1
|
Meaza I, Cahill CR, Speer RM, Kouokam JC, Wise JP. Particulate hexavalent chromium inhibits global transcription of genes in DNA repair pathways, particularly targeting homologous recombination repair, base excision repair, mismatch repair and microhomology-mediated end-joining. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136892. [PMID: 39706010 PMCID: PMC11794018 DOI: 10.1016/j.jhazmat.2024.136892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Hexavalent chromium [Cr(VI)] is a human lung carcinogen with widespread exposure. How Cr(VI) causes cancer is poorly understood, but chromosome instability plays a central role. Inhibition of DNA repair pathways leads to chromosome instability; however, despite the importance of these pathways in the mechanism of Cr(VI)-induced lung carcinogenesis, there are no data considering in-depth analysis on the transcriptional changes of genes involved in them. This study characterized the global transcriptional changes of mRNA expression after Cr(VI) exposure focusing on DNA repair pathways. The repair pathways considered included homologous recombination repair, non-homologous end joining, microhomology-directed end-joining, single strand annealing, mismatch repair, base excision repair, nucleotide excision repair and crosslink repair. Normal human lung fibroblast cells were exposed to increasing zinc chromate concentrations for 24, 72 or 120 h then RNA was extracted and sequenced. Our results indicate Cr(VI) causes differential expression of genes in lung cancer pathways and downregulates expression of some genes in all 8 DNA repair pathways. Homologous recombination repair, mismatch repair, base excision repair and microhomology-directed end-joining were the most affected pathways. This study provides a critical in-depth analysis of the effects of Cr(VI) on DNA repair pathways and contributes new insights into the mechanism of Cr(VI)-carcinogenesis.
Collapse
Affiliation(s)
- Idoia Meaza
- Wise Laboratory for Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States
| | - Caitlin R Cahill
- Wise Laboratory for Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States
| | - Rachel M Speer
- Wise Laboratory for Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States
| | - J Calvin Kouokam
- Wise Laboratory for Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States
| | - John Pierce Wise
- Wise Laboratory for Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, 500 S Preston Street, Building 55A, Room 1422, Louisville, KY 40292, United States.
| |
Collapse
|
2
|
Hossini H, Shafie B, Niri AD, Nazari M, Esfahlan AJ, Ahmadpour M, Nazmara Z, Ahmadimanesh M, Makhdoumi P, Mirzaei N, Hoseinzadeh E. A comprehensive review on human health effects of chromium: insights on induced toxicity. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:70686-70705. [PMID: 36042133 DOI: 10.1007/s11356-022-22705-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/20/2022] [Indexed: 05/13/2023]
Abstract
The growing use of heavy metals in most industrial activities has led to it being considered as the most important environmental pollutant that may cause harm and toxicity to animals and humans. Chromium has been found in the environment in different oxidation states such as Cr0, Cr(III), and Cr(VI) and is released from a variety of anthropogenic and natural activities. At among, trivalent and hexavalent chromium are the most stable forms. Considerably, Cr(VI) is frequently more toxic than Cr(III) because of its particular solubility and high mobility. Chronic exposure and bioaccumulation of chromium, as a heavy metal, can cause toxicity and numerous pathophysiological defects, including allergic reactions, anemia, burns, and sores especially in the stomach and small intestine, damage to sperm along with the male reproductive system, and affect various biological systems. Chromium pollution can have severe consequences for water and the soil environment. This article reviews the toxicological effects of Cr(VI) and Cr(III) and their mechanisms of toxicity and carcinogenicity.
Collapse
Affiliation(s)
- Hooshyar Hossini
- Department of Environmental Health Engineering, Faculty of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Behnaz Shafie
- Food and Drug Administration, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Dehghan Niri
- Department of Occupational Health Engineering, Shahid Sadoughi University of Medical Science, Yazd, Iran
| | - Mahboubeh Nazari
- Department of Medical and Surgical Nursing, School of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aylin Jahanban Esfahlan
- Department of Nursing, School of Nursing and Midwifery, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mohammad Ahmadpour
- Department of Public Health, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Zohreh Nazmara
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahnaz Ahmadimanesh
- Food and Drug Vice Presidency, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pouran Makhdoumi
- Student research committee, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Nezam Mirzaei
- Department of Environmental Health Engineering, Social Determinants of Health (SDH) Research Center, Kashan University of Medical Sciences, Kashan, Iran.
| | - Edris Hoseinzadeh
- Incubation and Innovation center, Saveh University of Medical Sciences, Saveh, Iran
| |
Collapse
|
3
|
Chappell GA, Wolf JC, Thompson CM. Crypt and Villus Transcriptomic Responses in Mouse Small Intestine Following Oral Exposure to Hexavalent Chromium. Toxicol Sci 2022; 186:43-57. [PMID: 34935971 PMCID: PMC8883354 DOI: 10.1093/toxsci/kfab152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Oral exposure to hexavalent chromium (Cr(VI)) induces tumors in the mouse duodenum. Previous microarray-based transcriptomic analyses of homogenized mouse duodenal tissue have demonstrated Cr(VI)-induced alterations in various cellular pathways and processes. However, X-ray fluorescence microscopy indicates that chromium localizes primarily to the duodenal villi following exposure to Cr(VI), suggesting that previous transcriptomic analyses of homogenized tissue provide an incomplete picture of transcriptomic responses in the duodenum. Herein, transcriptomic analyses were conducted separately on crypt and villus tissue from formalin-fixed paraffin-embedded transverse duodenal sections from the same study in which microarray-based analyses were previously conducted. A total of 28 groups (7 doses × 2 timepoints × 2 tissue compartments) were analyzed for differential gene expression, dose-response, and gene set enrichment. Tissue compartment isolation was confirmed by differences in expression of typical markers of crypt and villus compartments. Fewer than 21 genes were altered in the crypt compartment of mice exposed to 0.1-5 ppm Cr(VI) for 7 or 90 days, which increased to hundreds or thousands of genes at ≥20 ppm Cr(VI). Consistent with histological evidence for crypt proliferation, a significant, dose-dependent increase in genes that regulate mitotic cell cycle was prominent in the crypt, while subtle in the villus, when compared with samples from time-matched controls. Minimal transcriptomic evidence of DNA damage response in either the crypts or the villi is consistent with published in vivo genotoxicity data. These results are also discussed in the context of modes of action that have been proposed for Cr(VI)-induced small intestine tumors in mice.
