However, this nuclear receptor activation, assumed to regulate UDP-glucuronosyltransferases and sulfotransferases, appeared to have no effect on gT4 and sT4 formation in this human-derived hepatic cell line model. Finally, established single-tissue models were successfully co-cultured in a perfused two-organ chip for 21 days. In conclusion, this model presents a first step towards a complex multimodular human platform that will help to identify both direct and indirect thyroid disruptors that are relevant from a human safety perspective.


Subject(s)
Chemical Safety , Thyroid Gland , Animals , Humans , Thyroid Gland/metabolism , Microfluidics , Thyroid Hormones/metabolism , Thyroid Hormones/pharmacology , Liver , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/pharmacology
4.
ALTEX ; 40(1): 83-102, 2023.
Article in English | MEDLINE | ID: mdl-35791291

ABSTRACT

Endocrine disruption by environmental chemicals continues to be a concern for human safety. The rat, a widely used model organism in toxicology, is very sensitive to chemical-induced thyroid perturbation, e.g., histopathological alterations in thyroid tissue. Species differences in the susceptibility to thyroid perturbation lead to uncertainty in human safety risk assessments. Hazard identification and characterization of chemically induced thyroid perturbation would therefore benefit from in vitro models addressing different mechanisms of action in a single functional assay, ideally across species. We here introduce a rat thyroid-liver chip that enables simultaneous identification of direct and indirect (liver-mediated) thyroid perturbation on organ-level functions in vitro. A second manuscript describes our work toward a human thyroid-liver chip (Kühnlenz et al., 2022). The presented microfluidic model consisting of primary rat thyroid follicles and liver 3D spheroids maintains a tissue-specific phenotype for up to 21 days. More precisely, the thyroid model exhibits a follicular architecture expressing basolateral and apical markers and secretes T4. Likewise, liver spheroids retain hepatocellular characteristics, e.g., a stable release of albumin and urea, the presence of bile canalicular networks, and the formation of T4-glucuronide. Experiments with reference chemicals demonstrated proficiency to detect direct and indirect mechanisms of thyroid perturbation through decreased thyroid hormone secretion and increased gT4 formation, respectively. Prospectively this rat thyroid-liver chip model, together with its human counterpart, may support a species-specific quantitative in vitro to in vivo extrapolation to improve a data-driven and evidence-based human safety risk assessment with significant contributions to the 3R principles.


Subject(s)
Rodentia , Thyroid Gland , Humans , Rats , Animals , Animal Testing Alternatives , Liver
5.
Reprod Toxicol ; 115: 8-16, 2023 01.
Article in English | MEDLINE | ID: mdl-36375755

ABSTRACT

The zebrafish embryotoxicity test (ZET) is widely used in developmental toxicology. The analysis of gene expression regulation in ZET after chemical exposure provides mechanistic information about the effects of chemicals on morphogenesis in the test. The gene expression response magnitude has been shown to change with exposure duration. The objective of this work is to study the effect of the exposure duration on the magnitude of gene expression changes in the all-trans retinoic acid (ATRA) signaling pathway in the ZET. Retinoic acid regulation is a key driver of morphogenesis and is therefore employed here as an indicator for the regulation of developmental genes. A teratogenic concentration of 7.5 nM of ATRA was given at 3 hrs post fertilization (hpf) for a range of exposure durations until 120 hrs of development. The expression of a selection of genes related to ATRA signaling and downstream developmental genes was determined. The highest magnitudes of gene expression regulation were observed after 2-24 hrs exposure with an optimal response after 4 hrs. Longer exposures showed a decrease in the gene expression response, although continued exposure to 120 hpf caused malformations and lethality. This study shows that assessment of gene expression regulation at early time points after the onset of exposure in the ZET may be optimal for the prediction of developmental toxicity. We believe these results could help optimize sensitivity in future studies with ZET.


