1
|
Wu PC, Wen HJ, Huang KF, Huang SK, Liang MC. Transition metals and chemical compositions determine the oxidation capacity of atmospheric particulate matters. ENVIRONMENTAL RESEARCH 2025; 278:121661. [PMID: 40268221 DOI: 10.1016/j.envres.2025.121661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/07/2025] [Accepted: 04/20/2025] [Indexed: 04/25/2025]
Abstract
The knowledge of the causal relationship between exposure to airborne particulate matter (PM) and respiratory-related health issues remains unsatisfactory, owing to the complexities of physical and chemical characteristics in PM. One measure that greatly lifts the complexity is oxidative potential (OP), the overall production capacity of reactive oxygen species. We analyzed PM at different size fractions from three localities, exhibiting different source emission properties and photochemical aging states. We also investigated possible causes for their OPs, which were assessed using cellular and acellular assays. We found that higher PM mass did not always yield higher OP. Instead, chemical composition, modified by photochemical alteration (particle oxidation), played a critical role in the PM's reactivity. From a pollution hot spot to a downwind country town, the PM2.5 levels (mean ± SD) were 9.3 ± 4.5, 9.7 ± 4.9, and 6.6 ± 4.7 μg/m3, respectively. In contrast, the PM mass-normalized OP values in the downwind region were approximately 20 % higher than those in the upwind region based on the cellular assay and about three times higher from the acellular assay. Enhanced PM OP is associated with atmospheric oxidation, approximated by sulfur and nitrogen oxidation ratios. We further identified transition metals, particularly copper, a single most important species group, the primary determinant to the values of OP measured, contributing directly to OP and indirectly through metal-oxides enhanced photochemical alterations to PM.
Collapse
Affiliation(s)
- Po-Chao Wu
- Environmental Governance Research Center, National Environmental Research Academy, Taoyuan, Taiwan
| | - Hui-Ju Wen
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan; Institute of Earth Sciences, Academia Sinica, Taipei, Taiwan
| | - Kuo-Fang Huang
- Institute of Earth Sciences, Academia Sinica, Taipei, Taiwan
| | - Shau-Ku Huang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan; Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
| | - Mao-Chang Liang
- Institute of Earth Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
2
|
Eberle OF, Hartung F, Benndorf P, Haarmann-Stemmann T. Skin sensitizers enhance superoxide formation by polycyclic aromatic hydrocarbons via the aldo-keto reductase pathway. Free Radic Biol Med 2025; 230:50-57. [PMID: 39922325 DOI: 10.1016/j.freeradbiomed.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/23/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Exposure to combustion-derived airborne polycyclic aromatic hydrocarbons (PAHs) may harm human skin, exacerbate cutaneous inflammatory diseases and accelerate skin aging. The toxicity of PAHs is unleashed upon their metabolic activation by cytochrome P450 (CYP) 1 monooxygenases, resulting in the formation of reactive intermediates that form mutagenic DNA adducts. Moreover, PAHs cause oxidative stress, which is primarily due to aldo-keto reductases (AKRs), such as AKR1C3, which convert CYP1-derived PAH-trans-diols to PAH-catechols. The catechols undergo autooxidation leading to the formation of reactive oxygen species (ROS) and PAH-quinones. The latter are highly reactive, mitotoxic and are reduced back to PAH-catechols, thus facilitating redox cycling. As AKR1C expression is inducible by other NRF2-stimulating chemicals, we tested the hypothesis that co-exposure of HaCaT keratinocytes to skin sensitizers and the PAH benzo[a]pyrene (BaP) enhances ROS formation. We observed a synergistic effect of the skin sensitizers on the BaP-induced expression of the NRF2 target genes heme oxygenase-1, sulfiredoxin-1 and AKR1C3. In fact, co-exposure to the skin sensitizers also enhanced the BaP-induced formation of superoxide anions. Intriguingly, the co-exposure-related ROS formation was abolished upon inhibition of either CYP1A1 or AKR1C3. Testing of additional skin-sensitizing compounds, differing in their mode of action, indicated that especially potent Michael acceptors enhance the toxicity of BaP by increasing AKR1C3 expression and, presumably, downstream BaP-quinone formation. Our study reveals potential health risks associated with the simultaneous exposure to common skin-sensitizing substances and ubiquitous PAHs, and implies a role for NRF2 in mediating PAH toxicity.
Collapse
Affiliation(s)
- Oliver F Eberle
- IUF - Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany; Henkel AG & Co. KGaA, Henkelstraße 67, 40589, Düsseldorf, Germany
| | - Frederick Hartung
- IUF - Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany
| | - Paul Benndorf
- Henkel AG & Co. KGaA, Henkelstraße 67, 40589, Düsseldorf, Germany
| | - Thomas Haarmann-Stemmann
- IUF - Leibniz-Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany.
| |
Collapse
|
3
|
Rude CI, Wilson LB, La Du J, Lalli PM, Colby SM, Schultz KJ, Smith JN, Waters KM, Tanguay RL. Aryl hydrocarbon receptor-dependent toxicity by retene requires metabolic competence. Toxicol Sci 2024; 202:50-68. [PMID: 39107868 PMCID: PMC11514837 DOI: 10.1093/toxsci/kfae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are a class of organic compounds frequently detected in the environment with widely varying toxicities. Many PAHs activate the aryl hydrocarbon receptor (AHR), inducing the expression of a battery of genes, including xenobiotic metabolizing enzymes like cytochrome P450s (CYPs); however, not all PAHs act via this mechanism. We screened several parent and substituted PAHs in in vitro AHR activation assays to classify their unique activity. Retene (1-methyl-7-isopropylphenanthrene) displays Ahr2-dependent teratogenicity in zebrafish, but did not activate human AHR or zebrafish Ahr2, suggesting a retene metabolite activates Ahr2 in zebrafish to induce developmental toxicity. To investigate the role of metabolism in retene toxicity, studies were performed to determine the functional role of cyp1a, cyp1b1, and the microbiome in retene toxicity, identify the zebrafish window of susceptibility, and measure retene uptake, loss, and metabolite formation in vivo. Cyp1a-null fish were generated using CRISPR-Cas9. Cyp1a-null fish showed increased sensitivity to retene toxicity, whereas Cyp1b1-null fish were less susceptible, and microbiome elimination had no significant effect. Zebrafish required exposure to retene between 24 and 48 hours post fertilization (hpf) to exhibit toxicity. After static exposure, retene concentrations in zebrafish embryos increased until 24 hpf, peaked between 24 and 36 hpf, and decreased rapidly thereafter. We detected retene metabolites at 36 and 48 hpf, indicating metabolic onset preceding toxicity. This study highlights the value of combining molecular and systems biology approaches with mechanistic and predictive toxicology to interrogate the role of biotransformation in AHR-dependent toxicity.
Collapse
Affiliation(s)
- Christian I Rude
- Environmental and Molecular Toxicology Department, Oregon State University, Corvallis, OR 97333, United States
| | - Lindsay B Wilson
- Environmental and Molecular Toxicology Department, Oregon State University, Corvallis, OR 97333, United States
| | - Jane La Du
- Environmental and Molecular Toxicology Department, Oregon State University, Corvallis, OR 97333, United States
| | - Priscila M Lalli
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, United States
| | - Sean M Colby
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, United States
| | - Katherine J Schultz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, United States
| | - Jordan N Smith
- Environmental and Molecular Toxicology Department, Oregon State University, Corvallis, OR 97333, United States
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, United States
| | - Katrina M Waters
- Environmental and Molecular Toxicology Department, Oregon State University, Corvallis, OR 97333, United States
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, United States
| | - Robyn L Tanguay
- Environmental and Molecular Toxicology Department, Oregon State University, Corvallis, OR 97333, United States
| |
Collapse
|
4
|
Park M, Lee S, Lee H, Denna MCFJ, Jang J, Oh D, Bae MS, Jang KS, Park K. New health index derived from oxidative potential and cell toxicity of fine particulate matter to assess its potential health effect. Heliyon 2024; 10:e25310. [PMID: 38356560 PMCID: PMC10864913 DOI: 10.1016/j.heliyon.2024.e25310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
Toxicological data and exposure levels of fine particulate matters (PM2.5) are necessary to better understand their health effects. Simultaneous measurements of PM2.5 oxidative potential (OP) and cell toxicity in urban areas (Beijing, China and Gwangju, Korea) reveal their dependence on chemical composition. Notably, acids (Polar), benzocarboxylic acids, and Pb were the chemical components that affected both OP and cell toxicity. OP varied more significantly among different locations and seasons (winter and summer) than cell toxicity. Using the measured OP, cell toxicity, and PM2.5 concentration, a health index was developed to better assess the potential health effects of PM2.5. The health index was related to the sources of PM2.5 derived from the measured chemical components. The contributions of secondary organic aerosols and dust to the proposed health index were more significant than their contributions to PM2.5 mass. The developed regression equation was used to predict the health effect of PM2.5 without further toxicity measurements. This new index could be a valuable health metric that provides information beyond just the PM2.5 concentration level.
Collapse
Affiliation(s)
- Minhan Park
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Seunghye Lee
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Haebum Lee
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Ma. Cristine Faye J. Denna
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jiho Jang
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Dahye Oh
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Min-Suk Bae
- Department of Environmental Engineering, Mokpo National University, Muan, 58554, Republic of Korea
| | - Kyoung-Soon Jang
- Biomedical Omics Center, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Kihong Park
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| |
Collapse
|
5
|
Zhao C, Li A, Zhang G, Pan Y, Meng L, Yang R, Li Y, Zhang Q, Jiang G. Parent and Halogenated Polycyclic Aromatic Hydrocarbons in the Serum of Coal-Fired Power Plant Workers: Levels, Sex Differences, Accumulation Trends, and Risks. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:12431-12439. [PMID: 36001868 DOI: 10.1021/acs.est.2c03099] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Workers in coal-fired power plants are at a high risk of exposure to polycyclic aromatic hydrocarbons (PAHs) and their halogenated derivatives (HPAHs), yet no studies have investigated such exposure of HPAHs. In this study, 12 PAHs and 8 chlorinated PAHs, but no brominated PAHs, were detected in >80% of serum samples from workers of a coal-fired power plant in eastern China. Serum HPAH concentrations were higher in plant workers (16-273 ng/g lipid) than in people without occupational exposure (12-51 ng/g lipid), and serum PAH and HPAH concentrations both in male and female workers were positively correlated with the occupational exposure duration, with an estimated doubling time of 11-17 years. Correlations were found between concentrations of ∑8HPAHs and ∑12PAHs but not between 7-chlorobenz[a]anthracene (7-ClBaA) and 1-chloropyrene (1-ClPyr) and their respective parent PAHs. In males, total concentrations of PAHs and HPAHs were positively correlated with pulmonary hypofunction and hypertension but not with abnormal electrocardiogram. The benzo[a]pyrene equivalents ratio of ∑8HPAHs/∑12PAHs was 0.3 ± 0.1. Among the HPAHs in the serum, 9-chlorophenanthrene, 7-ClBaA, and 1-ClPyr showed high health risks. This study is the first report on HPAH exposure in coal-fired power plant workers and provides new evidence on the health risks of PAHs and HPAHs in humans.
Collapse
Affiliation(s)
- Chuxuan Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - An Li
- School of Public Health, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Gaoxin Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Key Laboratory of Eco-Environment-Related Polymer Materials Ministry of Education, Key Laboratory of Polymer Materials Ministry of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lan Zhou, Gansu 730070, China
| | - Yiyao Pan
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lingling Meng
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province 250014, China
| | - Ruiqiang Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingming Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qinghua Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
6
|
Almeida-Silva M, Cardoso J, Alemão C, Santos S, Monteiro A, Manteigas V, Marques-Ramos A. Impact of Particles on Pulmonary Endothelial Cells. TOXICS 2022; 10:toxics10060312. [PMID: 35736920 PMCID: PMC9227819 DOI: 10.3390/toxics10060312] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023]
Abstract
According to the WHO, air quality affects around 40 million people, contributing to around 21,000 premature deaths per year. Severe respiratory diseases, such as asthma and chronic obstructive pulmonary disorder, can be promoted by air pollution, which has already been documented; this is one of the reasons why air quality is a very relevant factor for human health and well-being. Aerosols are an aggregation of solid or liquid particles dispersed in the air and can be found in the form of dust or fumes. Aerosols can be easily inhaled or absorbed by the skin, which can lead to adverse health effects according to their sizes that range from the nanometre to the millimetre scale. Based on the PRISMA methodology and using the Rayyan QCRI platform, it was possible to assess more than four hundred research articles. This systematic review study aimed to understand the impact of particles on pulmonary endothelial cells, namely particulate matter in different sizes, cigarette smoke, diesel exhaust particles and carbon black. The main conclusions were that particles induce multiple health effects on endothelial cells, namely endothelial dysfunction, which can lead to apoptosis and necrosis, and it may also cause necroptosis in lung structure.