Collapse
|
4
|
Mezencev R, Auerbach SS. Inferred inactivation of the Cftr gene in the duodena of mice exposed to hexavalent chromium (Cr(VI)) in drinking water supports its tumor-suppressor status and implies its potential role in Cr(VI)-induced carcinogenesis of the small intestines. Toxicol Appl Pharmacol 2021; 433:115773. [PMID: 34688701 PMCID: PMC9659473 DOI: 10.1016/j.taap.2021.115773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/12/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022]
Abstract
Carcinogenicity of hexavalent chromium [Cr (VI)] has been supported by a number of epidemiological and animal studies; however, its carcinogenic mode of action is still incompletely understood. To identify mechanisms involved in cancer development, we analyzed gene expression data from duodena of mice exposed to Cr(VI) in drinking water. This analysis included (i) identification of upstream regulatory molecules that are likely responsible for the observed gene expression changes, (ii) identification of annotated gene expression data from public repositories that correlate with gene expression changes in duodena of Cr(VI)-exposed mice, and (iii) identification of hallmark and oncogenic signature gene sets relevant to these data. We identified the inactivated CFTR gene among the top scoring upstream regulators, and found positive correlations between the expression data from duodena of Cr(VI)-exposed mice and other datasets in public repositories associated with the inactivation of the CFTR gene. In addition, we found enrichment of signatures for oncogenic signaling, sustained cell proliferation, impaired apoptosis and tissue remodeling. Results of our computational study support the tumor-suppressor role of the CFTR gene. Furthermore, our results support human relevance of the Cr(VI)-mediated carcinogenesis observed in the small intestines of exposed mice and suggest possible groups that may be more vulnerable to the adverse outcomes associated with the inactivation of CFTR by hexavalent chromium or other agents. Lastly, our findings predict, for the first time, the role of CFTR inactivation in chemical carcinogenesis and expand the range of plausible mechanisms that may be operative in Cr(VI)-mediated carcinogenesis of intestinal and possibly other tissues.
Collapse
Affiliation(s)
- Roman Mezencev
- Center for Public Health and Environmental Assessment, Office of Research and Development, US EPA, Washington, DC, United States.
| | - Scott S Auerbach
- National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, United States
| |
Collapse
|
5
|
Speer RM, Toyoda JH, Croom-Perez TJ, Liu KJ, Wise JP. Particulate Hexavalent Chromium Inhibits E2F1 Leading to Reduced RAD51 Nuclear Foci Formation in Human Lung Cells. Toxicol Sci 2021; 181:35-46. [PMID: 33677506 DOI: 10.1093/toxsci/kfab019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is the leading cause of cancer death; however, the mechanisms of lung carcinogens are poorly understood. Metals, including hexavalent chromium [Cr(VI)], induce chromosome instability, an early event in lung cancer. Failure of homologous recombination repair is a key mechanism for chromosome instability. Particulate Cr(VI) causes DNA double-strand breaks and prolonged exposure impairs homologous recombination targeting a key effector protein in this pathway, RAD51. Reduced RAD51 protein is a key endpoint of particulate Cr(VI) exposure. It is currently unknown how Cr(VI) reduces RAD51 protein. E2F1 is the predominant transcription factor for RAD51. This study sought to identify if E2F1 modulates the RAD51 response to particulate Cr(VI). Particulate Cr(VI) reduced RAD51 protein and mRNA levels but had a minimal effect on RAD51 half-life. E2F1 protein and mRNA were also inhibited by particulate Cr(VI) exposure. To connect these two outcomes, we tested if modulating E2F1 affects RAD51 outcomes after particulate Cr(VI) exposure. E2F1 knockdown inhibited RAD51 nuclear foci formation after acute particulate Cr(VI) exposure. These data indicate reduced RAD51 protein levels after prolonged particulate Cr(VI) exposure are predominantly due to inhibited expression. Particulate Cr(VI) also inhibits E2F1 expression. However, although loss of E2F1 does not modulate RAD51 expression after particulate Cr(VI) exposure, RAD51 nuclear foci formation is inhibited. These findings suggest E2F1 is important for RAD51 localization to double-strand breaks, but not expression after particulate Cr(VI) exposure in human lung cells.
Collapse
Affiliation(s)
- Rachel M Speer
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40292, USA
| | - Jennifer H Toyoda
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40292, USA
| | - Tayler J Croom-Perez
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40292, USA
| | - Ke Jian Liu
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico, USA
| | - John Pierce Wise
- Wise Laboratory of Environmental and Genetic Toxicology, Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40292, USA
| |
Collapse
|
6
|
Chappell GA, Wikoff DS, Thompson CM. Assessment of Mechanistic Data for Hexavalent Chromium-Induced Rodent Intestinal Cancer Using the Key Characteristics of Carcinogens. Toxicol Sci 2021; 180:38-50. [PMID: 33404626 PMCID: PMC7916733 DOI: 10.1093/toxsci/kfaa187] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Oral exposure to hexavalent chromium (Cr[VI]) induces intestinal tumors in mice. Mutagenic and nonmutagenic modes of action (MOAs) have been accepted by different regulatory bodies globally, the latter involving cytotoxicity-induced regenerative cell proliferation. However, concerns persist that all possible MOAs have not been fully considered. To address the potential for alternative MOAs, mechanistic data not represented in the existing two MOAs were evaluated. Relevant data were identified and organized by key characteristics of carcinogens (KCCs); literature related to epigenetics, immunosuppression, receptor-mediated effects, and immortalization were reviewed to identify potential key events associated with an alternative MOA. Over 200 references were screened for these four KCCs and further prioritized based on relevance to the research objective (ie, in vivo, oral exposure, gastrointestinal tissue). Minimal data were available specific to the intestine for these KCCs, and there was no evidence of any underlying mechanisms or key events that are not already represented in the two proposed MOAs. For example, while epigenetic dysregulation of DNA repair genes has been demonstrated, epigenetic effects were not measured in intestinal tissue, and it has been shown that Cr(VI) does not cause DNA damage in intestinal tissue. High-throughput screening data related to the KCCs were also evaluated, with activity generally limited to the two recognized MOAs. Collectively, no plausible alternative MOAs (or key events) were identified in addition to those previously proposed for Cr(VI) small intestine tumors.
