Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Mutagenesis ; 35(6): 453-463, 2020 12 31.
Article in English | MEDLINE | ID: mdl-33399867

ABSTRACT

Chemicals in commerce or under development must be assessed for genotoxicity; assessment is generally conducted using validated assays (e.g. Tk mouse lymphoma assay) as part of a regulatory process. Currently, the MutaMouse FE1 cell mutagenicity assay is undergoing validation for eventual use as a standard in vitro mammalian mutagenicity assay. FE1 cells have been shown to be metabolically competent with respect to some cytochrome P450 (CYP) isozymes; for instance, they can convert the human carcinogen benzo[a]pyrene into its proximate mutagenic metabolite. However, some contradictory results have been noted for other genotoxic carcinogens that require two-step metabolic activation (e.g. 2-acetylaminofluorene and 2-amino-3-methylimidazo[4,5-f]quinoxaline). Here, we examined three known or suspected human carcinogens, namely acrylamide, 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) and 4-aminobiphenyl (4-ABP), together with their proximate metabolites (i.e. glycidamide, N-OH-PhIP and N-OH-4-ABP), to aid in the validation of the FE1 cell mutagenicity assay. Assessments of the parent compounds were conducted both in the presence and absence of an exogenous metabolic activation mixture S9; assessments of the metabolites were in the absence of S9. The most potent compound was N-OH-PhIP -S9, which elicited a mutant frequency (MF) level 5.3-fold over background at 5 µM. There was a 4.3-fold increase for PhIP +S9 at 5 µM, a 1.7-fold increase for glycidamide -S9 at 3.5 mM and a 1.5-fold increase for acrylamide +S9 at 4 mM. Acrylamide -S9 elicited a marginal 1.4-fold MF increase at 8 mM. Treatment with PhIP -S9, 4-ABP ±S9 and N-OH-4-ABP -S9 failed to elicit significant increases in lacZ MF with any of the treatment conditions tested. Gene expression of key CYP isozymes was quantified by RT-qPCR. Cyp1a1, 1a2 and 1b1 are required to metabolise PhIP and 4-ABP. Results showed that treatment with both compounds induced expression of Cyp1a1 and Cyp1b1 but not Cyp1a2. Cyp2e1, which catalyses the bioactivation of acrylamide to glycidamide, was not induced after acrylamide treatment. Overall, our results confirm that the FE1 cell mutagenicity assay has the potential for use alongside other, more traditional in vitro mutagenicity assays.


Subject(s)
Carcinogens, Environmental/pharmacology , Epithelial Cells/drug effects , Lung/drug effects , Mutagenesis/drug effects , Acrylamide/metabolism , Acrylamide/pharmacology , Acrylamide/toxicity , Animals , Carcinogens, Environmental/metabolism , Carcinogens, Environmental/toxicity , Cell Line , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1B1/genetics , Cytochrome P-450 CYP2E1/genetics , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Humans , Imidazoles/metabolism , Imidazoles/pharmacology , Imidazoles/toxicity , Lung/pathology , Metabolome/drug effects , Mice , Mutagenesis/genetics , Mutagenicity Tests , Quinoxalines/metabolism , Quinoxalines/pharmacology , Quinoxalines/toxicity
2.
Regul Toxicol Pharmacol ; 95: 75-90, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29475067

ABSTRACT

Acrylamide (AA) exposure causes increased incidence of forestomach, lung, and Harderian gland tumors in male mice. One hypothesized mode of action (MOA) for AA-carcinogenicity includes genotoxicity/mutagenicity as a key event, possibly resulting from AA metabolism to the direct genotoxic metabolite glycidamide. Alternatively, altered calcium signaling (CS) has been proposed as a central key event in the MOA. To examine the plausibility of these proposed MOAs, RNA-sequencing was performed on tumor target tissues: Harderian glands (the most sensitive tumor target tissue in the rodent 2-year cancer bioassay) and lungs of AA-exposed male CD-1 mice. Animals were exposed to 0.0, 1.5, 3.0, 6.0, 12.0, or 24.0 mg AA/kg bw-day in drinking water for 5, 15, or 31 days. We observed a pronounced effect on genes involved in CS and cytoskeletal processes in both tissues, but no evidence supporting a genotoxic MOA. Benchmark dose modeling suggests transcriptional points of departure (PODs) of 0.54 and 2.21 mg/kg bw-day for the Harderian glands and lungs, respectively. These are concordant with PODs of 0.17 and 1.27 mg/kg bw-day derived from the cancer bioassay data for these tissues in male mice, respectively. Overall, this study supports the involvement of CS in AA-induced mouse carcinogenicity, which is consistent with a recently proposed CS-based MOA in rat thyroid, and with other published reports of aberrant CS in malignant tumors in rodents and humans.


