143 results on '"Corton JC"'
Search Results
2. Peroxisome proliferator-activated receptor gamma coactivator 1 in caloric restriction and other models of longevity.
- Author
-
Corton JC, Brown-Borg HM, Corton, J Christopher, and Brown-Borg, Holly M
- Abstract
Dietary restriction of calories (caloric restriction [CR]) increases longevity in phylogenetically diverse species. CR retards or prevents age-dependent deterioration of tissues and an array of spontaneous and chemically induced diseases associated with obesity including cardiovascular disease, diabetes, and cancer. An understanding of the molecular mechanisms that underlie the beneficial effects of CR will help identify novel dietary, pharmacological, and lifestyle strategies for slowing the rate of aging and preventing these diseases as well as identify factors which modulate chemical toxicity. Here, we review the involvement of transcriptional coactivator proteins, peroxisome proliferator-activated receptor (PPAR) gamma coactivator 1 (PGC-1) alpha and beta, and regulated nuclear receptors (NR) in mediating the phenotypic changes found in models of longevity which include rodent CR models and mouse mutants in which insulin and/or insulin-like growth factor-I signaling is attenuated. PGC-1alpha is transcriptionally or posttranslationally regulated in mammals by: 1) forkhead box "other" (FoxO) transcription factors through an insulin/insulin-like growth factor-I -dependent pathway, 2) glucagon-stimulated cellular AMP (cAMP) response element binding protein, 3) stress-activated kinase signaling through p38 mitogen-activated protein kinase, and 4) the deacetylase and longevity factor sirtuin 1 (SIRT1). PGC-1alpha and PGC-1beta regulate the ligand-dependent and -independent activation of a large number of NR including PPARalpha and constitutive activated receptor (CAR). These NR regulate genes involved in nutrient and xenobiotic transport and metabolism as well as resistance to stress. CR reverses age-dependent decreases in PGC-1alpha, PPARalpha, and regulated genes. Strategies that target one or multiple PGC-1-regulated NR could be used to mimic the beneficial health effects found in models of longevity. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
3. Altering DNA-binding specificity of GAL4 requires sequences adjacent to the zinc finger
- Author
-
Corton Jc and Stephen Albert Johnston
- Subjects
chemistry.chemical_classification ,Zinc finger ,Multidisciplinary ,biology ,Saccharomyces cerevisiae ,Molecular Sequence Data ,biology.organism_classification ,DNA-binding protein ,Yeast ,DNA sequencing ,Recombinant Proteins ,Amino acid ,DNA-Binding Proteins ,Fungal Proteins ,Zinc ,Biochemistry ,chemistry ,Transcription (biology) ,Metalloproteins ,Mutation ,Transcriptional regulation ,Amino Acid Sequence ,Transcription Factors - Abstract
MANY eukaryotic proteins involved in transcriptional regulation contain within their DNA-binding domains a polypeptide loop (the zinc finger) which interacts with DNA1,2. In proteins possessing multiple zinc fingers, including TFIIIA3, Spl4,5, SWI56 and oestrogen/glucocorticoid receptors7, the region containing the zinc fingers confers DNA-binding specificity. By contrast, our results demonstrate that all but one of the 28 amino acids encompassing the single zinc-finger region of GAL4, the yeast transcriptional activator, can be replaced with the analogous zinc-finger region from another yeast-activator protein, PPR1, without changing the DNA-binding specificity of GAL4. A 14-amino-acid region adjacent to the zinc finger is necessary for determining specific recognition of DNA sequences.
- Published
- 1989
4. Consensus findings of an International Workshops on Genotoxicity Testing workshop on using transcriptomic biomarkers to predict genotoxicity.
- Author
-
Froetschl R, Corton JC, Li H, Aubrecht J, Auerbach SS, Caiment F, Doktorova TY, Fujita Y, Jennen D, Koyama N, Meier MJ, Mezencev R, Recio L, Suzuki T, and Yauk CL
- Abstract
Gene expression biomarkers have the potential to identify genotoxic and non-genotoxic carcinogens, providing opportunities for integrated testing and reducing animal use. In August 2022, an International Workshops on Genotoxicity Testing (IWGT) workshop was held to critically review current methods to identify genotoxicants using transcriptomic profiling. Here, we summarize the findings of the workgroup on the state of the science regarding the use of transcriptomic biomarkers to identify genotoxic chemicals in vitro and in vivo. A total of 1341 papers were examined to identify the biomarkers that show the most promise for identifying genotoxicants. This analysis revealed two independently derived in vivo biomarkers and three in vitro biomarkers that, when used in conjunction with standard computational techniques, can identify genotoxic chemicals in vivo (rat or mouse liver) or in human cells in culture using different gene expression profiling platforms, with predictive accuracies of ≥92%. These biomarkers have been validated to differing degrees but typically show high reproducibility across transcriptomic platforms and model systems. They offer several advantages for applications in different contexts of use in genotoxicity testing including: early signal detection, moderate-to-high-throughput screening capacity, adaptability to different cell types and tissues, and insights on mechanistic information on DNA-damage response. Workshop participants agreed on consensus statements to advance the regulatory adoption of transcriptomic biomarkers for genotoxicity. The participants agreed that transcriptomic biomarkers have the potential to be used in conjunction with other biomarkers in integrated test strategies in vitro and using short-term rodent exposures to identify genotoxic and non-genotoxic chemicals that may cause cancer and heritable genetic effects. Following are the consensus statements from the workgroup. Transcriptomic biomarkers for genotoxicity can be used in Weight of Evidence (WoE) evaluation to: determine potential genotoxic mechanisms and hazards; identify misleading positives from in vitro genotoxicity assays; serve as new approach methodologies (NAMs) integrated into the standard battery of genotoxicity tests. Several transcriptomic biomarkers have been developed from sufficiently robust training data sets, validated with external test sets, and have demonstrated performance in multiple laboratories. These transcriptomic biomarkers can be used following established study designs and models designated through existing validation exercises in WoE evaluation. Bridging studies using a selection of training and test chemicals are needed to deviate from the established protocols to confirm performance when a transcriptomic biomarker is being applied in other: tissues, cell models, or gene expression platforms. Top dose selection and time of gene expression analysis are critical and should be established during transcriptomic biomarker development. These conditions are the only ones suited for transcriptomic biomarker use unless additional bridging or pharmacokinetic studies are conducted. Temporal effects for genotoxicants that operate via distinct mechanisms should be considered in data interpretation. Fixed transcriptomic biomarker gene sets and analytical processes do not need to be independently rederived in biomarker validation. Validation should focus on the performance of the gene set in external test sets. Robust external testing should ensure a minimum of additional chemicals spanning genotoxic and non-genotoxic modes of action. Genes in the transcriptomic biomarker do not need to be known to be mechanistically involved in genotoxicity responses. Existing frameworks described for NAMs could be applied for validation of transcriptomic biomarkers. Reproducibility of bioinformatic analysis is critical for the regulatory application of transcriptomic biomarkers. A bioinformatics expert should be involved with creating reproducible methods for the qualification and application of each transcriptomic biomarker., (© 2025 The Author(s). Environmental and Molecular Mutagenesis published by Wiley Periodicals LLC on behalf of Environmental Mutagenesis and Genomics Society.)
- Published
- 2025
- Full Text
- View/download PDF
5. Outcome of IWGT workshop on transcriptomic biomarkers for genotoxicity: Key considerations for bioinformatics.
- Author
-
Meier MJ, Caiment F, Corton JC, Frötschl R, Fujita Y, Jennen D, Mezencev R, and Yauk C
- Abstract
As a part of the International Workshop on Genotoxicity Testing (IWGT) in 2022, a workgroup was formed to evaluate the level of validation and regulatory acceptance of transcriptomic biomarkers that identify genotoxic substances. Several such biomarkers have been developed using various molecular techniques and computational approaches. Within the IWGT workgroup on transcriptomic biomarkers, bioinformatics was a central topic of discussion, focusing on the current approaches used to process the underlying molecular data to distill a reliable predictive signal; that is, a gene set that is indicative of genotoxicity and can then be used in later studies to predict potential DNA damaging properties for uncharacterized chemicals. While early studies used microarray data, a technological shift occurred in the past decade to incorporate modern transcriptome measuring techniques such as high-throughput transcriptomics, which in turn is based on high-throughput sequencing. Herein, we present the workgroup's review of the current bioinformatic approaches to identify genes comprising transcriptomic biomarkers. Within the context of regulatory toxicology, the reproducibility of a given analysis is critical. Therefore, the workgroup provides consensus recommendations on how to facilitate sufficient reporting of experimental parameters for the analytical procedures used in a transcriptomic biomarker study, including the recommendation to develop a biomarker-specific reporting module within the OECD Omics Reporting Framework., (© 2024 His Majesty the King in Right of Canada and The Author(s). Environmental and Molecular Mutagenesis published by Wiley Periodicals LLC on behalf of Environmental Mutagenesis and Genomics Society. Reproduced with the permission of the Minister of Minister of Health. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.)
- Published
- 2024
- Full Text
- View/download PDF
6. A new approach methodology to identify tumorigenic chemicals using short-term exposures and transcript profiling.
- Author
-
Ledbetter V, Auerbach S, Everett LJ, Vallanat B, Lowit A, Akerman G, Gwinn W, Wehmas LC, Hughes MF, Devito M, and Corton JC
- Abstract
Current methods for cancer risk assessment are resource-intensive and not feasible for most of the thousands of untested chemicals. In earlier studies, we developed a new approach methodology (NAM) to identify liver tumorigens using gene expression biomarkers and associated tumorigenic activation levels (TALs) after short-term exposures in rats. The biomarkers are used to predict the six most common rodent liver cancer molecular initiating events. In the present study, we wished to confirm that our approach could be used to identify liver tumorigens at only one time point/dose and if the approach could be applied to (targeted) RNA-Seq analyses. Male rats were exposed for 4 days by daily gavage to 15 chemicals at doses with known chronic outcomes and liver transcript profiles were generated using Affymetrix arrays. Our approach had 75% or 85% predictive accuracy using TALs derived from the TG-GATES or DrugMatrix studies, respectively. In a dataset generated from the livers of male rats exposed to 16 chemicals at up to 10 doses for 5 days, we found that our NAM coupled with targeted RNA-Seq (TempO-Seq) could be used to identify tumorigenic chemicals with predictive accuracies of up to 91%. Overall, these results demonstrate that our NAM can be applied to both microarray and (targeted) RNA-Seq data generated from short-term rat exposures to identify chemicals, their doses, and mode of action that would induce liver tumors, one of the most common endpoints in rodent bioassays., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Ledbetter, Auerbach, Everett, Vallanat, Lowit, Akerman, Gwinn, Wehmas, Hughes, Devito and Corton.)
- Published
- 2024
- Full Text
- View/download PDF
7. A transcriptomic biomarker predictive of cell proliferation for use in adverse outcome pathway-informed testing and assessment.
- Author
-
Corton JC, Ledbetter V, Cohen SM, Atlas E, Yauk CL, and Liu J
- Subjects
- Humans, Animals, Mice, Adverse Outcome Pathways, Rats, MCF-7 Cells, Male, Female, Gene Expression Profiling, Biomarkers metabolism, Cell Proliferation drug effects, Transcriptome drug effects
- Abstract
High-throughput transcriptomics (HTTr) is increasingly being used to identify molecular targets of chemicals that can be linked to adverse outcomes. Cell proliferation (CP) is an important key event in chemical carcinogenesis. Here, we describe the construction and characterization of a gene expression biomarker that is predictive of the CP status in human and rodent tissues. The biomarker was constructed from 30 genes known to be increased in expression in prostate cancers relative to surrounding tissues and in cycling human MCF-7 cells after estrogen receptor (ER) agonist exposure. Using a large compendium of gene expression profiles to test utility, the biomarker could identify increases in CP in (i) 308 out of 367 tumor vs. normal surrounding tissue comparisons from 6 human organs, (ii) MCF-7 cells after activation of ER, (iii) after partial hepatectomy in mice and rats, and (iv) the livers of mice and rats after exposure to nongenotoxic hepatocarcinogens. The biomarker identified suppression of CP (i) under conditions of p53 activation by DNA damaging agents in human cells, (ii) in human A549 lung cells exposed to therapeutic anticancer kinase inhibitors (dasatinib, nilotnib), and (iii) in the mouse liver when comparing high levels of CP at birth to the low background levels in the adult. The responses using the biomarker were similar to those observed using conventional markers of CP including PCNA, Ki67, and BrdU labeling. The CP biomarker will be a useful tool for interpretation of HTTr data streams to identify CP status after exposure to chemicals in human cells or in rodent tissues., (Published by Oxford University Press on behalf of the Society of Toxicology 2024.)