Collapse
Affiliation(s)
- Marina Almeida-Silva
- HTRC-Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal; (M.A.-S.); (J.C.); (C.A.); (S.S.); (A.M.); (V.M.)
| | - Jéssica Cardoso
- HTRC-Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal; (M.A.-S.); (J.C.); (C.A.); (S.S.); (A.M.); (V.M.)
| | - Catarina Alemão
- HTRC-Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal; (M.A.-S.); (J.C.); (C.A.); (S.S.); (A.M.); (V.M.)
| | - Sara Santos
- HTRC-Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal; (M.A.-S.); (J.C.); (C.A.); (S.S.); (A.M.); (V.M.)
| | - Ana Monteiro
- HTRC-Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal; (M.A.-S.); (J.C.); (C.A.); (S.S.); (A.M.); (V.M.)
- Centro de Ciências e Tecnologias Nucleares (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, ao Km 139.7, 2695-066 Bobadela-Loures, Portugal
| | - Vítor Manteigas
- HTRC-Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal; (M.A.-S.); (J.C.); (C.A.); (S.S.); (A.M.); (V.M.)
- Centro de Ciências e Tecnologias Nucleares (C2TN), Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, ao Km 139.7, 2695-066 Bobadela-Loures, Portugal
| | - Ana Marques-Ramos
- HTRC-Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal; (M.A.-S.); (J.C.); (C.A.); (S.S.); (A.M.); (V.M.)
- Correspondence: ; Tel.: +351-966087971
| |
Collapse
|
7
|
Vogeley C, Rolfes KM, Krutmann J, Haarmann-Stemmann T. The Aryl Hydrocarbon Receptor in the Pathogenesis of Environmentally-Induced Squamous Cell Carcinomas of the Skin. Front Oncol 2022; 12:841721. [PMID: 35311158 PMCID: PMC8927079 DOI: 10.3389/fonc.2022.841721] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/09/2022] [Indexed: 01/05/2023] Open
Abstract
Cutaneous squamous cell carcinoma (SCC) is one of the most frequent malignancies in humans and academia as well as public authorities expect a further increase of its incidence in the next years. The major risk factor for the development of SCC of the general population is the repeated and unprotected exposure to ultraviolet (UV) radiation. Another important risk factor, in particular with regards to occupational settings, is the chronic exposure to polycyclic aromatic hydrocarbons (PAH) which are formed during incomplete combustion of organic material and thus can be found in coal tar, creosote, bitumen and related working materials. Importantly, both exposomal factors unleash their carcinogenic potential, at least to some extent, by activating the aryl hydrocarbon receptor (AHR). The AHR is a ligand-dependent transcription factor and key regulator in xenobiotic metabolism and immunity. The AHR is expressed in all cutaneous cell-types investigated so far and maintains skin integrity. We and others have reported that in response to a chronic exposure to environmental stressors, in particular UV radiation and PAHs, an activation of AHR and downstream signaling pathways critically contributes to the development of SCC. Here, we summarize the current knowledge about AHR's role in skin carcinogenesis and focus on its impact on defense mechanisms, such as DNA repair, apoptosis and anti-tumor immune responses. In addition, we discuss the possible consequences of a simultaneous exposure to different AHR-stimulating environmental factors for the development of cutaneous SCC.
Collapse
Affiliation(s)
- Christian Vogeley
- IUF - Leibniz-Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Katharina M Rolfes
- IUF - Leibniz-Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Jean Krutmann
- IUF - Leibniz-Research Institute for Environmental Medicine, Düsseldorf, Germany
| | | |
Collapse
|
8
|
Pandit S, Singh P, Parthasarathi R. Computational risk assessment framework for the hazard analysis of bisphenols and quinone metabolites. JOURNAL OF HAZARDOUS MATERIALS 2022; 426:128031. [PMID: 34933259 DOI: 10.1016/j.jhazmat.2021.128031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/17/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
Bisphenol A (BPA) is a widely used chemical in plastics but its proven harmful effects has led to the replacement and production of its analogs that might also induce hazard as well as associated risks. To elucidate the adverse impact of the BPA analogs, a comprehensive computational framework is developed which applies toxicogenomics aligned with Density Functional Theory (DFT) and Molecular Dynamics (MD) based approaches to understand the toxic potential of quinone metabolites of Bisphenol F (BPF) and 3,3'-dimethylbisphenol A (DMBPA). The obtained results indicate a similar chemical reactivity profile for these metabolites of bisphenols to BPA metabolite. MD simulation revealed that the quinone metabolites tend to interact with the DNA comprising hydrogen bonding, van der Waals forces, and electrostatic interactions as an onset for covalent binding to adduct formation. Structural analysis suggests that interactions with DC9, DG10, DG16, DA17, DA18, and DT19 play a crucial role in stabilizing the quinone metabolite in the interactive pocket of DNA. These observations are demonstrating that BPF and DMBPA have the potential to impose genotoxicity via forming the quinone metabolite adducts. Combination of DFT and MD-based computational approaches providing a structure-activity-toxicity spectrum of chemicals can serve for the purpose of risk assessment.
Collapse
Affiliation(s)
- Shraddha Pandit
- Computational Toxicology Facility, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Prakrity Singh
- Computational Toxicology Facility, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ramakrishnan Parthasarathi
- Computational Toxicology Facility, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
9
|
Bukowska B, Duchnowicz P. Molecular Mechanisms of Action of Selected Substances Involved in the Reduction of Benzo[a]pyrene-Induced Oxidative Stress. Molecules 2022; 27:molecules27041379. [PMID: 35209168 PMCID: PMC8878767 DOI: 10.3390/molecules27041379] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/07/2022] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
Benzo[a]pyrene (BaP) is a polycyclic aromatic hydrocarbon (PAH) primarily formed by burning of fossil fuels, wood and other organic materials. BaP as group I carcinogen shows mutagenic and carcinogenic effects. One of the important mechanisms of action of (BaP) is its free radical activity, the effect of which is the induction of oxidative stress in cells. BaP induces oxidative stress through the production of reactive oxygen species (ROS), disturbances of the activity of antioxidant enzymes, and the reduction of the level of non-enzymatic antioxidants as well as of cytokine production. Chemical compounds, such as vitamin E, curcumin, quercetin, catechin, cyanidin, kuromanin, berberine, resveratrol, baicalein, myricetin, catechin hydrate, hesperetin, rhaponticin, as well as taurine, atorvastatin, diallyl sulfide, and those contained in green and white tea, lower the oxidative stress induced by BaP. They regulate the expression of genes involved in oxidative stress and inflammation, and therefore can reduce the level of ROS. These substances remove ROS and reduce the level of lipid and protein peroxidation, reduce formation of adducts with DNA, increase the level of enzymatic and non-enzymatic antioxidants and reduce the level of pro-inflammatory cytokines. BaP can undergo chemical modification in the living cells, which results in more reactive metabolites formation. Some of protective substances have the ability to reduce BaP metabolism, and in particular reduce the induction of cytochrome (CYP P450), which reduces the formation of oxidative metabolites, and therefore decreases ROS production. The aim of this review is to discuss the oxidative properties of BaP, and describe protective activities of selected chemicals against BaP activity based on of the latest publications.
Collapse
|
10
|
Vogeley C, Sondermann NC, Woeste S, Momin AA, Gilardino V, Hartung F, Heinen M, Maaß SK, Mescher M, Pollet M, Rolfes KM, Vogel CFA, Rossi A, Lang D, Arold ST, Nakamura M, Haarmann-Stemmann T. Unraveling the differential impact of PAHs and dioxin-like compounds on AKR1C3 reveals the EGFR extracellular domain as a critical determinant of the AHR response. ENVIRONMENT INTERNATIONAL 2022; 158:106989. [PMID: 34991250 PMCID: PMC8852774 DOI: 10.1016/j.envint.2021.106989] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/14/2021] [Accepted: 11/16/2021] [Indexed: 06/14/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs), dioxin-like compounds (DLCs) and structurally-related environmental pollutants may contribute to the pathogenesis of various diseases and disorders, primarily by activating the aryl hydrocarbon receptor (AHR) and modulating downstream cellular responses. Accordingly, AHR is considered an attractive molecular target for preventive and therapeutic measures. However, toxicological risk assessment of AHR-modulating compounds as well as drug development is complicated by the fact that different ligands elicit remarkably different AHR responses. By elucidating the differential effects of PAHs and DLCs on aldo-keto reductase 1C3 expression and associated prostaglandin D2 metabolism, we here provide evidence that the epidermal growth factor receptor (EGFR) substantially shapes AHR ligand-induced responses in human epithelial cells, i.e. primary and immortalized keratinocytes and breast cancer cells. Exposure to benzo[a]pyrene (B[a]P) and dioxin-like polychlorinated biphenyl (PCB) 126 resulted in a rapid c-Src-mediated phosphorylation of EGFR. Moreover, both AHR agonists stimulated protein kinase C activity and enhanced the ectodomain shedding of cell surface-bound EGFR ligands. However, only upon B[a]P treatment, this process resulted in an auto-/paracrine activation of EGFR and a subsequent induction of aldo-keto reductase 1C3 and 11-ketoreduction of prostaglandin D2. Receptor binding and internalization assays, docking analyses and mutational amino acid exchange confirmed that DLCs, but not B[a]P, bind to the EGFR extracellular domain, thereby blocking EGFR activation by growth factors. Finally, nanopore long-read RNA-seq revealed hundreds of genes, whose expression is regulated by B[a]P, but not by PCB126, and sensitive towards pharmacological EGFR inhibition. Our data provide novel mechanistic insights into the ligand response of AHR signaling and identify EGFR as an effector of environmental chemicals.
Collapse
Affiliation(s)
- Christian Vogeley
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Natalie C Sondermann
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Selina Woeste
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Afaque A Momin
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia
| | - Viola Gilardino
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Frederick Hartung
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Markus Heinen
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Sophia K Maaß
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Melina Mescher
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Marius Pollet
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Katharina M Rolfes
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Christoph F A Vogel
- Department of Environmental Toxicology and Center for Health and the Environment, University of California, Davis, CA 95616, USA
| | - Andrea Rossi
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Dieter Lang
- Bayer AG, Pharmaceuticals, Research Center, 42096 Wuppertal, Germany
| | - Stefan T Arold
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal 23955-6900, Saudi Arabia; Centre de Biologie Structurale (CBS), INSERM, CNRS, Université de Montpellier, F-34090 Montpellier, France
| | - Motoki Nakamura
- IUF - Leibniz-Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany; Department of Environmental and Geriatric Dermatology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | | |
Collapse
|
11
|
Jeng HA, Sikdar S. Mixture Analyses on Polycyclic Aromatic Hydrocarbons: An Example Using Semen Quality Biomarkers and Bayesian Kernel Machine Regression. Polycycl Aromat Compd 2021. [DOI: 10.1080/10406638.2021.2005640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Hueiwang Anna Jeng
- School of Community and Environmental Health, College of Health Sciences, Old Dominion University, Norfolk, VA, USA
| | - Sinjini Sikdar
- Department of Mathematics and Statistics, College of Sciences, Old Dominion University, Norfolk, VA, USA
| |
Collapse
|
12
|
Fujii J, Homma T, Miyata S, Takahashi M. Pleiotropic Actions of Aldehyde Reductase (AKR1A). Metabolites 2021; 11:343. [PMID: 34073440 PMCID: PMC8227408 DOI: 10.3390/metabo11060343] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
We provide an overview of the physiological roles of aldehyde reductase (AKR1A) and also discuss the functions of aldose reductase (AKR1B) and other family members when necessary. Many types of aldehyde compounds are cytotoxic and some are even carcinogenic. Such toxic aldehydes are detoxified via the action of AKR in an NADPH-dependent manner and the resulting products may exert anti-diabetic and anti-tumorigenic activity. AKR1A is capable of reducing 3-deoxyglucosone and methylglyoxal, which are reactive intermediates that are involved in glycation, a non-enzymatic glycosylation reaction. Accordingly, AKR1A is thought to suppress the formation of advanced glycation end products (AGEs) and prevent diabetic complications. AKR1A and, in part, AKR1B are responsible for the conversion of d-glucuronate to l-gulonate which constitutes a process for ascorbate (vitamin C) synthesis in competent animals. AKR1A is also involved in the reduction of S-nitrosylated glutathione and coenzyme A and thereby suppresses the protein S-nitrosylation that occurs under conditions in which the production of nitric oxide is stimulated. As the physiological functions of AKR1A are currently not completely understood, the genetic modification of Akr1a could reveal the latent functions of AKR1A and differentiate it from other family members.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan;
| | - Takujiro Homma
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan;
| | - Satoshi Miyata
- Miyata Diabetes and Metabolism Clinic, 5-17-21 Fukushima, Fukushima-ku, Osaka 553-0003, Japan;
| | - Motoko Takahashi
- Department of Biochemistry, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan;
| |
Collapse
|
13
|
Ku CC, Wuputra K, Kato K, Lin WH, Pan JB, Tsai SC, Kuo CJ, Lee KH, Lee YL, Lin YC, Saito S, Noguchi M, Nakamura Y, Miyoshi H, Eckner R, Nagata K, Wu DC, Lin CS, Yokoyama KK. Jdp2-deficient granule cell progenitors in the cerebellum are resistant to ROS-mediated apoptosis through xCT/Slc7a11 activation. Sci Rep 2020; 10:4933. [PMID: 32188872 PMCID: PMC7080836 DOI: 10.1038/s41598-020-61692-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
The Jun dimerization protein 2 (Jdp2) is expressed predominantly in granule cell progenitors (GCPs) in the cerebellum, as was shown in Jdp2-promoter-Cre transgenic mice. Cerebellum of Jdp2-knockout (KO) mice contains lower number of Atoh-1 positive GCPs than WT. Primary cultures of GCPs from Jdp2-KO mice at postnatal day 5 were more resistant to apoptosis than GCPs from wild-type mice. In Jdp2-KO GCPs, the levels of both the glutamate‒cystine exchanger Sc7a11 and glutathione were increased; by contrast, the activity of reactive oxygen species (ROS) was decreased; these changes confer resistance to ROS-mediated apoptosis. In the absence of Jdp2, a complex of the cyclin-dependent kinase inhibitor 1 (p21Cip1) and Nrf2 bound to antioxidant response elements of the Slc7a11 promoter and provide redox control to block ROS-mediated apoptosis. These findings suggest that an interplay between Jdp2, Nrf2, and p21Cip1 regulates the GCP apoptosis, which is one of critical events for normal development of the cerebellum.