Subject(s)
Embryo, Nonmammalian , Zebrafish , Animals , Zebrafish/physiology , Tretinoin/metabolism , Morphogenesis , Gene Expression Regulation , Gene Expression Regulation, Developmental
6.
Arch Toxicol ; 96(12): 3407-3419, 2022 12.
Article in English | MEDLINE | ID: mdl-36063173

ABSTRACT

With an increasing need to incorporate new approach methodologies (NAMs) in chemical risk assessment and the concomitant need to phase out animal testing, the interpretation of in vitro assay readouts for quantitative hazard characterisation becomes more important. Physiologically based kinetic (PBK) models, which simulate the fate of chemicals in tissues of the body, play an essential role in extrapolating in vitro effect concentrations to in vivo bioequivalent exposures. As PBK-based testing approaches evolve, it will become essential to standardise PBK modelling approaches towards a consensus approach that can be used in quantitative in vitro-to-in vivo extrapolation (QIVIVE) studies for regulatory chemical risk assessment based on in vitro assays. Based on results of an ECETOC expert workshop, steps are recommended that can improve regulatory adoption: (1) define context and implementation, taking into consideration model complexity for building fit-for-purpose PBK models, (2) harmonise physiological input parameters and their distribution and define criteria for quality chemical-specific parameters, especially in the absence of in vivo data, (3) apply Good Modelling Practices (GMP) to achieve transparency and design a stepwise approach for PBK model development for risk assessors, (4) evaluate model predictions using alternatives to in vivo PK data including read-across approaches, (5) use case studies to facilitate discussions between modellers and regulators of chemical risk assessment. Proof-of-concepts of generic PBK modelling approaches are published in the scientific literature at an increasing rate. Working on the previously proposed steps is, therefore, needed to gain confidence in PBK modelling approaches for regulatory use.


Subject(s)
Models, Biological , Animals , Kinetics , Risk Assessment/methods
7.
Arch Toxicol ; 96(3): 743-766, 2022 03.
Article in English | MEDLINE | ID: mdl-35103819

ABSTRACT

The long-term investment in new approach methodologies (NAMs) within the EU and other parts of the world is beginning to result in an emerging consensus of how to use information from in silico, in vitro and targeted in vivo sources to assess the safety of chemicals. However, this methodology is being adopted very slowly for regulatory purposes. Here, we have developed a framework incorporating in silico, in vitro and in vivo methods designed to meet the requirements of REACH in which both hazard and exposure can be assessed using a tiered approach. The outputs from each tier are classification categories, safe doses, and risk assessments, and progress through the tiers depends on the output from previous tiers. We have exemplified the use of the framework with three examples. The outputs were the same or more conservative than parallel assessments based on conventional studies. The framework allows a transparent and phased introduction of NAMs in chemical safety assessment and enables science-based safety decisions which provide the same level of public health protection using fewer animals, taking less time, and using less financial and expert resource. Furthermore, it would also allow new methods to be incorporated as they develop through continuous selective evolution rather than periodic revolution.


Subject(s)
Chemical Safety/methods , Risk Assessment/methods , Toxicity Tests/methods , Animal Testing Alternatives , Animals , Chemical Safety/legislation & jurisprudence , Computer Simulation , Environmental Exposure/prevention & control , Humans , Risk Assessment/legislation & jurisprudence
8.
Crit Rev Toxicol ; 44(1): 64-82, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24180433

ABSTRACT

The constitutive androstane receptor (CAR) and pregnane X receptor (PXR) are important nuclear receptors involved in the regulation of cellular responses from exposure to many xenobiotics and various physiological processes. Phenobarbital (PB) is a non-genotoxic indirect CAR activator, which induces cytochrome P450 (CYP) and other xenobiotic metabolizing enzymes and is known to produce liver foci/tumors in mice and rats. From literature data, a mode of action (MOA) for PB-induced rodent liver tumor formation was developed. A MOA for PXR activators was not established owing to a lack of suitable data. The key events in the PB-induced liver tumor MOA comprise activation of CAR followed by altered gene expression specific to CAR activation, increased cell proliferation, formation of altered hepatic foci and ultimately the development of liver tumors. Associative events in the MOA include altered epigenetic changes, induction of hepatic CYP2B enzymes, liver hypertrophy and decreased apoptosis; with inhibition of gap junctional intercellular communication being an associative event or modulating factor. The MOA was evaluated using the modified Bradford Hill criteria for causality and other possible MOAs were excluded. While PB produces liver tumors in rodents, important species differences were identified including a lack of cell proliferation in cultured human hepatocytes. The MOA for PB-induced rodent liver tumor formation was considered to be qualitatively not plausible for humans. This conclusion is supported by data from a number of epidemiological studies conducted in human populations chronically exposed to PB in which there is no clear evidence for increased liver tumor risk.


