1
|
Canzler S, Schubert K, Rolle-Kampczyk UE, Wang Z, Schreiber S, Seitz H, Mockly S, Kamp H, Haake V, Huisinga M, Bergen MV, Buesen R, Hackermüller J. Evaluating the performance of multi-omics integration: a thyroid toxicity case study. Arch Toxicol 2024:10.1007/s00204-024-03876-2. [PMID: 39441382 DOI: 10.1007/s00204-024-03876-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Multi-omics data integration has been repeatedly discussed as the way forward to more comprehensively cover the molecular responses of cells or organisms to chemical exposure in systems toxicology and regulatory risk assessment. In Canzler et al. (Arch Toxicol 94(2):371-388. https://doi.org/10.1007/s00204-020-02656-y ), we reviewed the state of the art in applying multi-omics approaches in toxicological research and chemical risk assessment. We developed best practices for the experimental design of multi-omics studies, omics data acquisition, and subsequent omics data integration. We found that multi-omics data sets for toxicological research questions were generally rare, with no data sets comprising more than two omics layers adhering to these best practices. Due to these limitations, we could not fully assess the benefits of different data integration approaches or quantitatively evaluate the contribution of various omics layers for toxicological research questions. Here, we report on a multi-omics study on thyroid toxicity that we conducted in compliance with these best practices. We induced direct and indirect thyroid toxicity through Propylthiouracil (PTU) and Phenytoin, respectively, in a 28-day plus 14-day recovery oral rat toxicity study. We collected clinical and histopathological data and six omics layers, including the long and short transcriptome, proteome, phosphoproteome, and metabolome from plasma, thyroid, and liver. We demonstrate that the multi-omics approach is superior to single-omics in detecting responses at the regulatory pathway level. We also show how combining omics data with clinical and histopathological parameters facilitates the interpretation of the data. Furthermore, we illustrate how multi-omics integration can hint at the involvement of non-coding RNAs in post-transcriptional regulation. Also, we show that multi-omics facilitates grouping, and we assess how much information individual and combinations of omics layers contribute to this approach.
Collapse
Affiliation(s)
- Sebastian Canzler
- Helmholtz Centre for Environmental Research, UFZ, 04318, Leipzig, Germany.
| | - Kristin Schubert
- Helmholtz Centre for Environmental Research, UFZ, 04318, Leipzig, Germany
| | | | - Zhipeng Wang
- Helmholtz Centre for Environmental Research, UFZ, 04318, Leipzig, Germany
| | - Stephan Schreiber
- Helmholtz Centre for Environmental Research, UFZ, 04318, Leipzig, Germany
| | - Hervé Seitz
- Institut de Génétique Humaine UMR 9002 CNRS-Université de Montpellier, 34396, Montpellier Cedex 5, France
| | - Sophie Mockly
- Institut de Génétique Humaine UMR 9002 CNRS-Université de Montpellier, 34396, Montpellier Cedex 5, France
| | - Hennicke Kamp
- BASF Metabolome Solutions GmbH, 10589, Berlin, Germany
| | - Volker Haake
- BASF Metabolome Solutions GmbH, 10589, Berlin, Germany
| | - Maike Huisinga
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen, Germany
| | - Martin von Bergen
- Helmholtz Centre for Environmental Research, UFZ, 04318, Leipzig, Germany
| | - Roland Buesen
- Experimental Toxicology and Ecology, BASF SE, 67056, Ludwigshafen, Germany
| | - Jörg Hackermüller
- Helmholtz Centre for Environmental Research, UFZ, 04318, Leipzig, Germany.
| |
Collapse
|
2
|
Jiang L, Yang J, Yang H, Kong L, Ma H, Zhu Y, Zhao X, Yang T, Liu W. Advanced understanding of the polybrominated diphenyl ethers (PBDEs): Insights from total environment to intoxication. Toxicology 2024; 509:153959. [PMID: 39341352 DOI: 10.1016/j.tox.2024.153959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Polybrominated diphenyl ethers (PBDEs) are brominated compounds connected by ester bonds between two benzene rings. There are 209 congeners of PBDEs, classified according to the number and position of the bromine atoms. Due to their low cost and superior flame retardant properties, PBDEs have been extensively used as flame retardants in electronic products, plastics, textiles, and other materials since the 1970s. PBDEs are classified as persistent organic pollutants (POPs) under the Stockholm Convention because of their environmental persistence, bioaccumulation, and toxicity to both humans and wildlife. Due to their extensive use and significant quantities, PBDEs have been detected across a range of environments and biological organisms. These compounds are known to cause damage to the metabolic system, exhibit neurotoxicity, and pose reproductive hazards. This review investigates the environmental distribution and human exposure pathways of PBDEs. Using China-a country with significant PBDE use-as an example, it highlights substantial regional and temporal variations in PBDE concentrations and notes that certain environmental levels may pose risks to human health. The article then examines the toxic effects and mechanisms of PBDEs on several major target organs, summarizing recent research and the specific mechanisms underlying these toxic effects from multiple toxicological perspectives. This review enhances our understanding of PBDEs' environmental distribution, exposure pathways, and toxic mechanisms, offering valuable insights for further research and management strategies.
Collapse
Affiliation(s)
- Liujiangshan Jiang
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang 110122, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China
| | - Jing Yang
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang 110122, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China
| | - Huajie Yang
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang 110122, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China
| | - Lingxu Kong
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang 110122, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China
| | - Haonan Ma
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang 110122, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China
| | - Yapei Zhu
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang 110122, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China
| | - Xuan Zhao
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang 110122, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China
| | - Tianyao Yang
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang 110122, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China.
| | - Wei Liu
- Department of Environmental and Occupational Health, School of Public Health, China Medical University, Shenyang 110122, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China.
| |
Collapse
|
3
|
Baze A, Wiss L, Horbal L, Biemel K, Asselin L, Richert L. Comparison of in vitro thyroxine (T4) metabolism between Wistar rat and human hepatocyte cultures. Toxicol In Vitro 2024; 96:105763. [PMID: 38142784 DOI: 10.1016/j.tiv.2023.105763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/24/2023] [Accepted: 12/05/2023] [Indexed: 12/26/2023]
Abstract
In vitro assays remain relatively new in exploring human relevance of liver, in particular nuclear receptor-mediated perturbations of the hypothalamus-pituitary-thyroid axis seen in rodents, mainly in the rat. Consistent with in vivo data, we confirm that thyroid hormone thyroxine metabolism was 9 times higher in primary rat hepatocytes (PRH) than in primary human hepatocytes (PHH) cultured in a 2D sandwich (2Dsw) configuration. In addition, thyroxine glucuronide (T4-G) was by far the major metabolite formed in both species (99.1% in PRH and 69.7% in PHH) followed by thyroxine sulfate (T4-S, 0.7% in PRH and 18.1% in PHH) and triiodothyronine/reverse triiodothyronine (T3/rT3, 0.2% in PRH and 12.2% in PHH). After a 7-day daily exposure to orphan receptor-mediated liver inducers, T4 metabolism was strongly increased in PRH, almost exclusively through increased T4-G formation. These results were consistent with the inductions of glucuronosyltransferase Ugt2b1 and canalicular transporter Mrp2. PHH also responded to activation of the three nuclear receptors, with mainly induction of glucuronosyltransferase UGT1A1 and canalicular transporter MRP2. Despite this, T4 disappearance rate and secreted T4 metabolites were only slightly increased in PHH. Overall, our data highlight that cryopreserved hepatocytes in 2Dsw culture allowing long-term exposure and species comparison are of major interest in improving liver-mediated human safety assessment.
Collapse
Affiliation(s)
- Audrey Baze
- KaLy-Cell SAS, 20A rue du Général Leclerc, 67115 Plobsheim, France
| | - Lucille Wiss
- KaLy-Cell SAS, 20A rue du Général Leclerc, 67115 Plobsheim, France
| | - Liliia Horbal
- Pharmacelsus GmbH, Science Park 2, 66123 Saarbrüken, Germany
| | - Klaus Biemel
- Pharmacelsus GmbH, Science Park 2, 66123 Saarbrüken, Germany
| | - Laure Asselin
- KaLy-Cell SAS, 20A rue du Général Leclerc, 67115 Plobsheim, France
| | - Lysiane Richert
- KaLy-Cell SAS, 20A rue du Général Leclerc, 67115 Plobsheim, France; Zylan SAS, 8 rue de la Haute Corniche, 67210 Obernai, France.
| |
Collapse
|
4
|
Kamp H, Kocabas NA, Faulhammer F, Synhaeve N, Rushton E, Flick B, Giri V, Sperber S, Higgins LG, Penman MG, van Ravenzwaay B, Rooseboom M. Utility of in vivo metabolomics to support read-across for UVCB substances under REACH. Arch Toxicol 2024; 98:755-768. [PMID: 38265474 PMCID: PMC10861390 DOI: 10.1007/s00204-023-03638-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 11/13/2023] [Indexed: 01/25/2024]
Abstract
Structure-based grouping of chemicals for targeted testing and read-across is an efficient way to reduce resources and animal usage. For substances of unknown or variable composition, complex reaction products, or biological materials (UVCBs), structure-based grouping is virtually impossible. Biology-based approaches such as metabolomics could provide a solution. Here, 15 steam-cracked distillates, registered in the EU through the Lower Olefins Aromatics Reach Consortium (LOA), as well as six of the major substance constituents, were tested in a 14-day rat oral gavage study, in line with the fundamental elements of the OECD 407 guideline, in combination with plasma metabolomics. Beyond signs of clinical toxicity, reduced body weight (gain), and food consumption, pathological investigations demonstrated the liver, thyroid, kidneys (males only), and hematological system to be the target organs. These targets were confirmed by metabolome pattern recognition, with no additional targets being identified. While classical toxicological parameters did not allow for a clear distinction between the substances, univariate and multivariate statistical analysis of the respective metabolomes allowed for the identification of several subclusters of biologically most similar substances. These groups were partly associated with the dominant (> 50%) constituents of these UVCBs, i.e., indene and dicyclopentadiene. Despite minor differences in clustering results based on the two statistical analyses, a proposal can be made for the grouping of these UVCBs. Both analyses correctly clustered the chemically most similar compounds, increasing the confidence that this biological approach may provide a solution for the grouping of UVCBs.