Subject(s)
Acrylamide/toxicity , Calcium Signaling/drug effects , Harderian Gland/drug effects , Lung/drug effects , Neoplasms/chemically induced , Neoplasms/genetics , Animals , Calcium Signaling/genetics , Gene Expression Profiling , Harderian Gland/metabolism , Lung/metabolism , Male , Mice , Neoplasms/metabolism , Sequence Analysis, RNA , Transcriptome
3.
Arch Toxicol ; 92(4): 1453-1469, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29275510

ABSTRACT

Experimental and/or epidemiological studies suggest that prenatal exposure to bisphenol A (BPA) may delay fetal lung development and maturation and increase the susceptibility to childhood respiratory disease. However, the underlying mechanisms remain to be elucidated. In our previous study with cultured human fetal lung fibroblasts (HFLF), we demonstrated that 24-h exposure to 1 and 100 µM BPA increased GPR30 protein in the nuclear fraction. Exposure to 100 µM BPA had no effects on cell viability, but increased cytoplasmic expression of ERß and release of GDF-15, as well as decreased release of IL-6, ET-1, and IP-10 through suppression of NFκB phosphorylation. By performing global gene expression and pathway analysis in this study, we identified molecular pathways, gene networks, and key molecules that were affected by 100, but not 0.01 and 1 µM BPA in HFLF. Using multiple genomic and proteomic tools, we confirmed these changes at both gene and protein levels. Our data suggest that 100 µM BPA increased CYP1B1 and HSD17B14 gene and protein expression and release of endogenous estradiol, which was associated with increased ROS production and DNA double-strand breaks, upregulation of genes and/or proteins in steroid synthesis and metabolism, and activation of Nrf2-regulated stress response pathways. In addition, BPA activated ATM-p53 signaling pathway, resulting in increased cell cycle arrest at G1 phase, senescence and autophagy, and decreased cell proliferation in HFLF. The results suggest that prenatal exposure to BPA at certain concentrations may affect fetal lung development and maturation, and thereby affecting susceptibility to childhood respiratory diseases.


Subject(s)
17-Hydroxysteroid Dehydrogenases/genetics , Air Pollutants, Occupational/toxicity , Benzhydryl Compounds/toxicity , Cytochrome P-450 CYP1B1/genetics , Estradiol/metabolism , Lung/drug effects , NF-E2-Related Factor 2/metabolism , Phenols/toxicity , Tumor Suppressor Protein p53/metabolism , 17-Hydroxysteroid Dehydrogenases/metabolism , Autophagy , Cell Cycle Checkpoints , Cellular Senescence/drug effects , Cytochrome P-450 CYP1B1/metabolism , Fibroblasts/drug effects , Humans , Reactive Oxygen Species/metabolism , Up-Regulation
4.
Food Chem Toxicol ; 107(Pt A): 186-200, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28606764

ABSTRACT

Acrylamide (AA) exposure in 2-year cancer bioassays leads to thyroid, but not liver, adenomas and adenocarcinomas in rats. Hypothesized modes of action (MOAs) include genotoxicity/mutagenicity, or thyroid hormone dysregulation. To examine the plausibility of these two or any alternative MOAs, RNA-sequencing was performed on the thyroids and livers of AA-exposed rats, in parallel with measurement of genotoxicity (blood micronucleus and Pig-a mutant frequency) and serum thyroid hormone levels, following the exposure of male Fischer 344/DuCrl rats to 0.0, 0.5, 1.5, 3.0, 6.0, or 12.0 mg AA/kg bw-day in drinking water for 5, 15, or 31 days. Differentially expressed genes in both tissues provided marginal support for hormonal and genotoxic MOAs, which was consistent with negative/equivocal genotoxicity assay and marginal changes in thyroid hormone levels. Instead, there was a pronounced effect on calcium signaling/cytoskeletal genes in the thyroid. Benchmark dose modeling of RNA-sequencing data for the calcium signaling pathway suggests a point of departure (POD) of 0.68 mg/kg bw-day, which is consistent with a POD of 0.82 mg/kg bw-day derived from the thyroid 2-year cancer bioassay data. Overall, this study suggests a novel MOA for AA-induced thyroid carcinogenicity in male rats centered around perturbation of calcium signaling.