- Published
- 2024
- Full Text
- View/download PDF
8. A 50-gene biomarker identifies estrogen receptor-modulating chemicals in a microarray compendium.
- Author
-
Corton JC, Matteo G, Chorley B, Liu J, Vallanat B, Everett L, Atlas E, Meier MJ, Williams A, and Yauk CL
- Subjects
- Humans, MCF-7 Cells, Gene Expression Profiling, Oligonucleotide Array Sequence Analysis, Biomarkers metabolism, Estrogen Receptor Modulators pharmacology, Receptors, Estrogen metabolism, Receptors, Estrogen genetics, Benzhydryl Compounds toxicity, Phenols pharmacology, Phenols toxicity
- Abstract
High throughput transcriptomics (HTTr) profiling has the potential to rapidly and comprehensively identify molecular targets of environmental chemicals that can be linked to adverse outcomes. We describe here the construction and characterization of a 50-gene expression biomarker designed to identify estrogen receptor (ER) active chemicals in HTTr datasets. Using microarray comparisons, the genes in the biomarker were identified as those that exhibited consistent directional changes when ER was activated (4 ER agonists; 4 ESR1 gene constitutively active mutants) and opposite directional changes when ER was suppressed (4 antagonist treatments; 4 ESR1 knockdown experiments). The biomarker was evaluated as a predictive tool using the Running Fisher algorithm by comparison to annotated gene expression microarray datasets including those evaluating the transcriptional effects of hormones and chemicals in MCF-7 cells. Depending on the reference dataset used, the biomarker had a predictive accuracy for activation of up to 96%. To demonstrate applicability for HTTr data analysis, the biomarker was used to identify ER activators in a set of 15 chemicals that are considered potential bisphenol A (BPA) alternatives examined at up to 10 concentrations in MCF-7 cells and analyzed by full-genome TempO-Seq. Using benchmark dose (BMD) modeling, the biomarker genes stratified the ER potency of BPA alternatives consistent with previous studies. These results demonstrate that the ER biomarker can be used to accurately identify ER activators in transcript profile data derived from MCF-7 cells., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Crown Copyright © 2024. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. Hepatic Transcriptome Comparative In Silico Analysis Reveals Similar Pathways and Targets Altered by Legacy and Alternative Per- and Polyfluoroalkyl Substances in Mice.
- Author
-
Robarts DR, Dai J, Lau C, Apte U, and Corton JC
- Abstract
Per- and poly-fluoroalkyl substances (PFAS) are a large class of fluorinated carbon chains that include legacy PFAS, such as perfluorooctane sulfonate (PFOS), perfluorooctanoic acid (PFOA), perfluorononanoic acid (PFNA), and perfluorohexane sulfonate (PFHxS). These compounds induce adverse health effects, including hepatotoxicity. Potential alternatives to the legacy PFAS (HFPO-DA (GenX), HFPO4, HFPO-TA, F-53B, 6:2 FTSA, and 6:2 FTCA), as well as a byproduct of PFAS manufacturing (Nafion BP2), are increasingly being found in the environment. The potential hazards of these new alternatives are less well known. To better understand the diversity of molecular targets of the PFAS, we performed a comparative toxicogenomics analysis of the gene expression changes in the livers of mice exposed to these PFAS, and compared these to five activators of PPARα, a common target of many PFAS. Using hierarchical clustering, pathway analysis, and predictive biomarkers, we found that most of the alternative PFAS modulate molecular targets that overlap with legacy PFAS. Only three of the 11 PFAS tested did not appreciably activate PPARα (Nafion BP2, 6:2 FTSA, and 6:2 FTCA). Predictive biomarkers showed that most PFAS (PFHxS, PFOA, PFOS, PFNA, HFPO-TA, F-53B, HFPO4, Nafion BP2) activated CAR. PFNA, PFHxS, PFOA, PFOS, HFPO4, HFPO-TA, F-53B, Nafion BP2, and 6:2 FTSA suppressed STAT5b, activated NRF2, and activated SREBP. There was no apparent relationship between the length of the carbon chain, type of head group, or number of ether linkages and the transcriptomic changes. This work highlights the similarities in molecular targets between the legacy and alternative PFAS.
- Published
- 2023
- Full Text
- View/download PDF
10. Increased Cell Proliferation as a Key Event in Chemical Carcinogenesis: Application in an Integrated Approach for the Testing and Assessment of Non-Genotoxic Carcinogenesis.
- Author
-
Strupp C, Corvaro M, Cohen SM, Corton JC, Ogawa K, Richert L, and Jacobs MN
- Subjects
- Animals, Humans, Agrochemicals, Biological Assay, Cell Proliferation, Carcinogenesis, Carcinogens toxicity
- Abstract
In contrast to genotoxic carcinogens, there are currently no internationally agreed upon regulatory tools for identifying non-genotoxic carcinogens of human relevance. The rodent cancer bioassay is only used in certain regulatory sectors and is criticized for its limited predictive power for human cancer risk. Cancer is due to genetic errors occurring in single cells. The risk of cancer is higher when there is an increase in the number of errors per replication (genotoxic agents) or in the number of replications (cell proliferation-inducing agents). The default regulatory approach for genotoxic agents whereby no threshold is set is reasonably conservative. However, non-genotoxic carcinogens cannot be regulated in the same way since increased cell proliferation has a clear threshold. An integrated approach for the testing and assessment (IATA) of non-genotoxic carcinogens is under development at the OECD, considering learnings from the regulatory assessment of data-rich substances such as agrochemicals. The aim is to achieve an endorsed IATA that predicts human cancer better than the rodent cancer bioassay, using methodologies that equally or better protect human health and are superior from the view of animal welfare/efficiency. This paper describes the technical opportunities available to assess cell proliferation as the central gateway of an IATA for non-genotoxic carcinogenicity.
- Published
- 2023
- Full Text
- View/download PDF
11. Carcinogenicity of aspartame, methyleugenol, and isoeugenol.
- Author
-
Riboli E, Beland FA, Lachenmeier DW, Marques MM, Phillips DH, Schernhammer E, Afghan A, Assunção R, Caderni G, Corton JC, de Aragão Umbuzeiro G, de Jong D, Deschasaux-Tanguy M, Hodge A, Ishihara J, Levy DD, Mandrioli D, McCullough ML, McNaughton SA, Morita T, Nugent AP, Ogawa K, Pandiri AR, Sergi CM, Touvier M, Zhang L, Benbrahim-Tallaa L, Chittiboyina S, Cuomo D, DeBono NL, Debras C, de Conti A, El Ghissassi F, Fontvieille E, Harewood R, Kaldor J, Mattock H, Pasqual E, Rigutto G, Simba H, Suonio E, Viegas S, Wedekind R, Schubauer-Berigan MK, and Madia F
- Subjects
- Humans, Carcinogenicity Tests, Aspartame adverse effects, Eugenol
- Published
- 2023
- Full Text
- View/download PDF
12. Letter to the Editors regarding "10% body weight (gain) change as criterion for the maximum tolerated dose: A critical analysis".
- Author
-
Berry SCL, Cohen SM, Corton JC, de Camargo JLV, Eisenbrand G, Fukushima S, Greim H, Weber K, Rietjens IMCM, and Strupp C
- Subjects
- Humans, Maximum Tolerated Dose, Body Weight, Weight Gain
- Abstract
Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: SMC is on the editorial board of Regulatory Toxicology and Pharmacology. The opinions expressed in this letter are personal and do not represent a position of the journal. CS is currently chairman of the Human Health Expert Group of CropLife Europe. CC: The information in this document has been funded in part by the U.S. Environmental Protection Agency. It has been subjected to review by the Center for Computational Toxicology and Exposure and approved for publication. Approval does not signify that the contents reflect the views of the Agency, nor does mention of trade names or commercial products constitute endorsement or recommendation for use.
- Published
- 2023
- Full Text
- View/download PDF
13. Identifying Human Specific Adverse Outcome Pathways of Per- and Polyfluoroalkyl Substances Using Liver-Chimeric Humanized Mice.
- Author
-
Robarts DR, Paine-Cabrera D, Kotulkar M, Venneman KK, Gunewardena S, Corton JC, Lau C, Foquet L, Bial G, and Apte U
- Abstract
Background: Per- and polyfluoroalkyl substances (PFAS) are persistent organic pollutants with myriad adverse effects. While perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS) are the most common contaminants, levels of replacement PFAS, such as perfluoro-2-methyl-3-oxahexanoic acid (GenX), are increasing. In rodents, PFOA, PFOS, and GenX have several adverse effects on the liver, including nonalcoholic fatty liver disease., Objective: We aimed to determine human-relevant mechanisms of PFAS induced adverse hepatic effects using FRG liver-chimeric humanized mice with livers repopulated with functional human hepatocytes., Methods: Male humanized mice were treated with 0.067 mg/L of PFOA, 0.145 mg/L of PFOS, or 1 mg/L of GenX in drinking water for 28 days. Liver and serum were collected for pathology and clinical chemistry, respectively. RNA-sequencing coupled with pathway analysis was used to determine molecular mechanisms., Results: PFOS caused a significant decrease in total serum cholesterol and LDL/VLDL, whereas GenX caused a significant elevation in LDL/VLDL with no change in total cholesterol and HDL. PFOA had no significant changes in serum LDL/VLDL and total cholesterol. All three PFAS induced significant hepatocyte proliferation. RNA-sequencing with alignment to the human genome showed a total of 240, 162, and 619 differentially expressed genes after PFOA, PFOS, and GenX exposure, respectively. Upstream regulator analysis revealed inhibition of NR1D1, a transcriptional repressor important in circadian rhythm, as the major common molecular change in all PFAS treatments. PFAS treated mice had significant nuclear localization of NR1D1. In silico modeling showed PFOA, PFOS, and GenX potentially interact with the DNA-binding domain of NR1D1., Discussion: These data implicate PFAS in circadian rhythm disruption via inhibition of NR1D1. These studies show that FRG humanized mice are a useful tool for studying the adverse outcome pathways of environmental pollutants on human hepatocytes in situ., Competing Interests: Declaration of conflicts of interest: LF and GB are employed by Yecuris Corp., the company that sells FRG humanized mice. All other authors declare that they have no actual or potential competing financial interests. This study has been subjected to review by the Center for Computational Toxicology and Exposure, Office of Research and Development, EPA and approved for publication. Approval does not signify that the contents reflect the views of the Agency, nor does mention of trade names or commercial products constitute endorsement or recommendation for use.
- Published
- 2023
- Full Text
- View/download PDF
14. Determinants of gene expression in the human liver: Impact of aging and sex on xenobiotic metabolism.
- Author
-
Corton JC, Lee JS, Liu J, Ren H, Vallanat B, and DeVito M
- Subjects
- Male, Humans, Female, Aged, Liver metabolism, Aging genetics, Aging metabolism, Gene Expression, Xenobiotics metabolism, Xenobiotics toxicity, Cytochrome P-450 Enzyme System genetics, Cytochrome P-450 Enzyme System metabolism
- Abstract
There is a need to characterize the potential susceptibility of older adults to toxicity from environmental chemical exposures. Liver xenobiotic metabolizing enzymes (XMEs) play important roles in detoxifying and eliminating xenobiotics. We examined global gene expression in the livers of young (21-45 years) and old (69+ years) men and women. Differentially expressed genes (DEG) were identified using two-way ANOVA (p ≤ 0.05). We identified 1437 and 1670 DEGs between young and old groups in men and women, respectively. Only a minor number of the total number of genes overlapped (146 genes). Aging increased or decreased pathways involved in inflammation and intermediary metabolism, respectively. Aging led to numerous changes in the expression of XME genes or genes known to control their expression (~90 genes). Out of 10 cytochrome P450s activities examined, there were increased activities of CYP1A2 and CYP2C9 enzymes in the old groups. We also identified sex-dependent genes that were more numerous in the young group (1065) than in the old group (202) and included changes in XMEs. These studies indicate that the livers from aging humans when compared to younger adults exhibit changes in XMEs that may lead to differences in the metabolism of xenobiotics., (Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
15. A gene expression biomarker identifies inhibitors of two classes of epigenome effectors in a human microarray compendium.
- Author
-
Corton JC, Liu J, Williams A, Cho E, and Yauk CL
- Subjects
- Biomarkers, Cell Cycle Proteins metabolism, Genetic Markers, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylases metabolism, Humans, Nuclear Proteins metabolism, Oligonucleotide Array Sequence Analysis, Transcription Factors, Transcriptome, Epigenome, Gene Expression Profiling methods
- Abstract
Biomarkers predictive of molecular and toxicological effects are needed to interpret emerging high-throughput transcriptomics (HTTr) data streams. To address the limited approaches available for identifying epigenotoxicants, we previously developed and validated an 81-gene biomarker that accurately predicts histone deacetylase inhibition (HDACi) in transcript profiles derived from chemically-treated TK6 cells. In the present study, we sought to determine if this biomarker (TGx-HDACi) could be used to identify HDACi chemicals in other cell lines using the Running Fisher correlation test. Using microarray comparisons derived from human cells exposed to HDACi, we found considerable heterogeneity in correlation with the TGx-HDACi biomarker dependent on chemical exposure conditions and tissue from which the cell line was derived. Using a defined set of conditions that overlapped with our earlier study, the biomarker was able to accurately identify HDACi chemicals (90-100% balanced accuracy). In an in silico screen of 2427 chemicals in 9660 chemical versus control comparisons, the biomarker coupled with the Running Fisher test was able to identify 14 additional HDACi chemicals as well as other chemicals not previously associated with HDACi. Most notable were 12 inhibitors of bromodomain (BRD) and extraterminal (BET) family proteins including BRD4 that bind to acetylated histones. The BET protein inhibitors could be distinguished from the HDACi based on differences in the expression of a small set of biomarker genes. Our results indicate that the TGx-HDACi biomarker will be useful for identifying inhibitors of two classes of epigenome effectors in HTTr screening studies., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
16. Towards replacement of animal tests with in vitro assays: a gene expression biomarker predicts in vitro and in vivo estrogen receptor activity.