Collapse
Affiliation(s)
- H Kamp
- BASF Metabolome Solutions GmbH, Berlin, Germany
| | | | | | | | - E Rushton
- LyondellBasell, Rotterdam, The Netherlands
| | - B Flick
- BASF SE, Ludwigshafen, Germany
- NUVISAN ICB GmbH, Toxicology, 13353, Berlin, Germany
| | - V Giri
- BASF SE, Ludwigshafen, Germany
| | | | - L G Higgins
- LOA C/O Penman Consulting Ltd, Brussels, Belgium
| | - M G Penman
- LOA C/O Penman Consulting Ltd, Brussels, Belgium
| | | | - M Rooseboom
- Shell Global Solution International B.V, The Hague, The Netherlands
| |
Collapse
|
5
|
Tallon MJ, Child R. Subchronic oral toxicity assessment of a cannabis extract. Regul Toxicol Pharmacol 2023; 144:105496. [PMID: 37734651 DOI: 10.1016/j.yrtph.2023.105496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023]
Abstract
Cannabidiol (CBD) is present in Cannabis Sativa L. and has been used in medicines and foods to deliver beneficial health effects. Despite this, research on CBD safety utilising modern testing methods is lacking. Therefore three separate safety experiments were performed on a CBD isolate. Sprague-Dawley rats were used to investigate prenatal development, a 14-day toxicity sighting study, and an OECD compliant 90-day subchronic oral toxicity trial, with 35-day off-dose recovery. The prenatal screening study demonstrated reduced body weights and food consumption in the highest dose group, but no substance-related changes in pregnancy rate, maternal or placental gross abnormalities, or premature deliveries. The 14-day study indicated tolerance up to 460 mg/kg bw/d of CBD isolate. Based on these findings, a 90-day repeated dose oral toxicity study was performed at doses of 0, 30, 115, 230, and 460 mg/kg bw/d of CBD, followed by a 35-day off-dose recovery period. In the 90-day study, some non-adverse organ and tissue changes were observed. With the exception of the high dose group, these fully reversed during the recovery period. Based on these findings, sub-chronic consumption of highly purified isolate results in a CBD NOAEL of 460 mg/kg bw/d for males and 230 mg/kg bw/d for females.
Collapse
Affiliation(s)
- Mark J Tallon
- Legal Products Group Ltd, 26 St Thomas Pl, Ely, Cambridgeshire, CB7 4EX, UK.
| | - Robert Child
- Legal Products Group Ltd, 26 St Thomas Pl, Ely, Cambridgeshire, CB7 4EX, UK; School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
6
|
Amin MF, Zubair MS, Ammar M. A short review on the role of thyroxine in fast wound healing and tissue regeneration. Tissue Cell 2023; 82:102115. [PMID: 37244096 DOI: 10.1016/j.tice.2023.102115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 05/29/2023]
Abstract
Wound healing is a multiplex interaction process that involves extracellular matrix, blood vessels, proteases, cytokines, and chemokine. So far, a number of studies have been performed to understand the basis of the wound-healing process and multiple wound-healing products have been designed. However, significant morbidity and mortality incidents still occurred due to poor wound healing. Thus, there is a dire need to understand the effects of topical applications of various therapeutic options that lead to fast wound healing. Thyroxine is one great panacea for wound healing that has been vigorously mooted throughout the years but a conclusive result regarding its effectiveness is still not achieved. This review is intended to find a rational basis for its positive role in wound healing. To accomplish the objective, this review highlights the different aspects of thyroxine's role in wound healing like keratin synthesis, skin thickening, and pro-angiogenesis, the basis of controversy on its wound healing ability and its potential to be used as a wound healing agent. This study will be helpful for researchers and surgeons to assess the importance of thyroxine as a candidate to comprehensively research to develop a potent, effective, and affordable wound healing drug.
Collapse
Affiliation(s)
| | | | - Muhammad Ammar
- Princess Royal University Hospital, King College Hospital Trust, United Kingdom.
| |
Collapse
|
7
|
Kruger C, Symonds J, Conze D. Toxicological assessment of truebroc® glucoraphanin-rich broccoli seed extract. TOXICOLOGY RESEARCH AND APPLICATION 2022. [DOI: 10.1177/23978473211068834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
TrueBroc® Glucoraphanin-Rich Broccoli Seed Extract (TrueBroc®), is a hot water extract of Brassica oleracea var. Italica Plenck (Brassicaceae) seeds. The broccoli seeds are non-genetically modified and are tested and selected for their specific glucosinolate content. TrueBroc® contains greater than 99% of the aliphatic glucosinolate glucoraphanin and its precursor glucoerucin in the glucosinolate fraction. A battery of toxicology assays was conducted on TrueBroc® which includes in vitro and in vivo genotoxicity studies, an acute oral toxicity study in rats, a subchronic toxicology study in rats. The result from the subchronic rat study establishes a NOAEL of 1500 mg/kg/day.
Collapse
|
8
|
Vansell NR. Mechanisms by Which Inducers of Drug Metabolizing Enzymes Alter Thyroid Hormones in Rats. Drug Metab Dispos 2022; 50:508-517. [DOI: 10.1124/dmd.121.000498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 12/23/2021] [Indexed: 11/22/2022] Open
|
9
|
Parmentier C, Baze A, Untrau M, Kampkoetter A, Lasserre D, Richert L. Evaluation of human relevance of Nicofluprole-induced rat thyroid disruption. Toxicol Appl Pharmacol 2021; 435:115831. [PMID: 34922950 DOI: 10.1016/j.taap.2021.115831] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 10/19/2022]
Abstract
Nicofluprole is a novel insecticide of the phenylpyrazole class conferring selective antagonistic activity on insect GABA receptors. After repeated daily dietary administration to Wistar rats for 28/90 days, Nicofluprole induced increases in thyroid (and liver) weight, associated with histopathology changes. Nicofluprole did not inhibit thyroid peroxydase nor sodium/iodide symporter, two key players in the biosynthesis of thyroid hormones, indicating the absence of a direct thyroid effect. The results seen in rats suggested a mode of action of Nicofluprole driven by the molecular initiating event of CAR/PXR nuclear receptor activation in livers, with key events of increases in liver weight and hypertrophy, decreasing circulatory thyroid hormones, a compensatory increase in TSH release and follicular cell hypertrophy. To explore the relevance of these changes to humans, well established in vitro rat and human sandwich-cultured hepatocytes were exposed to Nicofluprole up to 7 days. A concentration-dependent CYP3A induction (PXR-activation), an increase in T4-glucuronoconjugation accompanied by UGT1A/2B inductions was observed in rat but not in human hepatocytes. The inductions seen with Nicofluprole in rat (in vivo and in vitro in hepatocytes) that were absent in human hepatocytes represent another example of species-selectivity of nuclear CAR/PXR receptor activators. Importantly, the different pattern observed in rat and human models demonstrate that Nicofluprole-related thyroid effects observed in the rat are with no human relevance.
Collapse
Affiliation(s)
- Céline Parmentier
- KaLy-Cell S.A.S, 20A rue du Général Leclerc, 67115 Plobsheim, France.
| | - Audrey Baze
- KaLy-Cell S.A.S, 20A rue du Général Leclerc, 67115 Plobsheim, France.
| | - Meiggie Untrau
- KaLy-Cell S.A.S, 20A rue du Général Leclerc, 67115 Plobsheim, France
| | - Andreas Kampkoetter
- Bayer Animal Health GmbH, An Elanco Animal Health Company, 50 Alfred-Nobel-Strasse, 40789 Monheim, Germany.
| | - Dominique Lasserre
- Bayer S.A.S. Bayer CropScience, 355 rue Dostoïevski, F-06560 Sophia Antipolis, France.
| | - Lysiane Richert
- KaLy-Cell S.A.S, 20A rue du Général Leclerc, 67115 Plobsheim, France.
| |
Collapse
|
10
|
Marty S, Beekhuijzen M, Charlton A, Hallmark N, Hannas BR, Jacobi S, Melching-Kollmuss S, Sauer UG, Sheets LP, Strauss V, Urbisch D, Botham PA, van Ravenzwaay B. Towards a science-based testing strategy to identify maternal thyroid hormone imbalance and neurodevelopmental effects in the progeny - part II: how can key events of relevant adverse outcome pathways be addressed in toxicological assessments? Crit Rev Toxicol 2021; 51:328-358. [PMID: 34074207 DOI: 10.1080/10408444.2021.1910625] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The current understanding of thyroid-related adverse outcome pathways (AOPs) with adverse neurodevelopmental outcomes in mammals has been reviewed. This served to establish if standard rodent toxicity test methods and in vitro assays allow identifying thyroid-related modes-of-action potentially leading to adverse neurodevelopmental outcomes, and the human relevance of effects - in line with the European Commission's Endocrine Disruptor Criteria. The underlying hypothesis is that an understanding of the key events of relevant AOPs provides insight into differences in incidence, magnitude, or species sensitivity of adverse outcomes. The rodent studies include measurements of serum thyroid hormones, thyroid gland pathology and neurodevelopmental assessments, but do not directly inform on specific modes-of-action. Opportunities to address additional non-routine parameters reflecting critical events of AOPs in toxicological assessments are presented. These parameters appear relevant to support the identification of specific thyroid-related modes-of-action, provided that prevailing technical limitations are overcome. Current understanding of quantitative key event relationships is often weak, but would be needed to determine if the triggering of a molecular initiating event will ultimately result in an adverse outcome. Also, significant species differences in all processes related to thyroid hormone signalling are evident, but the biological implications thereof (including human relevance) are often unknown. In conclusion, careful consideration of the measurement (e.g. timing, method) and interpretation of additional non-routine parameters is warranted. These findings will be used in a subsequent paper to propose a testing strategy to identify if a substance may elicit maternal thyroid hormone imbalance and potentially also neurodevelopmental effects in the progeny.