Subject(s)
Liver Neoplasms/pathology , Liver/drug effects , Phenobarbital/toxicity , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Aryl Hydrocarbon Hydroxylases , Cell Proliferation/drug effects , Constitutive Androstane Receptor , Cytochrome P-450 CYP2B6 , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver Neoplasms/chemically induced , Pregnane X Receptor , Receptors, Steroid/metabolism , Xenobiotics/toxicity
9.
Exp Toxicol Pathol ; 65(4): 397-407, 2013 May.
Article in English | MEDLINE | ID: mdl-22405603

ABSTRACT

Repeated exposure to 17-α-methyltestosterone (17MT) and estradiol benzoate (EB) for 28 or 90 days in rats induce similar ovarian atrophy. The objective of the present work was to identify and compare the early effects induced by 17MT and EB on the ovary using molecular and histopathological tools. Female rats were evaluated after 1, 3 or 7 days following an oral exposure by gavage at a daily dose of 600 mg/kg/day for 17MT and 5 mg/kg/day for EB. All animals were found to be acyclic after 3 or 7 days of treatment with 17MT and EB. Histopathological changes were present in the ovary, uterus, vagina and mammary gland after both treatments. Ovarian atrophy known as the long term effect of 17MT and EB was not yet detected after 7 days of treatment. But non regressive corpora lutea and cystic follicles were identically observed in the ovary of 17MT and EB treated females. Both compounds induced a decrease of LH transcripts together with an increase of plasma progesterone and prolactin levels. Differences in the profile of regulation of the aromatase were noted after 1 and 3 days of treatment in 17MT treated animals (upregulated) when compared to EB treated animals (downregulated). In summary, we have shown that despite the different nature of hormonal activity, EB and 17MT induce very early endocrine perturbation which presents several similarities. Our work indicated that the detection of early key hormonal markers in short term studies can help to predict the adverse long term effects on target tissues.


Subject(s)
Anabolic Agents/toxicity , Contraceptive Agents/toxicity , Estradiol/analogs & derivatives , Methyltestosterone/toxicity , Ovary/drug effects , Animals , Endocrine System/drug effects , Estradiol/toxicity , Estrous Cycle/drug effects , Female , Luteinizing Hormone/blood , Ovary/metabolism , Ovary/pathology , Pituitary Gland/drug effects , Polymerase Chain Reaction , Progesterone/blood , Prolactin/blood , Rats , Rats, Wistar
10.
Toxicol Lett ; 213(2): 275-84, 2012 Sep 03.
Article in English | MEDLINE | ID: mdl-22841810

ABSTRACT

1,3-Dinitrobenzene (DNB) causes testicular injury, particularly to Sertoli cells, and induces apoptosis in the surrounding germinal cells in rodents; however, the mechanisms causing this toxicity are poorly understood. Our studies, using standard and molecular tools, were conducted to better understand the pathogenesis of the testicular effects. Four daily oral doses of 0.1-8mg/kg/day caused marked testicular lesions in rats from 4mg/kg/day. Global transcriptomics revealed cell cycle and cell death as the major biological processes affected with the expression of genes associated with cell cycle progression ("mitotic roles of polo-like kinase") being particularly altered. In a single dose time course study (4mg/kg), no adverse changes were recorded; however, in contrast to the data from the multiple dose study, plasma testosterone and testicular steroidogenesis-related gene expression were affected. These steroid hormone effects were confirmed in vitro using the H295R steroidogenesis assay. With this global approach we show that DNB not only induces apoptosis and interferes with cell cycle in the testes but that DNB can also modulate steroid hormone biosynthesis, suggesting an interference with the endocrine system. However, the contribution of the endocrine changes to the severe testicular lesions is presently unknown and requires further investigation.