Collapse
|
7
|
Bhat VS, Cohen SM, Gordon EB, Wood CE, Cullen JM, Harris MA, Proctor DM, Thompson CM. An adverse outcome pathway for small intestinal tumors in mice involving chronic cytotoxicity and regenerative hyperplasia: a case study with hexavalent chromium, captan, and folpet. Crit Rev Toxicol 2020; 50:685-706. [PMID: 33146058 DOI: 10.1080/10408444.2020.1823934] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Small intestinal (SI) tumors are relatively uncommon outcomes in rodent cancer bioassays, and limited information regarding chemical-induced SI tumorigenesis has been reported in the published literature. Herein, we propose a cytotoxicity-mediated adverse outcome pathway (AOP) for SI tumors by leveraging extensive target species- and site-specific molecular, cellular, and histological mode of action (MOA) research for three reference chemicals, the fungicides captan and folpet and the transition metal hexavalent chromium (Cr(VI)). The gut barrier functions through highly efficient homeostatic regulation of SI epithelial cell sloughing, regenerative proliferation, and repair, which involves the replacement of up to 1011 cells per day. This dynamic turnover in the SI provides a unique local environment for a cytotoxicity mediated AOP/MOA. Upon entering the duodenum, cytotoxicity to the villous epithelium is the molecular initiating event, as indicated by crypt elongation, villous atrophy/blunting, and other morphologic changes. Over time, the regenerative capacity of the gut epithelium to compensate declines as epithelial loss accelerates, especially at higher exposures. The first key event (KE), sustained regenerative crypt proliferation/hyperplasia, requires sufficient durations, likely exceeding 6 or 12 months, due to extensive repair capacity, to create more opportunities for the second KE, spontaneous mutation/transformation, ultimately leading to proximal SI tumors. Per OECD guidance, biological plausibility, essentiality, and empirical support were assessed using modified Bradford Hill considerations. The weight-of-evidence also included a lack of induced mutations in the duodenum after up to 90 days of Cr(VI) or captan exposure. The extensive evidence for this AOP, along with the knowledge that human exposures are orders of magnitude below those associated with KEs in this AOP, supports its use for regulatory applications, including hazard identification and risk assessment.
Collapse
Affiliation(s)
| | - Samuel M Cohen
- Havlik-Wall Professor of Oncology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Charles E Wood
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - John M Cullen
- North Carolina State University, Raleigh, NC, USA.,EPL, Inc., Sterling, VA, USA
| | | | | | | |
Collapse
|
8
|
Thompson CM, Kirman CR, Hays SM, Suh M, Harvey SE, Proctor DM, Rager JE, Haws LC, Harris MA. Integration of mechanistic and pharmacokinetic information to derive oral reference dose and margin-of-exposure values for hexavalent chromium. J Appl Toxicol 2018; 38:351-365. [PMID: 29064106 PMCID: PMC5813206 DOI: 10.1002/jat.3545] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 08/28/2017] [Accepted: 09/05/2017] [Indexed: 01/18/2023]
Abstract
The current US Environmental Protection Agency (EPA) reference dose (RfD) for oral exposure to chromium, 0.003 mg kg-1 day-1 , is based on a no-observable-adverse-effect-level from a 1958 bioassay of rats exposed to ≤25 ppm hexavalent chromium [Cr(VI)] in drinking water. EPA characterizes the confidence in this RfD as "low." A more recent cancer bioassay indicates that Cr(VI) in drinking water is carcinogenic to mice at ≥30 ppm. To assess whether the existing RfD is health protective, neoplastic and non-neoplastic lesions from the 2 year cancer bioassay were modeled in a three-step process. First, a rodent physiological-based pharmacokinetic (PBPK) model was used to estimate internal dose metrics relevant to each lesion. Second, benchmark dose modeling was conducted on each lesion using the internal dose metrics. Third, a human PBPK model was used to estimate the daily mg kg-1 dose that would produce the same internal dose metric in both normal and susceptible humans. Mechanistic research into the mode of action for Cr(VI)-induced intestinal tumors in mice supports a threshold mechanism involving intestinal wounding and chronic regenerative hyperplasia. As such, an RfD was developed using incidence data for the precursor lesion diffuse epithelial hyperplasia. This RfD was compared to RfDs for other non-cancer endpoints; all RfD values ranged 0.003-0.02 mg kg-1 day-1 . The lowest of these values is identical to EPA's existing RfD value. Although the RfD value remains 0.003 mg kg-1 day-1 , the confidence is greatly improved due to the use of a 2-year bioassay, mechanistic data, PBPK models and benchmark dose modeling.
Collapse
Affiliation(s)
| | | | - Sean M Hays
- Summit Toxicology, LLP, Bozeman, MT, 59722, USA
| | - Mina Suh
- ToxStrategies, Inc., Mission Viejo, CA, 92692, USA
| | | | | | | | | | | |
Collapse
|
9
|
Thompson CM, Suh M, Proctor DM, Haws LC, Harris MA. Ten factors for considering the mode of action of Cr(VI)-induced gastrointestinal tumors in rodents. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2017; 823:45-57. [DOI: 10.1016/j.mrgentox.2017.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/26/2017] [Accepted: 08/28/2017] [Indexed: 12/28/2022]
|
10
|
Thompson CM, Young RR, Dinesdurage H, Suh M, Harris MA, Rohr AC, Proctor DM. Assessment of the mutagenic potential of hexavalent chromium in the duodenum of big blue® rats. Toxicol Appl Pharmacol 2017; 330:48-52. [DOI: 10.1016/j.taap.2017.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/01/2017] [Accepted: 07/03/2017] [Indexed: 02/07/2023]
|
11
|
Rager JE, Ring CL, Fry RC, Suh M, Proctor DM, Haws LC, Harris MA, Thompson CM. High-Throughput Screening Data Interpretation in the Context of In Vivo Transcriptomic Responses to Oral Cr(VI) Exposure. Toxicol Sci 2017; 158:199-212. [PMID: 28472532 PMCID: PMC5837509 DOI: 10.1093/toxsci/kfx085] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The toxicity of hexavalent chromium [Cr(VI)] in drinking water has been studied extensively, and available in vivo and in vitro studies provide a robust dataset for application of advanced toxicological tools to inform the mode of action (MOA). This study aimed to contribute to the understanding of Cr(VI) MOA by evaluating high-throughput screening (HTS) data and other in vitro data relevant to Cr(VI), and comparing these findings to robust in vivo data, including transcriptomic profiles in target tissues. Evaluation of Tox21 HTS data for Cr(VI) identified 11 active assay endpoints relevant to the Ten Key Characteristics of Carcinogens (TKCCs) that have been proposed by other investigators. Four of these endpoints were related to TP53 (tumor protein 53) activation mapping to genotoxicity (KCC#2), and four were related to cell death/proliferation (KCC#10). HTS results were consistent with other in vitro data from the Comparative Toxicogenomics Database. In vitro responses were compared to in vivo transcriptomic responses in the most sensitive target tissue, the duodenum, of mice exposed to ≤ 180 ppm Cr(VI) for 7 and 90 days. Pathways that were altered both in vitro and in vivo included those relevant to cell death/proliferation. In contrast, pathways relevant to p53/DNA damage were identified in vitro but not in vivo. Benchmark dose modeling and phenotypic anchoring of in vivo transcriptomic responses strengthened the finding that Cr(VI) causes cell stress/injury followed by proliferation in the mouse duodenum at high doses. These findings contribute to the body of evidence supporting a non-mutagenic MOA for Cr(VI)-induced intestinal cancer.