Collapse
Affiliation(s)
- Sue Marty
- The Dow Chemical Company, Midland, MI, USA
| | | | | | | | | | | | | | - Ursula G Sauer
- Scientific Consultancy - Animal Welfare, Neubiberg, Germany
| | | | | | | | | | | |
Collapse
|
11
|
Bomann W, Tinwell H, Jenkinson P, Kluxen FM. Metribuzin-induced non-adverse liver changes result in rodent-specific non-adverse thyroid effects via uridine 5'-diphospho-glucuronosyltransferase (UDPGT, UGT) modulation. Regul Toxicol Pharmacol 2021; 122:104884. [PMID: 33596450 DOI: 10.1016/j.yrtph.2021.104884] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 01/19/2021] [Accepted: 02/01/2021] [Indexed: 11/18/2022]
Abstract
Metribuzin is a herbicide that inhibits photosynthesis and has been used for over 40 years. Its main target organ is the liver and to some extent the kidney in rats, dogs, and rabbits. Metribuzin shows a specific thyroxine (T4) profile in rat studies with T4 increases at low doses and T4 decreases at higher doses. Only the T4 decreases occur together with histopathological changes in the thyroid and weight changes of liver and thyroid. A set of experiments was conducted to investigate metribuzin's endocrine disruptor potential according to European guidance and regulations. The results indicate that a liver enzyme modulation, i.e. of the uridine 5'-diphospho-glucuronosyltransferase (UDPGT, UGT), is most likely responsible for both increased and decreased plasma thyroxine level and for thyroid histopathological observations. Animals with high T4 levels show low UGT activity, while animals with low T4 levels show high UGT activity. A causal relationship was inferred, since other potentially human-relevant mode of action (MOA) pathways were excluded in dedicated studies, i.e. inhibition of deiodinases (DIO), inhibition of thyroid peroxidase (TPO) or of the sodium importer system (NIS). This liver metabolism-associated MOA is considered not relevant for human hazard assessment, due to species differences in thyroid homeostasis between humans and rats and, more importantly, based on experimental data showing that metribuzin affects UGT activity in rat but not in human hepatocytes. Further, we discuss whether or not increased T4 levels in the rat, in the absence of histopathological changes, should be considered as adverse and therefore used as an appropriate hazard model for humans. Based on a weight of evidence approach, metribuzin should not be classified as an endocrine disruptor with regard to the thyroid modality.
Collapse
Affiliation(s)
- Werner Bomann
- Toxconsult, 9393 W 110th Street, 51 Corporate Woods, Suite 500, Overland Park, KS, 66210, USA.
| | - Helen Tinwell
- Bayer.SAS, 16 rue Jean-Marie Leclair, 69009, Lyon, France
| | | | | |
Collapse
|
12
|
A review of species differences in the control of, and response to, chemical-induced thyroid hormone perturbations leading to thyroid cancer. Arch Toxicol 2021; 95:807-836. [PMID: 33398420 DOI: 10.1007/s00204-020-02961-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022]
Abstract
This review summarises the current state of knowledge regarding the physiology and control of production of thyroid hormones, the effects of chemicals in perturbing their synthesis and release that result in thyroid cancer. It does not consider the potential neurodevelopmental consequences of low thyroid hormones. There are a number of known molecular initiating events (MIEs) that affect thyroid hormone synthesis in mammals and many chemicals are able to activate multiple MIEs simultaneously. AOP analysis of chemical-induced thyroid cancer in rodents has defined the key events that predispose to the development of rodent cancer and many of these will operate in humans under appropriate conditions, if they were exposed to high enough concentrations of the affecting chemicals. There are conditions however that, at the very least, would indicate significant quantitative differences in the sensitivity of humans to these effects, with rodents being considerably more sensitive to thyroid effects by virtue of differences in the biology, transport and control of thyroid hormones in these species as opposed to humans where turnover is appreciably lower and where serum transport of T4/T3 is different to that operating in rodents. There is heated debate around claimed qualitative differences between the rodent and human thyroid physiology, and significant reservations, both scientific and regulatory, still exist in terms of the potential neurodevelopmental consequences of low thyroid hormone levels at critical windows of time. In contrast, the situation for the chemical induction of thyroid cancer, through effects on thyroid hormone production and release, is less ambiguous with both theoretical, and actual data, showing clear dose-related thresholds for the key events predisposing to chemically induced thyroid cancer in rodents. In addition, qualitative differences in transport, and quantitative differences in half life, catabolism and turnover of thyroid hormones, exist that would not operate under normal situations in humans.
Collapse
|
13
|
Garcia de Lomana M, Weber AG, Birk B, Landsiedel R, Achenbach J, Schleifer KJ, Mathea M, Kirchmair J. In Silico Models to Predict the Perturbation of Molecular Initiating Events Related to Thyroid Hormone Homeostasis. Chem Res Toxicol 2020; 34:396-411. [PMID: 33185102 PMCID: PMC7887800 DOI: 10.1021/acs.chemrestox.0c00304] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Disturbance of the thyroid hormone homeostasis has been associated with adverse health effects such as goiters and impaired mental development in humans and thyroid tumors in rats. In vitro and in silico methods for predicting the effects of small molecules on thyroid hormone homeostasis are currently being explored as alternatives to animal experiments, but are still in an early stage of development. The aim of this work was the development of a battery of in silico models for a set of targets involved in molecular initiating events of thyroid hormone homeostasis: deiodinases 1, 2, and 3, thyroid peroxidase (TPO), thyroid hormone receptor (TR), sodium/iodide symporter, thyrotropin-releasing hormone receptor, and thyroid-stimulating hormone receptor. The training data sets were compiled from the ToxCast database and related scientific literature. Classical statistical approaches as well as several machine learning methods (including random forest, support vector machine, and neural networks) were explored in combination with three data balancing techniques. The models were trained on molecular descriptors and fingerprints and evaluated on holdout data. Furthermore, multi-task neural networks combining several end points were investigated as a possible way to improve the performance of models for which the experimental data available for model training are limited. Classifiers for TPO and TR performed particularly well, with F1 scores of 0.83 and 0.81 on the holdout data set, respectively. Models for the other studied targets yielded F1 scores of up to 0.77. An in-depth analysis of the reliability of predictions was performed for the most relevant models. All data sets used in this work for model development and validation are available in the Supporting Information.
Collapse
Affiliation(s)
- Marina Garcia de Lomana
- BASF SE, 67063 Ludwigshafen am Rhein, Germany.,Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | | | | | | | | | | | | | - Johannes Kirchmair
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
14
|
Colnot T, Melching-Kollmuß S, Semino G, Dekant W. A flow scheme for cumulative assessment of pesticides for adverse liver effects. Regul Toxicol Pharmacol 2020; 116:104694. [PMID: 32621977 DOI: 10.1016/j.yrtph.2020.104694] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/15/2020] [Accepted: 05/28/2020] [Indexed: 12/17/2022]
Abstract
The European Food Safety Authority (EFSA) is developing approaches to cumulative risk assessment by assigning pesticides to cumulative assessment groups (CAGs). For assignment to CAGs, EFSA relies on common toxic effects (CTEs) on the target system. The developed flow scheme for assignment to liver CAGs sequentially assesses the consistency of the CTE, its adversity, its potential to be secondary to other toxicities, its human relevance, and the relation of the NOAEL for the CTE to the overall NOAEL. If the responses to all questions are "yes", allocation to a CAG is supported; "no" stops the process.
Collapse
Affiliation(s)
| | | | | | - Wolfgang Dekant
- Department of Toxicology, University of Würzburg, Versbacher Strasse 9, 97078, Würzburg, Germany.
| |
Collapse
|
15
|
Ramhøj L, Hass U, Gilbert ME, Wood C, Svingen T, Usai D, Vinggaard AM, Mandrup K, Axelstad M. Evaluating thyroid hormone disruption: investigations of long-term neurodevelopmental effects in rats after perinatal exposure to perfluorohexane sulfonate (PFHxS). Sci Rep 2020; 10:2672. [PMID: 32060323 PMCID: PMC7021709 DOI: 10.1038/s41598-020-59354-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/24/2020] [Indexed: 12/17/2022] Open
Abstract
Thyroid hormones are critical for mammalian brain development. Thus, chemicals that can affect thyroid hormone signaling during pregnancy are of great concern. Perfluorohexane sulfonate (PFHxS) is a widespread environmental contaminant found in human serum, breastmilk, and other tissues, capable of lowering serum thyroxine (T4) in rats. Here, we investigated its effects on the thyroid system and neurodevelopment following maternal exposure from early gestation through lactation (0.05, 5 or 25 mg/kg/day PFHxS), alone or in combination with a mixture of 12 environmentally relevant endocrine disrupting compounds (EDmix). PFHxS lowered thyroid hormone levels in both dams and offspring in a dose-dependent manner, but did not change TSH levels, weight, histology, or expression of marker genes of the thyroid gland. No evidence of thyroid hormone-mediated neurobehavioral disruption in offspring was observed. Since human brain development appear very sensitive to low T4 levels, we maintain that PFHxS is of potential concern to human health. It is our view that current rodent models are not sufficiently sensitive to detect adverse neurodevelopmental effects of maternal and perinatal hypothyroxinemia and that we need to develop more sensitive brain-based markers or measurable metrics of thyroid hormone-dependent perturbations in brain development.
Collapse
Affiliation(s)
- Louise Ramhøj
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800,, Denmark
| | - Ulla Hass
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800,, Denmark
| | - Mary E Gilbert
- Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Carmen Wood
- Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Terje Svingen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800,, Denmark
| | - Diana Usai
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800,, Denmark
| | - Anne Marie Vinggaard
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800,, Denmark
| | - Karen Mandrup
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800,, Denmark
| | - Marta Axelstad
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800,, Denmark.
| |
Collapse
|
16
|
Martin PGP, Dupouy V, Leghait J, Pineau T, Polizzi A, Lasserre F, Roques BB, Viguié C. Transcriptomic modifications of the thyroid gland upon exposure to phytosanitary-grade fipronil: Evidence for the activation of compensatory pathways. Toxicol Appl Pharmacol 2019; 389:114873. [PMID: 31881178 DOI: 10.1016/j.taap.2019.114873] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/16/2019] [Accepted: 12/21/2019] [Indexed: 10/25/2022]
Abstract
Fipronil is a phenylpyrazole insecticide used for the control of a variety of pest for domestic, veterinary and agricultural uses. Fipronil exposure is associated to thyroid disruption in the rat. It increases thyroid hormone (TH) hepatic clearance. The effect on thyroxine (T4) clearance is about four fold higher than the effect on T4 plasma concentrations suggesting that the thyroid gland might develop compensatory mechanisms. The aim of this study was to document the potential effects of fipronil treatment on the thyroid transcriptome together with its effects on TSH and TH blood levels under well characterized internal exposure to fipronil and its main metabolite fipronil sulfone. Fipronil (3 mg/kg/d by gavage for 14 days) clearance increased while its half-life decreased (about 10 fold) throughout treatment. Fipronil treatment in adult female rats significantly decreased total T4 and free triiodothyronine (T3) concentrations. Key genes related to thyroid hormone synthesis and/or cellular dynamic were modulated by fipronil exposure. RT-PCR confirmed that thyroglobulin gene expression was upregulated. A trend toward higher Na/I symporter expression was also noted, while sulfotransferase 1a1 gene expression was down-regulated. The expression of genes potentially involved in thyroid cell dynamic were upregulated (e.g. prostaglandin synthase 1, amphiregulin and Rhoa). Our results indicate that both pathways of TH synthesis and thyroid cell dynamics are transcriptional targets of fipronil and/or its main sulfone metabolite. The underlying mechanisms remain to be elucidated.