- Author
-
Corton JC, Liu J, Kleinstreuer N, Gwinn MR, and Ryan N
- Subjects
- Animals, Biomarkers, Female, Gene Expression, High-Throughput Screening Assays, Humans, Receptors, Estrogen genetics, Receptors, Estrogen metabolism, Breast Neoplasms genetics, Endocrine Disruptors
- Abstract
High-throughput transcriptomics (HTTr) has the potential to support efforts to reduce or replace some animal tests. In past studies, we described a computational approach utilizing a gene expression biomarker consisting of 46 genes to predict estrogen receptor (ER) activity after chemical exposure in ER-positive human breast cancer cells including the MCF-7 cell line. We hypothesized that the biomarker model could identify ER activities of chemicals examined by Endocrine Disruptor Screening Program (EDSP) Tier 1 screening assays in which transcript profiles of the same chemicals were examined in MCF-7 cells. For the 62 chemicals examined including 5 chemicals examined in this study using RNA-Seq, the ER biomarker model accuracy was 1) 97% for in vitro reference chemicals, 2) 76-85% for guideline uterotrophic assays, and 3) 87-88% for guideline and nonguideline uterotrophic assays. For the same chemicals, these accuracies were similar or slightly better than those of the ToxCast ER model based on 18 in vitro assays. The performance of the ER biomarker model indicates that HTTr interpreted using the ER biomarker correctly identifies active and inactive ER reference chemicals. As part of the HTTr screening program the approach could rapidly identify chemicals with potential ER bioactivities for additional screening and testing., (Copyright © 2022. Published by Elsevier B.V.)
- Published
- 2022
- Full Text
- View/download PDF
17. A Collaborative Initiative to Establish Genomic Biomarkers for Assessing Tumorigenic Potential to Reduce Reliance on Conventional Rodent Carcinogenicity Studies.
- Author
-
Corton JC, Mitchell CA, Auerbach S, Bushel P, Ellinger-Ziegelbauer H, Escobar PA, Froetschl R, Harrill AH, Johnson K, Klaunig JE, Pandiri AR, Podtelezhnikov AA, Rager JE, Tanis KQ, van der Laan JW, Vespa A, Yauk CL, Pettit SD, and Sistare FD
- Subjects
- Animals, Biomarkers, Tumor genetics, Carcinogenesis, Carcinogenicity Tests, Carcinogens toxicity, Genomics, Neoplasms chemically induced, Neoplasms genetics, Rodentia
- Abstract
There is growing recognition across broad sectors of the scientific community that use of genomic biomarkers has the potential to reduce the need for conventional rodent carcinogenicity studies of industrial chemicals, agrochemicals, and pharmaceuticals through a weight-of-evidence approach. These biomarkers fall into 2 major categories: (1) sets of gene transcripts that can identify distinct tumorigenic mechanisms of action; and (2) cancer driver gene mutations indicative of rapidly expanding growth-advantaged clonal cell populations. This call-to-action article describes a collaborative approach launched to develop and qualify biomarker gene expression panels that measure widely accepted molecular pathways linked to tumorigenesis and their activation levels to predict tumorigenic doses of chemicals from short-term exposures. Growing evidence suggests that application of such biomarker panels in short-term exposure rodent studies can identify both tumorigenic hazard and tumorigenic activation levels for chemical-induced carcinogenicity. In the future, this approach will be expanded to include methodologies examining mutations in key cancer driver gene mutation hotspots as biomarkers of both genotoxic and nongenotoxic chemical tumor risk. Analytical, technical, and biological validation studies of these complementary genomic tools are being undertaken by multisector and multidisciplinary collaborative teams within the Health and Environmental Sciences Institute. Success from these efforts will facilitate the transition from current heavy reliance on conventional 2-year rodent carcinogenicity studies to more rapid animal- and resource-sparing approaches for mechanism-based carcinogenicity evaluation supporting internal and regulatory decision-making., (© The Author(s) 2022. Published by Oxford University Press on behalf of the Society of Toxicology.)
- Published
- 2022
- Full Text
- View/download PDF
18. A gene expression biomarker for predictive toxicology to identify chemical modulators of NF-κB.
- Author
-
Korunes KL, Liu J, Huang R, Xia M, Houck KA, and Corton JC
- Subjects
- Cell Line, Databases, Chemical, Gene Expression Regulation drug effects, High-Throughput Screening Assays, Humans, I-kappa B Proteins genetics, I-kappa B Proteins metabolism, NF-kappa B agonists, NF-kappa B antagonists & inhibitors, NF-kappa B p50 Subunit deficiency, NF-kappa B p50 Subunit genetics, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, Tumor Necrosis Factor-alpha pharmacology, Biomarkers metabolism, Gene Expression Regulation genetics, NF-kappa B metabolism
- Abstract
The nuclear factor-kappa B (NF-κB) is a transcription factor with important roles in inflammation, immune response, and oncogenesis. Dysregulation of NF-κB signaling is associated with inflammation and certain cancers. We developed a gene expression biomarker predictive of NF-κB modulation and used the biomarker to screen a large compendia of gene expression data. The biomarker consists of 108 genes responsive to tumor necrosis factor α in the absence but not the presence of IκB, an inhibitor of NF-κB. Using a set of 450 profiles from cells treated with immunomodulatory factors with known NF-κB activity, the balanced accuracy for prediction of NF-κB activation was > 90%. The biomarker was used to screen a microarray compendium consisting of 12,061 microarray comparisons from human cells exposed to 2,672 individual chemicals to identify chemicals that could cause toxic effects through NF-κB. There were 215 and 49 chemicals that were identified as putative or known NF-κB activators or suppressors, respectively. NF-κB activators were also identified using two high-throughput screening assays; 165 out of the ~3,800 chemicals (ToxCast assay) and 55 out of ~7,500 unique compounds (Tox21 assay) were identified as potential activators. A set of 32 chemicals not previously associated with NF-κB activation and which partially overlapped between the different screens were selected for validation in wild-type and NFKB1-null HeLa cells. Using RT-qPCR and targeted RNA-Seq, 31 of the 32 chemicals were confirmed to be NF-κB activators. These results comprehensively identify a set of chemicals that could cause toxic effects through NF-κB., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2022
- Full Text
- View/download PDF
19. Genomic comparisons between hepatocarcinogenic and non-hepatocarcinogenic organophosphate insecticides in the mouse liver.
- Author
-
Rooney J, Wehmas LC, Ryan N, Chorley BN, Hester SD, Kenyon EM, Schmid JE, George BJ, Hughes MF, Sey YM, Tennant AH, Simmons JE, Wood CE, and Corton JC
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors agonists, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Cell Transformation, Neoplastic chemically induced, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, Chemical and Drug Induced Liver Injury etiology, Chemical and Drug Induced Liver Injury metabolism, Chemical and Drug Induced Liver Injury pathology, Constitutive Androstane Receptor agonists, Constitutive Androstane Receptor genetics, Constitutive Androstane Receptor metabolism, Cytochrome P-450 Enzyme System genetics, Cytochrome P-450 Enzyme System metabolism, Fenthion toxicity, Gene Expression Profiling, Liver metabolism, Liver pathology, Liver Neoplasms chemically induced, Liver Neoplasms metabolism, Liver Neoplasms pathology, Male, Mice, Organothiophosphorus Compounds toxicity, PPAR alpha agonists, PPAR alpha genetics, PPAR alpha metabolism, Parathion toxicity, Receptors, Aryl Hydrocarbon agonists, Receptors, Aryl Hydrocarbon genetics, Receptors, Aryl Hydrocarbon metabolism, STAT5 Transcription Factor genetics, STAT5 Transcription Factor metabolism, Cell Transformation, Neoplastic genetics, Chemical and Drug Induced Liver Injury genetics, Genomics, Insecticides toxicity, Liver drug effects, Liver Neoplasms genetics, Organophosphorus Compounds toxicity, Transcriptome drug effects
- Abstract
Short-term biomarkers of toxicity have an increasingly important role in the screening and prioritization of new chemicals. In this study, we examined early indicators of liver toxicity for three reference organophosphate (OP) chemicals, which are among the most widely used insecticides in the world. The OP methidathion was previously shown to increase the incidence of liver toxicity, including hepatocellular tumors, in male mice. To provide insights into the adverse outcome pathway (AOP) that underlies these tumors, effects of methidathion in the male mouse liver were examined after 7 and 28 day exposures and compared to those of two other OPs that either do not increase (fenthion) or possibly suppress liver cancer (parathion) in mice. None of the chemicals caused increases in liver weight/body weight or histopathological changes in the liver. Parathion decreased liver cell proliferation after 7 and 28 days while the other chemicals had no effects. There was no evidence for hepatotoxicity in any of the treatment groups. Full-genome microarray analysis of the livers from the 7 and 28 day treatments demonstrated that methidathion and fenthion regulated a large number of overlapping genes, while parathion regulated a unique set of genes. Examination of cytochrome P450 enzyme activities and use of predictive gene expression biomarkers found no consistent evidence for activation of AhR, CAR, PXR, or PPARα. Parathion suppressed the male-specific gene expression pattern through STAT5b, similar to genetic and dietary conditions that decrease liver tumor incidence in mice. Overall, these findings indicate that methidathion causes liver cancer by a mechanism that does not involve common mechanisms of liver cancer induction., (Published by Elsevier B.V.)
- Published
- 2022
- Full Text
- View/download PDF
20. Key Characteristics of Human Hepatotoxicants as a Basis for Identification and Characterization of the Causes of Liver Toxicity.
- Author
-
Rusyn I, Arzuaga X, Cattley RC, Corton JC, Ferguson SS, Godoy P, Guyton KZ, Kaplowitz N, Khetani SR, Roberts RA, Roth RA, and Smith MT
- Subjects
- Animals, Chemical and Drug Induced Liver Injury pathology, Humans, Liver drug effects, Liver pathology, Chemical and Drug Induced Liver Injury diagnosis
- Abstract
Hazard identification regarding adverse effects on the liver is a critical step in safety evaluations of drugs and other chemicals. Current testing paradigms for hepatotoxicity rely heavily on preclinical studies in animals and human data (epidemiology and clinical trials). Mechanistic understanding of the molecular and cellular pathways that may cause or exacerbate hepatotoxicity is well advanced and holds promise for identification of hepatotoxicants. One of the challenges in translating mechanistic evidence into robust decisions about potential hepatotoxicity is the lack of a systematic approach to integrate these data to help identify liver toxicity hazards. Recently, marked improvements were achieved in the practice of hazard identification of carcinogens, female and male reproductive toxicants, and endocrine disrupting chemicals using the key characteristics approach. Here, we describe the methods by which key characteristics of human hepatotoxicants were identified and provide examples for how they could be used to systematically identify, organize, and use mechanistic data when identifying hepatotoxicants., (© 2021 by the American Association for the Study of Liver Diseases.)
- Published
- 2021
- Full Text
- View/download PDF
21. A Gene Expression Biomarker Predicts Heat Shock Factor 1 Activation in a Gene Expression Compendium.
- Author
-
Cervantes PW and Corton JC
- Subjects
- Cell Line, Gene Expression Regulation drug effects, Heat-Shock Response drug effects, Humans, Toxicity Tests methods, Gene Expression Profiling methods, Heat Shock Transcription Factors genetics, Transcriptome drug effects
- Abstract
The United States Environmental Protection Agency (US EPA) recently developed a tiered testing strategy to use advances in high-throughput transcriptomics (HTTr) testing to identify molecular targets of thousands of environmental chemicals that can be linked to adverse outcomes. Here, we describe a method that uses a gene expression biomarker to predict chemical activation of heat shock factor 1 (HSF1), a transcription factor critical for proteome maintenance. The HSF1 biomarker was built from transcript profiles derived from A375 cells exposed to a HSF1-activating heat shock protein (HSP) 90 inhibitor in the presence or absence of HSF1 expression. The resultant 44 identified genes included those that (1) are dependent on HSF1 for regulation, (2) have direct interactions with HSF1 assessed by ChIP-Seq, and (3) are in the molecular chaperone family. To test for accuracy, the biomarker was compared in a pairwise manner to gene lists derived from treatments with known HSF1 activity (HSP and proteasomal inhibitors) using the correlation-based Running Fisher test; the balanced accuracy for prediction was 96%. A microarray compendium consisting of 12,092 microarray comparisons from human cells exposed to 2670 individual chemicals was screened using our approach; 112 and 19 chemicals were identified as putative HSF1 activators or suppressors, respectively, and most appear to be novel modulators. A large percentage of the chemical treatments that induced HSF1 also induced oxidant-activated NRF2 (∼46%). For five compounds or mixtures, we found that NRF2 activation occurred at lower concentrations or at earlier times than HSF1 activation, supporting the concept of a tiered cellular protection system dependent on the level of chemical-induced stress. The approach described here could be used to identify environmentally relevant chemical HSF1 activators in HTTr data sets.