Collapse
Affiliation(s)
| | | | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health
- Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27516
| | - Mina Suh
- ToxStrategies Inc, Mission Viejo, California 92692
| | | | | | | | | |
Collapse
|
12
|
Thompson CM, Rager JE, Suh M, Ring CL, Proctor DM, Haws LC, Fry RC, Harris MA. Transcriptomic responses in the oral cavity of F344 rats and B6C3F1 mice following exposure to Cr(VI): Implications for risk assessment. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2016; 57:706-716. [PMID: 27859739 PMCID: PMC5215477 DOI: 10.1002/em.22064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/27/2016] [Indexed: 05/13/2023]
Abstract
Exposure to hexavalent chromium [Cr(VI)] in drinking water was previously reported to increase oral tumor incidence in F344 rats. To investigate the mode of action for these tumors, transcriptomic profiles in oral mucosa samples of F344 rats and B6C3F1 mice were analyzed following exposure to 0.1-180 ppm Cr(VI) for 7 or 90 days. In rats, genome-wide microarray analyses identified no significantly differentially expressed genes (DEGs) at either time point. In mice, 14 and 1 DEGs were respectively identified after 7 and 90 days of exposure. Therefore, relaxed statistical criteria were employed to identify potential DEGs (pDEGs), followed by high-throughput benchmark dose modeling to identify responsive pDEGs for pathway enrichment analysis. This identified 288 and 168 pDEGs in the rat oral mucosa, of which only 20 and 7 showed evidence of dose-response. No significant pathway enrichment was obtained with either pDEG or dose-responsive pDEG lists. Similar results were obtained in mice. These analyses indicate a negligible transcriptional response in the oral mucosa of both species. Comparison of the total number of gene changes in the oral mucosa of rats and mice with responses in the duodenum of animals from the same study demonstrated remarkable dose-response concordance across tissues and species as a function of tissue chromium concentration. The low chromium levels in the oral mucosa and negligible transcript response are consistent with an absence of tissue lesions. These findings are used to compare the merits of linear and nonlinear approaches for deriving toxicity criteria based on the oral tumors in rats. Environ. Mol. Mutagen. 57:706-716, 2016. © 2016 The Authors. Environmental and Molecular Mutagenesis Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | | | - Mina Suh
- ToxStrategies, IncMission ViejoCalifornia
| | | | | | | | - Rebecca C. Fry
- Department of Environmental Sciences and EngineeringGillings School of Global Public HealthChapel HillNorth Carolina
- Curriculum in Toxicology, University of North Carolina at Chapel HillChapel HillNorth Carolina
| | | |
Collapse
|
13
|
Cullen JM, Ward JM, Thompson CM. Reevaluation and Classification of Duodenal Lesions in B6C3F1 Mice and F344 Rats from 4 Studies of Hexavalent Chromium in Drinking Water. Toxicol Pathol 2016; 44:279-89. [PMID: 26538584 PMCID: PMC4785997 DOI: 10.1177/0192623315611501] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Thirteen-week and 2-year drinking water studies conducted by the National Toxicology Program (NTP) reported that hexavalent chromium (Cr(VI)) induced diffuse epithelial hyperplasia in the duodenum of B6C3F1 mice but not F344 rats. In the 2-year study, Cr(VI) exposure was additionally associated with duodenal adenomas and carcinomas in mice only. Subsequent 13-week Cr(VI) studies conducted by another group demonstrated non-neoplastic duodenal lesions in B6C3F1 mice similar to those of the NTP study as well as mild duodenal hyperplasia in F344 rats. Because intestinal lesions in mice are the basis for proposed safety standards for Cr(VI), and the histopathology data are relevant to the mode of action, consistency (an important Hill criterion for causality) was assessed across the aforementioned studies. Two veterinary pathologists applied uniform diagnostic criteria to the duodenal lesions in rats and mice from the 4 repeated-dose studies. Comparable non-neoplastic intestinal lesions were evident in mice and rats from all 4 studies; however, the incidence and severity of intestinal lesions were greater in mice than rats. These findings demonstrate consistency across studies and species and highlight the importance of standardized nomenclature for intestinal pathology. The differences in the severity of non-neoplastic lesions also likely contribute to the differential tumor response.
Collapse
Affiliation(s)
- John M Cullen
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | | | | |
Collapse
|
14
|
Luczak MW, Green SE, Zhitkovich A. Different ATM Signaling in Response to Chromium(VI) Metabolism via Ascorbate and Nonascorbate Reduction: Implications for in Vitro Models and Toxicogenomics. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:61-6. [PMID: 25977998 PMCID: PMC4710604 DOI: 10.1289/ehp.1409434] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 05/13/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND Carcinogenic hexavalent chromium [Cr(VI)] requires cellular reduction to generate DNA damage. Metabolism of Cr(VI) by its principal reducer ascorbate (Asc) lacks a Cr(V) intermediate, which is abundant in reactions with a minor reducing agent, glutathione. Cultured cells are widely used in mechanistic studies of Cr(VI) toxicity; however, they typically contain < 1% of normal Asc levels. Asc deficiency is also expected to diminish protection against reactive oxygen species. OBJECTIVES We assessed how the presence of Asc in cells affects their stress signaling and survival responses to chromate. METHODS We investigated the effects of Asc restoration in human lung H460 cells and normal human lung fibroblasts on the activation and functional role of ATM kinase, which controls DNA damage responses involving several hundreds of proteins. RESULTS Treatment of standard cultures with Cr(VI) strongly activated ATM, as indicated by its automodification at Ser1981 and by phosphorylation of checkpoint kinase 2 (CHK2) and chromatin/transcription regulator KRAB-associated protein 1 (KAP1). Confirming the importance of activated ATM, its inhibition impaired replication recovery and clonogenic survival. In contrast, fully Asc-restored cells lacked ATM activation by Cr(VI), and ATM silencing produced no significant effects on p53 stabilization, apoptosis, replication recovery, or clonogenic survival. Dose dependence studies found a close correlation between ATM activation and the extent of Cr(VI) reduction by glutathione. CONCLUSIONS Asc restoration in cultured cells dramatically altered their stress responses to Cr(VI) by preventing activation of the oxidant-sensitive ATM network. We suggest that toxicogenomic and other cell response-based approaches likely underestimate Cr(VI) genotoxicity when standard ATM-activating carcinogens are used as references. CITATION Luczak MW, Green SE, Zhitkovich A. 2016. Different ATM signaling in response to chromium(VI) metabolism via ascorbate and nonascorbate reduction: implications for in vitro models and toxicogenomics. Environ Health Perspect 124:61-66; http://dx.doi.org/10.1289/ehp.1409434.