Collapse
Affiliation(s)
- Pascal G P Martin
- ToxAlim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 180 chemin de Tournefeuille, BP93173, F-31027 Toulouse Cedex 03, France
| | - Véronique Dupouy
- ToxAlim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 180 chemin de Tournefeuille, BP93173, F-31027 Toulouse Cedex 03, France; IntheRes, INRAE, ENVT, 23 chemin des Capelles, BP87614, F-31076 Toulouse Cedex 03, France
| | - Julien Leghait
- ToxAlim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 180 chemin de Tournefeuille, BP93173, F-31027 Toulouse Cedex 03, France
| | - Thierry Pineau
- ToxAlim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 180 chemin de Tournefeuille, BP93173, F-31027 Toulouse Cedex 03, France
| | - Arnaud Polizzi
- ToxAlim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 180 chemin de Tournefeuille, BP93173, F-31027 Toulouse Cedex 03, France
| | - Frédéric Lasserre
- ToxAlim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 180 chemin de Tournefeuille, BP93173, F-31027 Toulouse Cedex 03, France
| | - Béatrice B Roques
- ToxAlim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 180 chemin de Tournefeuille, BP93173, F-31027 Toulouse Cedex 03, France; IntheRes, INRAE, ENVT, 23 chemin des Capelles, BP87614, F-31076 Toulouse Cedex 03, France
| | - Catherine Viguié
- ToxAlim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, 180 chemin de Tournefeuille, BP93173, F-31027 Toulouse Cedex 03, France.
| |
Collapse
|
17
|
Kato Y, Tamaki S, Haraguchi K, Ikushiro SI, Fujii Y, Ohta C, Atobe K, Kimura O, Endo T, Koga N, Yamada S, Degawa M. Kanechlor 500-mediated changes in serum and hepatic thyroxine levels primarily occur in a transthyretin-unrelated manner. J Appl Toxicol 2019; 39:1701-1709. [PMID: 31498458 DOI: 10.1002/jat.3895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/19/2019] [Accepted: 07/24/2019] [Indexed: 11/08/2022]
Abstract
The effects of Kanechlor-500 (KC500) on the levels of serum total thyroxine (T4 ) and hepatic T4 in wild-type C57BL/6 (WT) and its transthyretin (TTR)-deficient (TTR-null) mice were comparatively examined. Four days after a single intraperitoneal injection with KC500 (100 mg/kg body weight), serum total T4 levels were significantly decreased in both WT and TTR-null mice. The KC500 pretreatment also promoted serum [125 I]T4 clearance in both strains of mice administrated with [125 I]T4 , and the promotion of serum [125 I]T4 clearance in WT mice occurred without inhibition of the [125 I]T4 -TTR complex formation. Furthermore, the KC500 pretreatment led to significant increases in liver weight, steady-state distribution volume of [125 I]T4 , hepatic accumulation level of [125 I]T4 , and concentration ratio of the liver to serum in both strains of mice. The present findings indicate that the KC500-mediated decrease in serum T4 level occurs in a TTR-unrelated manner and further suggest that KC500-promoted T4 accumulation in the liver occurs through the development of liver hypertrophy and the promotion of T4 transportation from serum to liver.
Collapse
Affiliation(s)
- Yoshihisa Kato
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Kagawa, Japan
| | - Sekihiro Tamaki
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | | | | | - Yukiko Fujii
- Daiichi College of Pharmaceutical Sciences, Fukuoka, Japan
| | - Chiho Ohta
- Faculty of Nutritional Sciences, Nakamura Gakuen University, Fukuoka, Japan
| | - Kazutaka Atobe
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki, Kagawa, Japan
| | - Osamu Kimura
- Faculty of Pharmaceutical Sciences, Health Science University of Hokkaido, Hokkaido, Japan
| | - Tetsuya Endo
- Faculty of Pharmaceutical Sciences, Health Science University of Hokkaido, Hokkaido, Japan
| | - Nobuyuki Koga
- Faculty of Nutritional Sciences, Nakamura Gakuen University, Fukuoka, Japan
| | - Shizuo Yamada
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Masakuni Degawa
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
18
|
Bartsch R, Brinkmann B, Jahnke G, Laube B, Lohmann R, Michaelsen S, Neumann I, Greim H. Human relevance of follicular thyroid tumors in rodents caused by non-genotoxic substances. Regul Toxicol Pharmacol 2018; 98:199-208. [PMID: 30076866 DOI: 10.1016/j.yrtph.2018.07.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/27/2018] [Accepted: 07/28/2018] [Indexed: 12/31/2022]
Abstract
Chronic stimulation of the thyroid gland of rodents by TSH leads to thyroid follicular hyperplasia and subsequently to thyroid follicular adenomas and carcinomas. However, the interpretations of rodent thyroid tumors are contradictory. The U.S. Food and Drug Administration (FDA) concluded that findings with drugs that lead to increased levels of thyroid-stimulating hormone (TSH) in rats are not relevant to humans, whereas the U.S. Environmental Protection Agency (US EPA) concluded that chemicals that produce rodent thyroid tumors may pose a carcinogenic hazard for humans although the thyroid of rodents appears to be more sensitive to a carcinogenic stimulus than that of humans. Meanwhile, based on the CLP Criteria of the European Chemicals Agency (ECHA), rodent thyroid tumors caused by the induction of uridine-diphosphate-glucuronosyl transferases (UDGT) were assessed as not relevant to humans. To clarify these discrepant positions, the function and regulation of the thyroid gland are described and the types of thyroid tumors and the causes of their development in humans and animals are examined. Based on these data and the evidence that so far, except radiation, no chemical is known to increase the incidence of thyroid tumors in humans, it is concluded that rodent thyroid tumors resulting from continuous stimulation of the thyroid gland by increased TSH levels are not relevant to humans. Consequently, compounds that induce such tumors do not warrant classification as carcinogenic.
Collapse
Affiliation(s)
- Ruediger Bartsch
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Britta Brinkmann
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Gunnar Jahnke
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Britta Laube
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Ruth Lohmann
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Sandra Michaelsen
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Ingrid Neumann
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Helmut Greim
- Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany.
| |
Collapse
|
19
|
Hu J, Webster D, Cao J, Shao A. The safety of green tea and green tea extract consumption in adults - Results of a systematic review. Regul Toxicol Pharmacol 2018; 95:412-433. [PMID: 29580974 DOI: 10.1016/j.yrtph.2018.03.019] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 03/17/2018] [Accepted: 03/20/2018] [Indexed: 12/22/2022]
Abstract
A systematic review of published toxicology and human intervention studies was performed to characterize potential hazards associated with consumption of green tea and its preparations. A review of toxicological evidence from laboratory studies revealed the liver as the target organ and hepatotoxicity as the critical effect, which was strongly associated with certain dosing conditions (e.g. bolus dose via gavage, fasting), and positively correlated with total catechin and epigallocatechingallate (EGCG) content. A review of adverse event (AE) data from 159 human intervention studies yielded findings consistent with toxicological evidence in that a limited range of concentrated, catechin-rich green tea preparations resulted in hepatic AEs in a dose-dependent manner when ingested in large bolus doses, but not when consumed as brewed tea or extracts in beverages or as part of food. Toxico- and pharmacokinetic evidence further suggests internal dose of catechins is a key determinant in the occurrence and severity of hepatotoxicity. A safe intake level of 338 mg EGCG/day for adults was derived from toxicological and human safety data for tea preparations ingested as a solid bolus dose. An Observed Safe Level (OSL) of 704 mg EGCG/day might be considered for tea preparations in beverage form based on human AE data.
Collapse
Affiliation(s)
- Jiang Hu
- Worldwide Scientific Affairs, Herbalife Nutrition, Torrance, CA 90502, USA.
| | - Donna Webster
- Product Science, Herbalife Nutrition, Torrance, CA 90502, USA.
| | - Joyce Cao
- Global Post Market Safety Surveillance, Herbalife Nutrition, Torrance, CA 90502, USA.
| | - Andrew Shao
- Independent Consultant, Rancho Palos Verdes, CA 90505, USA.
| |
Collapse
|
20
|
Funk J, Ebeling M, Singer T, Landes C. Image analysis for TSH mRNA in situ hybridization in pituitary glands from rats with thyroid follicular cell hypertrophy after treatment with three different test compounds. Res Vet Sci 2017. [PMID: 28646742 DOI: 10.1016/j.rvsc.2017.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The goal of this in situ hybridization and image analysis technique is to study the effects of new pharmacological/chemical entities on the thyroid and pituitary gland in rats, reveal the pathogenesis of thyroid follicular cell hypertrophy and to retrospectively exclude the risk of thyroid tumor development in humans. In the present study, we describe the increase of thyroid-stimulating hormone- (TSH-) beta subunit mRNA in the pars distalis of the pituitary gland and the quantitative measurement of TSH mRNA positive cells from rats of three 4-week toxicity studies treated with three different test compounds inducing thyroid follicular cell and hepatocellular hypertrophy in rats. Compared to immunohistochemistry (IHC), in situ hybridization (ISH) for TSH was found to be more sensitive. With this technique we are able to exclude a direct effect of the test compound on the thyroid gland by showing the activation of thyrotrope cells from the pituitary gland and therefore this technique retrospectively enables us to exclude a possible risk for humans at an early stage of drug development. Also in case blood serum samples for evaluation of TSH are not available anymore or hepatocellular hypertrophy is not present (close metabolic relationship between thyroid gland and liver in rodents), the described method allows retrospective investigations on thyroid follicular cell hypertrophy or hyperplasia. This can be of high relevance in human safety assessment for certain drugs in order to exclude a primary effect on the thyroid gland especially when it comes to thyroid neoplasia in rodents as previously described.