Subject(s)
Acrylamide/toxicity , Calcium Signaling , Thyroid Neoplasms/etiology , Thyroid Neoplasms/genetics , Animals , Humans , Liver/drug effects , Liver/metabolism , Male , Rats , Rats, Inbred F344 , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyroid Hormones/blood , Thyroid Neoplasms/blood , Thyroid Neoplasms/metabolism , Transcription, Genetic
5.
Int J Toxicol ; 36(2): 124-132, 2017.
Article in English | MEDLINE | ID: mdl-28403741

ABSTRACT

Acrylamide (AA) at high exposure levels is neurotoxic, induces testicular toxicity, and increases dominant lethal mutations in rats. RNA-sequencing in testes was used to identify differentially expressed genes (DEG), explore AA-induced pathway perturbations that could contribute to AA-induced testicular toxicity and then used to derive a benchmark dose (BMD). Male F344/DuCrl rats were administered 0.0, 0.5, 1.5, 3.0, 6.0, or 12.0 mg AA/kg bw/d in drinking water for 5, 15, or 31 days. The experimental design used exposure levels that spanned and exceeded the exposure levels used in the rat dominant lethal, 2-generation reproductive toxicology, and cancer bioassays. The time of sample collection was based on previous studies that developed gene expression-based BMD. At 12.0 mg/kg, there were 38, 33, and 65 DEG ( P value <.005; fold change >1.5) in the testes after 5, 15, or 31 days of exposure, respectively. At 31 days, there was a dose-dependent increase in the number of DEG, and at 12.0 mg/kg/d the top three functional clusters affected by AA exposure were actin filament organization, response to calcium ion, and regulation of cell proliferation. The BMD lower 95% confidence limit using DEG ranged from 1.8 to 6.8 mg/kg compared to a no-observed-adverse-effect-level of 2.0 mg/kg/d for male reproductive toxicity. These results are consistent with the known effects of AA on calcium signaling and cytoskeletal actin filaments leading to neurotoxicity and suggest that AA can cause rat dominant lethal mutations by these same mechanisms leading to impaired chromosome segregation during cell division.


Subject(s)
Acrylamide/toxicity , Calcium Signaling/drug effects , Cytoskeleton/drug effects , Gene Expression Regulation/drug effects , Testis/drug effects , Animals , Dose-Response Relationship, Drug , Male , Rats, Inbred F344 , Testis/metabolism
6.
Arch Toxicol ; 91(5): 2045-2065, 2017 May.
Article in English | MEDLINE | ID: mdl-27928627

ABSTRACT

There is increasing interest in the use of quantitative transcriptomic data to determine benchmark dose (BMD) and estimate a point of departure (POD) for human health risk assessment. Although studies have shown that transcriptional PODs correlate with those derived from apical endpoint changes, there is no consensus on the process used to derive a transcriptional POD. Specifically, the subsets of informative genes that produce BMDs that best approximate the doses at which adverse apical effects occur have not been defined. To determine the best way to select predictive groups of genes, we used published microarray data from dose-response studies on six chemicals in rats exposed orally for 5, 14, 28, and 90 days. We evaluated eight approaches for selecting genes for POD derivation and three previously proposed approaches (the lowest pathway BMD, and the mean and median BMD of all genes). The relationship between transcriptional BMDs derived using these 11 approaches and PODs derived from apical data that might be used in chemical risk assessment was examined. Transcriptional BMD values for all 11 approaches were remarkably aligned with corresponding apical PODs, with the vast majority of toxicogenomics PODs being within tenfold of those derived from apical endpoints. We identified at least four approaches that produce BMDs that are effective estimates of apical PODs across multiple sampling time points. Our results support that a variety of approaches can be used to derive reproducible transcriptional PODs that are consistent with PODs produced from traditional methods for chemical risk assessment.


Subject(s)
Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Risk Assessment/methods , Toxicogenetics/methods , Animals , Bromobenzenes/administration & dosage , Bromobenzenes/toxicity , Chlorophenols/administration & dosage , Chlorophenols/toxicity , Female , Humans , Male , Nitrosamines/administration & dosage , Nitrosamines/toxicity , Rats, Inbred F344 , Rats, Sprague-Dawley , Transcriptome
7.
Environ Mol Mutagen ; 57(5): 350-63, 2016 06.
Article in English | MEDLINE | ID: mdl-27195522