Subject(s)
Dinitrobenzenes/toxicity , Testis/drug effects , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dinitrobenzenes/administration & dosage , Dose-Response Relationship, Drug , Estradiol/metabolism , Gene Expression Profiling , Humans , Male , Progesterone/metabolism , Random Allocation , Rats , Rats, Wistar , Sertoli Cells/cytology , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Testis/cytology , Testis/metabolism , Testosterone/metabolism
11.
Regul Toxicol Pharmacol ; 64(1): 143-54, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22735369

ABSTRACT

The European regulation on plant protection products (1107/2009) (EC, 2009a), the revisions to the biocides Directive (COM[2009]267) (EC, 2009b), and the regulation concerning chemicals (Regulation (EC) No. 1907/2006 'REACH') (EC.2006) only support the marketing and use of chemical products on the basis that they do not induce endocrine disruption in humans or wildlife species. In the absence of agreed guidance on how to identify and evaluate endocrine activity and disruption within these pieces of legislation a European Centre for Ecotoxicology and Toxicology of Chemicals (ECETOC) task force was formed to provide scientific criteria that may be used within the context of these three legislative documents. The resulting ECETOC technical report (ECETOC, 2009a) and the associated workshop (ECETOC, 2009b) presented a science-based concept on how to identify endocrine activity and disrupting properties of chemicals for both human health and the environment. The synthesis of the technical report and the workshop report was published by the ECETOC task force (Bars et al., 2011a,b). Specific scientific criteria for the determination of endocrine activity and disrupting properties that integrate information from both regulatory (eco)toxicity studies and mechanistic/screening studies were proposed. These criteria combined the nature of the adverse effects detected in studies which give concern for endocrine toxicity with an understanding of the mode of action of toxicity so that adverse effects can be explained scientifically. A key element in the data evaluation is the consideration of all available information in a weight-of-evidence approach. However, to be able to discriminate chemicals with endocrine properties of low concern from those of higher concern (for regulatory purposes), the task force recognised that the concept needed further refinement. Following a discussion of the key factors at a second workshop of invited regulatory, academic and industry scientists (ECETOC, 2011), the task force developed further guidance, which is presented in this paper. For human health assessments these factors include the relevance to humans of the endocrine mechanism of toxicity, the specificity of the endocrine effects with respect to other potential toxic effects, the potency of the chemical to induce endocrine toxicity and consideration of exposure levels. For ecotoxicological assessments the key considerations include specificity and potency, but also extend to the consideration of population relevance and negligible exposure. It is intended that these complement and reinforce the approach originally described and previously published in this journal (Bars et al., 2011a,b).


Subject(s)
Drug and Narcotic Control , Endocrine Disruptors/toxicity , Toxicity Tests/standards , Toxicology/standards , Advisory Committees , Animals , Environmental Monitoring , European Union , Government Agencies , Government Regulation , Guidelines as Topic , Humans , International Agencies , Risk Assessment , Toxicology/legislation & jurisprudence
12.
Toxicol Sci ; 122(1): 52-63, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21525395

ABSTRACT

The safety assessment of chemicals for humans relies on identifying no-observed adverse effect levels (NOAELs) in animal toxicity studies using standard methods. With the advent of high information content technologies, especially microarrays, it is pertinent to determine the impact of molecular data on the NOAELs. Consequently, we conducted an integrative study to identify a no-transcriptomic effect dose using microarray analyses coupled with quantitative reverse transcriptase PCR (RT-qPCR) and determined how this correlated with the NOAEL. We assessed the testicular effects of the antiandrogen, flutamide (FM), in a rat 28-day toxicity study using doses of 0.2-30 mg/kg/day. Plasma testosterone levels and testicular histopathology indicated a NOAEL of 1 mg/kg/day. A no-effect dose of 0.2 mg/kg/day was established based on molecular data relevant to the phenotypic changes. We observed differential gene expression starting from 1 mg/kg/day and a deregulation of more than 1500 genes at 30 mg/kg/day. Dose-related changes were identified for the major pathways (e.g., fatty acid metabolism) associated with the testicular lesion (Leydig cell hyperplasia) that were confirmed by RT-qPCR. These data, along with protein accumulation profiles and FM metabolite concentrations in testis, supported the no-effect dose of 0.2 mg/kg/day. Furthermore, the microarray data indicated a dose-dependent change in the fatty acid catabolism pathway, a biological process described for the first time to be affected by FM in testicular tissue. In conclusion, the present data indicate the existence of a transcriptomic threshold, which must be exceeded to progress from a normal state to an adaptative state and subsequently to adverse toxicity.