Collapse
Affiliation(s)
- Juergen Funk
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland.
| | - Martin Ebeling
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland
| | - Thomas Singer
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland
| | - Christian Landes
- Roche Pharmaceutical Research & Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Switzerland
| |
Collapse
|
21
|
Movasseghi AR, Rodríguez-Estival J, Smits JEG. Thyroid pathology in deer mice (Peromyscus maniculata) from a reclaimed mine site on the athabasca oil sands. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 222:42-49. [PMID: 28104343 DOI: 10.1016/j.envpol.2017.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/29/2016] [Accepted: 01/06/2017] [Indexed: 06/06/2023]
Abstract
Information on naturally occurring thyroid disease in wild animals in general and in small mammals specifically is extremely limited. In the present field-based work, we investigated the structure and function of thyroid glands of deer mice (Peromyscus maniculata) studied as sentinels of ecosystem sustainability on reclaimed areas post-mining on the oil sands of northeastern Alberta, because of their greater sensitivity to contaminants relative to meadow voles (Microtus pennsylvanicus) on the same sites. Extraction of bitumen in the oil sands of northeastern Alberta, results in the release of contaminants including polycyclic aromatic compounds (PACs), metals, and metalloids to the environment that have a measurable biological cost to wildlife living in the affected areas. In previous investigations, deer mice exposed to pollution at reclaimed areas showed compromised ability to regenerate glutathione indicating oxidative stress, together with decreased testicular mass and body condition during the breeding season. In the present study, thyroid glands from those deer mice from the reclaimed site had markedly increased follicular cell proliferation and decreased colloid compared to animals from the reference site. This pathology was positively associated with the greater oxidative stress in the deer mice. Thyroid hormones, both thyroxine and triiodothyronine, were also higher in animals with greater oxidative stress indicating increased metabolic demands from contaminant related subclinical toxicity. This work emphasizes the value of using a combination of endocrinological, histological and oxidative stress biomarkers to provide sensitive measures of contaminant exposure in small mammals on the oil sands.
Collapse
Affiliation(s)
- Ahmad R Movasseghi
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, 3280 Hospital Drive NW Calgary, Alberta T2N 4Z6, Canada.
| | - Jaime Rodríguez-Estival
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, 3280 Hospital Drive NW Calgary, Alberta T2N 4Z6, Canada.
| | - Judit E G Smits
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, 3280 Hospital Drive NW Calgary, Alberta T2N 4Z6, Canada.
| |
Collapse
|
22
|
Abstract
Doxylamine succinate, a commonly used antihistamine, was administered as an admixture in the feed to groups of male and female B6C3F1 mice at dosage levels of 0,190,375, and 750 parts per million (ppm) (based on free amine) for 65 weeks (12 per group) or 2 years (48 per group). Survival to terminal sacrifice in the 2-year groups was 85–98% with no significant differences between groups of the same sex. Final body weights of the highest dose group were 3.4% and 8.7% less than controls in males and females, respectively. Doxylamine produced liver lesions in male mice including hepatocellular hypertrophy, atypical hepatocytes, clear cell and mixed cell foci, and necrosis. In females, doxylamine produced liver fatty change, hepatocellular hypertrophy, and necrosis. Doxylamine produced a significant increase in hepatocellular adenomas in the mid- and high-dosage groups of males and in the high-dosage group of females. Thyroid follicular cell hyperplasia and thyroid follicular cell adenomas also were increased in treated mice of both sexes. A treatment-related increase in cytoplasmic alteration of the parotid salivary gland in males and an increased incidence in hyperplasia of the pituitary gland in females were observed.
Collapse
|
23
|
Burns-Naas LA, Zorbas M, Jessen B, Evering W, Stevens G, Ivett JL, Ryan TE, Cook JC, Capen CC, Chen M, Furman G, Theiss JC, Webber S, Wu E, Shetty B, Gasser R, McClain RM. Increase in thyroid follicular cell tumors in nelfinavir-treated rats observed in a 2-year carcinogenicity study is consistent with a rat-specific mechanism of thyroid neoplasia. Hum Exp Toxicol 2016; 24:643-54. [PMID: 16408618 DOI: 10.1191/0960327105ht568oa] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The carcinogenic potential of nelfinavir mesylate (nelfinavir) was evaluated in a 2-year oral (gavage) study on Sprague-Dawley rats at dose levels of 0 (control), 0 (vehicle control), 100, 300 and 1000 mg/kg per day. At the end of the treatment, increased incidences of thyroid follicular cell hyperplasia and neoplasms were observed at 300 (males) and 1000 mg/kg per day (both sexes). There were no other treatment-related effects and no tumors at other sites. Results from previous studies indicated a number of effects in the liver and thyroid, as well as metabolic profiles that suggested nelfinavir might cause thyroid hyperplasia/neoplasia secondary to hormone imbalance by altering thyroid hormone disposition. To investigate this hypothesis, the effects of nelfinavir on gene expression in rat hepatocytes and liver slices (in vitro), thyroxine plasma clearance, and thyroid gland function were evaluated. Compared to controls, gene expression analyses demonstrated an increased expression of glucuronyltransferase (UDPGT) and CYP450 3A1 in nelfinavir-treated rat hepatocytes and liver slices. In rats treated with nelfinavir (1000 mg/kg per day) for 4 weeks, liver weights and centrilobular hepatocellular hypertrophy were increased and minimal to mild diffuse thyroid follicular cell hypertrophy and follicular cell hyperplasia were evident in the thyroid gland. Thyroid-stimulating hormone (TSH) levels were significantly increased (three-fold), while tri-iodothyronine (T3)/tetraiodothyronine (T4) and reverse T3(rT3) levels were unchanged, indicating that a compensated state to maintain homeostasis of T3/T4 had been achieved. Plasma 125I-thyroxine clearance was increased and the plasma thyroxine AUC0 48 was decreased (24%) compared to control. In conclusion, these data indicate that thyroid neoplasms observed in the nelfinavir-treated rats were secondary to thyroid hormone imbalance. Increased thyroxine clearance contributes to the effects of nelfinavir on thyroid gland function and is probably a result of UDPGT induction that leads to elevated TSH levels in the rat and eventual thyroid neoplasia. These results are consistent with a well-recognized rat-specific mechanism for thyroid neoplasms.
Collapse
Affiliation(s)
- L A Burns-Naas
- Pfizer Global Research and Development, San Diego, CA 92121, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
van der Laan JW, Kasper P, Silva Lima B, Jones DR, Pasanen M. Critical analysis of carcinogenicity study outcomes. Relationship with pharmacological properties. Crit Rev Toxicol 2016; 46:587-614. [DOI: 10.3109/10408444.2016.1163664] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | - Peter Kasper
- Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany
| | | | - David R. Jones
- Medicines and Healthcare Products Regulatory Agency, London, UK
| | - Markku Pasanen
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
25
|
Paul Friedman K, Watt ED, Hornung MW, Hedge JM, Judson RS, Crofton KM, Houck KA, Simmons SO. Tiered High-Throughput Screening Approach to Identify Thyroperoxidase Inhibitors Within the ToxCast Phase I and II Chemical Libraries. Toxicol Sci 2016; 151:160-80. [PMID: 26884060 DOI: 10.1093/toxsci/kfw034] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
High-throughput screening for potential thyroid-disrupting chemicals requires a system of assays to capture multiple molecular-initiating events (MIEs) that converge on perturbed thyroid hormone (TH) homeostasis. Screening for MIEs specific to TH-disrupting pathways is limited in the U.S. Environmental Protection Agency ToxCast screening assay portfolio. To fill 1 critical screening gap, the Amplex UltraRed-thyroperoxidase (AUR-TPO) assay was developed to identify chemicals that inhibit TPO, as decreased TPO activity reduces TH synthesis. The ToxCast phase I and II chemical libraries, comprised of 1074 unique chemicals, were initially screened using a single, high concentration to identify potential TPO inhibitors. Chemicals positive in the single-concentration screen were retested in concentration-response. Due to high false-positive rates typically observed with loss-of-signal assays such as AUR-TPO, we also employed 2 additional assays in parallel to identify possible sources of nonspecific assay signal loss, enabling stratification of roughly 300 putative TPO inhibitors based upon selective AUR-TPO activity. A cell-free luciferase inhibition assay was used to identify nonspecific enzyme inhibition among the putative TPO inhibitors, and a cytotoxicity assay using a human cell line was used to estimate the cellular tolerance limit. Additionally, the TPO inhibition activities of 150 chemicals were compared between the AUR-TPO and an orthogonal peroxidase oxidation assay using guaiacol as a substrate to confirm the activity profiles of putative TPO inhibitors. This effort represents the most extensive TPO inhibition screening campaign to date and illustrates a tiered screening approach that focuses resources, maximizes assay throughput, and reduces animal use.