ABSTRACT

Benzo[a]pyrene (BaP) is a genotoxic carcinogen and a neurotoxicant. The neurotoxicity of BaP is proposed to arise from either genotoxicity leading to neuronal cell death, or perturbed expression of N-methyl-d-aspartate receptor (NMDAR) subunits. To explore these hypotheses, we profiled hippocampal gene expression of adult male Muta(™) Mouse administered 0, 1, 35, or 70 mg BaP/kg bw per day by oral gavage for 3 days. Transcriptional profiles were examined by RNA-sequencing (RNA-seq), DNA microarrays, and real-time quantitative reverse transcription polymerase chain reaction (RT-PCR). BaP-DNA adducts in the cerebellum were quantified by (32) P-post-labeling to measure genotoxicity. RNA-seq revealed altered expression of 0, 260, and 219 genes (P-value < 0.05, fold-change ≥ ± 1.5) following exposure to the low, medium, and high doses, respectively; 54 genes were confirmed by microarrays. Microarray and RT-PCR analysis showed increased expression of NMDAR subunits Grina and Grin2a. In contrast, no effects on DNA-damage response genes were observed despite comparable BaP-DNA adduct levels in the cerebellum and in the lungs and livers of mice at similar BaP doses in previous studies. The results suggest that DNA-damage response does not play a major role in BaP-induced adult neurotoxicity. Meta-analysis revealed that BaP-induced transcriptional profiles are highly correlated with those from the hippocampus of transgenic mice exhibiting similar neurotoxicity outcomes to BaP-exposed mice and rats (i.e., defects in learning and memory). Overall, we suggest that BaP-induced neurotoxicity is more likely to be a consequence of NMDAR perturbation than genotoxicity, and identify other important genes potentially mediating this adverse outcome. Environ. Mol. Mutagen. 57:350-363, 2016. © 2016 Her Majesty the Queen in Right of Canada. Environmental and Molecular Mutagenesis © 2016 Environmental Mutagen Society.


Subject(s)
Benzo(a)pyrene/toxicity , DNA Damage , Hippocampus/drug effects , Neurotoxicity Syndromes/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Transcriptome , Animals , Benzo(a)pyrene/metabolism , DNA Adducts/metabolism , Dose-Response Relationship, Drug , Gene Expression Profiling , Hippocampus/metabolism , Male , Mice, Inbred Strains , Real-Time Polymerase Chain Reaction , Receptors, N-Methyl-D-Aspartate/metabolism
8.
Toxicol Sci ; 149(1): 251-68, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26496743

ABSTRACT

Dibenzo[def,p]chrysene (DBC) is the most carcinogenic polycyclic aromatic hydrocarbon (PAH) examined to date. We investigated the immunotoxicity of DBC, manifested as spleen atrophy, following acute exposure of adult MutaMouse males by oral gavage. Mice were exposed to 0, 2.0, 6.2, or 20.0 mg DBC /kg-bw per day, for 3 days. Genotoxic endpoints (DBC-DNA adducts and lacZ mutant frequency in spleen and bone marrow, and red blood cell micronucleus frequency) and global gene expression changes were measured. All of the genotoxicity measures increased in a dose-dependent manner in spleen and bone marrow. Gene expression analysis showed that DBC activates p53 signaling pathways related to cellular growth and proliferation, which was evident even at the low dose. Strikingly, the expression profiles of DBC exposed mouse spleens were highly inversely correlated with the expression profiles of the only published toxicogenomics dataset of enlarged mouse spleen. This analysis suggested a central role for Bnip3l, a pro-apoptotic protein involved in negative regulation of erythroid maturation. RT-PCR confirmed expression changes in several genes related to apoptosis, iron metabolism, and aryl hydrocarbon receptor signaling that are regulated in the opposite direction during spleen atrophy versus benzo[a]pyrene-mediated splenomegaly. In addition, benchmark dose modeling of toxicogenomics data yielded toxicity estimates that are very close to traditional toxicity endpoints. This work illustrates the power of toxicogenomics to reveal rich mechanistic information for immunotoxic compounds and its ability to provide information that is quantitatively similar to that derived from standard toxicity methods in health risk assessment.


Subject(s)
Benzopyrenes/toxicity , Carcinogens/toxicity , Gene Expression Profiling , Spleen/drug effects , Animals , Atrophy/chemically induced , Benchmarking , Benzopyrenes/metabolism , Bone Marrow/drug effects , Bone Marrow/metabolism , DNA Adducts/analysis , Dose-Response Relationship, Drug , Male , Membrane Proteins/analysis , Mice , Mice, Transgenic , Mitochondrial Proteins/analysis , Organ Specificity , Reticulocytes/drug effects , Reticulocytes/ultrastructure , Sequence Analysis, RNA , Spleen/metabolism , Spleen/pathology , Toxicogenetics
9.
J Vis Exp ; (102): e52697, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26273842