- Published
- 2021
- Full Text
- View/download PDF
22. Why is elevation of serum cholesterol associated with exposure to perfluoroalkyl substances (PFAS) in humans? A workshop report on potential mechanisms.
- Author
-
Andersen ME, Hagenbuch B, Apte U, Corton JC, Fletcher T, Lau C, Roth WL, Staels B, Vega GL, Clewell HJ 3rd, and Longnecker MP
- Subjects
- Alkanesulfonic Acids adverse effects, Alkanesulfonic Acids toxicity, Animals, Caprylates adverse effects, Caprylates toxicity, Endpoint Determination, Fluorocarbons adverse effects, Humans, Cholesterol blood, Environmental Exposure adverse effects, Environmental Pollutants blood, Fluorocarbons toxicity
- Abstract
Serum concentrations of cholesterol are positively correlated with exposure to perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS) in humans. The associated change in cholesterol is small across a broad range of exposure to PFOA and PFOS. Animal studies generally have not indicated a mechanism that would account for the association in humans. The extent to which the relationship is causal is an open question. Nonetheless, the association is of particular importance because increased serum cholesterol has been considered as an endpoint to derive a point of departure in at least one recent risk assessment. To gain insight into potential mechanisms for the association, both causal and non-causal, an expert workshop was held Oct 31 and Nov 1, 2019 to discuss relevant data and propose new studies. In this report, we summarize the relevant background data, the discussion among the attendees, and their recommendations for further research., (Copyright © 2021 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
23. Development and validation of the TGx-HDACi transcriptomic biomarker to detect histone deacetylase inhibitors in human TK6 cells.
- Author
-
Cho E, Rowan-Carroll A, Williams A, Corton JC, Li HH, Fornace AJ Jr, Hobbs CA, and Yauk CL
- Subjects
- Apoptosis, Cell Line, Computational Biology, DNA Damage, Gene Expression Profiling, Genetic Markers, Humans, Lymphocytes, Mutagens, Repressor Proteins, Toxicogenetics, Transcriptome, Histone Deacetylase Inhibitors metabolism, Histone Deacetylases metabolism
- Abstract
Transcriptomic biomarkers can be used to inform molecular initiating and key events involved in a toxicant's mode of action. To address the limited approaches available for identifying epigenotoxicants, we developed and assessed a transcriptomic biomarker of histone deacetylase inhibition (HDACi). First, we assembled a set of ten prototypical HDACi and ten non-HDACi reference compounds. Concentration-response experiments were performed for each chemical to collect TK6 human lymphoblastoid cell samples after 4 h of exposure and to assess cell viability following a 20-h recovery period in fresh media. One concentration was selected for each chemical for whole transcriptome profiling and transcriptomic signature derivation, based on cell viability at the 24-h time point and on maximal induction of HDACi-response genes (RGL1, NEU1, GPR183) or cellular stress-response genes (ATF3, CDKN1A, GADD45A) analyzed by TaqMan qPCR assays after 4 h of exposure. Whole transcriptomes were profiled after 4 h exposures by Templated Oligo-Sequencing (TempO-Seq). By applying the nearest shrunken centroid (NSC) method to the whole transcriptome profiles of the reference compounds, we derived an 81-gene toxicogenomic (TGx) signature, referred to as TGx-HDACi, that classified all 20 reference compounds correctly using NSC classification and the Running Fisher test. An additional 4 HDACi and 7 non-HDACi were profiled and analyzed using TGx-HDACi to further assess classification performance; the biomarker accurately classified all 11 compounds, including 3 non-HDACi epigenotoxicants, suggesting a promising specificity toward HDACi. The availability of TGx-HDACi increases the diversity of tools that can facilitate mode of action analysis of toxicants using gene expression profiling.
- Published
- 2021
- Full Text
- View/download PDF
24. Expression of cytochrome P450 isozyme transcripts and activities in human livers.
- Author
-
Liu J, Lu YF, Corton JC, and Klaassen CD
- Subjects
- Black or African American, Aged, Child, Female, Hispanic or Latino, Humans, Isoenzymes genetics, Male, White People, Cytochrome P-450 Enzyme System genetics, Liver enzymology
- Abstract
Individual differences in cytochrome P450 (CYP) enzymes contribute to responses to drugs and environmental chemicals. The expression of CYPs is influenced by sex, age, and ethnicity. Human CYP studies are often conducted with human liver microsomes and liver cells to evaluate chemical induction and drug interactions. However, the basal or constitutive expression of CYP transcripts and enzyme activities in the intact liver are also important in our understanding of individual variation in CYPs. This study utilised 100 human liver samples to profile the constitutive expression of CYP1A2, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, 3A4, and 4A11 enzyme activity and transcript levels. The mRNA expression of the CYPs and xenobiotic receptors AhR , CAR , and PXR was examined via qPCR. Results showed that there was greater inter-individual variation in mRNA expression than in enzyme activities, except for CYP2C19. Females had higher CYP3A4 activity than males. Children had lower CYP4A14 activity, while elderly had lower P450 oxidoreductase activity. Compared to Caucasians, Hispanics had higher CYP2C8 activity and higher CYP2B6 , CYP2C9 , and CYP2C19 mRNA expression, whereas African Americans had lower CYP2D6 mRNA expression. These results add to our understanding of individual variations in xenobiotic metabolism and toxicology.
- Published
- 2021
- Full Text
- View/download PDF
25. A Gene Expression Biomarker Identifies Chemical Modulators of Estrogen Receptor α in an MCF-7 Microarray Compendium.
- Author
-
Rooney J, Ryan N, Liu J, Houtman R, van Beuningen R, Hsieh JH, Chang G, Chen S, and Corton JC
- Subjects
- Estrogen Receptor Modulators pharmacology, Estrogen Receptor alpha agonists, Estrogen Receptor alpha antagonists & inhibitors, Female, Gene Expression Profiling, Humans, MCF-7 Cells, Tumor Cells, Cultured, Biomarkers, Tumor genetics, Estrogen Receptor Modulators analysis, Estrogen Receptor alpha genetics
- Abstract
Identification of chemicals that affect hormone-regulated systems will help to predict endocrine disruption. In our previous study, a 46 gene biomarker was found to be an accurate predictor of estrogen receptor (ER) α modulation in chemically treated MCF-7 cells. Here, potential ERα modulators were identified using the biomarker by screening a microarray compendium consisting of ∼1600 gene expression comparisons representing exposure to ∼1200 chemicals. A total of ∼170 chemicals were identified as potential ERα modulators. In the Connectivity Map 2.0 collection, 75 and 39 chemicals were predicted to activate or suppress ERα, and they included 12 and six known ERα agonists and antagonists/selective ERα modulators, respectively. Nineteen and eight of the total number were also identified as active in an ERα transactivation assay carried out in an MCF-7-derived cell line used to screen the Tox21 10K chemical library in agonist or antagonist modes, respectively. Chemicals predicted to modulate ERα in MCF-7 cells were examined further using global and targeted gene expression in wild-type and ERα-null cells, transactivation assays, and cell-free ERα coregulator interaction assays. Environmental chemicals classified as weak and very weak agonists were confirmed to activate ERα including apigenin, kaempferol, and oxybenzone. Novel activators included digoxin, nabumetone, ivermectin, and six progestins. Novel suppressors included emetine, mifepristone, niclosamide, and proscillaridin. Our strategy will be useful to identify environmentally relevant ERα modulators in future high-throughput transcriptomic screens.
- Published
- 2021
- Full Text
- View/download PDF
26. Sex-, Age-, and Race/Ethnicity-Dependent Variations in Drug-Processing and NRF2-Regulated Genes in Human Livers.
- Author
-
Liu J, Cui JY, Lu YF, Corton JC, and Klaassen CD
- Subjects
- Adult, Age Factors, Aged, Child, Cytochrome P-450 Enzyme System genetics, Female, Gene Expression Profiling methods, Humans, Male, Membrane Transport Proteins genetics, Pharmacogenomic Testing methods, Pharmacokinetics, Race Factors, Receptors, Cytoplasmic and Nuclear genetics, Sex Factors, Hepatobiliary Elimination physiology, Liver metabolism, Liver Diseases drug therapy, Liver Diseases metabolism, NF-E2-Related Factor 2 genetics, NF-E2-Related Factor 2 metabolism, Pharmaceutical Preparations metabolism
- Abstract
Individual variations in xenobiotic metabolism affect the sensitivity to diseases. In this study, the impacts of sex, age, and race/ethnicity on drug-processing genes and nuclear factor erythroid 2-related factor 2 (NRF2) genes in human livers were examined via QuantiGene multiplex suspension array (226 samples) and quantitative polymerase chain reaction (qPCR) (247 samples) to profile the expression of nuclear receptors, cytochrome P450s, conjugation enzymes, transporters, bile acid metabolism, and NRF2-regulated genes. Sex differences were found in expression of about half of the genes, but in general the differences were not large. For example, females had higher transcript levels of catalase , glutamate-cysteine ligase catalytic subunit ( GCLC ) , heme oxygenase 1 ( HO-1 ) , Kelch-like ECH-associated protein 1 ( KEAP1 ) , superoxide dismutase 1, and thioredoxin reductase-1 compared with males via qPCR. There were no apparent differences due to age, except children had higher glutamate-cysteine ligase modifier subunit ( GCLM ) and elderly had higher multidrug resistance protein 3. African Americans had lower expression of farnesoid X receptor ( FXR ) but higher expression of HO-1 , Caucasians had higher expression of organic anion transporter 2, and Hispanics had higher expression of FXR, SULT2A1 , small heterodimer partner, and bile salt export pump. An examination of 34 diseased and control human liver samples showed that compared with disease-free livers, fibrotic livers had higher NAD(P)H-quinone oxidoreductase 1 ( NQO1 ) , GCLC, GCLM , and NRF2 ; hepatocellular carcinoma had higher transcript levels of NQO1 and KEAP1 ; and steatotic livers had lower GCLC , GCLM , and HO-1 expression. In summary, in drug-processing gene and NRF2 genes, sex differences were the major findings, and there were no apparent age differences, and race/ethnicity differences occurred for a few genes. These descriptive findings could add to our understanding of the sex-, age-, and race/ethnicity-dependent differences in drug-processing genes as well as NRF2 genes in normal and diseased human livers. SIGNIFICANCE STATEMENT: In human liver drug-processing and nuclear factor erythroid 2-related factor 2 genes, sex differences were the main finding. There were no apparent differences due to age, except children had higher glutamate-cysteine ligase modifier subunit, and elderly had higher multidrug resistance protein 3. African Americans had lower expression of farnesoid X receptor ( FXR ) but higher expression of heme oxygenase 1, Caucasians had higher expression of organic anion transporter 2, and Hispanics had higher expression of FXR, small heterodimer partner , SULT2A1 , and bile salt export pump., Competing Interests: Disclaimer: The information in this document has been funded in part by the U.S. Environmental Protection Agency. It has been subjected to review by the Center for Computational Toxicology and Exposure and approved for publication. Approval does not signify that the contents reflect the views of the Agency, nor does mention of trade names or commercial products constitute endorsement or recommendation for use., (Copyright © 2020 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2021
- Full Text
- View/download PDF
27. Thresholds Derived From Common Measures in Rat Studies Are Predictive of Liver Tumorigenic Chemicals.
- Author
-
Corton JC, Korunes KL, Abedini J, El-Masri H, Brown J, Paul-Friedman K, Liu Y, Martini C, He S, and Rooney J
- Subjects
- Alanine Transaminase, Animals, Aspartate Aminotransferases, Liver, Rats, Carcinogenesis, Liver Neoplasms chemically induced
- Abstract
We hypothesized that typical tissue and clinical chemistry (ClinChem) end points measured in rat toxicity studies exhibit chemical-independent biological thresholds beyond which cancer occurs. Using the rat in vivo TG-GATES study, 75 chemicals were examined across chemical-dose-time comparisons that could be linked to liver tumor outcomes. Thresholds for liver weight to body weight (LW/BW) and 21 serum ClinChem end points were defined as the maximum and minimum values for those exposures that did not lead to liver tumors in rats. Upper thresholds were identified for LW/BW (117%), aspartate aminotransferase (195%), alanine aminotransferase (141%), alkaline phosphatase (152%), and total bilirubin (115%), and lower thresholds were identified for phospholipids (82%), relative albumin (93%), total cholesterol (82%), and total protein (94%). Thresholds derived from the TG-GATES data set were consistent across other acute and subchronic rat studies. A training set of ClinChem and LW/BW thresholds derived from a 38 chemical training set from TG-GATES was predictive of liver tumor outcomes for a test set of 37 independent TG-GATES chemicals (91%). The thresholds were most predictive when applied to 7d treatments (98%). These findings provide support that biological thresholds for common end points in rodent studies can be used to predict chemical tumorigenic potential.