Subject(s)
Androgen Antagonists/toxicity , Flutamide/toxicity , Leydig Cells/drug effects , Testicular Diseases/pathology , Animals , Dose-Response Relationship, Drug , Gene Expression Regulation , Leydig Cells/pathology , Lipid Metabolism/drug effects , Male , Microarray Analysis/methods , No-Observed-Adverse-Effect Level , Phenotype , Proteins/genetics , Proteins/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Testicular Diseases/chemically induced , Testosterone/blood , Toxicity Tests/methods , Transcriptome
13.
Regul Toxicol Pharmacol ; 59(1): 37-46, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20858523

ABSTRACT

The European legislation on plant protection products (Regulation (EC) No. 1107/2009) and biocides (Directive 98/8/EC), as well as the regulation concerning chemicals (Regulation (EC) No. 1907/2006 'REACH') only support the marketing and use of chemical products on the basis that they do not induce endocrine disruption in humans or non-target species. However, there is currently no agreed guidance on how to identify and evaluate endocrine activity and disruption. Consequently, an ECETOC task force was formed to provide scientific criteria that may be used within the context of these three legislative documents. Specific scientific criteria for the determination of endocrine disrupting properties that integrate information from both regulatory (eco)toxicity studies and mechanistic/screening studies are proposed. These criteria combine the nature of the adverse effects detected in studies which give concern for endocrine toxicity with an understanding of the mode of action of toxicity so that adverse effects can be explained scientifically. The criteria developed are presented in the form of flow charts for assessing relevant effects for both humans and wildlife species. In addition, since not all chemicals with endocrine disrupting properties are of equal hazard, assessment of potency is also proposed to discriminate chemicals of high concern from those of lower concern. The guidance presented in this paper includes refinements made to an initial proposal following discussion of the criteria at a workshop of invited regulatory, academic and industry scientists.


Subject(s)
Endocrine Disruptors/toxicity , Toxicity Tests/standards , Toxicology/standards , Advisory Committees , Animals , Ecotoxicology/legislation & jurisprudence , Ecotoxicology/standards , Europe , Government Regulation , Guidelines as Topic , Humans , International Agencies , Risk Assessment , Toxicology/legislation & jurisprudence
14.
Drug Metab Dispos ; 38(9): 1598-604, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20551240

ABSTRACT

Cryopreserved rat hepatocytes entrapped within an alginate matrix, commercially available as Liverbeads, were evaluated for their relevance as a screening tool for gene induction in vitro, using quantitative real-time reverse transcriptase-polymerase chain reaction. They were treated with the reference compounds beta-naphthoflavone (BNF), phenobarbital (PB), pregnenolone 16alpha-carbonitrile (PCN), and clofibric acid (CLO) and analyzed for mRNA levels of Cyp1a1, Cyp2b1, Cyp3a1, Cyp4a1, Ugt1a6, and Ugt2b1. In addition, for PB and PCN, the results were compared with those obtained in rat liver in vivo. For each inducer, the gene induction profiles obtained with the Liverbeads in vitro model were time- and dose-dependent. The in vitro gene expression profiles confirmed the corresponding known P450 and UGT induction by each reference compound. In particular, the most strongly induced genes were Cyp1a1 by BNF, Cyp2b1 by PB, Cyp3a1 and Ugt2b1 by PCN, and Cyp4a1 and Cyp2b1 by CLO. Other genes investigated were also induced by the reference compounds, but the expression levels were lower, and increases were seen only after prolonged treatment. In particular, Ugt1a6 and Cyp2b1 were increased by BNF, Cyp1a1, Cyp3a1, and Ugt2b1 by PB, and Cyp3a1 and Ugt2b1 by CLO. All of these results correlated well with published in vitro data and our in vivo data. In conclusion, our results suggest that Liverbeads is a relevant and useful in vitro screening tool for determining gene induction profiles of new molecules. In addition, because Liverbeads from different species are available, this tool offers the possibility to conduct interspecies comparisons.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/drug effects , Models, Theoretical , Animals , Hepatocytes/enzymology , Hepatocytes/metabolism , Male , RNA/genetics , RNA/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
15.
Toxicol Sci ; 109(1): 59-65, 2009 May.
Article in English | MEDLINE | ID: mdl-19299419