ABSTRACT

Oxidative stress is associated with many physiological and pathological processes, as well as xenobiotic metabolism, leading to the oxidation of biomacromolecules, including DNA. Therefore, efficient detection of DNA oxidation is important for a variety of research disciplines, including medicine and toxicology. A common biomarker of oxidatively damaged DNA is 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxo-dGuo; often erroneously referred to as 8-hydroxy-2'-deoxyguanosine (8-OH-dGuo or 8-oxo-dG)). Several protocols for 8-oxo-dGuo measurement by high pressure liquid chromatography with electrochemical detection (HPLC-ED) have been described. However, these were mainly applied to purified DNA treated with pro-oxidants. In addition, due to methodological differences between laboratories, mainly due to differences in analytical equipment, the adoption of published methods for detection of 8-oxo-dGuo by HPLC-ED requires careful optimization by each laboratory. A comprehensive protocol, describing such an optimization process, is lacking. Here, a detailed protocol is described for the detection of 8-oxo-dGuo by HPLC-ED, in DNA from cultured cells or animal tissues. It illustrates how DNA sample preparation can be easily and rapidly optimized to minimize undesirable DNA oxidation that can occur during sample preparation. This protocol shows how to detect 8-oxo-dGuo in cultured human alveolar adenocarcinoma cells (i.e., A549 cells) treated with the oxidizing agent KBrO3, and from the spleen of mice exposed to the polycyclic aromatic hydrocarbon dibenzo(def,p)chrysene (DBC, formerly known as dibenzo(a,l)pyrene, DalP). Overall, this work illustrates how an HPLC-ED methodology can be readily optimized for the detection of 8-oxo-dGuo in biological samples.


Subject(s)
Chromatography, High Pressure Liquid/methods , DNA/chemistry , Deoxyguanosine/analogs & derivatives , 8-Hydroxy-2'-Deoxyguanosine , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cell Line , Cells, Cultured , DNA/metabolism , DNA Damage , Deoxyguanosine/analysis , Deoxyguanosine/metabolism , Humans , Male , Mice , Oxidants/chemistry , Oxidants/pharmacology , Oxidation-Reduction , Spleen/chemistry , Spleen/drug effects
10.
Biochim Biophys Acta ; 1849(10): 1260-76, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26254094

ABSTRACT

The "cap'n'collar" bZIP transcription factor Nrf1 heterodimerizes with small Maf proteins to bind to the Antioxidant Response Element/Electrophile Response Element to transactivate antioxidant enzyme, phase 2 detoxification enzyme and proteasome subunit gene expression. Nrf1 specifically regulates pathways in lipid metabolism, amino acid metabolism, proteasomal degradation, the citric acid cycle, and the mitochondrial respiratory chain. Nrf1 is maintained in the endoplasmic reticulum (ER) in an inactive glycosylated state. Activation involves retrotranslocation from the ER lumen to the cytoplasm, deglycosylation and partial proteolytic processing to generate the active forms of Nrf1. Recent evidence has revealed how this factor is regulated and its involvement in various metabolic diseases. This review outlines Nrf1 structure, function, regulation and its links to insulin resistance, diabetes and inflammation. The glycosylation/deglycosylation of Nrf1 is controlled by glucose levels. Nrf1 glycosylation affects its control of glucose transport, glycolysis, gluconeogenesis and lipid metabolism.


Subject(s)
Diabetes Mellitus/genetics , Glucose/metabolism , Inflammation/genetics , Nuclear Respiratory Factor 1/genetics , Amino Acids/genetics , Amino Acids/metabolism , Citric Acid Cycle/genetics , Diabetes Mellitus/metabolism , Glycosylation , Humans , Inflammation/metabolism , Insulin Resistance/genetics , Lipid Metabolism/genetics , Nuclear Respiratory Factor 1/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis
11.
Mutat Res Rev Mutat Res ; 764: 64-89, 2015.
Article in English | MEDLINE | ID: mdl-26041267

ABSTRACT

Benzo[a]pyrene (BaP) is a well-studied environmental compound that requires metabolic activation to have a carcinogenic effect. The neurotoxicity of BaP has received considerably less attention than its carcinogenicity. Environmental exposure to BaP correlates with impaired learning and memory in adults, and poor neurodevelopment in children. We carried out a comprehensive literature review to examine the neurotoxicity of BaP. The data were used to identify potential point of departure (POD) values for cancer and neurotoxicity endpoints using benchmark dose (BMD) modelling to compare the utility of both endpoints in the risk assessment of BaP. The POD for neurotoxicity in rodents, based on a standard behavioural test (Morris water maze), was 0.025 mg BaP/kg-bw-day compared to 0.54 mg BaP/kg-bw-day for rodent forestomach carcinogenicity, suggesting that neurotoxic endpoints are more sensitive than cancer endpoints for health risks associated with BaP exposure. Using the limited number of published studies on this topic, we propose a preliminary mode of action (MOA) to explain BaP-induced neurotoxicity in rodents. The MOA includes: (1) BaP binding to the aryl hydrocarbon receptor (AHR); (2) AHR-dependent modulation of the transcription of N-methyl-d-aspartate glutamate receptor (NMDAR) subunits; (3) NMDAR-mediated loss of neuronal activity and decreased long-term potentiation; and (4) compromised learning and memory. More data are needed to explore the proposed neurotoxic MOA. In addition, we consider alternative MOAs, including the hypothesis that BaP-mediated DNA damage may lead to either carcinogenicity or neurotoxicity, depending on the tissue. Our proposed MOA is intended to serve as a basis for hypothesis testing in future studies. We emphasise that further studies are needed to validate the proposed MOA, to evaluate its human relevance, and to explore other potential mechanisms of BaP neurotoxicity.