Collapse
Affiliation(s)
- Michal W. Luczak
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Samantha E. Green
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Anatoly Zhitkovich
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
15
|
Consideration of non-linear, non-threshold and threshold approaches for assessing the carcinogenicity of oral exposure to hexavalent chromium. Regul Toxicol Pharmacol 2015; 73:834-52. [DOI: 10.1016/j.yrtph.2015.10.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/19/2015] [Accepted: 10/15/2015] [Indexed: 12/28/2022]
|
16
|
Thompson CM, Young RR, Suh M, Dinesdurage HR, Elbekai RH, Harris MA, Rohr AC, Proctor DM. Assessment of the mutagenic potential of Cr(VI) in the oral mucosa of Big Blue® transgenic F344 rats. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2015; 56:621-628. [PMID: 26010270 DOI: 10.1002/em.21952] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 03/07/2015] [Accepted: 03/15/2015] [Indexed: 06/04/2023]
Abstract
Exposure to high concentrations of hexavalent chromium [Cr(VI)] in drinking water was associated with an increased incidence of oral tumors in F344 rats in a 2-year cancer bioassay conducted by the National Toxicology Program. These tumors primarily occurred at 180 ppm Cr(VI) and appeared to originate from the gingival mucosa surrounding the upper molar teeth. To investigate whether these tumors could have resulted from a mutagenic mode of action (MOA), a transgenic mutation assay based on OECD Test Guideline 488 was conducted in Big Blue(®) TgF344 rats. The mutagenic oral carcinogen 4-nitroquinoline-1-oxide (4-NQO) served as a positive control. Mutant frequency was measured in the inner gingiva with adjacent palate, and outer gingiva with adjacent buccal tissue. Exposure to 10 ppm 4-NQO in drinking water for 28 days increased mutant frequency in the cII transgene significantly, from 39.1 ± 7.5 × 10(-6) to 688 ± 250 × 10(-6) in the gingival/buccal region, and from 49.8 ± 17.8 × 10(-6) to 1818 ± 362 × 10(-6) in the gingival/palate region. Exposure to 180 ppm Cr(VI) in drinking water for 28 days did not significantly increase the mutant frequency in the gingival/buccal (44.4 ± 25.4 × 10(-6)) or the gingival/palate (57.8 ± 9.1 × 10(-6)) regions relative to controls. These data indicate that high (∼180,000 times expected human exposure), tumorigenic concentrations of Cr(VI) did not significantly increase mutations in the gingival epithelium, and suggest that Cr(VI) does not act by a mutagenic MOA in the rat oral cavity.
Collapse
Affiliation(s)
| | | | - Mina Suh
- ToxStrategies, Inc., Mission Viejo, California
| | | | | | | | - Annette C Rohr
- Electric Power Research Institute, Palo Alto, California
| | | |
Collapse
|
17
|
Thompson CM, Kirman CR, Proctor DM, Haws LC, Suh M, Hays SM, Hixon JG, Harris MA. A chronic oral reference dose for hexavalent chromium-induced intestinal cancer. J Appl Toxicol 2014; 34:525-36. [PMID: 23943231 PMCID: PMC4282340 DOI: 10.1002/jat.2907] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 05/15/2013] [Accepted: 06/02/2013] [Indexed: 01/18/2023]
Abstract
High concentrations of hexavalent chromium [Cr(VI)] in drinking water induce villous cytotoxicity and compensatory crypt hyperplasia in the small intestines of mice (but not rats). Lifetime exposure to such cytotoxic concentrations increases intestinal neoplasms in mice, suggesting that the mode of action for Cr(VI)-induced intestinal tumors involves chronic wounding and compensatory cell proliferation of the intestine. Therefore, we developed a chronic oral reference dose (RfD) designed to be protective of intestinal damage and thus intestinal cancer. A physiologically based pharmacokinetic model for chromium in mice was used to estimate the amount of Cr(VI) entering each intestinal tissue section (duodenum, jejunum and ileum) from the lumen per day (normalized to intestinal tissue weight). These internal dose metrics, together with corresponding incidences for diffuse hyperplasia, were used to derive points of departure using benchmark dose modeling and constrained nonlinear regression. Both modeling techniques resulted in similar points of departure, which were subsequently converted to human equivalent doses using a human physiologically based pharmacokinetic model. Applying appropriate uncertainty factors, an RfD of 0.006 mg kg(-1) day(-1) was derived for diffuse hyperplasia-an effect that precedes tumor formation. This RfD is protective of both noncancer and cancer effects in the small intestine and corresponds to a safe drinking water equivalent level of 210 µg l(-1). This concentration is higher than the current federal maximum contaminant level for total Cr (100 µg l(-1)) and well above levels of Cr(VI) in US drinking water supplies (typically ≤ 5 µg l(-1)).