- Published
- 2020
- Full Text
- View/download PDF
28. A set of six Gene expression biomarkers and their thresholds identify rat liver tumorigens in short-term assays.
- Author
-
Lewis RW, Hill T 3rd, and Corton JC
- Subjects
- Animals, Biological Assay, Gene Expression, Male, Rats, Sprague-Dawley, Biomarkers, Tumor genetics, Carcinogens toxicity, Liver Neoplasms chemically induced, Liver Neoplasms genetics
- Abstract
Traditional methods for cancer risk assessment are retrospective, resource-intensive, and not feasible for the vast majority of environmental chemicals. In earlier studies, we used a set of six biomarkers to accurately identify liver tumorigens in transcript profiles derived from chemically-treated rats using either a Toxicological Priority Index (ToxPi) approach or using derived biomarker thresholds for cancer. The biomarkers consisting of 7-113 genes are used to predict the most common liver cancer molecular initiating events: genotoxicity, cytotoxicity and activation of the xenobiotic receptors AhR, CAR, ER, and PPARα. In the present study, we apply and evaluate the performance of these methods for cancer prediction in an independent rat liver study of 44 chemicals (6 h-7d exposures) examined by Affymetrix arrays. In the first approach, ToxPi ranking of biomarker scores consistently gave the highest scores to tumorigenic chemical-dose pairs; balanced accuracies for identification of liver tumorigenic chemicals were up to 89 %. The second approach used tumorigenic thresholds derived in the present study or from our earlier study that were set at the maximum value for chemical-dose exposures without detectable liver tumor outcomes. Using these thresholds, balanced accuracies were up to 90 %. Both approaches identified all tumorigenic chemicals. Almost all of the tumorigenic chemicals activated more than one MIE. We also compared biomarker responses between two types of profiling platforms (Affymetrix full-genome array, TempO-Seq 1500+ array containing ∼2600 genes) and found that the lack of the full set of biomarker genes on the 1500+ array resulted in decreased ability to identify chemicals that activate the MIEs. Overall, these results demonstrate that predictive approaches based on the 6 biomarkers could be used in short-term assays to identify chemicals and their doses that induce liver tumors, the most common endpoint in rodent bioassays., (Published by Elsevier B.V.)
- Published
- 2020
- Full Text
- View/download PDF
29. Mining a human transcriptome database for chemical modulators of NRF2.
- Author
-
Rooney JP, Chorley B, Hiemstra S, Wink S, Wang X, Bell DA, van de Water B, and Corton JC
- Subjects
- Biomarkers metabolism, Hep G2 Cells, Humans, Data Mining, Databases, Genetic, NF-E2-Related Factor 2 genetics, NF-E2-Related Factor 2 metabolism, Transcriptome
- Abstract
Nuclear factor erythroid-2 related factor 2 (NRF2) encoded by the NFE2L2 gene is a transcription factor critical for protecting cells from chemically-induced oxidative stress. We developed computational procedures to identify chemical modulators of NRF2 in a large database of human microarray data. A gene expression biomarker was built from statistically-filtered gene lists derived from microarray experiments in primary human hepatocytes and cancer cell lines exposed to NRF2-activating chemicals (oltipraz, sulforaphane, CDDO-Im) or in which the NRF2 suppressor Keap1 was knocked down by siRNA. Directionally consistent biomarker genes were further filtered for those dependent on NRF2 using a microarray dataset from cells after NFE2L2 siRNA knockdown. The resulting 143-gene biomarker was evaluated as a predictive tool using the correlation-based Running Fisher algorithm. Using 59 gene expression comparisons from chemically-treated cells with known NRF2 activating potential, the biomarker gave a balanced accuracy of 93%. The biomarker was comprised of many well-known NRF2 target genes (AKR1B10, AKR1C1, NQO1, TXNRD1, SRXN1, GCLC, GCLM), 69% of which were found to be bound directly by NRF2 using ChIP-Seq. NRF2 activity was assessed across ~9840 microarray comparisons from ~1460 studies examining the effects of ~2260 chemicals in human cell lines. A total of 260 and 43 chemicals were found to activate or suppress NRF2, respectively, most of which have not been previously reported to modulate NRF2 activity. Using a NRF2-responsive reporter gene in HepG2 cells, we confirmed the activity of a set of chemicals predicted using the biomarker. The biomarker will be useful for future gene expression screening studies of environmentally-relevant chemicals., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
30. Identification of novel activators of the metal responsive transcription factor (MTF-1) using a gene expression biomarker in a microarray compendium.
- Author
-
Jackson AC, Liu J, Vallanat B, Jones C, Nelms MD, Patlewicz G, and Corton JC
- Subjects
- DNA-Binding Proteins genetics, Gene Expression drug effects, Gene Expression Profiling, Gene Expression Regulation drug effects, Humans, MCF-7 Cells, Transcription Factors genetics, Transcription Factor MTF-1, DNA-Binding Proteins agonists, Drug Discovery, Transcription Factors agonists
- Abstract
Environmental exposure to metals is known to cause a number of human toxicities including cancer. Metal-responsive transcription factor 1 (MTF-1) is an important component of metal regulation systems in mammalian cells. Here, we describe a novel method to identify chemicals that activate MTF-1 based on microarray profiling data. MTF-1 biomarker genes were identified that exhibited consistent, robust expression across 10 microarray comparisons examining the effects of metals (zinc, nickel, lead, arsenic, mercury, and silver) on gene expression in human cells. A subset of the resulting 81 biomarker genes was shown to be altered by knockdown of the MTF1 gene including metallothionein family members and a zinc transporter. The ability to correctly identify treatment conditions that activate MTF-1 was determined by comparing the biomarker to microarray comparisons from cells exposed to reference metal activators of MTF-1 using the rank-based Running Fisher algorithm. The balanced accuracy for prediction was 93%. The biomarker was then used to identify organic chemicals that activate MTF-1 from a compendium of 11 725 human gene expression comparisons representing 2582 chemicals. There were 700 chemicals identified that included those known to interact with cellular metals, such as clioquinol and disulfiram, as well as a set of novel chemicals. All nine of the novel chemicals selected for validation were confirmed to activate MTF-1 biomarker genes in MCF-7 cells and to lesser extents in MTF1-null cells by qPCR and targeted RNA-Seq. Overall, our work demonstrates that the biomarker for MTF-1 coupled with the Running Fisher test is a reliable strategy to identify novel chemical modulators of metal homeostasis using gene expression profiling.
- Published
- 2020
- Full Text
- View/download PDF
31. Gene Expression Thresholds Derived From Short-term Exposures Identify Rat Liver Tumorigens.
- Author
-
Hill T, Rooney J, Abedini J, El-Masri H, Wood CE, and Corton JC
- Subjects
- Animals, Carcinogenesis, Gene Expression, Rats, Retrospective Studies, DNA Damage, Liver
- Abstract
Traditional methods for cancer risk assessment are resource-intensive, retrospective, and not feasible for the vast majority of environmental chemicals. In this study, we investigated whether quantitative genomic data from short-term studies may be used to set protective thresholds for potential tumorigenic effects. We hypothesized that gene expression biomarkers measuring activation of the key early events in established pathways for rodent liver cancer exhibit cross-chemical thresholds for tumorigenesis predictive for liver cancer risk. We defined biomarker thresholds for 6 major liver cancer pathways using training sets of chemicals with short-term genomic data (3-29 days of exposure) from the TG-GATES (n = 77 chemicals) and DrugMatrix (n = 86 chemicals) databases and then tested these thresholds within and between datasets. The 6 pathway biomarkers represented genotoxicity, cytotoxicity, and activation of xenobiotic, steroid, and lipid receptors (aryl hydrocarbon receptor, constitutive activated receptor, estrogen receptor, and peroxisome proliferator-activated receptor α). Thresholds were calculated as the maximum values derived from exposures without detectable liver tumor outcomes. We identified clear response values that were consistent across training and test sets. Thresholds derived from the TG-GATES training set were highly predictive (97%) in a test set of independent chemicals, whereas thresholds derived from the DrugMatrix study were 96%-97% predictive for the TG-GATES study. Threshold values derived from an abridged gene list (2/biomarker) also exhibited high predictive accuracy (91%-94%). These findings support the idea that early genomic changes can be used to establish threshold estimates or "molecular tipping points" that are predictive of later-life health outcomes., (Published by Oxford University Press on behalf of the Society of Toxicology 2020. This work is written by US Government employees and is in the public domain in the US.)
- Published
- 2020
- Full Text
- View/download PDF
32. A Set of Six Gene Expression Biomarkers Identify Rat Liver Tumorigens in Short-term Assays.
- Author
-
Corton JC, Hill T, Sutherland JJ, Stevens JL, and Rooney J
- Subjects
- Animals, Biomarkers metabolism, Carcinogenesis, Gene Expression, Rats, Biological Assay, Liver
- Abstract
Chemical-induced liver cancer occurs in rodents through well-characterized adverse outcome pathways. We hypothesized that measurement of the 6 most common molecular initiating events (MIEs) in liver cancer adverse outcome pathways in short-term assays using only gene expression will allow early identification of chemicals and their associated doses that are likely to be tumorigenic in the liver in 2-year bioassays. We tested this hypothesis using transcript data from a rat liver microarray compendium consisting of 2013 comparisons of 146 chemicals administered at doses with previously established effects on rat liver tumor induction. Five MIEs were measured using previously characterized gene expression biomarkers composed of gene sets predictive for genotoxicity and activation of 1 or more xenobiotic receptors (aryl hydrocarbon receptor, constitutive activated receptor, estrogen receptor, and peroxisome proliferator-activated receptor α). Because chronic injury can be important in tumorigenesis, we also developed a biomarker for cytotoxicity that had a 96% balanced accuracy. Characterization of the genes in each biomarker set using the unsupervised TXG-MAP network model demonstrated that the genes were associated with distinct functional coexpression modules. Using the Toxicological Priority Index to rank chemicals based on their ability to activate the MIEs showed that chemicals administered at tumorigenic doses clearly gave the highest ranked scores. Balanced accuracies using thresholds derived from either TG-GATES or DrugMatrix data sets to predict tumorigenicity in independent sets of chemicals were up to 93%. These results show that a MIE-directed approach using only gene expression biomarkers could be used in short-term assays to identify chemicals and their doses that cause tumors., (Published by Oxford University Press on behalf of the Society of Toxicology 2020. This work is written by US Government employees and is in the public domain in the US.)
- Published
- 2020
- Full Text
- View/download PDF
33. Transplacental arsenic exposure produced 5-methylcytosine methylation changes and aberrant microRNA expressions in livers of male fetal mice.
- Author
-
Liu J, Gunewardena S, Yue Cui J, Klaassen CD, Chorley BN, and Corton JC
- Subjects
- Animals, Cell Transformation, Neoplastic chemically induced, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, Female, Gene Expression Regulation, Neoplastic drug effects, Gestational Age, Liver embryology, Liver metabolism, Liver Neoplasms chemically induced, Liver Neoplasms genetics, Liver Neoplasms metabolism, Liver Neoplasms pathology, Male, Maternal Exposure, Mice, Inbred C3H, MicroRNAs genetics, Pregnancy, 5-Methylcytosine metabolism, Arsenites toxicity, DNA Methylation drug effects, Epigenesis, Genetic drug effects, Gene Expression Regulation, Developmental drug effects, Liver drug effects, MicroRNAs metabolism, Sodium Compounds toxicity
- Abstract
Arsenic is a known human carcinogen. Early-life exposure to inorganic arsenic induces tumors in humans and in C3H mice. We hypothesized that arsenic exposure in utero may induce epigenetic changes at the level of DNA methylation and miRNA alterations that could lead to greater postnatal susceptibility to cancer. To test this hypothesis, pregnant C3H mice were given sodium arsenite at doses known to cause liver cancer (42.5 and 85 ppm in the drinking water) from gestation day 8-19, and the livers from male fetal mice were collected for analysis. The antibody against 5-methylcytosine was used to perform chromatin-immunoprecipitation coupled with sequencing (ChIP-Seq) to determine genome-wide methylation alterations. In utero arsenic exposure produced global DNA hypomethylation and an array of gene-specific DNA methylation changes, including hypomethylation of Cyclin D1 and hypermethylation of Tp53. Illumina Correlation Engine analysis revealed 260 methylation alterations that would affect 143 microRNAs. MicroRNA array further revealed 140 aberrantly expressed miRNAs out of the 718 miRNAs. The increased expression of miR-205, miR-203, miR-215, miR-34a, and decreased expression of miR-217 were confirmed by qPCR. Comparison of the methylation changes to those of microarray analyses indicates little if any correspondence between gene methylation and gene expression. The increased expression of Xist, Prrc2, Krit1, Nish, and decreased expression of Prss2, Spp1, Col1a2, and Lox were confirmed by qPCR. In summary, in utero arsenic exposure induced global alterations in DNA methylation and aberrant miRNA expression that might contribute to adult adverse outcomes including liver cancer., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020. Published by Elsevier B.V.)