Subject(s)
Benzo(a)pyrene/toxicity , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/pathology , Animals , DNA Damage , Dose-Response Relationship, Drug , Humans , Learning/drug effects , Neoplasms/chemically induced , Neurotoxicity Syndromes/metabolism
12.
Crit Rev Toxicol ; 45(1): 44-52, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25605027

ABSTRACT

The use of short-term toxicogenomic tests to predict cancer (or other health effects) offers considerable advantages relative to traditional toxicity testing methods. The advantages include increased throughput, increased mechanistic data, and significantly reduced costs. However, precisely how toxicogenomics data can be used to support human health risk assessment (RA) is unclear. In a companion paper ( Moffat et al. 2014 ), we present a case study evaluating the utility of toxicogenomics in the RA of benzo[a]pyrene (BaP), a known human carcinogen. The case study is meant as a proof-of-principle exercise using a well-established mode of action (MOA) that impacts multiple tissues, which should provide a best case example. We found that toxicogenomics provided rich mechanistic data applicable to hazard identification, dose-response analysis, and quantitative RA of BaP. Based on this work, here we share some useful lessons for both research and RA, and outline our perspective on how toxicogenomics can benefit RA in the short- and long-term. Specifically, we focus on (1) obtaining biologically relevant data that are readily suitable for establishing an MOA for toxicants, (2) examining the human relevance of an MOA from animal testing, and (3) proposing appropriate quantitative values for RA. We describe our envisioned strategy on how toxicogenomics can become a tool in RA, especially when anchored to other short-term toxicity tests (apical endpoints) to increase confidence in the proposed MOA, and emphasize the need for additional studies on other MOAs to define the best practices in the application of toxicogenomics in RA.


Subject(s)
Benzo(a)pyrene/toxicity , Risk Assessment/methods , Toxicogenetics/methods , Animals , Carcinogens/toxicity , Dose-Response Relationship, Drug , Humans , Neoplasms/chemically induced , Toxicity Tests
13.
J Appl Toxicol ; 34(11): 1115-21, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25244189

ABSTRACT

Reliable quantification of gene and protein expression has potential to contribute significantly to the characterization of hypothesized modes of action (MOA) or adverse outcome pathways for critical effects of toxicants. Quantitative analysis of gene expression by benchmark dose (BMD) modeling has been facilitated by the development of effective software tools. In contrast, protein expression is still generally quantified by a less robust effect level (no or lowest [adverse] effect levels) approach, which minimizes its potential utility in the consideration of dose-response and temporal concordance for key events in hypothesized MOAs. BMD modeling is applied here to toxicological data on testicular toxicity to investigate its potential utility in analyzing protein expression relevant to the proposed MOA to inform human health risk assessment. The results illustrate how the BMD analysis of protein expression in animal tissues in response to toxicant exposure: (1) complements other toxicity data, and (2) contributes to consideration of the empirical concordance of dose-response relationships, as part of the weight of evidence for hypothesized MOAs to facilitate consideration and application in regulatory risk assessment. Lack of BMD analysis in proteomics has likely limited its use for these purposes. This paper illustrates the added value of BMD modeling to support and strengthen hypothetical MOAs as a basis to facilitate the translation and uptake of the results of proteomic research into risk assessment.


Subject(s)
Environmental Pollutants/toxicity , Gene Expression , Proteomics , Testis/drug effects , Animals , Databases, Factual , Dose-Response Relationship, Drug , Humans , Male , Mice , Models, Theoretical , Rats , Risk Assessment/methods , Testis/pathology
14.
Redox Biol ; 1: 183-9, 2013.
Article in English | MEDLINE | ID: mdl-24024152