Collapse
Affiliation(s)
- Katie Paul Friedman
- *Oak Ridge Institute for Science Education Postdoctoral Fellow, Oak Ridge, TN, 37831 Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711
| | - Eric D Watt
- *Oak Ridge Institute for Science Education Postdoctoral Fellow, Oak Ridge, TN, 37831 National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711
| | - Michael W Hornung
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Duluth, MN, 55804
| | - Joan M Hedge
- Integrated Systems Toxicology Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711
| | - Richard S Judson
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711
| | - Kevin M Crofton
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711
| | - Keith A Houck
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711
| | - Steven O Simmons
- National Center for Computational Toxicology, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, 27711,
| |
Collapse
|
26
|
Kinouchi H, Matsuyama K, Kitagawa H, Kamimori H. Surface plasmon resonance assay of inhibition by pharmaceuticals for thyroxine hormone binging to transport proteins. Anal Biochem 2016; 492:43-8. [DOI: 10.1016/j.ab.2015.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/28/2015] [Accepted: 09/07/2015] [Indexed: 11/16/2022]
|
27
|
Bus JS, Banton MI, Faber WD, Kirman CR, McGregor DB, Pourreau DB. Human health screening level risk assessments of tertiary-butyl acetate (TBAC): Calculated acute and chronic reference concentration (RfC) and Hazard Quotient (HQ) values based on toxicity and exposure scenario evaluations. Crit Rev Toxicol 2015; 45:142-71. [DOI: 10.3109/10408444.2014.980884] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
28
|
Derivation of an oral reference dose (RfD) for the plasticizer, di-(2-propylheptyl)phthalate (Palatinol® 10-P). Regul Toxicol Pharmacol 2014; 70:65-74. [DOI: 10.1016/j.yrtph.2014.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/30/2014] [Accepted: 06/01/2014] [Indexed: 11/19/2022]
|
29
|
Nambiar PR, Morton D, Dochterman LW, Houle C, Thomford PJ, Fate G, Bailey SA, Finch GL. Two-year Carcinogenicity Study in Rats with a Nonnucleoside Reverse Transcriptase Inhibitor. Toxicol Pathol 2014; 43:354-65. [DOI: 10.1177/0192623314544381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Administration of lersivirine, a nonnucleotide reverse transcriptase inhibitor, daily by oral gavage to Sprague-Dawley rats for up to 2 yr was associated with decreased survival, decreased body weights, and an increase in neoplasms and related proliferative lesions in the liver, thyroid, kidney, and urinary bladder. Thyroid follicular adenoma and carcinoma, the associated thyroid follicular hypertrophy/hyperplasia, hepatocellular adenoma/adenocarcinoma, altered cell foci, and hepatocellular hypertrophy were consistent with lersivirine-related induction of hepatic microsomal enzymes. Renal tubular adenoma and renal tubular hyperplasia were attributed to the lersivirine-related exacerbation of chronic progressive nephropathy (CPN), while urinary bladder hyperplasia and transitional cell carcinoma in the renal pelvis and urinary bladder were attributed to urinary calculi. Renal tubular neoplasms associated with increased incidence and severity of CPN, neoplasms of transitional epithelium attributed to crystalluria, and thyroid follicular and hepatocellular neoplasms related to hepatic enzyme induction have low relevance for human risk assessment.
Collapse
|
30
|
Mechanistic analysis of metabolomics patterns in rat plasma during administration of direct thyroid hormone synthesis inhibitors or compounds increasing thyroid hormone clearance. Toxicol Lett 2014; 225:240-51. [DOI: 10.1016/j.toxlet.2013.12.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 12/12/2013] [Accepted: 12/13/2013] [Indexed: 12/11/2022]
|
31
|
Benigni R, Bossa C, Tcheremenskaia O. Nongenotoxic carcinogenicity of chemicals: mechanisms of action and early recognition through a new set of structural alerts. Chem Rev 2013; 113:2940-57. [PMID: 23469814 DOI: 10.1021/cr300206t] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Romualdo Benigni
- Istituto Superiore di Sanita' Environment and Health Department, Viale Regina Elena 299, 00161 Rome, Italy.
| | | | | |
Collapse
|
32
|
van der Laan JW, DeGeorge JJ, Sistare F, Moggs J. Toward More Scientific Relevance in Carcinogenicity Testing. GLOBAL APPROACH IN SAFETY TESTING 2013. [DOI: 10.1007/978-1-4614-5950-7_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
33
|
Kato Y, Tamaki S, Haraguchi K, Ikushiro SI, Sekimoto M, Ohta C, Endo T, Koga N, Yamada S, Degawa M. Comparative study on 2,2′,4,5,5′-pentachlorobiphenyl-mediated decrease in serum thyroxine level between C57BL/6 and its transthyretin-deficient mice. Toxicol Appl Pharmacol 2012; 263:323-9. [DOI: 10.1016/j.taap.2012.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Revised: 07/05/2012] [Accepted: 07/06/2012] [Indexed: 11/29/2022]
|
34
|
Wu KM, Powley MW, Ghantous H. Timing of carcinogenicity studies and predictability of genotoxicity for tumorigenicity in anti-HIV drug development. Int J Toxicol 2012; 31:211-21. [PMID: 22550047 DOI: 10.1177/1091581812439585] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The timing of carcinogenicity studies in parallel with the clinical development of anti-human immunodeficiency virus (HIV) drugs has been flexible for most cases in the past. This includes postponement of the initiation of the studies and submission of final audited reports to the US Food and Drug Administration (FDA) for a new drug application (NDA) approval. We address this regulatory practice for anti-HIV drugs for which, in the past, there had been no effective treatment. We also examine the correlation of genotoxicity data with carcinogenicity data for the varied subclasses of anti-HIV drugs. We suggest that this regulatory policy regarding the timing of carcinogenicity testing does not compromise the safety standards of FDA's drug evaluation and the approval process. The policy does facilitate availability of these agents to meet the medical needs of the target population. Our analysis on the profile of carcinogenicity findings of anti-HIV drugs shows trends of class effects. Additionally, both carcinogenicity and genotoxicity data show significant correlations, which provide useful insights into issues involving these 2 important areas of toxicological investigations.
Collapse
Affiliation(s)
- Kuei-Meng Wu
- Division of Antiviral Products, Office of Antimicrobial Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA.
| | | | | |
Collapse
|
35
|
Weber S, Halm S, Du Vall M, Whitney K, Blaich G, Bury D. N-vinylpyrrolidone dimer (VPD), a novel excipient for oral drugs: Repeat-dose oral toxicity in Sprague–Dawley rats. Toxicol Lett 2012; 210:324-31. [DOI: 10.1016/j.toxlet.2012.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 02/01/2012] [Accepted: 02/02/2012] [Indexed: 10/14/2022]
|
36
|
Lindblom P, Berg AL, Zhang H, Westerberg R, Tugwood J, Lundgren H, Marcusson-Ståhl M, Sjögren N, Blomgren B, Öhman P, Skånberg I, Evans J, Hellmold H. Tesaglitazar, a dual PPAR-α/γ agonist, hamster carcinogenicity, investigative animal and clinical studies. Toxicol Pathol 2011; 40:18-32. [PMID: 22131108 DOI: 10.1177/0192623311429972] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Tesaglitazar was developed as a dual peroxisome proliferator-activated receptor (PPARα/γ). To support the clinical program, a hamster carcinogenicity study was performed. The only neoplastic findings possibly related to treatment with tesaglitazar were low incidences of hemangioma and hemangiosarcoma in the liver of male animals. A high-power, two-year investigative study with interim necropsies was performed to further elucidate these findings. Treatment with tesaglitazar resulted in changes typical for exaggerated PPARα pharmacology in rodents, such as hepatocellular hypertrophy and hepatocellular carcinoma, but not an increased frequency of hemangiosarcomas. At the highest dose level, there was an increased incidence of sinusoidal dilatation and hemangiomas. No increased endothelial cell (EC) proliferation was detected in vivo, which was confirmed by in vitro administration to ECs. Immunohistochemistry and gene expression analyses indicated increased cellular stress and vascular endothelial growth factor (VEGF) expression in the liver, which may have contributed to the sinusoidal dilatation. A two-fold increase in the level of circulating VEGF was detected in the hamster at all dose levels, whereas no effect on VEGF was observed in patients treated with tesaglitazar. In conclusion, investigations have demonstrated that tesaglitazar does not produce hemangiosarcomas in hamster despite a slight effect on vascular morphology in the liver.
Collapse
|
37
|
Sistare FD, Morton D, Alden C, Christensen J, Keller D, Jonghe SD, Storer RD, Reddy MV, Kraynak A, Trela B, Bienvenu JG, Bjurström S, Bosmans V, Brewster D, Colman K, Dominick M, Evans J, Hailey JR, Kinter L, Liu M, Mahrt C, Marien D, Myer J, Perry R, Potenta D, Roth A, Sherratt P, Singer T, Slim R, Soper K, Fransson-Steen R, Stoltz J, Turner O, Turnquist S, van Heerden M, Woicke J, DeGeorge JJ. An analysis of pharmaceutical experience with decades of rat carcinogenicity testing: support for a proposal to modify current regulatory guidelines. Toxicol Pathol 2011; 39:716-44. [PMID: 21666103 DOI: 10.1177/0192623311406935] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Data collected from 182 marketed and nonmarketed pharmaceuticals demonstrate that there is little value gained in conducting a rat two-year carcinogenicity study for compounds that lack: (1) histopathologic risk factors for rat neoplasia in chronic toxicology studies, (2) evidence of hormonal perturbation, and (3) positive genetic toxicology results. Using a single positive result among these three criteria as a test for outcome in the two-year study, fifty-two of sixty-six rat tumorigens were correctly identified, yielding 79% test sensitivity. When all three criteria were negative, sixty-two of seventy-six pharmaceuticals (82%) were correctly predicted to be rat noncarcinogens. The fourteen rat false negatives had two-year study findings of questionable human relevance. Applying these criteria to eighty-six additional chemicals identified by the International Agency for Research on Cancer as likely human carcinogens and to drugs withdrawn from the market for carcinogenicity concerns confirmed their sensitivity for predicting rat carcinogenicity outcome. These analyses support a proposal to refine regulatory criteria for conducting a two-year rat study to be based on assessment of histopathologic findings from a rat six-month study, evidence of hormonal perturbation, genetic toxicology results, and the findings of a six-month transgenic mouse carcinogenicity study. This proposed decision paradigm has the potential to eliminate over 40% of rat two-year testing on new pharmaceuticals without compromise to patient safety.
Collapse
Affiliation(s)
- Frank D Sistare
- Merck and Co., Inc., West Point, Pennsylvania 19486-0004, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Li Y, Kumazawa T, Ishiguro T, Kawakami Y, Nishitani H, Tagawa Y, Matsumoto Y. Hypothyroidism caused by phenobarbital affects patterns of estrous cyclicity in rats. Congenit Anom (Kyoto) 2011; 51:55-61. [PMID: 21595753 DOI: 10.1111/j.1741-4520.2011.00314.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We found that repeated treatment with phenobarbital (PB), a thyroid modulator, resulted in a persistent estrous stage in the present study. Although the effects of PB in blocking the surge release of luteinizing hormone (LH), inducing anovulation and prolonging the diestrous period has been well established, there is still no research describing the appearance of persistent estrous states in normal cycling rats dosed with PB. To further study this phenomenon, female rats exhibiting regular estrous cycle were administered an oral dose of PB for 14 consecutive days. Consecutively, vaginal smears were observed and rats from all the groups were sacrificed and serum hormone levels for prolactin, progesterone, estradiol, triiodothyronin (T3), thyroxine (T4) and thyroid stimulating hormone (TSH) were measured. Pituitary, thyroid, liver, uteri and ovaries were excised, weighed and further subjected to histological observations. We found that PB induced irregular estrous cycles, especially persistent estrus in rats. Histopathologically, the persistent estrous stages are characterized by persistent vaginal cornification in the vagina, cystic follicles and anovulation in the ovaries. Endocrinologically, serum T3 and T4 levels were significantly lower, and TSH was higher in treated-female rats compared to control females. The serum estradiol level and the estradiol/progesterone ratio tend to increase in treated-females. Furthermore, PB-treated animals with irregular estrous cycle were reduced by T4 replacement. Our data indicate that treatment with PB resulted in hypothyroidism and irregular estrous cycle, particularly a persistent estrous stage in normal cycling female rats.