Collapse
Affiliation(s)
| | | | | | | | - Mina Suh
- ToxStrategies, Inc.Rancho Santa Margarita, CA, 92688, USA
| | - Sean M Hays
- Summit Toxicology, LLPAllenspark, CO, 80510, USA
| | | | | |
Collapse
|
18
|
Scientific Opinion on the risks to public health related to the presence of chromium in food and drinking water. EFSA J 2014. [DOI: 10.2903/j.efsa.2014.3595] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
19
|
Martini CN, Brandani JN, Gabrielli M, Vila MDC. Effect of hexavalent chromium on proliferation and differentiation to adipocytes of 3T3-L1 fibroblasts. Toxicol In Vitro 2014; 28:700-6. [PMID: 24576443 DOI: 10.1016/j.tiv.2014.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 02/05/2014] [Accepted: 02/15/2014] [Indexed: 10/25/2022]
Abstract
Heavy metals contamination has become an important risk factor for public health and the environment. Chromium is a frequent industrial contaminant and is also used in orthopaedic joint replacements made from cobalt-chromium-alloy. Since hexavalent chromium (Cr(VI)) was reported as genotoxic and carcinogenic in different mammals, to further evaluate its cytotoxicity, we investigated the effect of this heavy metal in the proliferation and differentiation to adipocytes of 3T3-L1 fibroblasts. These cells, after the addition of a mixture containing insulin, dexamethasone and methylisobutylxanthine, first proliferate, a process known as mitotic clonal expansion (MCE), and then differentiate to adipocytes. In this differentiation process a key transcription factor is induced: peroxisome proliferator-activated receptor gamma (PPAR gamma). We found that treatment of 3T3-L1 fibroblasts with potassium chromate inhibited proliferation in exponentially growing cells and MCE as well as differentiation. A decrease in PPAR gamma content, evaluated by western blot and immunofluorescence, was found in cells differentiated in the presence of chromium. On the other hand, after inhibition of differentiation with chromium, when the metal was removed, differentiation was recovered, which indicates that this may be a reversible effect. We also found an increase in the number of micronucleated cells after treatment with Cr(VI) which is associated with genotoxic effects. According to our results, Cr(VI) is able to inhibit proliferation and differentiation to adipocytes of 3T3-L1 fibroblasts and to increase micronucleated cells, which are all indicative of alterations in cellular physiology and therefore, contributes to further elucidate the cytotoxic effects of this heavy metal.
Collapse
Affiliation(s)
- Claudia N Martini
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, 1428 Buenos Aires, Argentina
| | - Javier N Brandani
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, 1428 Buenos Aires, Argentina
| | - Matías Gabrielli
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, 1428 Buenos Aires, Argentina
| | - María del C Vila
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Pabellón 2, Ciudad Universitaria, 1428 Buenos Aires, Argentina.
| |
Collapse
|
20
|
Suh M, Thompson CM, Kirman CR, Carakostas MC, Haws LC, Harris MA, Proctor DM. High concentrations of hexavalent chromium in drinking water alter iron homeostasis in F344 rats and B6C3F1 mice. Food Chem Toxicol 2014; 65:381-8. [PMID: 24418189 DOI: 10.1016/j.fct.2014.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 01/03/2014] [Accepted: 01/05/2014] [Indexed: 11/16/2022]
Abstract
Hexavalent chromium [Cr(VI)] induces hematological signs of microcytic anemia in rodents. Considering that Cr(VI) can oxidize ferrous (Fe(2+)) to ferric (Fe(3+)) iron, and that only the former is transported across the duodenum, we hypothesize that, at high concentrations, Cr(VI) oxidizes Fe(2+) in the lumen of the small intestine and perturbs iron absorption. Herein we report that 90-day exposure to Cr(VI) in drinking water resulted in dose-dependent decreases in Fe levels in the duodenum, liver, serum, and bone marrow. Toxicogenomic analyses from the duodenum indicate responses consistent with Fe deficiency, including significant induction of divalent metal transporter 1 (DMT1, Slc11a2) and transferrin receptor 1 (TFR1, Tfr1). In addition, at ⩾20mg Cr(VI)/L in drinking water, Cr RBC:plasma ratios in rats were increased and exceeded unity, indicating saturation of reductive capacity and intracellular absorption of Cr(VI) into red blood cells (RBCs). These effects occurred in both species but were generally more severe in rats. These data suggest that high concentrations of Cr(VI) in drinking limit Fe absorption and alter iron homeostasis. Furthermore, some effects observed at high doses in recent Cr(VI) chronic and subchronic bioassays may be explained, at least in part, by iron deficiency and disruption of homeostasis.
Collapse
Affiliation(s)
- Mina Suh
- ToxStrategies, Inc., Mission Viejo, CA 92692, United States.
| | | | | | | | - Laurie C Haws
- ToxStrategies, Inc., Austin, TX 78759, United States.
| | | | | |
Collapse
|
21
|
Black MB, Parks BB, Pluta L, Chu TM, Allen BC, Wolfinger RD, Thomas RS. Comparison of microarrays and RNA-seq for gene expression analyses of dose-response experiments. Toxicol Sci 2013; 137:385-403. [PMID: 24194394 DOI: 10.1093/toxsci/kft249] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Relative to microarrays, RNA-seq has been reported to offer higher precision estimates of transcript abundance, a greater dynamic range, and detection of novel transcripts. However, previous comparisons of the 2 technologies have not covered dose-response experiments that are relevant to toxicology. Male F344 rats were exposed for 13 weeks to 5 doses of bromobenzene, and liver gene expression was measured using both microarrays and RNA-seq. Multiple normalization methods were evaluated for each technology, and gene expression changes were statistically analyzed using both analysis of variance and benchmark dose (BMD). Fold-change values were highly correlated between the 2 technologies, whereas the -log p values showed lower correlation. RNA-seq detected fewer statistically significant genes at lower doses, but more significant genes based on fold change except when a negative binomial transformation was applied. Overlap in genes significant by both p value and fold change was approximately 30%-40%. Random sampling of the RNA-seq data showed an equivalent number of differentially expressed genes compared with microarrays at ~5 million reads. Quantitative RT-PCR of differentially expressed genes uniquely identified by each technology showed a high degree of confirmation when both fold change and p value were considered. The mean dose-response expression of each gene was highly correlated between technologies, whereas estimates of sample variability and gene-based BMD values showed lower correlation. Differences in BMD estimates and statistical significance may be due, in part, to differences in the dynamic range of each technology and the degree to which normalization corrects genes at either end of the scale.