ABSTRACT

Although the Nrf2 (nuclear factor-erythroid 2 p45 subunit-related factor 2) regulated expression of multiple antioxidant and cytoprotective genes through the electrophile responsive element (EpRE) is well established, interaction of Nrf2/EpRE with Nrf1, a closely-related transcription factor, is less well understood. Due to either proteolysis or alternative translation, Nrf1 has been found as proteins of varying size, p120, p95, and p65, which have been described as either activators of EpRE or competitive inhibitors of Nrf2. We investigated the effect of Nrf1 on EpRE-regulated gene expression using the catalytic and modifier subunits of glutamate cysteine ligase (GCLC and GCLM) as models and explored the potential role of Nrf1 in altering their expression in aging and upon chronic exposure to airborne nano-sized particulate matter (nPM). Nrf1 knockout resulted in the increased expression of GCLC and GCLM in human bronchial epithelial (HBE1) cells. Overexpression Nrf2 in combination with either p120 or p65 diminished or failed to further increase the GCLC- and GLCM-EpRE luciferase activity. All known forms of Nrf1 protein, remained unchanged in the lungs of mice with age or in response to nPM. Our study shows that Nrf1 could inhibit EpRE activity in vitro, whereas the precise role of Nrf1 in vivo requires further investigations. We conclude that Nrf1 may not be directly responsible for the loss of Nrf2-dependent inducibility of antioxidant and cytoprotective genes observed in aged animals.


Subject(s)
Bronchi/metabolism , Glutamate-Cysteine Ligase/genetics , NF-E2 Transcription Factor, p45 Subunit/genetics , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Aging/genetics , Animals , Antioxidants/metabolism , Bronchi/cytology , Cell Line , Epithelial Cells/metabolism , Gene Expression Regulation , Gene Knockout Techniques , Glutamate-Cysteine Ligase/metabolism , HEK293 Cells , Humans , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , NF-E2 Transcription Factor, p45 Subunit/metabolism , Particulate Matter/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Response Elements
15.
Chem Res Toxicol ; 26(3): 498-506, 2013 Mar 18.
Article in English | MEDLINE | ID: mdl-23360430

ABSTRACT

Bisphenol A (BPA) is used in the production of polycarbonate plastics and epoxy resins for baby bottles, liners of canned food, and many other consumer products. Previously, BPA has been shown to reduce the activity of several antioxidant enzymes, which may contribute to oxidative stress. However, the underlying mechanism of the BPA-mediated effect upon antioxidant enzyme activity is unknown. Antioxidant and phase II metabolizing enzymes protect cells from oxidative stress and are transcriptionally activated by Nrf1 and Nrf2 factors through their cis-regulatory antioxidant response elements (AREs). In this work, we have assessed the effect of BPA on the Nrf1/2-ARE pathway in cultured human embryonic kidney (HEK) 293 cells. Surprisingly, glutathione and reactive oxygen species (ROS) assays revealed that BPA application created a more reduced intracellular environment in cultured HEK 293 cells. Furthermore, BPA increased the transactivation activity of ectopic Nrf1 and Nrf2 and increased the expression of ARE-target genes ho-1 and nqo1 at high (100-200 µM) BPA concentrations only. Our study suggests that BPA activates the Nrf1/2-ARE pathway at high (>10 µM) micromolar concentrations.


Subject(s)
Air Pollutants, Occupational/metabolism , Antioxidants/metabolism , Benzhydryl Compounds/metabolism , NF-E2-Related Factor 2/metabolism , Nuclear Respiratory Factor 1/metabolism , Phenols/metabolism , Signal Transduction/drug effects , Glutathione/metabolism , HEK293 Cells , Humans , Reactive Oxygen Species/metabolism
16.
Free Radic Biol Med ; 50(6): 645-66, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21185934

ABSTRACT

Constituting an integral part of a heme's porphyrin ring, iron is essential for supplying cells and tissues with oxygen. Given tight links between oxygen delivery and iron availability, it is not surprising that iron deprivation and oxygen deprivation (hypoxia) have very similar consequences at the molecular level. Under hypoxia, the expression of major iron homeostasis genes including transferrin, transferrin receptor, ceruloplasmin, and heme oxygenase-1 is activated by hypoxia-inducible factors to provide increased iron availability for erythropoiesis in an attempt to enhance oxygen uptake and delivery to hypoxic cells. Iron-response proteins (IRP1 and IRP2) and "cap-n-collar" bZIP transcriptional factors (NE-F2 p45; Nrf1, 2, and 3; Bach1 and 2) also control gene and protein expression of the key iron homeostasis proteins. In this article, we give an overview of the mechanisms by which iron pathways are regulated by hypoxia at multiple levels. In addition, potential clinical benefits of manipulating iron pathways in the hypoxia-related conditions anemia and ischemia are discussed.