Collapse
Affiliation(s)
- Yumei Li
- Pharmaceutical Research Laboratories, Sanwa Kagaku Kenkyusho Co., Ltd, Hokusei, Inabe, Japan
| | | | | | | | | | | | | |
Collapse
|
39
|
Zabka TS, Fielden MR, Garrido R, Tao J, Fretland AJ, Fretland JL, Albassam MA, Singer T, Kolaja KL. Characterization of Xenobiotic-Induced Hepatocellular Enzyme Induction in Rats. Toxicol Pathol 2011; 39:664-77. [DOI: 10.1177/0192623311406934] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
During routine safety evaluation of RO2910, a non-nucleoside reverse transcriptase inhibitor for HIV infection, histopathology findings concurrent with robust hepatocellular induction occurred in multiple organs, including a unique, albeit related, finding in the pituitary gland. For fourteen days, male and female rats were administered, by oral gavage vehicle, 100, 300, or 1000 mg/kg/day of RO2910. Treated groups had elevated serum thyroid-stimulating hormone and decreased total thyroxine, and hypertrophy in the liver, thyroid gland, and pituitary pars distalis. These were considered consequences of hepatocellular induction and often were dose dependent and more pronounced in males than in females. Hepatocellular centrilobular hypertrophy corresponded with increased expression of cytochrome P450s 2B1/2, 3A1, and 3A2 and UGT 2B1. Bilateral thyroid follicular cell hypertrophy occurred concurrent to increased mitotic activity and sometimes colloid depletion, which were attributed to changes in thyroid hormone levels. Males had hypertrophy of thyroid-stimulating hormone–producing cells (thyrotrophs) in the pituitary pars distalis. All findings were consistent with the well-established adaptive physiologic response of rodents to xenobiotic-induced hepatocellular microsomal enzyme induction. Although the effects on the pituitary gland following hepatic enzyme induction-mediated hypothyroidism have not been reported previously, other models of stress and thyroid depletion leading to pituitary stimulation support such a shared pathogenesis.
Collapse
Affiliation(s)
- Tanja S. Zabka
- Roche Pharmaceuticals, Nonclinical Safety, Nutley, New Jersey, USA
| | | | - Rosario Garrido
- Roche Pharmaceuticals, Nonclinical Safety, Nutley, New Jersey, USA
| | - Jianhua Tao
- Genentech, South San Francisco, California, USA
| | | | | | | | - Thomas Singer
- Roche Pharmaceuticals, Nonclinical Safety, Nutley, New Jersey, USA
| | - Kyle L. Kolaja
- Roche Pharmaceuticals, Nonclinical Safety, Nutley, New Jersey, USA
| |
Collapse
|
40
|
Abstract
Tert-Butanol is an important intermediate in industrial chemical synthesis, particularly of fuel oxygenates. Human exposure to tert-butanol may occur following fuel oxygenate metabolism or biodegradation. It is poorly absorbed through skin, but is rapidly absorbed upon inhalation or ingestion and distributed to tissues throughout the body. Elimination from blood is slower and the half-life increases with dose. It is largely metabolised by oxidation via 2-methyl-1,2-propanediol to 2-hydroxyisobutyrate, the dominant urinary metabolites. Conjugations also occur and acetone may be found in urine at high doses. The single-dose systemic toxicity of tert-butanol is low, but it is irritant to skin and eyes; high oral doses produce ataxia and hypoactivity and repeated exposure can induce dependence. Tert-Butanol is not definable as a genotoxin and has no effects specific for reproduction or development; developmental delay occurred only with marked maternal toxicity. Target organs for toxicity clearly identified are kidney in male rats and urinary bladder, particularly in males, of both rats and mice. Increased tumour incidences observed were renal tubule cell adenomas in male rats and thyroid follicular cell adenomas in female mice and, non-significantly, at an intermediate dose in male mice. The renal adenomas were associated with alpha(2u)-globulin nephropathy and, to a lesser extent, exacerbation of chronic progressive nephropathy. Neither of these modes of action can function in humans. The thyroid tumour response could be strain-specific. No thyroid toxicity was observed and a study of hepatic gene expression and enzyme induction and thyroid hormone status has suggested a possible mode of action.
Collapse
Affiliation(s)
- Douglas McGregor
- Toxicity Evaluation Consultants, Aberdour, Scotland, United Kingdom.
| |
Collapse
|
41
|
A possible mechanism for the decrease in serum thyroxine level by phenobarbital in rodents. Toxicol Appl Pharmacol 2010; 249:238-46. [DOI: 10.1016/j.taap.2010.09.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 09/03/2010] [Accepted: 09/27/2010] [Indexed: 11/17/2022]
|
42
|
Amacher DE. The effects of cytochrome P450 induction by xenobiotics on endobiotic metabolism in pre-clinical safety studies. Toxicol Mech Methods 2010; 20:159-66. [DOI: 10.3109/15376511003690307] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
43
|
Paul KB, Hedge JM, DeVito MJ, Crofton KM. Short-term exposure to triclosan decreases thyroxine in vivo via upregulation of hepatic catabolism in Young Long-Evans rats. Toxicol Sci 2009; 113:367-79. [PMID: 19910387 DOI: 10.1093/toxsci/kfp271] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Triclosan (5-chloro-2-(2,4-dichlorophenoxy)-phenol) is a chlorinated phenolic antibacterial compound found in consumer products. In vitro human pregnane X receptor activation, hepatic phase I enzyme induction, and decreased in vivo total thyroxine (T4) suggest adverse effects on thyroid hormone homeostasis. Current research tested the hypothesis that triclosan decreases circulating T4 via upregulation of hepatic catabolism and transport. Weanling female Long-Evans rats received triclosan (0-1000 mg/kg/day) by gavage for 4 days. Whole blood and liver were collected 24 h later. Total serum T4, triiodothyronine (T3), and thyroid-stimulating hormone (TSH) were measured by radioimmunoassay. Hepatic microsomal assays measured ethoxyresorufin-O-deethylase, pentoxyresorufin-O-deethylase (PROD), and uridine diphosphate glucuronyltransferase enzyme activities. The messenger RNA (mRNA) expression of cytochrome P450s 1a1, 2b1/2, and 3a1/23; UGTs 1a1, 1a6, and 2b5; sulfotransferases 1c1 and 1b1; and hepatic transporters Oatp1a1, Oatp1a4, Mrp2, and Mdr1b was measured by quantitative reverse transcriptase PCR. Total T4 decreased dose responsively, down to 43% of control at 1000 mg/kg/day. Total T3 was decreased to 89 and 75% of control at 300 and 1000 mg/kg/day. TSH did not change. Triclosan dose dependently increased PROD activity up to 900% of control at 1000 mg/kg/day. T4 glucuronidation increased nearly twofold at 1000 mg/kg/day. Cyp2b1/2 and Cyp3a1/23 mRNA expression levels were induced twofold and fourfold at 300 mg/kg/day. Ugt1a1 and Sult1c1 mRNA expression levels increased 2.2-fold and 2.6-fold at 300 mg/kg/day. Transporter mRNA expression levels were unchanged. These data denote important key events in the mode of action for triclosan-induced hypothyroxinemia in rats and suggest that this effect may be partially due to upregulation of hepatic catabolism but not due to mRNA expression changes in the tested hepatic transporters.
Collapse
Affiliation(s)
- Katie B Paul
- Curriculum in Toxicology, University of North Carolina, Chapel Hill, North Carolina 27514, USA
| | | | | | | |
Collapse
|
44
|
Blanck O, Fowles J, Schorsch F, Pallen C, Espinasse-Lormeau H, Schulte-Koerne E, Totis M, Banton M. Tertiary butyl alcohol in drinking water induces phase I and II liver enzymes with consequent effects on thyroid hormone homeostasis in the B6C3F1 female mouse. J Appl Toxicol 2009; 30:125-32. [PMID: 19760798 DOI: 10.1002/jat.1478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Tertiary butyl alcohol (TBA) was administered to groups of 15 female B6C3F1 mice in drinking water at concentrations of 0, 2.0 or 20 mg TBA ml(-1), for 14 days, for assessment of gross and histological changes in the liver and thyroid, thyroid hormones (T3, T4, and TSH), total hepatic cytochrome P450 (Cyp) content, specific Cyp activities and quantitative PCR analysis of specific Cyp enzymes (Cyp1a1, Cyp2b9, Cyp2b10, Cyp3a11), sulfuryltransferases (ST1a1, ST2a2, and STn) and glucuronyltransferases (UGT1a1, UGT2b1, and UGT2b5). Phenobarbital (PB) was administered to a positive control group by oral gavage at a daily dose of 80 mg kg(-1). TBA caused, on day 14, a reduction in circulating T3 (12-15% decrease) and a dose-dependent reduction in T4 (13-22% decrease), with no evidence of thyroid pathology. Two of five livers examined in the 20 mg TBA ml(-1) dose group showed mild, diffuse centrilobular hypertrophy. On day 14, Cyp 7-benzoxyresorufin-O-debenzylase activity was significantly induced 12-fold by TBA at 20 mg ml(-1), and 1.8-fold at the 2.0 mg TBA ml(-1) concentration. Cyp 7-pentoxyresorufin-O-dealkylase activity was slightly induced (2.1-fold) by 20 mg TBA ml(-1) on day 14. Quantitative PCR analysis of gene transcripts showed a significant induction of Cyp2b10 and ST1a1 with both TBA concentrations, and a slight induction of Cyp2b9 at 20 mg TBA ml(-1) only. PB induced all phase I and phase II gene transcripts except for Cyp1a1 and Cyp2b9. These findings suggest that TBA, at and below doses used in chronic studies, is an inducer of phase I and phase II liver enzymes, with resulting decreases in circulating thyroid hormones in B6C3F1 mice.