Collapse
Affiliation(s)
- Michael B Black
- * The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709
| | | | | | | | | | | | | |
Collapse
|
22
|
Physiologically based pharmacokinetic model for humans orally exposed to chromium. Chem Biol Interact 2013; 204:13-27. [DOI: 10.1016/j.cbi.2013.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 02/28/2013] [Accepted: 04/02/2013] [Indexed: 12/15/2022]
|
23
|
Thompson CM, Proctor DM, Suh M, Haws LC, Kirman CR, Harris MA. Assessment of the mode of action underlying development of rodent small intestinal tumors following oral exposure to hexavalent chromium and relevance to humans. Crit Rev Toxicol 2013; 43:244-74. [PMID: 23445218 PMCID: PMC3604738 DOI: 10.3109/10408444.2013.768596] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 01/16/2013] [Accepted: 01/17/2013] [Indexed: 12/13/2022]
Abstract
Abstract Chronic exposure to high concentrations of hexavalent chromium (Cr(VI)) in drinking water causes intestinal adenomas and carcinomas in mice, but not in rats. Cr(VI) causes damage to intestinal villi and crypt hyperplasia in mice after only one week of exposure. After two years of exposure, intestinal damage and crypt hyperplasia are evident in mice (but not rats), as are intestinal tumors. Although Cr(VI) has genotoxic properties, these findings suggest that intestinal tumors in mice arise as a result of chronic mucosal injury. To better understand the mode of action (MOA) of Cr(VI) in the intestine, a 90-day drinking water study was conducted to collect histological, biochemical, toxicogenomic and pharmacokinetic data in intestinal tissues. Using MOA analyses and human relevance frameworks proposed by national and international regulatory agencies, the weight of evidence supports a cytotoxic MOA with the following key events: (a) absorption of Cr(VI) from the intestinal lumen, (b) toxicity to intestinal villi, (c) crypt regenerative hyperplasia and (d) clonal expansion of mutations within the crypt stem cells, resulting in late onset tumorigenesis. This article summarizes the data supporting each key event in the MOA, as well as data that argue against a mutagenic MOA for Cr(VI)-induced intestinal tumors.
Collapse
|
24
|
Witt KL, Stout MD, Herbert RA, Travlos GS, Kissling GE, Collins BJ, Hooth MJ. Mechanistic Insights from the NTP Studies of Chromium. Toxicol Pathol 2013; 41:326-42. [DOI: 10.1177/0192623312469856] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Hexavalent chromium (Cr(VI)) is a contaminant of water and soil and is a human lung carcinogen. Trivalent chromium (Cr(III)), a proposed essential element, is ingested by humans in the diet and in dietary supplements such as chromium picolinate (CP). The National Toxicology Program (NTP) demonstrated that Cr(VI) is also carcinogenic in rodents when administered in drinking water as sodium dichromate dihydrate (SDD), inducing neoplasms of the oral cavity and small intestine in rats and mice, respectively. In contrast, there was no definitive evidence of toxicity or carcinogenicity following exposure to Cr(III) administered in feed as CP monohydrate (CPM). Cr(VI) readily enters cells via nonspecific anion channels, in contrast to Cr(III), which cannot easily pass through the cell membrane. Extracellular reduction of Cr(VI) to Cr(III), which occurs primarily in the stomach, is considered a mechanism of detoxification, while intracellular reduction is thought to be a mechanism of genotoxicity and carcinogenicity. Tissue distribution studies in additional groups of male rats and female mice demonstrated higher Cr concentrations in tissues following exposure to Cr(VI) compared to controls and Cr(III) exposure at a similar external dose, indicating that some of the Cr(VI) escaped gastric reduction and was distributed systemically. The multiple potential pathways of Cr-induced genotoxicity will be discussed.
Collapse
Affiliation(s)
- Kristine L. Witt
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Matthew D. Stout
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Ronald A. Herbert
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Gregory S. Travlos
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Grace E. Kissling
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Bradley J. Collins
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Michelle J. Hooth
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| |
Collapse
|
25
|
Prokopec SD, Watson JD, Waggott DM, Smith AB, Wu AH, Okey AB, Pohjanvirta R, Boutros PC. Systematic evaluation of medium-throughput mRNA abundance platforms. RNA (NEW YORK, N.Y.) 2013; 19:51-62. [PMID: 23169800 PMCID: PMC3527726 DOI: 10.1261/rna.034710.112] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 10/10/2012] [Indexed: 05/22/2023]
Abstract
Profiling of mRNA abundances with high-throughput platforms such as microarrays and RNA-seq has become an important tool in both basic and biomedical research. However, these platforms remain prone to systematic errors and have challenges in clinical and industrial applications. As a result, it is standard practice to validate a subset of key results using alternate technologies. Similarly, clinical and industrial applications typically involve transitions from a high-throughput discovery platform to medium-throughput validation ones. These medium-throughput validation platforms have high technical reproducibility and reduced sample input needs, and low sensitivity to sample quality (e.g., for processing FFPE specimens). Unfortunately, while medium-throughput platforms have proliferated, there are no comprehensive comparisons of them. Here we fill that gap by comparing two key medium-throughput platforms--NanoString's nCounter Analysis System and ABI's OpenArray System--to gold-standard quantitative real-time RT-PCR. We quantified 38 genes and positive and negative controls in 165 samples. Signal:noise ratios, correlations, dynamic range, and detection accuracy were compared across platforms. All three measurement technologies showed good concordance, but with divergent price/time/sensitivity trade-offs. This study provides the first detailed comparison of medium-throughput RNA quantification platforms and provides a template and a standard data set for the evaluation of additional technologies.