Collapse
Affiliation(s)
- Chia-Chen Ku
- Graduate Institute of Medicine, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.).,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.)
| | - Kenly Wuputra
- Graduate Institute of Medicine, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.).,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.)
| | - Kohsuke Kato
- Department of Infection Biology, Graduate School of Comprehensive Human Sciences, The University of Tsukuba, 305-8577, Tsukuba, Ibaraki, Japan
| | - Wen-Hsin Lin
- Graduate Institute of Medicine, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.).,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.)
| | - Jia-Bin Pan
- Graduate Institute of Medicine, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.).,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.)
| | - Shih-Chieh Tsai
- National Laboratory Animal Center, National Applied Research Laboratories (NARLabs), Xinshi Dist., 74147, Tainan, Taiwan (R.O.C.).,Founder of Gecoll Biomedicine Co. Ltd., Xinshi Dist., 744, Tainan, Taiwan (R.O.C.)
| | - Che-Jung Kuo
- National Laboratory Animal Center, National Applied Research Laboratories (NARLabs), Xinshi Dist., 74147, Tainan, Taiwan (R.O.C.)
| | - Kan-Hung Lee
- National Laboratory Animal Center, National Applied Research Laboratories (NARLabs), Nangang Dist., 11599, Taipei, Taiwan (R.O.C.)
| | - Yan-Liang Lee
- Welgene Biotech., Inc., 11503, Taipei, Taiwan (R.O.C.)
| | - Ying-Chu Lin
- School of Dentistry, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan
| | - Shigeo Saito
- Saito Laboratory of Cell Technology, Yaita, 329-2192, Tochigi, Japan.,Waseda Research Institute for Science & Engineering, Waseda University, 169-0051, Tokyo, Japan
| | - Michiya Noguchi
- Cell Engineering Division, RIKEN BioResource Research Center, 305-0074, Tsukuba, Ibaraki, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Research Center, 305-0074, Tsukuba, Ibaraki, Japan
| | - Hiroyuki Miyoshi
- Graduate Institute of Medicine, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.).,Department of Physiology, Keio University School of Medicine, Shinanaomachi, 168-8582, Tokyo, Japan
| | - Richard Eckner
- Departent of. Biochemistry & Molecular Biology, Rutgers New Jersey Medical School, The State University of New Jersey, 07-103, Newark, NJ, USA
| | - Kyosuke Nagata
- Department of Infection Biology, Graduate School of Comprehensive Human Sciences, The University of Tsukuba, 305-8577, Tsukuba, Ibaraki, Japan
| | - Deng-Chyang Wu
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.).,Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, 80708, Kaohsiung, Taiwan (R.O.C.)
| | - Chang-Shen Lin
- Graduate Institute of Medicine, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.). .,Department of Biological Sciences, National Sun Yat-sen University, 80424, Kaohsiung, Taiwan (R.O.C.).
| | - Kazunari K Yokoyama
- Graduate Institute of Medicine, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.). .,Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan (R.O.C.). .,Department of Molecular Preventive Medicine, Graduate School of Medicine, The University of Tokyo, 113-8655, Tokyo, Japan.
| |
Collapse
|
14
|
Interactions of oxidative DNA damage and CYP1A gene expression with the liver enzymes in Klunzinger's mullet exposed to benzo[ a]pyrene. Toxicol Rep 2019; 6:1097-1103. [PMID: 31720230 PMCID: PMC6839019 DOI: 10.1016/j.toxrep.2019.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/30/2022] Open
Abstract
Benzo[a]pyrene (B[a]P) is an important contaminant whose liver biotransformation is dependent on the species, the route of exposure and the concentration. The goal of this study was to assess the interactions of oxidative DNA damage and CYP1A gene expression with the liver enzymes in Klunzinger’s mullet (Liza klunzingeri) exposed to benzo[a]pyrene. Sublethal doses of B[a]P (5, 10 and 50 mg/kg) were intraperitoneally administered to the fish for 14 days. The alterations in antioxidant enzymes’ activity (SOD, CAT, and GPX), hepatic enzymes’ activity (ALT, AST and ALP), DNA damage (measured by comet assay and cellProfiler software) and CYP1A gene expression in the fish liver were studied on the 1st, 3rd, 7th and 14th days. The determination of these parameters in the liver showed that most of these parameters significantly increased mostly in a time-dependent manner. Multiple regression analysis showed that DNA damage and CYP1A gene expression had positive correlations with the liver enzymes in this fish species intraperitoneally exposed to these concentrations. Moreover, these interactions indicated that theses parameters are sensitive biomarkers for the exposure to B[a]P in Klunzinger's mullet. However, other possible factors and B[a]P metabolites should be considered in future studies for better elucidating the biotransformation mechanisms and introducing better biomarkers of B[a]P.
Collapse
|
15
|
Wang Y, Peng A, Chen Z, Jin X, Gu C. Transformation of gaseous 2-bromophenol on clay mineral dust and the potential health effect. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 250:686-694. [PMID: 31035151 DOI: 10.1016/j.envpol.2019.04.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/22/2019] [Accepted: 04/15/2019] [Indexed: 06/09/2023]
Abstract
Iron-bearing clays are ubiquitously distributed as mineral dusts in the atmosphere. Bromophenols were reported as the major products from thermal decomposition of the widely used brominated flame retardants (BFRs). However, little information is available for the reactivity of iron associated with mineral dusts to interact with the atmospheric bromophenols and the subsequent toxic effects. Herein, three common clay minerals (montmorillonite, illite and kaolinite) were used to simulate mineral dusts, and the reactions with gaseous 2-bromophenol were systematically investigated under environmentally relevant atmospheric conditions. Our results demonstrate that structural Fe(III) in montmorillonite and Fe(III) from iron oxide in illite mediated the dimerization of 2-bromophenol to form hydroxylated polybrominated biphenyl and hydroxylated polybrominated diphenyl ether. The surface reaction is favored to occur at moisture environment, since water molecules formed complex with 2-bromophenol and the reaction intermediates via hydrogen bond to significantly lower the reaction energy and promote the dimerization reaction. More importantly, the formed dioxin-like products on clay mineral dust increased the toxicity of the particles to A549 lung cell by decreasing cell survival and damaging cellular membrane and proteins. The results of this study indicate that not only mineral dust itself but also the associated surface reaction should be fully considered to accurately evaluate the toxic effect of mineral dust on human health.
Collapse
Affiliation(s)
- Yi Wang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Anping Peng
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Zeyou Chen
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Xin Jin
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Cheng Gu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu, 210023, PR China.
| |
Collapse
|
16
|
Tylichová Z, Neča J, Topinka J, Milcová A, Hofmanová J, Kozubík A, Machala M, Vondráček J. n-3 Polyunsaturated fatty acids alter benzo[a]pyrene metabolism and genotoxicity in human colon epithelial cell models. Food Chem Toxicol 2018; 124:374-384. [PMID: 30572064 DOI: 10.1016/j.fct.2018.12.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 11/30/2018] [Accepted: 12/14/2018] [Indexed: 12/23/2022]
Abstract
Dietary carcinogens, such as benzo[a]pyrene (BaP), are suspected to contribute to colorectal cancer development. n-3 Polyunsaturated fatty acids (PUFAs) decrease colorectal cancer risk in individuals consuming diets rich in PUFAs. Here, we investigated the impact of eicosapentaenoic (EPA) and docosahexaenoic (DHA) acid on metabolism and genotoxicity of BaP in human cell models derived from the colon: HT-29 and HCT-116 cell lines. Both PUFAs reduced levels of excreted BaP metabolites, in particular BaP-tetrols and hydroxylated BaP metabolites, as well as formation of DNA adducts in HT-29 and HCT-116 cells. However, EPA appeared to be a more potent inhibitor of formation of some intracellular BaP metabolites, including BaP-7,8-dihydrodiol. EPA also reduced phosphorylation of histone H2AX (Ser139) in HT-29 cells, which indicated that it may reduce further forms of DNA damage, including DNA double strand breaks. Both PUFAs inhibited induction of CYP1 activity in colon cells determined as 7-ethoxyresorufin-O-deethylase (EROD); this was at least partly linked with inhibition of induction of CYP1A1, 1A2 and 1B1 mRNAs. The downregulation and/or inhibition of CYP1 enzymes by PUFAs could thus alter metabolism and reduce genotoxicity of BaP in human colon cells, which might contribute to known chemopreventive effects of PUFAs in colon epithelium.
Collapse
Affiliation(s)
- Zuzana Tylichová
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Jiří Neča
- Veterinary Research Institute, Brno, Czech Republic
| | - Jan Topinka
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alena Milcová
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiřina Hofmanová
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Alois Kozubík
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | | | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic.
| |
Collapse
|
17
|
Deng C, Dang F, Gao J, Zhao H, Qi S, Gao M. Acute benzo[a]pyrene treatment causes different antioxidant response and DNA damage in liver, lung, brain, stomach and kidney. Heliyon 2018; 4:e00898. [PMID: 30456321 PMCID: PMC6234523 DOI: 10.1016/j.heliyon.2018.e00898] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 05/08/2018] [Accepted: 10/25/2018] [Indexed: 01/22/2023] Open
Abstract
Acute effects of oxidative damage induced by benzo[a]pyrene (B[a]P) on various organs are still not clear. In this study, we investigated oxidative stress and DNA damage in liver, lung, stomach, brain and kidney of ICR male mice induced by acute B[a]P treatment. B[a]P treatment led to a significant decrease at the different doses in body weight. For the variations of superoxide dismutase (SOD), catalase (CAT), glutathione S-transferase (GST), glutathione (GSH) and GSH/GSSG, significant increases were observed at 24 h, then decreased till 72 h after B[a]P injection. The increase percent indicated in a dose- dependent decrease manner. However, glutathione peroxidase (GPx), GSSG and MDA were significantly increased in a time- and dose-dependent increase manner. DNA damage showed the significant and top levels at 24 h, and increased in proportion to the doses of B[a]P treatment. The total induction could be indicated by the variation of MDA at 24 h after B[a]P injection and showed the following order of predominance: lung > liver > kidney = stomach > brain. This was further certificated by histopathological changes in the examined organs. Additionally, the levels of serum glutamic-oxaloacetic transaminase (GOT), glutamic-pyruvic transaminase (GPT), and blood urea nitrogen (UN), creatinine were also significantly increased at 24 h after B[a]P injection. These findings suggested the disturbance of antioxidant responses and aggravation of DNA damages, and the different responses on various organs induced by acute B[a]P treatment in organism.