ABSTRACT

An important step in the safety assessment of chemicals for humans is to determine the no observed adverse effect level (NOAEL) in toxicity studies conducted in animal models. With the increasing use of molecular tools in toxicity studies, a question often posed is how a NOAEL derived from molecular data compares to a NOAEL established using standard methods. The objective of the present study was to address this question when considering testicular toxicity. To do this, we assessed the effects of the reference antiandrogen flutamide on rat testes in a standard 28-day toxicity study using doses of 0.04-150 mg/kg/day. At necropsy, blood samples were collected for testosterone measurements. The testes were collected for histopathological assessment as well as for the evaluation of gene expression changes using quantitative PCR analyses. Results showed that increases in plasma testosterone level and Leydig cell hyperplasia were detected from 6 mg/kg/day. An alteration in the level of accumulation of a selection of genes was also detected from 6 mg/kg/day. This was the case for genes functionally associated with the testicular lesion, such as lipid metabolism and cell death/cell proliferation, as well as for genes not functionally associated with the lesion. Contrary to the misgivings, these data show that, using a standard 28-day toxicity study and a well-characterized adverse effect, the NOAEL based on transcript changes is similar to the NOAELs based on testosterone levels and histopathological examination.


Subject(s)
Androgen Antagonists/toxicity , Flutamide/toxicity , Gene Expression/drug effects , Testis/drug effects , Toxicity Tests/methods , Animals , Cell Death/drug effects , Cell Proliferation/drug effects , Data Interpretation, Statistical , Gene Expression Profiling , Histocytochemistry , Hyperplasia , Leydig Cells/metabolism , Leydig Cells/pathology , Lipid Metabolism/drug effects , Male , No-Observed-Adverse-Effect Level , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Testis/metabolism , Testosterone/blood , Toxicogenetics
16.
Regul Toxicol Pharmacol ; 54(3 Suppl): S52-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19100305

ABSTRACT

Genetically modified crops convey many benefits to world population. However, a rigorous safety assessment procedure, including an evaluation of the allergenic potential, is fundamental before their release into the food chain. As an integral part of the safety assessment process, regulatory authorities worldwide strongly recommend the use of tests that can predict the allergenic potential of the novel proteins. All guidance documents are based on an array of tests that have been proposed in 2003 by the Codex Alimentarius. Although the animal model is not a requirement of the Codex Alimentarius weight of evidence approach, allergenic hazard of novel proteins could only be evaluated by an in vivo model that can potentially identify and distinguish commonly allergenic proteins from rarely allergenic proteins. Therefore, food allergy experts encourage its development. During the 2007 International Life Science Institute (ILSI) workshop (Nice, France), worldwide experts shared their latest research results on rodent models to evaluate the allergenic potential of proteins and foods. This review presents the most promising rodent models for assessing food protein allergenicity that were evaluated during this ILSI workshop.


Subject(s)
Allergens/immunology , Food Hypersensitivity/etiology , Models, Animal , Proteins/immunology , Animals , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Immunoglobulin E/biosynthesis , Immunoglobulin G/biosynthesis , Mice , Risk Assessment
17.
Toxicol Sci ; 100(1): 54-65, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17686919