Collapse
Affiliation(s)
- O Blanck
- Bayer CropScience S.A., Sophia Antipolis, France
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Hoshi S, Hoshi N, Okamoto M, Paiz J, Kusakabe T, Ward JM, Kimura S. Role of NKX2-1 in N-bis(2-hydroxypropyl)-nitrosamine-induced thyroid adenoma in mice. Carcinogenesis 2009; 30:1614-9. [PMID: 19581346 DOI: 10.1093/carcin/bgp167] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
NKX2-1 is a homeodomain transcription factor that is critical for genesis of the thyroid and transcription of the thyroid-specific genes. Nkx2-1-thyroid-conditional hypomorphic mice were previously developed in which Nkx2-1 gene expression is lost in 50% of the thyroid cells. Using this mouse line as compared with wild-type and Nkx2-1 heterozygous mice, a thyroid carcinogenesis study was carried out using the genotoxic carcinogen N-bis(2-hydroxypropyl)-nitrosamine (DHPN), followed by sulfadimethoxine (SDM) or the non-genotoxic carcinogen amitrole (3-amino-1,2,4-triazole). A significantly higher incidence of adenomas was obtained in Nkx2-1-thyroid-conditional hypomorphic mice as compared with the other two groups of mice only when they were treated with DHPN + SDM, but not amitrole. A bromodeoxyuridine incorporation study revealed that thyroids of the Nkx2-1-thyroid-conditional hypomorphic mice had >2-fold higher constitutive cell proliferation rate than the other two groups of mice, suggesting that this may be at least partially responsible for the increased incidence of adenoma in this mouse line after genotoxic carcinogen exposure. Thus, NKX2-1 may function to control the proliferation of thyroid follicular cells following damage by a genotoxic carcinogen.
Collapse
Affiliation(s)
- Sayuri Hoshi
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Safety of smoke flavour Primary Product Zesti Smoke Code 10. EFSA J 2009. [DOI: 10.2903/j.efsa.2009.982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
47
|
Abstract
When methyl tertiary-butyl ether (MTBE) in gasoline was first introduced to reduce vehicle exhaust emissions and comply with the Clean Air Act, in the United States, a pattern of complaints emerged characterised by seven "key symptoms." Later, carefully controlled volunteer studies did not confirm the existence of the specific key symptoms, although one study of self-reported sensitive (SRS) people did suggest that a threshold at about 11-15% MTBE in gasoline may exist for SRSs in total symptom scores. Neurobehavioral and psychophysiological studies on volunteers, including SRSs, found no adverse responses associated with MTBE at likely exposure levels. MTBE is well and rapidly absorbed following oral and inhalation exposures. Cmax values for MTBE are achieved almost immediately after oral dosing and within 2 h of continuous inhalation. It is rapidly eliminated, either by exhalation as unchanged MTBE or by urinary excretion of its less volatile metabolites. Metabolism is more rapid humans than in rats, for both MTBE and tert-butyl alcohol (TBA), its more persistent primary metabolite. The other primary metabolite, formaldehyde, is detoxified at a rate very much greater than its formation from MTBE. MTBE has no specific effects on reproduction or development, or on genetic material. Neurological effects were observed only at very high concentrations. In carcinogenicity studies of MTBE, TBA, and methanol (included as an endogenous precursor of formaldehyde, without the presence of TBA), some increases in tumor incidence have been observed, but consistency of outcome was lacking and even some degree of replication was observed in only three cases, none of which had human relevance: alpha(2u)-globulin nephropathy-related renal tubule cell adenoma in male rats; Leydig-cell adenoma in male rats, but not in mice, which provide the better model of the human disease; and B-cell-derived lymphoma/leukemia of doubtful pathogenesis that arose mainly in lungs of orally dosed female rats. In addition, hepatocellular adenomas were significantly higher in female CD-1 mice and thyroid follicular-cell adenomas were increased in female B6C3F1 mice treated with TBA, but these results lack any independent confirmation, which would have been possible from a number of other studies.
Collapse
|
48
|
Moser GJ, Foley J, Burnett M, Goldsworthy TL, Maronpot R. Furan-induced dose-response relationships for liver cytotoxicity, cell proliferation, and tumorigenicity (furan-induced liver tumorigenicity). ACTA ACUST UNITED AC 2008; 61:101-11. [PMID: 18809303 DOI: 10.1016/j.etp.2008.06.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Accepted: 06/05/2008] [Indexed: 10/21/2022]
Abstract
Rodent studies of furan are associated with liver cell necrosis, release of liver-associated enzymes, increased hepatocyte proliferation, and hepatocarcinogenesis. For carcinogens whose proposed mode of action is cytolethality, it is hypothesized that the dose-response curve for tumor development would parallel the dose-response curve for cell death with compensatory proliferation in the target organ. To prospectively test this hypothesis, female B6C3F(1) mice were exposed to furan at carcinogenic doses and lower for 3 weeks or 2 years. At 3 weeks and in the 2-year study, there were dose-dependent and significant increases in hepatic cytotoxicity at 1.0, 2.0, 4.0, and 8.0mg furan/kg. For cell proliferation as measured by 5-bromo-2'-deoxyuridine (BrdU) labeling index (LI), there was a statistically significant trend with increasing dose levels of furan and increased LI at 8.0mg/kg. There was an increased incidence of foci of altered hepatocytes, hepatocellular adenomas, and adenomas or carcinomas at 4.0 and 8.0mg/kg and carcinomas at 8.0mg/kg. The multiplicity of microscopic tumors was increased and latency was decreased in mice exposed to 8.0mg/kg. Prevalence of hepatic nodules at necropsy was increased in mice exposed to 4.0 and 8.0mg/kg. Data demonstrate an association among furan-induced hepatic cytotoxicity, compensatory cell replication, and liver tumor formation in mice; at high doses >or=4.0mg/kg, furan induced hepatotoxicity, compensatory cell replication and tumorigenesis in a dose-related manner, while furan did not produce tumors at cytotoxic doses of 1.0 and 2.0mg/kg.
Collapse
Affiliation(s)
- Glenda J Moser
- Integrated Laboratory Systems, Inc., Research Triangle Park, NC 27709, USA.
| | | | | | | | | |
Collapse
|
49
|
Safety evaluation to support first-in-man investigations II: Toxicology studies. Regul Toxicol Pharmacol 2008; 51:237-43. [DOI: 10.1016/j.yrtph.2008.04.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 04/11/2008] [Indexed: 11/22/2022]
|
50
|
Buckman AH, Fisk AT, Parrott JL, Solomon KR, Brown SB. PCBs can diminish the influence of temperature on thyroid indices in rainbow trout (Oncorhynchus mykiss). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2007; 84:366-378. [PMID: 17681619 DOI: 10.1016/j.aquatox.2007.06.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2006] [Revised: 06/25/2007] [Accepted: 06/26/2007] [Indexed: 05/16/2023]
Abstract
The influence of PCBs on the thyroid status of rainbow trout was assessed at various temperatures to identify if PCB mixtures, as well OH-PCBs produced via biotransformation of parent PCBs, can illicit thyroid effects in fish. Juvenile rainbow trout (Oncorhynchus mykiss) held at 8, 12 or 16 degrees C were exposed to dietary concentrations of an environmentally relevant mixture of PCBs for 30 days followed by a depuration phase. Two additional treatments at 12 degrees C included higher concentrations of PCBs (congeners 77, 126 and 169) known to induce CYP1A in fish (referred to as CYP1A treatment) and PCBs (congeners 87, 99, 101, 153, 180, 183 and 194) known to induce CYP2B in mammals (referred to as CYP2 treatment), to assess the influence of more biologically relevant PCB congeners on thyroid indices in fish. Growth rate and liver somatic index varied with water temperature (p<0.05) but did not differ between PCB exposed and control fish (p>0.05) and mortality was low in all treatments. Changes in some measures of thyroid status were apparent in PCB-exposed fish held in the 12 and 16 degrees C treatments while other measures showed no change in any treatment. The natural inverse relationship between thyroid epithelial cell height (TECH) and temperature, was diminished after 30 days of exposure to PCBs as the epithelial cell height in PCB-exposed fish was significantly augmented in the 12 and 16 degrees C treatments compared to controls at these temperatures (p<0.05). However, after 20 days of depuration, TECH values in the PCB exposed fish returned to control values. The natural linear gradient between T(4) outer-ring deiodinase activity (ORD) and temperature was also diminished after 30 days of exposure to PCBs. PCB-exposed fish from the 16 degrees C treatment had significantly lower deiodinase activities (p<0.05) compared to controls at this temperature, but deiodinase activities returned to normal by day 20 of depuration. No differences were observed in T(3) inner-ring deiodinase (IRD) activities and plasma concentrations of T(3) and T(4) in any of the treatments (p>0.05). EROD activity in fish from the CYP1A and CYP2 treatments were elevated compared to control and high dose PCB-exposed treatments (p<0.05), but the inclusion of CYP inducing congeners did not appear to influence any index of thyroid status. Results of this study suggest that exposure of rainbow trout to high concentrations of PCBs and/or OH-PCBs may alter some indices of thyroid status when water temperatures are high, but these changes are within the compensatory scope of the thyroid system based on no change in circulating hormone concentrations, growth rates or mortality.
Collapse
Affiliation(s)
- Andrea H Buckman
- Department of Environmental Biology, University of Guelph, Guelph, Ont., Canada N1G 2W1; National Waters Research Institute, Environment Canada, Burlington, Ont., Canada L7R 4A6
| | - Aaron T Fisk
- Great Lakes Institute for Environmental Research, University of Windsor, 401 Sunset Avenue, Windsor, Ont., Canada N9B 3P4.
| | - Joanne L Parrott
- National Waters Research Institute, Environment Canada, Burlington, Ont., Canada L7R 4A6
| | - Keith R Solomon
- Department of Environmental Biology, University of Guelph, Guelph, Ont., Canada N1G 2W1
| | - Scott B Brown
- National Waters Research Institute, Environment Canada, Burlington, Ont., Canada L7R 4A6
| |
Collapse
|