Collapse
Affiliation(s)
- Chun Deng
- Department of Biological Science and Engineering, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710049, China
| | - Fan Dang
- Department of Biological Science and Engineering, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710049, China
| | - Jianghong Gao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Deparment of Preventive Dentistry, Colleage of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710004, China
| | - Hongyan Zhao
- Department of Biological Science and Engineering, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710049, China
| | - Shuyan Qi
- Department of Biological Science and Engineering, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710049, China
| | - Meili Gao
- Department of Biological Science and Engineering, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710049, China
| |
Collapse
|
18
|
Andersen MHG, Saber AT, Clausen PA, Pedersen JE, Løhr M, Kermanizadeh A, Loft S, Ebbehøj N, Hansen ÅM, Pedersen PB, Koponen IK, Nørskov EC, Møller P, Vogel U. Association between polycyclic aromatic hydrocarbon exposure and peripheral blood mononuclear cell DNA damage in human volunteers during fire extinction exercises. Mutagenesis 2018; 33:105-115. [PMID: 29045708 DOI: 10.1093/mutage/gex021] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 09/11/2017] [Indexed: 11/12/2022] Open
Abstract
This study investigated a number of biomarkers, associated with systemic inflammation as well as genotoxicity, in 53 young and healthy subjects participating in a course to become firefighters, while wearing personal protective equipment (PPE). The exposure period consisted of a 3-day training course where the subjects participated in various live-fire training exercises. The subjects were instructed to extinguish fires of either wood or wood with electrical cords and mattresses. The personal exposure was measured as dermal polycyclic aromatic hydrocarbon (PAH) concentrations and urinary excretion of 1-hydroxypyrene (1-OHP). The subjects were primarily exposed to particulate matter (PM) in by-stander positions, since the self-contained breathing apparatus effectively prevented pulmonary exposure. There was increased dermal exposure to pyrene (68.1%, 95% CI: 52.5%, 83.8%) and sum of 16 polycyclic aromatic hydrocarbons (ƩPAH; 79.5%, 95% CI: 52.5%, 106.6%), and increased urinary excretion of 1-OHP (70.4%, 95% CI: 52.5%; 106.6%) after the firefighting exercise compared with the mean of two control measurements performed 2 weeks before and 2 weeks after the firefighting course, respectively. The level of Fpg-sensitive sites in peripheral blood mononuclear cells (PBMCs) was increased by 8.0% (95% CI: 0.02%, 15.9%) compared with control measurements. The level of DNA strand breaks was positively associated with dermal exposure to pyrene and ƩPAHs, and urinary excretion of 1-OHP. Fpg-sensitive sites were only associated positively with PAHs. Biomarkers of inflammation and lung function showed no consistent response. In summary, the study demonstrated that PAH exposure during firefighting activity was associated with genotoxicity in PBMCs.
Collapse
Affiliation(s)
- Maria Helena Guerra Andersen
- Department of Public Health, Section of Environmental Health, University of Copenhagen, Øster Farimagsgade, Copenhagen K, Denmark
| | - Anne Thoustrup Saber
- The National Research Centre for the Working Environment, Lersø Parkalle, Copenhagen Ø, Denmark
| | - Per Axel Clausen
- The National Research Centre for the Working Environment, Lersø Parkalle, Copenhagen Ø, Denmark
| | - Julie Elbæk Pedersen
- The National Research Centre for the Working Environment, Lersø Parkalle, Copenhagen Ø, Denmark
| | - Mille Løhr
- Department of Public Health, Section of Environmental Health, University of Copenhagen, Øster Farimagsgade, Copenhagen K, Denmark
| | - Ali Kermanizadeh
- Department of Public Health, Section of Environmental Health, University of Copenhagen, Øster Farimagsgade, Copenhagen K, Denmark
| | - Steffen Loft
- Department of Public Health, Section of Environmental Health, University of Copenhagen, Øster Farimagsgade, Copenhagen K, Denmark
| | - Niels Ebbehøj
- Department of Occupational and Environmental Medicine, Bispebjerg Hospital, Bispebjerg Bakke, Copenhagen NV, Denmark
| | - Åse Marie Hansen
- The National Research Centre for the Working Environment, Lersø Parkalle, Copenhagen Ø, Denmark.,Department of Public Health, Section of Social Medicine, University of Copenhagen, Øster Farimagsgade, Copenhagen K, Denmark
| | - Peter Bøgh Pedersen
- Danish Technological Institute, Teknologiparken, Kongsvang Allé, Aarhus C, Denmark
| | - Ismo Kalevi Koponen
- The National Research Centre for the Working Environment, Lersø Parkalle, Copenhagen Ø, Denmark
| | - Eva-Carina Nørskov
- Danish Technological Institute, Teknologiparken, Kongsvang Allé, Aarhus C, Denmark
| | - Peter Møller
- Department of Public Health, Section of Environmental Health, University of Copenhagen, Øster Farimagsgade, Copenhagen K, Denmark
| | - Ulla Vogel
- Department of Micro- and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
19
|
A lincRNA-p21/miR-181 family feedback loop regulates microglial activation during systemic LPS- and MPTP- induced neuroinflammation. Cell Death Dis 2018; 9:803. [PMID: 30038357 PMCID: PMC6056543 DOI: 10.1038/s41419-018-0821-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/12/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022]
Abstract
The role of microglial-mediated sustained neuroinflammation in the onset and progression of Parkinson’s disease (PD) is well established, but the mechanisms contributing to microglial activation remain unclear. LincRNA-p21, a well studied long intergenic noncoding RNA (lincRNA), plays pivotal roles in diverse biological processes and diseases. Its role in microglial activation and inflammation-induced neurotoxicity, however, has not yet been fully elucidated. Here, we report that lincRNA-p21 promotes microglial activation through a p53-dependent transcriptional pathway. We further demonstrate that lincRNA-p21 competitively binds to the miR-181 family and induces microglial activation through the miR-181/PKC-δ pathway. Moreover, PKC-δ induction further increases the expression of p53/lincRNA-p21 and thus forms a circuit. Taken together, our results suggest that p53/lincRNA-p21, together with miR-181/PKC-δ, form a double-negative feedback loop that facilitates sustained microglial activation and the deterioration of neurodegeneration.
Collapse
|
20
|
Zhou Y, Liu Y, Sun H, Ma J, Xiao L, Cao L, Li W, Wang B, Yuan J, Chen W. Associations of urinary polycyclic aromatic hydrocarbon metabolites with fractional exhaled nitric oxide and exhaled carbon monoxide: A cross-sectional study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 618:542-550. [PMID: 29149738 DOI: 10.1016/j.scitotenv.2017.10.294] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/26/2017] [Accepted: 10/28/2017] [Indexed: 06/07/2023]
Abstract
Exposure to Polycyclic aromatic hydrocarbons (PAHs) has been associated with inflammatory responses. Fractional exhaled nitric oxide (FeNO) and exhaled carbon monoxide (eCO) are both important inflammatory mediators especially in airways. However, few studies have investigated associations of PAH exposures with FeNO or eCO. Therefore, we aimed to quantify the associations of urinary PAH metabolites with FeNO and eCO levels, and investigate their potential effect modifiers by linear mixed models among 4133 participants from the Wuhan-Zhuhai cohort in China. We further performed stratified analyses to estimate effect modification. We found significant associations of increased urinary PAH metabolites with elevated eCO and FeNO. Among all participants, each 1% increase of 1-hydroxynaphthalene, 2-hydroxynaphthalene, 2-hydroxyfluorene, 4-hydroxyphenanthrene, 3-hydroxyphenanthrene, and total PAH metabolites was significantly associated with a 12.6% (95% confidence interval: 9.3%, 15.9%), 9.7% (6.5%, 12.9%), 7.5% (4.1%, 10.9%), 3.2% (0.2%, 6.2%), 2.7% (0.1%, 5.3%), and 6.5% (2.7%, 10.4%) increased eCO level, respectively; while each 1% increase of urinary 1-hydroxynaphthalene, 9-hydroxyphenanthrene, 3-hydroxyphenanthrene, and 2-hydroxyphenanthrene was associated with a -3.0% (-5.8%, -0.2%), 2.9% (0.3%, 5.6%), 3.2% (1.0%, 5.4%), and 4.5% (2.2%, 6.9%) change of FeNO level, respectively. Positive associations between certain urinary PAH metabolites and eCO were observed among both ever-smokers and non-smokers, and the associations were stronger among ever-smokers than that among non-smokers. Increased urinary PAH metabolites were associated with decreased FeNO among ever-smokers and elevated FeNO levels among non-smokers. Our findings suggest that PAH exposures may impair airway through inducing inflammatory response, especially among ever-smokers.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yuewei Liu
- Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, Wuhan, Hubei 430079, China
| | - Huizhen Sun
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jixuan Ma
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Lili Xiao
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Limin Cao
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wei Li
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Bin Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jing Yuan
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Weihong Chen
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
21
|
Bai H, Wu M, Zhang H, Tang G. Chronic polycyclic aromatic hydrocarbon exposure causes DNA damage and genomic instability in lung epithelial cells. Oncotarget 2017; 8:79034-79045. [PMID: 29108285 PMCID: PMC5668018 DOI: 10.18632/oncotarget.20891] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 08/26/2017] [Indexed: 11/25/2022] Open
Abstract
Cell exposure to atmospheric polycyclic aromatic hydrocarbons (PAHs) is closely associated with DNA damage and genomic instability. We assessed the mechanisms of chronic and acute PAH exposure-induced genotoxicity in two human lung epithelial cell lines, A549 and NC-H1975. We sampled atmospheric PAHs at the Xixi Campus, Zhejiang University in Hangzhou, China during August (non-haze episode) and November (haze episode) 2015. We identified vehicle emissions as a dominant anthropogenic PAH source in our study. PAHs were extracted according to the United States Environmental Protection Agency Method TO-13A. We found that chronic PAH exposure saturated lung cell xenobiotic metabolic pathways, promoting intercellular reactive oxygen species production and accumulation. Chronic alteration of the cellular redox status resulted in DNA damage and genomic instability. Chronic PAH exposure also perturbed the cellular DNA damage response, inducing S phase arrest and inhibiting apoptosis. Dysregulation of PAH metabolism and the DNA damage response altered cellular homeostasis and increased cell susceptibility to subsequent PAH exposures, thereby enhancing the likelihood of genomic mutation and instability.
Collapse
Affiliation(s)
- Hongzhen Bai
- State Key Laboratory of Industrial Control Technology, College of Control Science and Engineering, Zhejiang University, Hangzhou 310028, China.,Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou 310028, China
| | - Min Wu
- Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou 310028, China
| | - Hongjian Zhang
- State Key Laboratory of Industrial Control Technology, College of Control Science and Engineering, Zhejiang University, Hangzhou 310028, China
| | - Guping Tang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou 310028, China
| |
Collapse
|
22
|
Penning TM. Genotoxicity of ortho-quinones: reactive oxygen species versus covalent modification. Toxicol Res (Camb) 2017. [PMID: 29527287 DOI: 10.1039/c7tx00223h] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
o-Quinones are formed metabolically from natural and synthetic estrogens as well as upon exposure to polycyclic aromatic hydrocarbons (PAH) and contribute to estrogen and PAH carcinogenesis by genotoxic mechanisms. These mechanisms include the production of reactive oxygen species to produce DNA strand breaks and oxidatively damaged nucleobases; and the formation of covalent depurinating and stable DNA adducts. Unrepaired DNA-lesions can lead to mutation in critical growth control genes and cellular transformation. The genotoxicity of the o-quinones is exacerbated by nuclear translocation of estrogen o-quinones by the estrogen receptor and by the nuclear translocation of PAH o-quinones by the aryl hydrocarbon receptor. The properties of o-quinones, their formation and detoxication mechanisms, quinone-mediated DNA lesions and their mutagenic properties support an important role in hormonal and chemical carcinogenesis.