- Published
- 2020
- Full Text
- View/download PDF
34. Identification of potential endocrine disrupting chemicals using gene expression biomarkers.
- Author
-
Corton JC, Kleinstreuer NC, and Judson RS
- Subjects
- Animals, Biomarkers analysis, Gene Expression, Humans, Endocrine Disruptors toxicity, Receptors, Androgen genetics, Receptors, Estrogen genetics
- Abstract
Recent technological advances have moved the field of toxicogenomics from reliance on microarray platforms to high-throughput transcriptomic (HTTr) technologies that measure global gene expression. Gene expression biomarkers are emerging as useful tools for interpreting gene expression profiles to identify perturbations of targets of xenobiotic chemicals including those that act as endocrine disrupting chemicals (EDCs). Gene expression biomarkers are lists of similarly-regulated genes identified in global gene expression comparisons of cells or tissues 1) exposed to known agonists or antagonists of the transcription factor (TF) and 2) after expression of the TF itself is knocked down/knocked out or overexpressed. Estrogen receptor α (ERα) and androgen receptor (AR) biomarkers have been shown to be very accurate at identifying both agonists (94-97%) and antagonists (93-98%) in microarray data derived from human breast or prostate cancer cell lines. Importantly, the biomarkers have been shown to accurately replicate the results of computational models that predict ERα or AR modulation using multiple ToxCast HT screening assays. An integrated screening strategy using sets of biomarkers that simultaneously predict various EDC targets in relevant cell lines should simplify chemical screening without sacrificing accuracy. The biomarker predictions can be put into the context of the adverse outcome pathway framework to help prioritize chemicals with the greatest risk of potential adverse outcomes in the endocrine systems of animals and people., (Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
35. Identification of p53 Activators in a Human Microarray Compendium.
- Author
-
Corton JC, Witt KL, and Yauk CL
- Subjects
- Cell Line, Tumor, Databases, Nucleic Acid statistics & numerical data, Gene Expression Profiling, Gene Knockdown Techniques, High-Throughput Screening Assays, Humans, Oligonucleotide Array Sequence Analysis statistics & numerical data, Protein Binding drug effects, Proto-Oncogene Proteins c-mdm2 metabolism, Tumor Suppressor Protein p53 metabolism, DNA Damage drug effects, Gene Expression drug effects, Genetic Markers physiology, Organic Chemicals pharmacology, Tumor Suppressor Protein p53 agonists
- Abstract
Biomarkers predictive of molecular and toxicological effects are needed to interpret emerging high-throughput transcriptomic data streams. The previously characterized 63 gene TGx-DDI biomarker that includes 20 genes known to be regulated by p53 was previously shown to accurately predict DNA damage in chemically treated cells. We comprehensively evaluated whether the molecular basis of the DDI predictions was based on a p53-dependent response. The biomarker was compared to microarray data in a compendium derived from human cells using the Running Fisher test, a nonparametric correlation test. Using the biomarker, we identified conditions that led to p53 activation, including exposure to the chemical nutlin-3 which disrupts interactions between p53 and the negative regulator MDM2 or by knockdown of MDM2 . The expression of most of the genes in the biomarker (75%) were found to depend on p53 activation status based on gene behavior after TP53 overexpression or knockdown. The biomarker identified DDI chemicals that were strong inducers of p53 in wild-type cells; these p53 responses were decreased or abolished in cells after p53 knockdown by siRNAs. Using the biomarker, we screened ∼1950 chemicals in ∼9800 human cell line chemical vs control comparisons and identified ∼100 chemicals that caused p53 activation. Among the positive chemicals were many that are known to activate p53 through direct and indirect DNA damaging mechanisms. These results contribute to the evidence that the TGx-DDI biomarker is useful for identifying chemicals that cause DDI and activate p53.
- Published
- 2019
- Full Text
- View/download PDF
36. Hepatic carboxylesterases are differentially regulated in PPARα-null mice treated with perfluorooctanoic acid.
- Author
-
Wen X, Baker AA, Klaassen CD, Corton JC, Richardson JR, and Aleksunes LM
- Subjects
- Animals, Carboxylesterase genetics, Cytochrome P450 Family 4 genetics, Cytochrome P450 Family 4 metabolism, Gene Expression Regulation, Enzymologic, Liver enzymology, Male, Mice, Inbred C57BL, Mice, Knockout, PPAR alpha agonists, PPAR alpha genetics, Caprylates toxicity, Carboxylesterase metabolism, Environmental Pollutants toxicity, Fluorocarbons toxicity, Liver drug effects, PPAR alpha deficiency
- Abstract
Hepatic carboxylesterases (Ces) catalyze the metabolism of drugs, environmental toxicants, and endogenous lipids and are known to be regulated by multiple nuclear receptors. Perfluorooctanoic acid (PFOA) is a synthetic fluorochemical that has been associated with dyslipidemia in exposed populations. In liver, PFOA can activate nuclear receptors such as PPARα, and alter the metabolism and excretion of chemicals. Here, we sought to test the ability of PFOA to modulate Ces expression and activity in the presence and absence of the PPARα receptor. For this purpose, male C57BL/6 NCrl mice were administered PFOA (1 or 3 mg/kg, po, 7 days) and livers collected for assessment of Ces expression and activity. PFOA increased Ces1 and 2 protein and activity. Notably, PFOA increased Ces1d, 1e, 1f, 1 g, 2c, and 2e mRNAs between 1.5- and 2.5-fold, while it decreased Ces1c and 2b. Activation of PPARα by PFOA was confirmed by up-regulation of Cyp4a14 mRNA. In a separate study of PFOA-treated wild-type (WT) and PPARα-null mice, induction of Ces 1e and 1f mRNA and in turn, Ces1 protein, was PPARα-dependent. Interestingly, in PPARα-null mice, Ces1c, 1d, 1 g, 2a, 2b, and 2e mRNAs and Ces2 protein were up-regulated by PFOA which contributed to sustained up-regulation of Ces activity, although to a lower extent than observed in WT mice. Activation of the CAR and PXR receptors likely accounted for up-regulation of select Ces1 and 2 subtypes in PPARα-null mice. In conclusion, the environmental contaminant PFOA modulates the expression and function of hepatic Ces enzymes, in part through PPARα., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
37. Assessment of the performance of the TGx-DDI biomarker to detect DNA damage-inducing agents using quantitative RT-PCR in TK6 cells.
- Author
-
Cho E, Buick JK, Williams A, Chen R, Li HH, Corton JC, Fornace AJ Jr, Aubrecht J, and Yauk CL
- Subjects
- Canada, Cell Line, DNA Damage drug effects, Gene Expression Profiling, Humans, Oligonucleotide Array Sequence Analysis, Transcriptome drug effects, Transcriptome genetics, DNA Damage genetics, Gene Expression drug effects, Genetic Markers, Mutagens toxicity
- Abstract
Gene expression biomarkers are now available for application in the identification of genotoxic hazards. The TGx-DDI transcriptomic biomarker can accurately distinguish DNA damage-inducing (DDI) from non-DDI exposures based on changes in the expression of 64 biomarker genes. The 64 genes were previously derived from whole transcriptome DNA microarray profiles of 28 reference agents (14 DDI and 14 non-DDI) after 4 h treatments of TK6 human lymphoblastoid cells. To broaden the applicability of TGx-DDI, we tested the biomarker using quantitative RT-PCR (qPCR), which is accessible to most molecular biology laboratories. First, we selectively profiled the expression of the 64 biomarker genes using TaqMan qPCR assays in 96-well arrays after exposing TK6 cells to the 28 reference agents for 4 h. To evaluate the classification capability of the qPCR profiles, we used the reference qPCR signature to classify 24 external validation chemicals using two different methods-a combination of three statistical analyses and an alternative, the Running Fisher test. The qPCR results for the reference set were comparable to the original microarray biomarker; 27 of the 28 reference agents (96%) were accurately classified. Moreover, the two classification approaches supported the conservation of TGx-DDI classification capability using qPCR; the combination of the two approaches accurately classified 21 of the 24 external validation chemicals, demonstrating 100% sensitivity, 81% specificity, and 91% balanced accuracy. This study demonstrates that qPCR can be used when applying the TGx-DDI biomarker and will improve the accessibility of TGx-DDI for genotoxicity screening. Environ. Mol. Mutagen. 60: 122-133, 2019. © 2018 Her Majesty the Queen in Right of Canada Environmental and Molecular Mutagenesis., (© 2018 Her Majesty the Queen in Right of Canada Environmental and Molecular Mutagenesis.)
- Published
- 2019
- Full Text
- View/download PDF
38. Chemical Activation of the Constitutive Androstane Receptor Leads to Activation of Oxidant-Induced Nrf2.
- Author
-
Rooney JP, Oshida K, Kumar R, Baldwin WS, and Corton JC
- Subjects
- Animals, Aryl Hydrocarbon Hydroxylases genetics, Aryl Hydrocarbon Hydroxylases metabolism, Biomarkers metabolism, Constitutive Androstane Receptor, Cytochrome P450 Family 2 genetics, Cytochrome P450 Family 2 metabolism, Enzyme Induction, Gene Expression drug effects, Mice, Inbred C57BL, Mice, Knockout, Microsomes, Liver metabolism, NF-E2-Related Factor 2 genetics, PPAR alpha genetics, PPAR alpha metabolism, Phenobarbital metabolism, Receptors, Cytoplasmic and Nuclear genetics, Steroid Hydroxylases genetics, Steroid Hydroxylases metabolism, Xenobiotics metabolism, Microsomes, Liver drug effects, NF-E2-Related Factor 2 metabolism, Oxidative Stress drug effects, Phenobarbital toxicity, Receptors, Cytoplasmic and Nuclear metabolism, Xenobiotics toxicity
- Abstract
Exposure to environmentally relevant chemicals that activate the xenobiotic receptors aryl hydrocarbon receptor (AhR), constitutive androstane receptor (CAR), and peroxisome proliferator-activated receptor alpha (PPARα) in rodent test systems often leads to increases in oxidative stress (OS) that contributes to liver cancer induction. We hypothesized that activation of the oxidant-induced transcription factor Nrf2 could be used as a surrogate endpoint for increases in OS. We examined the relationships between activation of xenobiotic receptors and Nrf2 using previously characterized gene expression biomarkers that accurately predict modulation. Using a correlation approach (Running Fisher Test), the biomarkers were compared with microarray profiles in a mouse liver gene expression compendium. Out of the 163 chemicals examined, 47% from 53 studies activated Nrf2. We found consistent coupling between CAR and Nrf2 activation. Out of the 41 chemicals from 32 studies that activated CAR, 90% also activated Nrf2. CAR was activated earlier and at lower doses than Nrf2, indicating CAR activation preceded Nrf2 activation. Nrf2 activation by 2 CAR activators was abolished in CAR-null mice. We hypothesized that Nrf2 is activated by reactive oxygen species from the increased activity of enzymes encoded by Cyp2b family members. However, Nrf2 was similarly activated in the livers of both TCPOBOP-treated wild-type and Cyp2b9/10/13-null mice. This study provides evidence that Nrf2 activation (1) often occurs after exposure to xenobiotic chemicals, (2) is tightly linked to activation of CAR, and (3) does not require induction of 3 Cyp2b genes secondary to CAR activation.
- Published
- 2019
- Full Text
- View/download PDF
39. Frequent Modulation of the Sterol Regulatory Element Binding Protein (SREBP) by Chemical Exposure in the Livers of Rats.
- Author
-
Corton JC
- Abstract
Inappropriate activation of sterol regulatory element-binding proteins (SREBPs) can lead to non-alcoholic fatty liver disease (NAFLD). To link chemical exposure to SREBP activity, a previously characterized gene expression biomarker (Rooney et al., 2019) was used to identify microarray comparisons from rat liver that exhibited significant positive or negative correlation to the biomarker. The effects of 620 chemicals on SREBP activity were examined across 9305 chemical-dose-time microarray comparisons. SREBP was found to be frequently modulated by chemical exposure with 54% of the chemicals affecting SREBP activity. Activators included inhibitors of cholesterogenesis that act to inhibit HMG-CoA reductase (statins) or inhibit Cyp51 (conazoles). Most chemical effects were transient, lasting usually no more than 2-4 days. Modulation of SREBP in most cases led to coordinated increases or decreases in lipogenic and cholesterogenic genes. However, 570 chemical exposure conditions were identified in which regulation was uncoupled. Most of these conditions affected cholesterogenic genes in the absence of parallel effects on lipogenic genes. Together, these findings show that SREBP is a frequent target of chemical exposure and expression of lipogenic and cholesterogenic genes can be uncoupled.
- Published
- 2019
- Full Text
- View/download PDF
40. Using a gene expression biomarker to identify DNA damage-inducing agents in microarray profiles.
- Author
-
Corton JC, Williams A, and Yauk CL
- Subjects
- Cell Line, Computational Biology methods, Hepatocytes, Humans, Reproducibility of Results, DNA Damage drug effects, Drug Discovery methods, Gene Expression Profiling methods, Genetic Markers, Mutagens pharmacology, Transcriptome
- Abstract
High-throughput transcriptomic technologies are increasingly being used to screen environmental chemicals in vitro to provide mechanistic context for regulatory testing. The TGx-DDI biomarker is a 64-gene expression profile generated from testing 28 model chemicals or treatments (13 that cause DNA damage and 15 that do not) in human TK6 cells. While the biomarker is very accurate at predicting DNA damage inducing (DDI) potential using the nearest shrunken centroid method, the broad utility of the biomarker using other computational methods is not fully known. Here, we determined the accuracy of the biomarker used with the Running Fisher test, a nonparametric correlation test. In TK6 cells, the methods could readily differentiate DDI and non-DDI compounds with balanced accuracies of 87-97%, depending on the threshold for determining DDI positives. The methods identified DDI agents in the metabolically competent hepatocyte cell line HepaRG (accuracy = 90%) but not in HepG2 cells or hepatocytes derived from embryonic stem cells (60 and 80%, respectively). DDI was also accurately classified when the gene expression changes were derived using the nCounter technology (accuracy = 89%). In addition, we found: (1) not all genes contributed equally to the correlations; (2) the minimal overlap in genes between the biomarker and the individual comparisons required for significant positive correlation was 10 genes, but usually was much higher; and (3) different sets of genes in the biomarker can by themselves contribute to the significant correlations. Overall, these results demonstrate the utility of the biomarker to accurately classify DDI agents. Environ. Mol. Mutagen. 59:772-784, 2018. Published 2018. This article is a U.S. Government work and is in the public domain in the USA., (Published 2018. This article is a U.S. Government work and is in the public domain in the USA.)