Collapse
Affiliation(s)
- Stephenie D. Prokopec
- Informatics and Biocomputing Platform, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - John D. Watson
- Informatics and Biocomputing Platform, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Daryl M. Waggott
- Informatics and Biocomputing Platform, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Ashley B. Smith
- Informatics and Biocomputing Platform, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Alexander H. Wu
- Informatics and Biocomputing Platform, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
| | - Allan B. Okey
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Raimo Pohjanvirta
- Laboratory of Toxicology, Department of Environmental Health, National Public Health Institute, FIN-70701 Kuopio, Finland
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, FI-00014 Helsinki, Finland
| | - Paul C. Boutros
- Informatics and Biocomputing Platform, Ontario Institute for Cancer Research, Toronto, Ontario M5G 0A3, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 2M9, Canada
- Corresponding authorE-mail
| |
Collapse
|
26
|
Kirman CR, Hays SM, Aylward LL, Suh M, Harris MA, Thompson CM, Haws LC, Proctor DM. Physiologically based pharmacokinetic model for rats and mice orally exposed to chromium. Chem Biol Interact 2012; 200:45-64. [PMID: 22981460 DOI: 10.1016/j.cbi.2012.08.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 08/14/2012] [Accepted: 08/17/2012] [Indexed: 11/16/2022]
Abstract
A multi-compartment physiologically based pharmacokinetic (PBPK) model was developed to describe the behavior of Cr(III) and Cr(VI) in rats and mice following long-term oral exposure. Model compartments were included for GI lumen, oral mucosa, forestomach/stomach, small intestinal mucosa (duodenum, jejunum, ileum), blood, liver, kidney, bone, and a combined compartment for remaining tissues. Data from ex vivo Cr(VI) reduction studies were used to characterize reduction of Cr(VI) in fed rodent stomach fluid as a second-order, pH-dependent process. For model development, tissue time-course data for total chromium were collected from rats and mice exposed to Cr(VI) in drinking water for 90 days at six concentrations ranging from 0.1 to 180 mg Cr(VI)/L. These data were used to supplement the tissue time-course data collected in other studies with oral administration of Cr(III) and Cr(VI), including that from recent NTP chronic bioassays. Clear species differences were identified for chromium delivery to the target tissue (small intestines), with higher concentrations achieved in mice than in rats, consistent with small intestinal tumor formation, which was observed upon chronic exposures in mice but not in rats. Erythrocyte:plasma chromium ratios suggest that Cr(VI) entered portal circulation at drinking water concentrations equal to and greater than 60 mg/L in rodents. Species differences are described for distribution of chromium to the liver and kidney, with liver:kidney ratios higher in mice than in rats. Overall, the PBPK model provides a good description of chromium toxicokinetics, with model predictions for tissue chromium within a factor of 3 for greater than 80% of measurements evaluated. The tissue data and PBPK model predictions indicate a concentration gradient in the small intestines (duodenum > jejunum > ileum), which will be useful for assessing the tumor response gradient observed in mouse small intestines in terms of target tissue dose. The rodent PBPK model presented here, when used in conjunction with a human PBPK model for Cr(VI), should provide a more robust characterization of species differences in toxicokinetic factors for assessing the potential risks associated with low-dose exposures of Cr(VI) in human populations.
Collapse
Affiliation(s)
- C R Kirman
- Summit Toxicology, 29449 Pike Drive, Orange Village, OH 44022, USA.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Thompson CM, Fedorov Y, Brown DD, Suh M, Proctor DM, Kuriakose L, Haws LC, Harris MA. Assessment of Cr(VI)-induced cytotoxicity and genotoxicity using high content analysis. PLoS One 2012; 7:e42720. [PMID: 22905163 PMCID: PMC3414448 DOI: 10.1371/journal.pone.0042720] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 07/10/2012] [Indexed: 11/18/2022] Open
Abstract
Oral exposure to high concentrations of hexavalent chromium [Cr(VI)] induces intestinal redox changes, villus cytotoxicity, crypt hyperplasia, and intestinal tumors in mice. To assess the effects of Cr(VI) in a cell model relevant to the intestine, undifferentiated (proliferating) and differentiated (confluent) Caco-2 cells were treated with Cr(VI), hydrogen peroxide or rotenone for 2-24 hours. DNA damage was then assessed by nuclear staining intensity of 8-hydroxydeoxyguanosine (8-OHdG) and phosphorylated histone variant H2AX (γ-H2AX) measured by high content analysis methods. In undifferentiated Caco-2, all three chemicals increased 8-OHdG and γ-H2AX staining at cytotoxic concentrations, whereas only 8-OHdG was elevated at non-cytotoxic concentrations at 24 hr. Differentiated Caco-2 were more resistant to cytotoxicity and DNA damage than undifferentiated cells, and there were no changes in apoptotic markers p53 or annexin-V. However, Cr(VI) induced a dose-dependent translocation of the unfolded protein response transcription factor ATF6 into the nucleus. Micronucleus (MN) formation was assessed in CHO-K1 and A549 cell lines. Cr(VI) increased MN frequency in CHO-K1 only at highly cytotoxic concentrations. Relative to the positive control Mitomycin-C, Cr(VI) only slightly increased MN frequency in A549 at mildly cytotoxic concentrations. The results demonstrate that Cr(VI) genotoxicity correlates with cytotoxic concentrations, and that H2AX phosphorylation occurs at higher concentrations than oxidative DNA damage in proliferating Caco-2 cells. The findings suggest that in vitro genotoxicity of Cr(VI) is primarily oxidative in nature at low concentrations. Implications for in vivo intestinal toxicity of Cr(VI) will be discussed.
Collapse
|
28
|
Thompson CM, Gregory Hixon J, Proctor DM, Haws LC, Suh M, Urban JD, Harris MA. Assessment of genotoxic potential of Cr(VI) in the mouse duodenum: an in silico comparison with mutagenic and nonmutagenic carcinogens across tissues. Regul Toxicol Pharmacol 2012; 64:68-76. [PMID: 22705708 DOI: 10.1016/j.yrtph.2012.05.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 05/15/2012] [Accepted: 05/16/2012] [Indexed: 01/09/2023]
Abstract
In vitro studies on hexavalent chromium [Cr(VI)] indicate that reduced forms of this metal can interact with DNA and cause mutations. Recently, Cr(VI) was shown to induce intestinal tumors in mice; however, Cr(VI) elicited redox changes, cytotoxicity and hyperplasia - suggesting involvement of tissue injury rather than direct mutagenesis. Moreover, toxicogenomic analyses indicated limited evidence for DNA damage responses. Herein, we extend these toxicogenomic analyses by comparing the gene expression patterns elicited by Cr(VI) with those of four mutagenic and four nonmutagenic carcinogens. To date, toxicogenomic profiles for mutagenic and nonmutagenic duodenal carcinogens do not exist, thus duodenal gene changes in mice were compared to those elicited by hepatocarcinogens. Specifically, duodenal gene changes in mice following exposure to Cr(VI) in drinking water were compared to hepatic gene changes previously identified as potentially discriminating mutagenic and nonmutagenic hepatocarcinogens. Using multivariate statistical analyses (including logistic regression classification), the Cr(VI) gene responses clustered apart from mutagenic carcinogens and closely with nonmutagenic carcinogens. These findings are consistent with other intestinal data supporting a nonmutagenic mode of action (MOA). These findings may be useful as part of a full weight of evidence MOA evaluation for Cr(VI)-induced intestinal carcinogenesis. Limitations to this analysis will also be discussed.
Collapse
|