Collapse
Affiliation(s)
- Trevor M Penning
- Center of Excellence in Environmental Toxicology and Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| |
Collapse
|
23
|
Marabini L, Ozgen S, Turacchi S, Aminti S, Arnaboldi F, Lonati G, Fermo P, Corbella L, Valli G, Bernardoni V, Dell’Acqua M, Vecchi R, Becagli S, Caruso D, Corrado GL, Marinovich M. Ultrafine particles (UFPs) from domestic wood stoves: genotoxicity in human lung carcinoma A549 cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2017; 820:39-46. [DOI: 10.1016/j.mrgentox.2017.06.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/01/2017] [Accepted: 06/05/2017] [Indexed: 10/19/2022]
|
24
|
Penning TM. Aldo-Keto Reductase Regulation by the Nrf2 System: Implications for Stress Response, Chemotherapy Drug Resistance, and Carcinogenesis. Chem Res Toxicol 2017; 30:162-176. [PMID: 27806574 PMCID: PMC5241174 DOI: 10.1021/acs.chemrestox.6b00319] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human aldo-keto reductases (AKRs) are NAD(P)H-dependent oxidoreductases that convert aldehydes and ketones to primary and secondary alcohols for subsequent conjugation reactions and can be referred to as "phase 1" enzymes. Among all the human genes regulated by the Keap1/Nrf2 pathway, they are consistently the most overexpressed in response to Nrf2 activators. Although these enzymes play clear cytoprotective roles and deal effectively with carbonyl stress, their upregulation by the Keap1/Nrf2 pathway also has a potential dark-side, which can lead to chemotherapeutic drug resistance and the metabolic activation of lung carcinogens (e.g., polycyclic aromatic hydrocarbons). They also play determinant roles in 4-(methylnitrosoamino)-1-(3-pyridyl)-1-butanone metabolism to R- and S-4-(methylnitrosoamino)-1-(3-pyridyl)-1-butanol. The overexpression of AKR genes as components of the "smoking gene" battery raises the issue as to whether this is part of a smoking stress response or acquired susceptibility to lung cancer. Human AKR genes also regulate retinoid, prostaglandin, and steroid hormone metabolism and can regulate the local concentrations of ligands available for nuclear receptors (NRs). The prospect exists that signaling through the Keap1/Nrf2 system can also effect NR signaling, but this has remained largely unexplored. We present the case that chemoprevention through the Keap1/Nrf2 system may be context dependent and that the Nrf2 "dose-response curve" for electrophilic and redox balance may not be monotonic.
Collapse
Affiliation(s)
- Trevor M. Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
25
|
Bolton JL, Dunlap T. Formation and Biological Targets of Quinones: Cytotoxic versus Cytoprotective Effects. Chem Res Toxicol 2016; 30:13-37. [PMID: 27617882 PMCID: PMC5241708 DOI: 10.1021/acs.chemrestox.6b00256] [Citation(s) in RCA: 282] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Quinones represent a class of toxicological intermediates, which can create a variety of hazardous effects in vivo including, acute cytotoxicity, immunotoxicity, and carcinogenesis. In contrast, quinones can induce cytoprotection through the induction of detoxification enzymes, anti-inflammatory activities, and modification of redox status. The mechanisms by which quinones cause these effects can be quite complex. The various biological targets of quinones depend on their rate and site of formation and their reactivity. Quinones are formed through a variety of mechanisms from simple oxidation of catechols/hydroquinones catalyzed by a variety of oxidative enzymes and metal ions to more complex mechanisms involving initial P450-catalyzed hydroxylation reactions followed by two-electron oxidation. Quinones are Michael acceptors, and modification of cellular processes could occur through alkylation of crucial cellular proteins and/or DNA. Alternatively, quinones are highly redox active molecules which can redox cycle with their semiquinone radical anions leading to the formation of reactive oxygen species (ROS) including superoxide, hydrogen peroxide, and ultimately the hydroxyl radical. Production of ROS can alter redox balance within cells through the formation of oxidized cellular macromolecules including lipids, proteins, and DNA. This perspective explores the varied biological targets of quinones including GSH, NADPH, protein sulfhydryls [heat shock proteins, P450s, cyclooxygenase-2 (COX-2), glutathione S-transferase (GST), NAD(P)H:quinone oxidoreductase 1, (NQO1), kelch-like ECH-associated protein 1 (Keap1), IκB kinase (IKK), and arylhydrocarbon receptor (AhR)], and DNA. The evidence strongly suggests that the numerous mechanisms of quinone modulations (i.e., alkylation versus oxidative stress) can be correlated with the known pathology/cytoprotection of the parent compound(s) that is best described by an inverse U-shaped dose-response curve.
Collapse
Affiliation(s)
- Judy L Bolton
- Department of Medicinal Chemistry and Pharmacognosy (M/C 781), College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612-7231, United States
| | - Tareisha Dunlap
- Department of Medicinal Chemistry and Pharmacognosy (M/C 781), College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612-7231, United States
| |
Collapse
|
26
|
Ji X, Li Y, He J, Shah W, Xue X, Feng G, Zhang H, Gao M. Depletion of mitochondrial enzyme system in liver, lung, brain, stomach and kidney induced by benzo(a)pyrene. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 43:83-93. [PMID: 26970059 DOI: 10.1016/j.etap.2016.03.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 02/25/2016] [Accepted: 03/02/2016] [Indexed: 06/05/2023]
Abstract
Mitochondrial dysfunction has recently received considerable attention as it plays an important role in adult human pathology caused by various drugs, endogenous agents and environmental agents. Benzo(a)pyrene (BaP), is a ubiquitous environmental contaminant mainly derived from anthropogenic activity during incomplete combustion of organic materials from various sources. The present study aimed to evaluate the effects of benzo(a)pyrene (BaP) on mitochondrial enzymes in the multiple organs including liver, lung, brain, stomach and kidney. ICR mice were exposed to different doses of BaP (2.5, 5 and 10mg/kg body weight) through oral gavage and intraperitoneal injection treatment for 13 weeks consecutively. The induced mitochondrial damage in the examined organs was assayed in terms of significant increase in lipid peroxidation (LPO) and prominent decrease in antioxidant enzymes. Non enzymatic antioxidants and Krebs cycle's enzymes were also significantly decreased in mitochondria. Additionally, BaP induced the body growth retardation and decrease in relative liver weight, increase in relative lung, stomach, kidney and brain weights, and this was further certified through histopathological lesions. Liver and lungs were more prominently damaged by BaP. The mitochondrial depletion increased in BaP dose-dependent manner.
Collapse
Affiliation(s)
- Xiaoying Ji
- Department of Biological Science and Engineering, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, China
| | - Yongfei Li
- School of Materials and Chemical Engineering, Xi'an Technological University, Xi'an 710032, China
| | - Jianlong He
- Xi'an Jiaotong University, Xi'an 710049, China
| | - Walayat Shah
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Xiaochang Xue
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Guodong Feng
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 17 Changle West Road, Shaanxi, Xi'an 710032, China
| | - Huqin Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Meili Gao
- Department of Biological Science and Engineering, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, 710049, China.
| |
Collapse
|
27
|
Gao M, Li Y, Ji X, Xue X, Chen L, Feng G, Zhang H, Wang H, Shah W, Hou Z, Kong Y. Disturbance of Bcl-2, Bax, Caspase-3, Ki-67 and C-myc expression in acute and subchronic exposure to benzo(a)pyrene in cervix. Acta Histochem 2016; 118:63-73. [PMID: 26709117 DOI: 10.1016/j.acthis.2015.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 10/31/2015] [Accepted: 11/02/2015] [Indexed: 12/18/2022]
Abstract
Epidemiological studies have demonstrated that cigarette smoking is an important cofactor or an independent risk factor for the development of cervical cancer. Benzo(a)pyrene (BaP) is one of the most potent tobacco smoke carcinogens in tobacco smoke. BaP induced DNA damage and over expression in p53 cervical tissue of mice as demonstrated in our previous study. Here we present the findings of exposure to BaP on the expression of Bcl-2, C-myc, Ki-67, Caspase-3 and Bax genes in mouse cervix. Acute intraperitoneal administration of BaP (12.5, 25, 50, 100mg/kg body weight) to ICR female mice induced a significant increase in Bcl-2, C-myc, Ki-67 mRNA and protein level till 72h except in Bcl-2 at 24h with 12.5, 25, 50mg/kg as well as at 48h with 12.5mg/kg body weight post treatment. A significant increase was also seen in Caspase-3 and Bax mRNA and protein level with peak level at 24h and gradual decrease till 72h, however, the expression of caspase-3 increased while that of Bax decreased with increasing dose of Bap after 24h. In sub chronic intraperitoneal and oral gavage administration of BaP (2.5, 5, 10mg/kg body weight), similar significant increase was observed for all the examined genes as compared to the control and vehicle groups, however the expression of Bax decreased in a dose dependent manner. The findings of this study will help in further understanding the molecular mechanism of BaP induced carcinogenesis of cervical cancer.
Collapse
|
28
|
Toxicity of wood smoke particles in human A549 lung epithelial cells: the role of PAHs, soot and zinc. Arch Toxicol 2016; 90:3029-3044. [DOI: 10.1007/s00204-016-1659-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 01/04/2016] [Indexed: 10/22/2022]
|
29
|
He J, Ji X, Li Y, Xue X, Feng G, Zhang H, Wang H, Gao M. Subchronic exposure of benzo(a)pyrene interferes with the expression of Bcl-2, Ki-67, C-myc and p53, Bax, Caspase-3 in sub-regions of cerebral cortex and hippocampus. ACTA ACUST UNITED AC 2016; 68:149-56. [DOI: 10.1016/j.etp.2015.11.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/05/2015] [Accepted: 11/25/2015] [Indexed: 12/23/2022]
|
30
|
Song X, Li L, Shi Q, Lehmler HJ, Fu J, Su C, Xia X, Song E, Song Y. Polychlorinated Biphenyl Quinone Metabolite Promotes p53-Dependent DNA Damage Checkpoint Activation, S-Phase Cycle Arrest and Extrinsic Apoptosis in Human Liver Hepatocellular Carcinoma HepG2 Cells. Chem Res Toxicol 2015; 28:2160-9. [PMID: 26451628 DOI: 10.1021/acs.chemrestox.5b00320] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Polychlorinated biphenyls (PCBs) are a group of persistent organic pollutants. The toxic behavior and mechanism of PCBs individuals and congeners have been extensively investigated. However, there is only limited information on their metabolites. Our previous studies have shown that a synthetic PCB metabolite, PCB29-pQ, causes oxidative damage with the evidence of cytotoxicity, genotoxicity, and mitochondrial-derived intrinsic apoptosis. Here, we investigate the effects of PCB29-pQ on DNA damage checkpoint activation, cell cycle arrest, and death receptor-related extrinsic apoptosis in human liver hepatocellular carcinoma HepG2 cells. Our results illustrate that PCB29-pQ increases the S-phase cell population by down-regulating cyclins A/D1/E, cyclin-dependent kinases (CDK 2/4/6), and cell division cycle 25A (CDC25A) and up-regulating p21/p27 protein expressions. PCB29-pQ also induces apoptosis via the up-regulation of Fas/FasL and the activation of caspase 8/3. Moreover, p53 plays a pivotal role in PCB29-pQ-induced cell cycle arrest and apoptosis via the activation of ATM/Chk2 and ATR/Chk1 checkpoints. Cell cycle arrest and apoptotic cell death were attenuated by the pretreatment with antioxidant N-acetyl-cysteine (NAC). Taken together, these results demonstrate that PCB29-pQ induces oxidative stress and promotes p53-dependent DNA damage checkpoint activation, S-phase cycle arrest, and extrinsic apoptosis in HepG2 cells.