Subject(s)
Cell Hypoxia , Hypoxia/metabolism , Iron Deficiencies , Iron/metabolism , Oxygen , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Ceruloplasmin/genetics , Erythropoiesis , Gene Expression , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Iron-Regulatory Proteins/genetics , Iron-Regulatory Proteins/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Transferrin/genetics , Transferrin/metabolism
17.
PLoS One ; 6(12): e29167, 2011.
Article in English | MEDLINE | ID: mdl-22216197

ABSTRACT

BACKGROUND: Nrf1 (nuclear factor-erythroid 2 p45 subunit-related factor 1) is a transcription factor mediating cellular responses to xenobiotic and pro-oxidant stress. Nrf1 regulates the transcription of many stress-related genes through the electrophile response elements (EpREs) located in their promoter regions. Despite its potential importance in human health, the mechanisms controlling Nrf1 have not been addressed fully. PRINCIPAL FINDINGS: We found that proteasomal inhibitors MG-132 and clasto-lactacystin-ß-lactone stabilized the protein expression of full-length Nrf1 in both COS7 and WFF2002 cells. Concomitantly, proteasomal inhibition decreased the expression of a smaller, N-terminal Nrf1 fragment, with an approximate molecular weight of 23 kDa. The EpRE-luciferase reporter assays revealed that proteasomal inhibition markedly inhibited the Nrf1 transactivational activity. These results support earlier hypotheses that the 26 S proteasome processes Nrf1 into its active form by removing its inhibitory N-terminal domain anchoring Nrf1 to the endoplasmic reticulum. Immunoprecipitation demonstrated that Nrf1 is ubiquitinated and that proteasomal inhibition increased the degree of Nrf1 ubiquitination. Furthermore, Nrf1 protein had a half-life of approximately 5 hours in COS7 cells. In contrast, hypoxia (1% O(2)) significantly increased the luciferase reporter activity of exogenous Nrf1 protein, while decreasing the protein expression of p65, a shorter form of Nrf1, known to act as a repressor of EpRE-controlled gene expression. Finally, the protein phosphatase inhibitor okadaic acid activated Nrf1 reporter activity, while the latter was repressed by the PKC inhibitor staurosporine. CONCLUSIONS: Collectively, our data suggests that Nrf1 is controlled by several post-translational mechanisms, including ubiquitination, proteolytic processing and proteasomal-mediated degradation as well as by its phosphorylation status.


Subject(s)
Cell Hypoxia , Nuclear Respiratory Factor 1/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Phosphorylation , Proteolysis , Ubiquitination
18.
J Enzyme Inhib Med Chem ; 24(6): 1319-31, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19795928

ABSTRACT

In mammals, aging is linked to a decline in the activity of citrate synthase (CS; E.C. 2.3.3.1), the first enzyme of the citric acid cycle. We used 2,2'-azobis(2-amidinopropane) dihydrochloride (AAPH), a water-soluble generator of peroxyl and alkoxyl radicals, to investigate the susceptibility of CS to oxidative damage. Treatment of isolated mitochondria with AAPH for 8-24 h led to CS inactivation; however, the activity of aconitase, a mitochondrial enzyme routinely used as an oxidative stress marker, was unaffected. In addition to enzyme inactivation, AAPH treatment of purified CS resulted in dityrosine formation, increased protein surface hydrophobicity, and loss of tryptophan fluorescence. Propyl gallate, 1,8-naphthalenediol, 2,3-naphthalenediol, ascorbic acid, glutathione, and oxaloacetate protected CS from AAPH-mediated inactivation, with IC(50) values of 9, 14, 34, 37, 150, and 160 muM, respectively. Surprisingly, the antioxidant epigallocatechin gallate offered no protection against AAPH, but instead caused CS inactivation. Our results suggest that the current practice of using the enzymatic activity of CS as an index of mitochondrial abundance and the use of aconitase activity as an oxidative stress marker may be inappropriate, especially in oxidative stress-related studies, during which alkyl peroxyl and alkoxyl radicals can be generated.


Subject(s)
Antioxidants/pharmacology , Citrate (si)-Synthase/metabolism , Oxidants/pharmacology , Oxidative Stress/drug effects , Peroxides/pharmacology , Aconitate Hydratase/antagonists & inhibitors , Aconitate Hydratase/metabolism , Amidines/pharmacology , Amidines/toxicity , Ascorbic Acid/pharmacology , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/toxicity , Citrate (si)-Synthase/antagonists & inhibitors , Enzyme Activation , Glutathione/pharmacology , Inhibitory Concentration 50 , Mitochondria/enzymology , Naphthols/pharmacology , Oxaloacetic Acid/pharmacology , Oxidants/toxicity , Oxidation-Reduction/drug effects , Oxidative Stress/physiology , Peroxides/toxicity , Propyl Gallate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...