- Published
- 2018
- Full Text
- View/download PDF
41. Identification of Androgen Receptor Modulators in a Prostate Cancer Cell Line Microarray Compendium.
- Author
-
Rooney JP, Chorley B, Kleinstreuer N, and Corton JC
- Subjects
- Cell Line, Tumor, Gene Expression Profiling, High-Throughput Screening Assays, Humans, Male, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism, Androgen Receptor Antagonists toxicity, Androgens toxicity, Gene Expression Regulation, Neoplastic drug effects, Receptors, Androgen genetics, Transcriptome drug effects
- Abstract
High-throughput transcriptomic (HTTr) technologies are increasingly being used to screen environmental chemicals in vitro to identify molecular targets and provide mechanistic context for regulatory testing. Here, we describe the development and validation of a novel gene expression biomarker to identify androgen receptor (AR)-modulating chemicals using a pattern matching method. Androgen receptor biomarker genes were identified by their consistent expression after exposure to 4 AR agonists and 4 AR antagonists and included only those genes that were regulated by AR. The 51 gene biomarker was evaluated as a predictive tool using the fold-change, rank-based Running Fisher algorithm. Using 158 comparisons from cells treated with 95 chemicals, the biomarker gave balanced accuracies for prediction of AR activation or AR suppression of 97% or 98%, respectively. The biomarker correctly classified 16 out of the 17 AR reference antagonists including those that are "weak" and "very weak". Predictions based on microarray profiles from AR-positive LAPC-4 cells treated with 28 chemicals in antagonist mode were compared with those from an AR pathway model which used 11 in vitro HT assays. The balanced accuracy for suppression was 93%. Using our approach, we identified conditions in which AR was modulated in a large collection of microarray profiles from prostate cancer cell lines including (1) constitutively active mutants or knockdown of AR, (2) decreases in availability of androgens by castration or removal from media, and (3) exposure to chemical modulators that work through indirect mechanisms including suppression of AR expression. These results demonstrate that the AR gene expression biomarker could be a useful tool in HTTr to identify AR modulators.
- Published
- 2018
- Full Text
- View/download PDF
42. Adverse outcome pathway-driven identification of rat liver tumorigens in short-term assays.
- Author
-
Rooney J, Hill T 3rd, Qin C, Sistare FD, and Corton JC
- Subjects
- Animals, Biomarkers, Tumor metabolism, Body Weight drug effects, Carcinogens classification, DNA Damage drug effects, Gene Expression Regulation, Neoplastic drug effects, Liver Neoplasms, Experimental pathology, Organ Size drug effects, Rats, Rats, Sprague-Dawley, Adverse Outcome Pathways, Carcinogenicity Tests methods, Carcinogens toxicity, Liver Neoplasms, Experimental chemically induced
- Abstract
Chemicals induce liver cancer in rodents through well characterized adverse outcome pathways (AOPs). We hypothesized that measurement of molecular initiating events (MIEs) and downstream key events (KEs) in liver cancer AOPs in short-term assays will allow early identification of chemicals and their associated doses that are likely to be tumorigenic in the liver in two-year bioassays. We tested this hypothesis using the rat in vivo TG-GATES study data to measure MIEs (genotoxicity, cytotoxicity, AhR, CAR, ER, PPARα) and associated KEs (oxidative stress, cell proliferation, liver to body weights) across 77 chemicals that could be linked to doses with previously established effects on rat liver tumor induction. Gene expression biomarkers for MIEs generally considered to be rodent specific and human irrelevant (CAR, PPARα) and for MIEs that would be considered of greater risk at human relevant exposures (ER, AhR) were built using microarray comparisons from the livers of rats treated with prototypical activators of the receptors. The genotoxicity biomarker, also a potentially human relevant MIE, was comprised of 7 p53-responsive genes known to be induced upon DNA damage. The ability of the biomarkers to accurately predict MIE activation ranged from 91% to 98%. The Toxicological Priority Index (ToxPi) was used to rank chemicals based on their ability to activate MIEs/KEs. Chemicals administered at tumorigenic doses clearly gave the highest ranked scores. Our AOP-directed approach could be used in short term assays to identify chemicals and their doses that would be predicted to cause liver tumors in rats., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
43. Activation of Nrf2 in the liver is associated with stress resistance mediated by suppression of the growth hormone-regulated STAT5b transcription factor.
- Author
-
Rooney J, Oshida K, Vasani N, Vallanat B, Ryan N, Chorley BN, Wang X, Bell DA, Wu KC, Aleksunes LM, Klaassen CD, Kensler TW, and Corton JC
- Subjects
- Animals, Disease Resistance, Female, Hormones metabolism, Kelch-Like ECH-Associated Protein 1 deficiency, Kelch-Like ECH-Associated Protein 1 genetics, Male, Mice, Transgenic, NF-E2-Related Factor 2 genetics, Oxidants adverse effects, Sex Characteristics, Transcriptome, Liver metabolism, NF-E2-Related Factor 2 metabolism, Oxidative Stress physiology, STAT5 Transcription Factor metabolism
- Abstract
The transcription factor Nrf2 (encoded by Nfe2l2) induces expression of numerous detoxifying and antioxidant genes in response to oxidative stress. The cytoplasmic protein Keap1 interacts with and represses Nrf2 function. Computational approaches were developed to identify factors that modulate Nrf2 in a mouse liver gene expression compendium. Forty-eight Nrf2 biomarker genes were identified using profiles from the livers of mice in which Nrf2 was activated genetically in Keap1-null mice or chemically by a potent activator of Nrf2 signaling. The rank-based Running Fisher statistical test was used to determine the correlation between the Nrf2 biomarker genes and a test set of 81 profiles with known Nrf2 activation status demonstrating a balanced accuracy of 96%. For a large number of factors examined in the compendium, we found consistent relationships between activation of Nrf2 and feminization of the liver transcriptome through suppression of the male-specific growth hormone (GH)-regulated transcription factor STAT5b. The livers of female mice exhibited higher Nrf2 activation than male mice in untreated or chemical-treated conditions. In male mice, Nrf2 was activated by treatment with ethinyl estradiol, whereas in female mice, Nrf2 was suppressed by treatment with testosterone. Nrf2 was activated in 5 models of disrupted GH signaling containing mutations in Pit1, Prop1, Ghrh, Ghrhr, and Ghr. Out of 59 chemical treatments that activated Nrf2, 36 exhibited STAT5b suppression in the male liver. The Nrf2-STAT5b coupling was absent in in vitro comparisons of chemical treatments. Treatment of male and female mice with 11 chemicals that induce oxidative stress led to activation of Nrf2 to greater extents in females than males. The enhanced basal and inducible levels of Nrf2 activation in females relative to males provides a molecular explanation for the greater resistance often seen in females vs. males to age-dependent diseases and chemical-induced toxicity., Competing Interests: We have the following interests: Keiyu Oshida is employed by the commercial company, Toray Industries (https://www.toray.com/). There are no patents, products in development or marketed products to declare. This does not alter our adherence to all the PLOS ONE policies on sharing data and materials. Kai Wu was not employed by Janssen Pharmaceuticals (https://www.janssen.com/us/) at the time of the study, but is currently employed by the company.
- Published
- 2018
- Full Text
- View/download PDF
44. Human relevance of rodent liver tumors: Key insights from a Toxicology Forum workshop on nongenotoxic modes of action.
- Author
-
Felter SP, Foreman JE, Boobis A, Corton JC, Doi AM, Flowers L, Goodman J, Haber LT, Jacobs A, Klaunig JE, Lynch AM, Moggs J, and Pandiri A
- Subjects
- Animals, Constitutive Androstane Receptor, Humans, Liver metabolism, Liver pathology, Liver Neoplasms metabolism, PPAR alpha metabolism, Receptors, Cytoplasmic and Nuclear metabolism, Risk Assessment, Rodentia, Liver Neoplasms pathology
- Abstract
The Toxicology Forum sponsored a workshop in October 2016, on the human relevance of rodent liver tumors occurring via nongenotoxic modes of action (MOAs). The workshop focused on two nuclear receptor-mediated MOAs (Constitutive Androstane Receptor (CAR) and Peroxisome Proliferator Activated Receptor-alpha (PPARα), and on cytotoxicity. The goal of the meeting was to review the state of the science to (1) identify areas of consensus and differences, data gaps and research needs; (2) identify reasons for inconsistencies in current regulatory positions; and (3) consider what data are needed to demonstrate a specific MOA, and when additional research is needed to rule out alternative possibilities. Implications for quantitative risk assessment approaches were discussed, as were implications of not considering MOA and dose in hazard characterization and labeling schemes. Most, but not all, participants considered the CAR and PPARα MOAs as not relevant to humans based on quantitative and qualitative differences. In contrast, cytotoxicity is clearly relevant to humans, but a threshold applies. Questions remain for all three MOAs concerning what data are necessary to determine the MOA and to what extent it is necessary to exclude other MOAs., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
45. The PPARα-dependent rodent liver tumor response is not relevant to humans: addressing misconceptions.
- Author
-
Corton JC, Peters JM, and Klaunig JE
- Subjects
- Adverse Outcome Pathways, Animals, Apoptosis drug effects, Cell Proliferation drug effects, Diethylhexyl Phthalate toxicity, Dose-Response Relationship, Drug, Guinea Pigs, Hepatocytes drug effects, Hepatocytes metabolism, Hepatocytes pathology, Humans, Liver drug effects, Liver metabolism, Liver Neoplasms pathology, Macaca fascicularis, Mice, Oxidative Stress drug effects, Oxidative Stress physiology, Species Specificity, Liver Neoplasms chemically induced, Liver Neoplasms metabolism, PPAR alpha metabolism, Rodentia
- Abstract
A number of industrial chemicals and therapeutic agents cause liver tumors in rats and mice by activating the nuclear receptor peroxisome proliferator-activated receptor α (PPARα). The molecular and cellular events by which PPARα activators induce rodent hepatocarcinogenesis have been extensively studied elucidating a number of consistent mechanistic changes linked to the increased incidence of liver neoplasms. The weight of evidence relevant to the hypothesized mode of action (MOA) for PPARα activator-induced rodent hepatocarcinogenesis is summarized here. Chemical-specific and mechanistic data support concordance of temporal and dose-response relationships for the key events associated with many PPARα activators. The key events (KE) identified in the MOA are PPARα activation (KE1), alteration in cell growth pathways (KE2), perturbation of hepatocyte growth and survival (KE3), and selective clonal expansion of preneoplastic foci cells (KE4), which leads to the apical event-increases in hepatocellular adenomas and carcinomas (KE5). In addition, a number of concurrent molecular and cellular events have been classified as modulating factors, because they potentially alter the ability of PPARα activators to increase rodent liver cancer while not being key events themselves. These modulating factors include increases in oxidative stress and activation of NF-kB. PPARα activators are unlikely to induce liver tumors in humans due to biological differences in the response of KEs downstream of PPARα activation. This conclusion is based on minimal or no effects observed on cell growth pathways and hepatocellular proliferation in human primary hepatocytes and absence of alteration in growth pathways, hepatocyte proliferation, and tumors in the livers of species (hamsters, guinea pigs and cynomolgus monkeys) that are more appropriate human surrogates than mice and rats at overlapping dose levels. Despite this overwhelming body of evidence and almost universal acceptance of the PPARα MOA and lack of human relevance, several reviews have selectively focused on specific studies that, as discussed, contradict the consensus opinion and suggest uncertainty. In the present review, we systematically address these most germane suggested weaknesses of the PPARα MOA.