Collapse
Affiliation(s)
- Xiufang Song
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University , Chongqing 400715, People's Republic of China
| | - Lingrui Li
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University , Chongqing 400715, People's Republic of China
| | - Qiong Shi
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University , Chongqing 400715, People's Republic of China
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, College of Public Health, The University of Iowa , Iowa City, Iowa 52242, United States
| | - Juanli Fu
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University , Chongqing 400715, People's Republic of China
| | - Chuanyang Su
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University , Chongqing 400715, People's Republic of China
| | - Xiaomin Xia
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University , Chongqing 400715, People's Republic of China
| | - Erqun Song
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University , Chongqing 400715, People's Republic of China
| | - Yang Song
- Key Laboratory of Luminescence and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University , Chongqing 400715, People's Republic of China
| |
Collapse
|
31
|
Tebay LE, Robertson H, Durant ST, Vitale SR, Penning TM, Dinkova-Kostova AT, Hayes JD. Mechanisms of activation of the transcription factor Nrf2 by redox stressors, nutrient cues, and energy status and the pathways through which it attenuates degenerative disease. Free Radic Biol Med 2015; 88:108-146. [PMID: 26122708 PMCID: PMC4659505 DOI: 10.1016/j.freeradbiomed.2015.06.021] [Citation(s) in RCA: 635] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) regulates the basal and stress-inducible expression of a battery of genes encoding key components of the glutathione-based and thioredoxin-based antioxidant systems, as well as aldo-keto reductase, glutathione S-transferase, and NAD(P)H quinone oxidoreductase-1 drug-metabolizing isoenzymes along with multidrug-resistance-associated efflux pumps. It therefore plays a pivotal role in both intrinsic resistance and cellular adaptation to reactive oxygen species (ROS) and xenobiotics. Activation of Nrf2 can, however, serve as a double-edged sword because some of the genes it induces may contribute to chemical carcinogenesis by promoting futile redox cycling of polycyclic aromatic hydrocarbon metabolites or confer resistance to chemotherapeutic drugs by increasing the expression of efflux pumps, suggesting its cytoprotective effects will vary in a context-specific fashion. In addition to cytoprotection, Nrf2 also controls genes involved in intermediary metabolism, positively regulating those involved in NADPH generation, purine biosynthesis, and the β-oxidation of fatty acids, while suppressing those involved in lipogenesis and gluconeogenesis. Nrf2 is subject to regulation at multiple levels. Its ability to orchestrate adaptation to oxidants and electrophiles is due principally to stress-stimulated modification of thiols within one of its repressors, the Kelch-like ECH-associated protein 1 (Keap1), which is present in the cullin-3 RING ubiquitin ligase (CRL) complex CRLKeap1. Thus modification of Cys residues in Keap1 blocks CRLKeap1 activity, allowing newly translated Nrf2 to accumulate rapidly and induce its target genes. The ability of Keap1 to repress Nrf2 can be attenuated by p62/sequestosome-1 in a mechanistic target of rapamycin complex 1 (mTORC1)-dependent manner, thereby allowing refeeding after fasting to increase Nrf2-target gene expression. In parallel with repression by Keap1, Nrf2 is also repressed by β-transducin repeat-containing protein (β-TrCP), present in the Skp1-cullin-1-F-box protein (SCF) ubiquitin ligase complex SCFβ-TrCP. The ability of SCFβ-TrCP to suppress Nrf2 activity is itself enhanced by prior phosphorylation of the transcription factor by glycogen synthase kinase-3 (GSK-3) through formation of a DSGIS-containing phosphodegron. However, formation of the phosphodegron in Nrf2 by GSK-3 is inhibited by stimuli that activate protein kinase B (PKB)/Akt. In particular, PKB/Akt activity can be increased by phosphoinositide 3-kinase and mTORC2, thereby providing an explanation of why antioxidant-responsive element-driven genes are induced by growth factors and nutrients. Thus Nrf2 activity is tightly controlled via CRLKeap1 and SCFβ-TrCP by oxidative stress and energy-based signals, allowing it to mediate adaptive responses that restore redox homeostasis and modulate intermediary metabolism. Based on the fact that Nrf2 influences multiple biochemical pathways in both positive and negative ways, it is likely its dose-response curve, in terms of susceptibility to certain degenerative disease, is U-shaped. Specifically, too little Nrf2 activity will lead to loss of cytoprotection, diminished antioxidant capacity, and lowered β-oxidation of fatty acids, while conversely also exhibiting heightened sensitivity to ROS-based signaling that involves receptor tyrosine kinases and apoptosis signal-regulating kinase-1. By contrast, too much Nrf2 activity disturbs the homeostatic balance in favor of reduction, and so may have deleterious consequences including overproduction of reduced glutathione and NADPH, the blunting of ROS-based signal transduction, epithelial cell hyperplasia, and failure of certain cell types to differentiate correctly. We discuss the basis of a putative U-shaped Nrf2 dose-response curve in terms of potentially competing processes relevant to different stages of tumorigenesis.
Collapse
Affiliation(s)
- Lauren E Tebay
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - Holly Robertson
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - Stephen T Durant
- AstraZeneca Oncology Innovative Medicines, Bioscience, 33F197 Mereside, Alderley Park, Cheshire SK10 4TG, UK
| | - Steven R Vitale
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6160, USA
| | - Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6160, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK.
| |
Collapse
|
32
|
Thomson PF, Parrish D, Pradhan P, Lakshman MK. Modular, Metal-Catalyzed Cycloisomerization Approach to Angularly Fused Polycyclic Aromatic Hydrocarbons and Their Oxidized Derivatives. J Org Chem 2015; 80:7435-46. [PMID: 26196673 PMCID: PMC4541800 DOI: 10.1021/acs.joc.5b00931] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Palladium-catalyzed cross-coupling reactions of 2-bromobenzaldehyde and 6-bromo-2,3-dimethoxybenzaldehyde with 4-methyl-1-naphthaleneboronic acid and acenaphthene-5-boronic acid gave corresponding o-naphthyl benzaldehydes. Corey-Fuchs olefination followed by reaction with n-BuLi led to various 1-(2-ethynylphenyl)naphthalenes. Cycloisomerization of individual 1-(2-ethynylphenyl)naphthalenes to various benzo[c]phenanthrene (BcPh) analogues was accomplished smoothly with catalytic PtCl2 in PhMe. In the case of 4,5-dihydrobenzo[l]acephenanthrylene, oxidation with DDQ gave benzo[l]acephenanthrylene. The dimethoxy-substituted benzo[c]phenanthrenes were demethylated with BBr3 and oxidized to the o-quinones with PDC. Reduction of these quinones with NaBH4 in THF/EtOH in an oxygen atmosphere gave the respective dihydrodiols. Exposure of the dihydrodiols to N-bromoacetamide in THF-H2O led to bromohydrins that were cyclized with Amberlite IRA 400 HO(-) to yield the series 1 diol epoxides. Epoxidation of the dihydrodiols with mCPBA gave the isomeric series 2 diol epoxides. All of the hydrocarbons as well as the methoxy-substituted ones were crystallized and analyzed by X-ray crystallography, and these data are compared to other previously studied BcPh derivatives. The methodology described is highly modular and can be utilized for the synthesis of a wide variety of angularly fused polycyclic aromatic hydrocarbons and their putative metabolites and/or other derivatives.
Collapse
Affiliation(s)
- Paul F. Thomson
- Department of Chemistry, The City College and The City University of New York, 160 Convent Avenue, New York, New York 10031, United States
| | - Damon Parrish
- Naval Research Laboratory, Code 6030, 4555 Overlook Avenue, Washington D.C. 20375, United States
| | - Padmanava Pradhan
- Department of Chemistry, The City College and The City University of New York, 160 Convent Avenue, New York, New York 10031, United States
| | - Mahesh K. Lakshman
- Department of Chemistry, The City College and The City University of New York, 160 Convent Avenue, New York, New York 10031, United States
| |
Collapse
|
33
|
Human aldo-keto reductases and the metabolic activation of polycyclic aromatic hydrocarbons. Chem Res Toxicol 2014; 27:1901-17. [PMID: 25279998 PMCID: PMC4237494 DOI: 10.1021/tx500298n] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
![]()
Aldo-keto reductases (AKRs) are promiscuous
NAD(P)(H) dependent
oxidoreductases implicated in the metabolic activation of polycyclic
aromatic hydrocarbons (PAH). These enzymes catalyze the oxidation
of non-K-region trans-dihydrodiols to the corresponding o-quinones with the concomitant production of reactive oxygen
species (ROS). The PAH o-quinones are Michael acceptors
and can form adducts but are also redox-active and enter into futile
redox cycles to amplify ROS formation. Evidence exists to support
this metabolic pathway in humans. The human recombinant AKR1A1 and
AKR1C1–AKR1C4 enzymes all catalyze the oxidation of PAH trans-dihydrodiols to PAH o-quinones. Many
human AKRs also catalyze the NADPH-dependent reduction of the o-quinone products to air-sensitive catechols, exacerbating
ROS formation. Moreover, this pathway of PAH activation occurs in
a panel of human lung cell lines, resulting in the production of ROS
and oxidative DNA damage in the form of 8-oxo-2′-deoxyguanosine.
Using stable-isotope dilution liquid chromatography tandem mass spectrometry,
this pathway of benzo[a]pyrene (B[a]P) metabolism was found to contribute equally with the diol-epoxide
pathway to the activation of this human carcinogen in human lung cells.
Evaluation of the mutagenicity of anti-B[a]P-diol epoxide with B[a]P-7,8-dione on
p53 showed that the o-quinone produced by AKRs was
the more potent mutagen, provided that it was permitted to redox cycle,
and that the mutations observed were G to T transversions, reminiscent
of those observed in human lung cancer. It is concluded that there
is sufficient evidence to support the role of human AKRs in the metabolic
activation of PAH in human lung cell lines and that they may contribute
to the causation of human lung cancer.
Collapse
|
34
|
Graupner A, Instanes C, Dertinger SD, Andersen JM, Lindeman B, Rongved TD, Brunborg G, Olsen AK. Single cell gel electrophoresis (SCGE) and Pig-a mutation assay in vivo-tools for genotoxicity testing from a regulatory perspective: a study of benzo[a]pyrene in Ogg1(-/-) mice. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2014; 772:34-41. [PMID: 25308545 DOI: 10.1016/j.mrgentox.2014.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 07/25/2014] [Accepted: 07/29/2014] [Indexed: 01/14/2023]
Abstract
The OECD has developed test guidelines (TG) to identify agents with genotoxic effects. The in vivo alkaline single cell gel electrophoresis (SCGE) assay is currently being prepared to become such a TG. The performance of a combined SCGE/Pig-a gene mutation study was evaluated with the prototypical genotoxicant benzo[a]pyrene (BaP) at an exposure level known to induce germ cell mutation. We aimed to better understand (i) the strengths and weaknesses of the two methods applied in blood and their potential to predict germ cell mutagenicity, and (ii) the involvement of reactive oxygen species (ROS) following in vivo BaP-exposure. To explore the involvement of ROS on BaP genotoxicity, we utilised a mouse model deficient in a DNA glycosylase. Specifically, C57BL/6 mice (Ogg1(+/+) and Ogg1(-/-)) were treated for three consecutive days with 50 mg BaP/kg/day. DNA damage in nucleated blood cells was measured four hours after the last treatment with the SCGE assay, with and without formamidopyrimidine DNA glycosylase (Fpg). Pig-a mutant phenotype blood erythrocytes were analysed two and four weeks after treatment. BaP-induced DNA lesions were not significantly increased in either version of the SCGE assay. The phenotypic mutation frequencies for immature and mature erythrocytes were significantly increased after two weeks. These effects were not affected by genotype, suggesting oxidative damage may have a minor role in BaP genotoxicity, at least in the acute exposure situation studied here. While both assays are promising tools for risk assessment, these results highlight the necessity of understanding the limitations regarding each assay's ability to detect chemicals' genotoxic potential.
Collapse
Affiliation(s)
- Anne Graupner
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway
| | - Christine Instanes
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway
| | | | - Jill Mari Andersen
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway
| | - Birgitte Lindeman
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway
| | - Tonje Danielsen Rongved
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway
| | - Gunnar Brunborg
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway
| | - Ann-Karin Olsen
- Department of Chemicals and Radiation, Norwegian Institute of Public Health, Oslo 0403, Norway.
| |
Collapse
|
35
|
Molecular profiles of non-small cell lung cancers in cigarette smoking and never-smoking patients. Adv Med Sci 2014; 58:196-206. [PMID: 24451080 DOI: 10.2478/ams-2013-0025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE Molecular features of non-small cell lung cancer (NSCLC) in never-smokers are not well recognized. We assessed the expression of genes potentially related to lung cancer etiology in smoking vs. never-smoking NSCLC patients. METHODS We assayed frozen tumor samples from surgically resected 31 never-smoking and 54 clinically pair-matched smoking NSCLC patients, and from corresponding normal lung tissue from 27 and 43 patients, respectively. Expression of 21 genes, including cell membrane kinases, sex hormone receptors, transcription factors, growth factors and others was assessed by reverse transcription - quantitative PCR. RESULTS Expression of 5 genes was significantly higher in tumors of non-smokers vs. smokers: CSF1R (p<0.0001), RRAD (p<0.0001), PR (p=0.0004), TGFBR2 (p=0.0027) and EPHB6 (p=0.0033). Expression of AKR1B10 (p<0.0001), CDKN2A (p<0.0001), CHRNA6 (p<0.0001), SOX9 (p<0.0001), survivin (p<0.0001) and ER2 (p=0.002) was significantly higher in tumors compared to normal lung tissue. Expression of AR (p<0.0001), EPHB6 (p<0.0001), PR (p<0.0001), TGFBR2 (p<0.0001), TGFBR3 (p<0.0001), ER1 (p=0.0006) and DLG1 (p=0.0016) was significantly lower in tumors than in normal lung tissue. Expression of IGF2 was higher in tumors than in healthy lung tissue in never-smokers (p=0.003), and expression of AHR (p<0.0001), CSF1R (p<0.0001) and RRAD (p<0.0001) was lower in tumors than in healthy lung tissue in smokers. CONCLUSION Expression of several genes in NSCLC is strongly related to smoking history. Lower expression of PR and higher expression of ER2 in tumors suggests a possibility of hormonal therapeutic intervention in selected NSCLC patients. Distinct molecular features of NSCLC in never-smokers, e.g. CHRNA6 upregulation, may prompt new treatment strategies.