ABSTRACT

The Organization for Economic Cooperation and Development (OECD) is currently funding the validation of the Hershberger assay as a rapid in vivo means of identifying (anti-) androgens. However, as the assay measures weight changes in the androgen-sensitive tissues of castrated rats, the evaluation of the androgen-stimulated intact weanling as a more ethical model to use in the assay has been requested. As part of the OECD validation exercise two weak antiandrogens, 1,1-dichloro-2,2-bis(4 chlorophenyl)ethane (DDE) and linuron (LIN), were investigated in our laboratory at several dose levels in the testosterone propionate (TP)-stimulated weanling using flutamide (FM) as a positive control. In addition to weight measurements (sex accessory tissues [SATs], epididymides, and testes), histopathological assessment of the seminal vesicles, prostate, and testes was conducted for vehicle control, TP-stimulated, and TP-stimulated animals treated with FM or the top dose level of DDE or LIN. The modulation of a novel prostate protein associated with apoptosis, L-amino acid oxidase (LAO), was evaluated in these same treatment groups. Our gravimetric data (supported by the histopathology data) indicated that the weanling assay can detect SAT and epididymal weight changes induced by the antiandrogens evaluated. Inconsistent and variable data were recorded for the testicular weight and histopathological effects, suggesting that the testis is of little value in the identification of antiandrogens using this model. Three isoforms of LAO were identified, and all were regulated by TP. Modulation of LAO by the antiandrogens indicated that this protein could be a biomarker for endocrine disruption in male rodents.


Subject(s)
Androgen Antagonists/toxicity , Dichlorodiphenyl Dichloroethylene/toxicity , Endocrine Disruptors/toxicity , Flutamide/toxicity , Genitalia, Male/drug effects , Linuron/toxicity , Proteomics , Toxicity Tests/methods , Adrenal Glands/drug effects , Animals , Biomarkers, Pharmacological/metabolism , Body Weight/drug effects , Dose-Response Relationship, Drug , Epididymis/drug effects , Genitalia, Male/metabolism , Genitalia, Male/pathology , Kidney/drug effects , L-Amino Acid Oxidase/metabolism , Liver/drug effects , Male , Organ Size/drug effects , Prostate/drug effects , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Seminal Vesicles/drug effects , Testis/drug effects , Testosterone Propionate/pharmacology , Time Factors , Weaning
18.
Toxicol Sci ; 97(1): 81-93, 2007 May.
Article in English | MEDLINE | ID: mdl-17311803

ABSTRACT

In conventional rodent toxicity studies the characterization of the adverse effects of a chemical relies primarily on gravimetric, and histopathological data. The aim of this study was to evaluate if the use of two-dimensional gel electrophoresis could generate protein accumulation profiles, which were in accordance with conventional toxicological findings by investigating a model antiandrogen, flutamide (FM), whose toxic effects, as measured using standard approaches, are well characterized. Male Sprague-Dawley rats were orally exposed to FM (0, 6, 30, and 150 mg/kg/day) for 28 days. The expected inhibition of androgen-dependent tissue stimulation, increased luteinizing hormone and testosterone plasma levels, and Leydig cell hyperplasia were observed. Changes in testicular protein accumulation profiles were evaluated in rats exposed to 150 mg/kg/day FM. Several proteins involved in steroidogenesis (e.g., StAR, ApoE, Hmgcs1, Idi1), cell cycle, and cancer (e.g., Ddx1, Hspd1) were modulated by FM, and these data provided molecular evidence for the hormonal and testicular histopathology changes recorded. Changes in proteins associated with spermatogenesis were also recorded, and these are discussed within the context of the testicular phenotype observed following FM treatment (i.e., normal spermatogenesis but Leydig cell hyperplasia). Overall, our data indicate that the combination of conventional toxicology measurements with omic observations has the potential to improve our global understanding of the toxicity of a compound.


Subject(s)
Androgen Antagonists/toxicity , Flutamide/toxicity , Genitalia, Male/drug effects , Proteins/metabolism , Proteomics/methods , Toxicity Tests/methods , Animals , Biomarkers/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Gel, Two-Dimensional , Gene Regulatory Networks/drug effects , Genitalia, Male/metabolism , Genitalia, Male/pathology , Hyperplasia , Leydig Cells/drug effects , Leydig Cells/metabolism , Luteinizing Hormone/blood , Male , Organ Size/drug effects , Proteins/genetics , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Testis/drug effects , Testis/metabolism , Testosterone/blood
19.
Mol Cell Proteomics ; 5(11): 2031-43, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16837577