- Published
- 2018
- Full Text
- View/download PDF
46. From the Cover: Genomic Effects of Androstenedione and Sex-Specific Liver Cancer Susceptibility in Mice.
- Author
-
Rooney JP, Ryan N, Chorley BN, Hester SD, Kenyon EM, Schmid JE, George BJ, Hughes MF, Sey YM, Tennant A, MacMillan DK, Simmons JE, McQueen CA, Pandiri A, Wood CE, and Corton JC
- Subjects
- Animals, Biomarkers, Tumor metabolism, Cell Proliferation drug effects, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, Cytochrome P-450 Enzyme System genetics, Cytochrome P-450 Enzyme System metabolism, Dose-Response Relationship, Drug, Ethinyl Estradiol toxicity, Female, Gene Expression Regulation, Neoplastic, Genetic Predisposition to Disease, Liver metabolism, Liver pathology, Liver Neoplasms, Experimental metabolism, Liver Neoplasms, Experimental pathology, Male, Mice, Phenotype, Prednisone toxicity, STAT5 Transcription Factor genetics, STAT5 Transcription Factor metabolism, Sex Factors, Time Factors, Transcriptome, Androstenedione toxicity, Biomarkers, Tumor genetics, Cell Transformation, Neoplastic chemistry, Cell Transformation, Neoplastic genetics, Liver drug effects, Liver Neoplasms, Experimental chemically induced, Liver Neoplasms, Experimental genetics
- Abstract
Current strategies for predicting carcinogenic mode of action for nongenotoxic chemicals are based on identification of early key events in toxicity pathways. The goal of this study was to evaluate short-term key event indicators resulting from exposure to androstenedione (A4), an androgen receptor agonist and known liver carcinogen in mice. Liver cancer is more prevalent in men compared with women, but androgen-related pathways underlying this sex difference have not been clearly identified. Short-term hepatic effects of A4 were compared with reference agonists of the estrogen receptor (ethinyl estradiol, EE) and glucocorticoid receptor (prednisone, PRED). Male B6C3F1 mice were exposed for 7 or 28 days to A4, EE, or PRED. EE increased and PRED suppressed hepatocyte proliferation, while A4 had no detectable effects. In a microarray analysis, EE and PRED altered >3000 and >670 genes, respectively, in a dose-dependent manner, whereas A4 did not significantly alter any genes. Gene expression was subsequently examined in archival liver samples from male and female B6C3F1 mice exposed to A4 for 90 days. A4 altered more genes in females than males and did not alter expression of genes linked to activation of the mitogenic xenobiotic receptors AhR, CAR, and PPARα in either sex. A gene expression biomarker was used to show that in female mice, the high dose of A4 activated the growth hormone-regulated transcription factor STAT5b, which controls sexually dimorphic gene expression in the liver. These findings suggest that A4 induces subtle age-related effects on STAT5b signaling that may contribute to the higher risk of liver cancer in males compared with females., (Published by Oxford University Press on behalf of the Society of Toxicology 2017. This work is written by US Government employees and is in the public domain in the United States.)
- Published
- 2017
- Full Text
- View/download PDF
47. Evaluation of estrogen receptor alpha activation by glyphosate-based herbicide constituents.
- Author
-
Mesnage R, Phedonos A, Biserni M, Arno M, Balu S, Corton JC, Ugarte R, and Antoniou MN
- Subjects
- Benzhydryl Compounds pharmacology, Cell Line, Tumor, Dose-Response Relationship, Drug, Down-Regulation, Estradiol pharmacology, Gene Expression Regulation drug effects, Glycine administration & dosage, Glycine pharmacology, Humans, Oligonucleotide Array Sequence Analysis, Phenols pharmacology, Polyethylene Glycols pharmacology, Sequence Analysis, RNA, Transcriptome, Up-Regulation, Glyphosate, Estrogen Receptor alpha metabolism, Glycine analogs & derivatives, Herbicides pharmacology
- Abstract
The safety, including the endocrine disruptive capability, of glyphosate-based herbicides (GBHs) is a matter of intense debate. We evaluated the estrogenic potential of glyphosate, commercial GBHs and polyethoxylated tallowamine adjuvants present as co-formulants in GBHs. Glyphosate (≥10,000 μg/L or 59 μM) promoted proliferation of estrogen-dependent MCF-7 human breast cancer cells. Glyphosate also increased the expression of an estrogen response element-luciferase reporter gene (ERE-luc) in T47D-KBluc cells, which was blocked by the estrogen antagonist ICI 182,780. Commercial GBH formulations or their adjuvants alone did not exhibit estrogenic effects in either assay. Transcriptomics analysis of MCF-7 cells treated with glyphosate revealed changes in gene expression reflective of hormone-induced cell proliferation but did not overlap with an ERα gene expression biomarker. Calculation of glyphosate binding energy to ERα predicts a weak and unstable interaction (-4.10 kcal mol
-1 ) compared to estradiol (-25.79 kcal mol-1 ), which suggests that activation of this receptor by glyphosate is via a ligand-independent mechanism. Induction of ERE-luc expression by the PKA signalling activator IBMX shows that ERE-luc is responsive to ligand-independent activation, suggesting a possible mechanism of glyphosate-mediated activation. Our study reveals that glyphosate, but not other components present in GBHs, can activate ERα in vitro, albeit at relatively high concentrations., (Copyright © 2017 The Authors. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
48. Editor's Highlight: Transcriptome Profiling Reveals Bisphenol A Alternatives Activate Estrogen Receptor Alpha in Human Breast Cancer Cells.
- Author
-
Mesnage R, Phedonos A, Arno M, Balu S, Corton JC, and Antoniou MN
- Subjects
- Benzhydryl Compounds chemistry, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Cell Proliferation, Female, Genes, Reporter, Humans, Luciferases genetics, Oligonucleotide Array Sequence Analysis, Phenols chemistry, Benzhydryl Compounds toxicity, Breast Neoplasms metabolism, Estrogen Receptor alpha agonists, Gene Expression Profiling, Phenols toxicity, Transcriptome
- Abstract
Plasticizers with estrogenic activity, such as bisphenol A (BPA), have potential adverse health effects in humans. Due to mounting evidence of these health effects, BPA is being phased out and replaced by other bisphenol variants in "BPA-free" products. We have compared estrogenic activity of BPA with 6 bisphenol analogues [bisphenol S (BPS); bisphenol F (BPF); bisphenol AP (BPAP); bisphenol AF (BPAF); bisphenol Z (BPZ); bisphenol B (BPB)] in 3 human breast cancer cell lines. Estrogenicity was assessed (10-11-10-4 M) by cell growth in an estrogen receptor (ER)-mediated cell proliferation assay, and by the induction of estrogen response element-mediated transcription in a luciferase assay. BPAF was the most potent bisphenol, followed by BPB > BPZ ∼ BPA > BPF ∼ BPAP > BPS. The addition of ICI 182,780 antagonized the activation of ERs. Data mining of ToxCast high-throughput screening assays confirm our results but also show divergence in the sensitivities of the assays. Gene expression profiles were determined in MCF-7 cells by microarray analysis. The comparison of transcriptome profile alterations resulting from BPA alternatives with an ERα gene expression biomarker further indicates that all BPA alternatives act as ERα agonists in MCF-7 cells. These results were confirmed by Illumina-based RNA sequencing. In conclusion, BPA alternatives are not necessarily less estrogenic than BPA in human breast cancer cells. BPAF, BPB, and BPZ were more estrogenic than BPA. These findings point to the importance of better understanding the risk of adverse effects from exposure to BPA alternatives, including hormone-dependent breast cancer., (© The Author 2017. Published by Oxford University Press on behalf of the Society of Toxicology.)
- Published
- 2017
- Full Text
- View/download PDF
49. PPARα-independent transcriptional targets of perfluoroalkyl acids revealed by transcript profiling.
- Author
-
Rosen MB, Das KP, Rooney J, Abbott B, Lau C, and Corton JC
- Subjects
- Animals, Anticholesteremic Agents pharmacology, Chemical and Drug Induced Liver Injury genetics, Chemical and Drug Induced Liver Injury metabolism, Chemical and Drug Induced Liver Injury pathology, Computational Biology, Constitutive Androstane Receptor, Databases, Genetic, Estrogen Receptor alpha agonists, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Estrogens pharmacology, Fatty Acids, Gene Expression Regulation, Hepatomegaly chemically induced, Hepatomegaly genetics, Hepatomegaly metabolism, Hepatomegaly pathology, Humans, Liver metabolism, Liver pathology, Male, Mice, 129 Strain, Mice, Knockout, PPAR alpha deficiency, PPAR alpha genetics, PPAR gamma agonists, PPAR gamma genetics, PPAR gamma metabolism, Pyrimidines pharmacology, Receptors, Cytoplasmic and Nuclear agonists, Receptors, Cytoplasmic and Nuclear genetics, Receptors, Cytoplasmic and Nuclear metabolism, STAT5 Transcription Factor metabolism, Signal Transduction drug effects, Fluorocarbons toxicity, Gene Expression Profiling methods, Liver drug effects, Oligonucleotide Array Sequence Analysis, PPAR alpha agonists, Sulfonic Acids toxicity, Transcription, Genetic drug effects
- Abstract
Perfluoroalkyl acids (PFAAs) are ubiquitous and persistent environmental contaminants. Compounds such as perfluoroocanoic acid (PFOA), perfluorooctane sulfonate (PFOS), perfluorononanoic acid (PFNA), and perfluorohexane sulfonate (PFHxS) are readily found in the tissues of humans and wildlife. While PFOA and PFOS have been the subject of numerous studies since they were first described over a decade ago, less is known about the biological activity of PFHxS and PFNA. Most PFAAs are activators of peroxisome proliferator-activated receptor α (PPARα), although the biological effects of these compounds are likely mediated by other factors in addition to PPARα. To evaluate the effects of PFHxS and PFNA, male wild-type and Pparα-null mice were dosed by oral gavage with PFHxS (3 or 10mg/kg/day), PFNA (1 or 3mg/kg/day), or vehicle for 7days, and liver gene expression was evaluated by full-genome microarrays. Gene expression patterns were then compared to historical in-house data for PFOA and PFOS in addition to the experimental hypolipidemic agent, WY-14,643. While WY-14,643 altered most genes in a PPARα-dependent manner, approximately 11-24% of regulated genes in PFAA-treated mice were independent of PPARα. The possibility that PFAAs regulate gene expression through other molecular pathways was evaluated. Using data available through a microarray database, PFAA gene expression profiles were found to exhibit significant similarity to profiles from mouse tissues exposed to agonists of the constitutive activated receptor (CAR), estrogen receptor α (ERα), and PPARγ. Human PPARγ and ERα were activated by all four PFAAs in trans-activation assays from the ToxCast screening program. Predictive gene expression biomarkers showed that PFAAs activate CAR in both genotypes and cause feminization of the liver transcriptome through suppression of signal transducer and activator of transcription 5B (STAT5B). These results indicate that, in addition to activating PPARα as a primary target, PFAAs also have the potential to activate CAR, PPARγ, and ERα as well as suppress STAT5B., (Published by Elsevier B.V.)
- Published
- 2017
- Full Text
- View/download PDF
50. Compensatory changes in CYP expression in three different toxicology mouse models: CAR-null, Cyp3a-null, and Cyp2b9/10/13-null mice.
- Author
-
Kumar R, Mota LC, Litoff EJ, Rooney JP, Boswell WT, Courter E, Henderson CM, Hernandez JP, Corton JC, Moore DD, and Baldwin WS
- Subjects
- Animals, Aryl Hydrocarbon Hydroxylases metabolism, CRISPR-Cas Systems, Constitutive Androstane Receptor, Cytochrome P-450 CYP3A, Cytochrome P-450 Enzyme System metabolism, Cytochrome P450 Family 2 metabolism, Female, Gene Deletion, Gene Expression, Mice, Mice, Inbred C57BL, Mice, Knockout, Pharmacological and Toxicological Phenomena, Receptors, Cytoplasmic and Nuclear metabolism, Steroid Hydroxylases metabolism, Aryl Hydrocarbon Hydroxylases genetics, Cytochrome P-450 Enzyme System genetics, Cytochrome P450 Family 2 genetics, Receptors, Cytoplasmic and Nuclear genetics, Steroid Hydroxylases genetics
- Abstract
Targeted mutant models are common in mechanistic toxicology experiments investigating the absorption, metabolism, distribution, or elimination (ADME) of chemicals from individuals. Key models include those for xenosensing transcription factors and cytochrome P450s (CYP). Here we investigated changes in transcript levels, protein expression, and steroid hydroxylation of several xenobiotic detoxifying CYPs in constitutive androstane receptor (CAR)-null and two CYP-null mouse models that have subfamily members regulated by CAR; the Cyp3a-null and a newly described Cyp2b9/10/13-null mouse model. Compensatory changes in CYP expression that occur in these models may also occur in polymorphic humans, or may complicate interpretation of ADME studies performed using these models. The loss of CAR causes significant changes in several CYPs probably due to loss of CAR-mediated constitutive regulation of these CYPs. Expression and activity changes include significant repression of Cyp2a and Cyp2b members with corresponding drops in 6α- and 16β-testosterone hydroxylase activity. Further, the ratio of 6α-/15α-hydroxylase activity, a biomarker of sexual dimorphism in the liver, indicates masculinization of female CAR-null mice, suggesting a role for CAR in the regulation of sexually dimorphic liver CYP profiles. The loss of Cyp3a causes fewer changes than CAR. Nevertheless, there are compensatory changes including gender-specific increases in Cyp2a and Cyp2b. Cyp2a and Cyp2b were down-regulated in CAR-null mice, suggesting activation of CAR and potentially PXR following loss of the Cyp3a members. However, the loss of Cyp2b causes few changes in hepatic CYP transcript levels and almost no significant compensatory changes in protein expression or activity with the possible exception of 6α-hydroxylase activity. This lack of a compensatory response in the Cyp2b9/10/13-null mice is probably due to low CYP2B hepatic expression, especially in male mice. Overall, compensatory and regulatory CYP changes followed the order CAR-null > Cyp3a-null > Cyp2b-null mice.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.