Collapse
|
36
|
Matsunaga T, Morikawa Y, Haga M, Endo S, Soda M, Yamamura K, El-Kabbani O, Tajima K, Ikari A, Hara A. Exposure to 9,10-phenanthrenequinone accelerates malignant progression of lung cancer cells through up-regulation of aldo-keto reductase 1B10. Toxicol Appl Pharmacol 2014; 278:180-9. [PMID: 24813866 DOI: 10.1016/j.taap.2014.04.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 04/21/2014] [Accepted: 04/26/2014] [Indexed: 01/13/2023]
Abstract
Inhalation of 9,10-phenanthrenequinone (9,10-PQ), a major quinone in diesel exhaust, exerts fatal damage against a variety of cells involved in respiratory function. Here, we show that treatment with high concentrations of 9,10-PQ evokes apoptosis of lung cancer A549 cells through production of reactive oxygen species (ROS). In contrast, 9,10-PQ at its concentrations of 2 and 5 μM elevated the potentials for proliferation, invasion, metastasis and tumorigenesis, all of which were almost completely inhibited by addition of an antioxidant N-acetyl-l-cysteine, inferring a crucial role of ROS in the overgrowth and malignant progression of lung cancer cells. Comparison of mRNA expression levels of six aldo-keto reductases (AKRs) in the 9,10-PQ-treated cells advocated up-regulation of AKR1B10 as a major cause contributing to the lung cancer malignancy. In support of this, the elevation of invasive, metastatic and tumorigenic activities in the 9,10-PQ-treated cells was significantly abolished by the addition of a selective AKR1B10 inhibitor oleanolic acid. Intriguingly, zymographic and real-time PCR analyses revealed remarkable increases in secretion and expression, respectively, of matrix metalloproteinase 2 during the 9,10-PQ treatment, and suggested that the AKR1B10 up-regulation and resultant activation of mitogen-activated protein kinase cascade are predominant mechanisms underlying the metalloproteinase induction. In addition, HPLC analysis and cytochrome c reduction assay in in vitro 9,10-PQ reduction by AKR1B10 demonstrated that the enzyme catalyzes redox-cycling of this quinone, by which ROS are produced. Collectively, these results suggest that AKR1B10 is a key regulator involved in overgrowth and malignant progression of the lung cancer cells through ROS production due to 9,10-PQ redox-cycling.
Collapse
Affiliation(s)
- Toshiyuki Matsunaga
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan.
| | - Yoshifumi Morikawa
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Mariko Haga
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Midori Soda
- Laboratory of Clinical Pharmacy, School of Pharmacy, Aichi Gakuin University, Nagoya 464-8650, Japan
| | - Keiko Yamamura
- Laboratory of Clinical Pharmacy, School of Pharmacy, Aichi Gakuin University, Nagoya 464-8650, Japan
| | - Ossama El-Kabbani
- Monash Institute of Pharmaceutical Sciences, Monash University, Victoria 3052, Australia
| | - Kazuo Tajima
- Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa 920-1181, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Akira Hara
- Faculty of Engineering, Gifu University, Gifu 501-1193, Japan
| |
Collapse
|
37
|
Cooke SL, Shlien A, Marshall J, Pipinikas CP, Martincorena I, Tubio JM, Li Y, Menzies A, Mudie L, Ramakrishna M, Yates L, Davies H, Bolli N, Bignell GR, Tarpey PS, Behjati S, Nik-Zainal S, Papaemmanuil E, Teixeira VH, Raine K, O’Meara S, Dodoran MS, Teague JW, Butler AP, Iacobuzio-Donahue C, Santarius T, Grundy RG, Malkin D, Greaves M, Munshi N, Flanagan AM, Bowtell D, Martin S, Larsimont D, Reis-Filho JS, Boussioutas A, Taylor JA, Hayes ND, Janes SM, Futreal PA, Stratton MR, McDermott U, Campbell PJ. Processed pseudogenes acquired somatically during cancer development. Nat Commun 2014; 5:3644. [PMID: 24714652 PMCID: PMC3996531 DOI: 10.1038/ncomms4644] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 03/13/2014] [Indexed: 12/14/2022] Open
Abstract
Cancer evolves by mutation, with somatic reactivation of retrotransposons being one such mutational process. Germline retrotransposition can cause processed pseudogenes, but whether this occurs somatically has not been evaluated. Here we screen sequencing data from 660 cancer samples for somatically acquired pseudogenes. We find 42 events in 17 samples, especially non-small cell lung cancer (5/27) and colorectal cancer (2/11). Genomic features mirror those of germline LINE element retrotranspositions, with frequent target-site duplications (67%), consensus TTTTAA sites at insertion points, inverted rearrangements (21%), 5' truncation (74%) and polyA tails (88%). Transcriptional consequences include expression of pseudogenes from UTRs or introns of target genes. In addition, a somatic pseudogene that integrated into the promoter and first exon of the tumour suppressor gene, MGA, abrogated expression from that allele. Thus, formation of processed pseudogenes represents a new class of mutation occurring during cancer development, with potentially diverse functional consequences depending on genomic context.
Collapse
Affiliation(s)
- Susanna L. Cooke
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Adam Shlien
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - John Marshall
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | | | - Inigo Martincorena
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Jose M.C. Tubio
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Yilong Li
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Andrew Menzies
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Laura Mudie
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Manasa Ramakrishna
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Lucy Yates
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Helen Davies
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Niccolo Bolli
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
- University of Cambridge, Cambridge CB2 0XY, UK
| | - Graham R. Bignell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Patrick S. Tarpey
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Sam Behjati
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
- University of Cambridge, Cambridge CB2 0XY, UK
| | - Serena Nik-Zainal
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Elli Papaemmanuil
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Vitor H. Teixeira
- Lungs for Living Research Centre, Rayne Institute, University College London, London WC1E 6JF, UK
| | - Keiran Raine
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Sarah O’Meara
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Maryam S. Dodoran
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Jon W. Teague
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Adam P. Butler
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | | | | | - Richard G. Grundy
- Children’s Brain Tumour Research Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - David Malkin
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada M5G 1X8
| | - Mel Greaves
- Institute for Cancer Research, Sutton, London SM2 5NG, UK
| | - Nikhil Munshi
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Adrienne M. Flanagan
- Lungs for Living Research Centre, Rayne Institute, University College London, London WC1E 6JF, UK
- Royal National Orthopaedic Hospital, Middlesex HA7 4LP, UK
| | - David Bowtell
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3002, Australia
| | - Sancha Martin
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Denis Larsimont
- Department of Pathology, Jules Bordet Institute, 1000 Brussels, Belgium
| | - Jorge S. Reis-Filho
- Department of Pathology, Memorial-Sloan-Kettering Cancer Center, New York, New York 10065, USA
| | - Alex Boussioutas
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3002, Australia
- Department of Gastroenterology, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Jack A. Taylor
- National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27713, USA
| | - Neil D. Hayes
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Sam M. Janes
- Lungs for Living Research Centre, Rayne Institute, University College London, London WC1E 6JF, UK
| | - P. Andrew Futreal
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Michael R. Stratton
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Ultan McDermott
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
- Addenbrooke’s NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Peter J. Campbell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
- University of Cambridge, Cambridge CB2 0XY, UK
- Addenbrooke’s NHS Foundation Trust, Cambridge CB2 0QQ, UK
| |
Collapse
|
38
|
Jarvis IWH, Dreij K, Mattsson Å, Jernström B, Stenius U. Interactions between polycyclic aromatic hydrocarbons in complex mixtures and implications for cancer risk assessment. Toxicology 2014; 321:27-39. [PMID: 24713297 DOI: 10.1016/j.tox.2014.03.012] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 03/28/2014] [Accepted: 03/30/2014] [Indexed: 01/27/2023]
Abstract
In this review we discuss the effects of exposure to complex PAH mixtures in vitro and in vivo on mechanisms related to carcinogenesis. Of particular concern regarding exposure to complex PAH mixtures is how interactions between different constituents can affect the carcinogenic response and how these might be included in risk assessment. Overall the findings suggest that the responses resulting from exposure to complex PAH mixtures is varied and complicated. More- and less-than additive effects on bioactivation and DNA damage formation have been observed depending on the various mixtures studied, and equally dependent on the different test systems that are used. Furthermore, the findings show that the commonly used biological end-point of DNA damage formation is insufficient for studying mixture effects. At present the assessment of the risk of exposure to complex PAH mixtures involves comparison to individual compounds using either a surrogate or a component-based potency approach. We discuss how future risk assessment strategies for complex PAH mixtures should be based around whole mixture assessment in order to account for interaction effects. Inherent to this is the need to incorporate different experimental approaches using robust and sensitive biological endpoints. Furthermore, the emphasis on future research should be placed on studying real life mixtures that better represent the complex PAH mixtures that humans are exposed to.
Collapse
Affiliation(s)
- Ian W H Jarvis
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden.
| | - Kristian Dreij
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden
| | - Åse Mattsson
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden
| | - Bengt Jernström
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden
| | - Ulla Stenius
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden
| |
Collapse
|
39
|
Šmerdová L, Neča J, Svobodová J, Topinka J, Schmuczerová J, Kozubík A, Machala M, Vondráček J. Inflammatory mediators accelerate metabolism of benzo[a]pyrene in rat alveolar type II cells: The role of enhanced cytochrome P450 1B1 expression. Toxicology 2013; 314:30-8. [DOI: 10.1016/j.tox.2013.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 08/29/2013] [Accepted: 09/02/2013] [Indexed: 12/15/2022]
|
40
|
Jun dimerization protein 2 is a critical component of the Nrf2/MafK complex regulating the response to ROS homeostasis. Cell Death Dis 2013; 4:e921. [PMID: 24232097 PMCID: PMC3847324 DOI: 10.1038/cddis.2013.448] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Revised: 10/10/2013] [Accepted: 10/14/2013] [Indexed: 02/06/2023]
Abstract
Oxidative stress and reactive oxygen species (ROS) are associated with diseases such as cancer, cardiovascular complications, inflammation and neurodegeneration. Cellular defense systems must work constantly to control ROS levels and to prevent their accumulation. We report here that the Jun dimerization protein 2 (JDP2) has a critical role as a cofactor for transcription factors nuclear factor-erythroid 2-related factor 2 (Nrf2) and small Maf protein family K (MafK) in the regulation of the antioxidant-responsive element (ARE) and production of ROS. Chromatin immunoprecipitation–quantitative PCR (qPCR), electrophoresis mobility shift and ARE-driven reporter assays were carried out to examine the role of JDP2 in ROS production. JDP2 bound directly to the ARE core sequence, associated with Nrf2 and MafK (Nrf2–MafK) via basic leucine zipper domains, and increased DNA-binding activity of the Nrf2–MafK complex to the ARE and the transcription of ARE-dependent genes. In mouse embryonic fibroblasts from Jdp2-knockout (Jdp2 KO) mice, the coordinate transcriptional activation of several ARE-containing genes and the ability of Nrf2 to activate expression of target genes were impaired. Moreover, intracellular accumulation of ROS and increased thickness of the epidermis were detected in Jdp2 KO mice in response to oxidative stress-inducing reagents. These data suggest that JDP2 is required to protect against intracellular oxidation, ROS activation and DNA oxidation. qPCR demonstrated that several Nrf2 target genes such as heme oxygenase-1, glutamate–cysteine ligase catalytic and modifier subunits, the notch receptor ligand jagged 1 and NAD(P)H dehydrogenase quinone 1 are also dependent on JDP2 for full expression. Taken together, these results suggest that JDP2 is an integral component of the Nrf2–MafK complex and that it modulates antioxidant and detoxification programs by acting via the ARE.
Collapse
|
41
|
Zhang L, Huang M, Blair IA, Penning TM. Interception of benzo[a]pyrene-7,8-dione by UDP glucuronosyltransferases (UGTs) in human lung cells. Chem Res Toxicol 2013; 26:1570-8. [PMID: 24047243 PMCID: PMC3829198 DOI: 10.1021/tx400268q] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Polycyclic
aromatic hydrocarbons (PAHs) are environmental and tobacco
carcinogens. Proximate carcinogenic PAH trans-dihydrodiols
are activated by human aldo-keto reductases (AKRs) to yield electrophilic
and redox-active o-quinones. Interconversion among
benzo[a]pyrene (B[a]P)-7,8-dione,
a representative PAH o-quinone, and its corresponding
catechol generates a futile redox-cycle with the concomitant production
of reactive oxygen species (ROS). We investigated whether glucuronidation
of B[a]P-7,8-catechol by human UDP glucuronosyltransferases
(UGTs) could intercept the catechol in three different human lung
cells. RT-PCR showed that UGT1A1, 1A3, and 2B7 were only expressed
in human lung adenocarcinoma A549 cells. The corresponding recombinant
UGTs were examined for their kinetic constants and product profile
using B[a]P-7,8-catechol as a substrate. B[a]P-7,8-dione was reduced to B[a]P-7,8-catechol
by dithiothreitol under anaerobic conditions and then further glucuronidated
by the UGTs in the presence of uridine-5′-diphosphoglucuronic
acid as a glucuronic acid group donor. UGT1A1 catalyzed the glucuronidation
of B[a]P-7,8-catechol and generated two isomeric O-monoglucuronsyl-B[a]P-7,8-catechol products
that were identified by RP-HPLC and by LC-MS/MS. By contrast, UGT1A3
and 2B7 catalyzed the formation of only one monoglucuronide, which
was identical to that formed in A549 cells. The kinetic profiles of
three UGTs followed Michaelis–Menten kinetics. On the basis
of the expression levels of UGT1A3 and UGT2B7 and the observation
that a single monoglucuronide was produced in A549 cells, we suggest
that the major UGT isoforms in A549 cells that can intercept B[a]P-7,8-catechol are UGT1A3 and 2B7.