ABSTRACT

To better understand the effects of antiandrogens on the prostate, we investigated the changes in the proteome of rat ventral prostate (VP) following treatment with a well characterized 5alpha-reductase inhibitor, finasteride. Sprague-Dawley rats were treated daily by gavage with finasteride at 0, 1, 5, 25, and 125 mg/kg/day. Changes in plasma hormone levels as well as the weight and histology of sex accessory tissues were determined after 28 days of treatment and showed a dose-related decrease of VP weights together with a marked atrophy of the tissue visible at the macroscopic and microscopic levels. In addition, significant reductions in seminal vesicle and epididymis weights were noted. VP proteins were analyzed by two-dimensional gel electrophoresis: 37 proteins, mainly involved in protein synthesis, processing, and cellular trafficking and in metabolism, detoxification, and oxidative stress, were identified as modulated by finasteride. The prominent feature of this study is the demonstration of finasteride dose-dependent up-regulation of a protein similar to l-amino-acid oxidase 1 (Lao1). An up-regulation of this protein was also observed with the antiandrogen flutamide. Lao1 expression occurred as early as 48 h after antiandrogen administration and persisted throughout the treatment duration. Immunohistochemistry showed that this protein was only detectable in epithelial cells and secretory vesicles. Altogether these data point to a potential use of Lao1 to reveal antiandrogen-induced prostate injury.


Subject(s)
Enzyme Inhibitors/administration & dosage , Finasteride/administration & dosage , Prostate/drug effects , Protein Array Analysis , Proteins/analysis , 5-alpha Reductase Inhibitors , Animals , Electrophoresis, Gel, Two-Dimensional , Epithelial Cells/enzymology , L-Amino Acid Oxidase/analysis , L-Amino Acid Oxidase/metabolism , Male , Organ Size/drug effects , Phosphorylation , Prostate/cytology , Prostate/metabolism , Rats , Rats, Sprague-Dawley , Secretory Vesicles/enzymology , Tyrosine/metabolism
20.
Endocrinology ; 146(12): 5135-43, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16166221

ABSTRACT

In utero exposure to chemicals with antiandrogen activity induces undescended testis, hypospadias, and sub- or infertility. The hypospermatogenesis observed in the adult rat testis exposed in utero to the antiandrogen flutamide has been reported to be a result of a long-term apoptotic cell death process in mature germ cells. However, little if anything is known about the upstream signaling mechanisms controlling this apoptosis. In the present study, we have investigated the possibility that the TGF-beta signaling pathway may be at play in this control of the apoptotic germ cell death process. By using a model of adult rat exposed in utero to 0, 0.4, 2, or 10 mg/kg.d flutamide, we observed that pro-TGF-beta signaling members, such as the three isoforms of TGF-beta ligands (TGF-beta1-3), the two TGF-beta receptors (TGF-betaRI and -RII) and the R-Smads Smad 1, Smad 2, Smad 3, and Smad 5 were inhibited at the mRNA and protein levels, whereas the anti-TGF-beta signaling member Smad 7 was overexpressed. Furthermore, we report that the overexpression of Smad 7 mRNA could induce an activation of c-Jun N-terminal kinase, because of the observed c-Jun overexpression, activation, and nuclear translocation leading to an increase in the transcription of the proapoptotic factor Fas-L. Together, the alterations of TGF-beta signaling may represent upstream mechanisms underlying the adult germ cell apoptotic process evidenced in adult rat testis exposed in utero to antiandrogenic compounds such as flutamide.


Subject(s)
Androgen Antagonists/pharmacology , Apoptosis/drug effects , Fetus/drug effects , Signal Transduction , Spermatozoa/physiology , Transforming Growth Factor beta/metabolism , Aging/metabolism , Androgen Antagonists/administration & dosage , Animals , Dose-Response Relationship, Drug , Fas Ligand Protein , Female , Flutamide/administration & dosage , Flutamide/pharmacology , Male , Membrane Glycoproteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Transforming Growth Factor beta/metabolism , Smad Proteins/genetics , Smad1 Protein/metabolism , Smad7 Protein/metabolism , Testis/cytology , Testis/metabolism , Time Factors , Tissue Distribution , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tumor Necrosis Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...