Collapse
Affiliation(s)
- Li Zhang
- Center of Excellence in Environmental Toxicology and ‡Center for Cancer Pharmacology, Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania 19104-6160, United States
| | | | | | | |
Collapse
|
42
|
Peluso MEM, Munnia A, Srivatanakul P, Jedpiyawongse A, Sangrajrang S, Ceppi M, Godschalk RWL, van Schooten FJ, Boffetta P. DNA adducts and combinations of multiple lung cancer at-risk alleles in environmentally exposed and smoking subjects. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2013; 54:375-383. [PMID: 23797975 DOI: 10.1002/em.21788] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 04/23/2013] [Accepted: 04/24/2013] [Indexed: 06/02/2023]
Abstract
Interindividual variation in DNA adduct levels in individuals exposed to similar amounts of environmental carcinogens may be due to genetic variability. We analysed the influence of genes involved in determining/modifying DNA damage, including microsomal epoxide hydrolase1 (EPHX1) His139Arg, N-acetyl-transferase, NAD(P)H:quinone oxidoreductase1 (NQO1) Pro187Ser, manganese superoxide dismutase2 (MnSOD2) Val16Ala, and apurinic/apyrimidinic endonuclease1 (APE1) Asp148Glu polymorphisms in blood of 120 smokers. Subsequently, we examined the effects of the combinations of the variant alleles of EPHX, NQO1 and MnSOD2 together with the wild type allele of APE1 on DNA damage by calculating the "sum of at-risk alleles." We reviewed the studies examining the relationships of DNA adducts with at-risk alleles in environmentally exposed subjects. Our findings showed that smokers carrying the EPHX1-139Arg and the NQO1-187Ser variants were significantly more likely to have higher adduct levels. Null associations were found with the other variants. Nevertheless, DNA adduct levels in smokers with ≥5 at-risk alleles were significantly different from those with fewer than two alleles. A similar picture emerged from studies of DNA adducts and at-risk alleles in environmentally exposed and smoking subjects. Certain at-risk allele combinations may confer a greater likelihood of increased levels of adducts after environmental insults. The increase in DNA adduct levels in susceptible subjects exposed to environmental carcinogens may reflect changes in the mechanisms that protect cells from the accumulation of genetic damage. Alterations of the physiological processes designed to maintain homeostasis may reduce the individual "genotoxic tolerance" to environmental challenges and result in phenotypes characterized by high levels of DNA adducts.
Collapse
Affiliation(s)
- Marco E M Peluso
- Cancer Risk Factor Branch, Cancer Prevention and Research Institute, Florence, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Huang M, Blair IA, Penning TM. Identification of stable benzo[a]pyrene-7,8-dione-DNA adducts in human lung cells. Chem Res Toxicol 2013; 26:685-92. [PMID: 23587017 PMCID: PMC3660951 DOI: 10.1021/tx300476m] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
![]()
Metabolic
activation of the proximate carcinogen benzo[a]pyrene-7,8-trans-dihydrodiol (B[a]P-7,8-trans-dihydrodiol) by aldo-keto
reductases (AKRs) leads to B[a]P-7,8-dione that is
both electrophilic and redox-active. B[a]P-7,8-dione
generates reactive oxygen species resulting in oxidative DNA damage
in human lung cells. However, information on the formation of stable
B[a]P-7,8-dione-DNA adducts in these cells is lacking.
We studied stable DNA adduct formation of B[a]P-7,8-dione
in human lung adenocarcinoma A549 cells, human bronchoalveolar H358
cells, and immortalized human bronchial epithelial HBEC-KT cells.
After treatment with 2 μM B[a]P-7,8-dione,
the cellular DNA was extracted from the cell pellets subjected to
enzyme hydrolysis and subsequent analysis by LC-MS/MS. Several stable
DNA adducts of B[a]P-7,8-dione were only detected
in A549 and HBEC-KT cells. In A549 cells, the structures of stable
B[a]P-7,8-dione-DNA adducts were identified as hydrated-B[a]P-7,8-dione-N2-2′-deoxyguanosine
and hydrated-B[a]P-7,8-dione-N1-2′-deoxyguanosine.
In HBEC-KT cells, the structures of stable B[a]P-7,8-dione-DNA
adducts were identified as hydrated-B[a]P-7,8-dione-2′-deoxyadenosine,
hydrated-B[a]P-7,8-dione-N1- or N3-2′-deoxyadenosine,
and B[a]P-7,8-dione-N1- or N3-2′-deoxyadenosine.
In each case, adduct structures were characterized by MSn spectra. Adduct structures were also compared to
those synthesized from reactions of B[a]P-7,8-dione
with either deoxyribonucleosides or salmon testis DNA in vitro but were found to be different.
Collapse
Affiliation(s)
- Meng Huang
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania 19104-6160, United States
| | | | | |
Collapse
|
44
|
Abedin Z, Louis-Juste M, Stangl M, Field J. The role of base excision repair genes OGG1, APN1 and APN2 in benzo[a]pyrene-7,8-dione induced p53 mutagenesis. Mutat Res 2013; 750:121-8. [PMID: 23117049 PMCID: PMC3931135 DOI: 10.1016/j.mrgentox.2012.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 10/17/2012] [Accepted: 10/18/2012] [Indexed: 12/17/2022]
Abstract
Lung cancer is primarily caused by exposure to tobacco smoke. Tobacco smoke contains numerous carcinogens, including polycyclic aromatic hydrocarbons (PAH). The most common PAH studied is benzo[a]pyrene (B[a]P). B[a]P is metabolically activated through multiple routes, one of which is catalyzed by aldo-keto reductase (AKR) to B[a]P-7,8-dione (BPQ). BPQ undergoes a futile redox cycle in the presence of NADPH to generate reactive oxygen species (ROS). ROS, in turn, damages DNA. Studies with a yeast p53 mutagenesis system found that the generation of ROS by PAH o-quinones may contribute to lung carcinogenesis because of similarities between the patterns (types of mutations) and spectra (location of mutations) and those seen in lung cancer. The patterns were dominated by G to T transversions, and the spectra in the experimental system have mutations at lung cancer hotspots. To address repair mechanisms that are responsible for BPQ induced damage we observed the effect of mutating two DNA repair genes OGG1 and APE1 (APN1 in yeast) and tested them in a yeast reporter system for p53 mutagenesis. There was an increase in both the mutant frequency and the number of G:C/T:A transversions in p53 treated with BPQ in ogg1 yeast but not in apn1 yeast. Knocking out APN2 increased mutagenesis in the apn1 cells. In addition, we did not find a strand bias on p53 treated with BPQ in ogg1 yeast. These studies suggest that Ogg1 is involved in repairing the oxidative damage caused by BPQ, Apn1 and Apn2 have redundant functions and that the stand bias seen in lung cancer may not be due to impaired repair of oxidative lesions.
Collapse
Affiliation(s)
- Zahidur Abedin
- Department of Pharmacology, Centers for Excellence in Environmental Toxicology and Cancer Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6084, USA
| | - Melissa Louis-Juste
- Department of Pharmacology, Centers for Excellence in Environmental Toxicology and Cancer Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6084, USA
| | - Melissa Stangl
- Department of Pharmacology, Centers for Excellence in Environmental Toxicology and Cancer Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6084, USA
| | - Jeffrey Field
- Department of Pharmacology, Centers for Excellence in Environmental Toxicology and Cancer Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6084, USA
| |
Collapse
|
45
|
Sen S, Field JM. Genotoxicity of Polycyclic Aromatic Hydrocarbon Metabolites. ADVANCES IN MOLECULAR TOXICOLOGY 2013. [DOI: 10.1016/b978-0-444-62645-5.00003-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
46
|
Kolšek K, Sollner Dolenc M, Mavri J. Computational Study of the Reactivity of Bisphenol A-3,4-quinone with Deoxyadenosine and Glutathione. Chem Res Toxicol 2012. [DOI: 10.1021/tx300411d] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Katra Kolšek
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7,
1000 Ljubljana, Slovenia
| | | | - Janez Mavri
- Laboratory for Biocomputing
and Bioinformatics, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
- EN-FIST Centre of Excellence, Dunajska 156, 1000, Ljubljana, Slovenia
| |
Collapse
|
47
|
Zhang L, Jin Y, Huang M, Penning TM. The Role of Human Aldo-Keto Reductases in the Metabolic Activation and Detoxication of Polycyclic Aromatic Hydrocarbons: Interconversion of PAH Catechols and PAH o-Quinones. Front Pharmacol 2012; 3:193. [PMID: 23162467 PMCID: PMC3499756 DOI: 10.3389/fphar.2012.00193] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 10/27/2012] [Indexed: 11/13/2022] Open
Abstract
Polycyclic aromatic hydrocarbons (PAH) are ubiquitous environmental pollutants. They are procarcinogens requiring metabolic activation to elicit their deleterious effects. Aldo-keto reductases (AKR) catalyze the oxidation of proximate carcinogenic PAH trans-dihydrodiols to yield electrophilic and redox-active PAH o-quinones. AKRs are also found to be capable of reducing PAH o-quinones to form PAH catechols. The interconversion of o-quinones and catechols results in the redox-cycling of PAH o-quinones to give rise to the generation of reactive oxygen species and subsequent oxidative DNA damage. On the other hand, PAH catechols can be intercepted through phase II metabolism by which PAH o-quinones could be detoxified and eliminated. The aim of the present review is to summarize the role of human AKRs in the metabolic activation/detoxication of PAH and the relevance of phase II conjugation reactions to human lung carcinogenesis.
Collapse
Affiliation(s)
- Li Zhang
- Center of Excellence in Environmental Toxicology, Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA
| | | | | | | |
Collapse
|
48
|
Peluso M, Srivatanakul P, Jedpiyawongse A, Sangrajrang S, Munnia A, Piro S, Ceppi M, Boffetta P, Godschalk RWL, van Schooten FJ. Aromatic DNA adducts and number of lung cancer risk alleles in Map-Ta-Phut Industrial Estate workers and nearby residents. Mutagenesis 2012; 28:57-63. [PMID: 22987024 DOI: 10.1093/mutage/ges053] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The Map-Ta-Phut Industrial Estate (MIE) in Rayong, Thailand, is the location of one of the largest industrial complexes in southeastern Asia. The MIE complex produces a mixture of air pollutants, including polycyclic aromatic hydrocarbons, compounds capable to induce the generation of DNA adducts. DNA adducts are considered to be a biomarker of carcinogen exposure; however, its production can be modulated by genetic susceptibility. Thus, we analysed the influence of EPHX1 His139Arg (A>G, rs2234922) and NQO1 Pro187Ser (C>T, rs1800566) involved in the metabolism of polycyclic aromatic hydrocarbons; MnSOD(2) Val16Ala (C>T, rs1799725) a gene that acts against the free radical generation; APE1/Ref-1 Asp148Glu (T>G, rs3136820) a gene involved in the repair of DNA, and in the control of cell-cycle and apoptosis on leucocyte DNA adducts in 77 MIE workers, 69 Map-Ta-Phut residents, and 50 rural controls, Rayong, Thailand. We searched for associations with the 'sum of at-risk alleles' by combining the variant alleles of EPHX1, NQO1 and MnSOD(2) together with the wild-type allele of APE1, since they appeared to influence lung cancer risk. Although our findings revealed significant associations between DNA adducts and the EPHX1 His139Arg and NQO1 Pro187Ser polymorphisms, the combination of at-risk alleles was found to affect DNA damage much stronger. DNA adducts were significantly increased in the individuals bearing 4 and ≥ 5 at-risk alleles [mean ratio (MR) = 1.55, 95% CI 1.10-2.18, P = 0.012, and MR = 2.11, 95% CI 1.27-3.51, P = 0.004, respectively)]. After correction for residence/employment categorisation, a significant increment was present in the MIE workers with ≥ 5 alleles (MR = 2.88, 95% CI 1.46-5.71, P = 0.003). Our data indicate relationships between the generation of DNA adducts and the enzymatic activities of EPHX and NQO1. The combination of unfavourable genetic variants seems to determine the individuals' susceptibility, rather than a single polymorphism.
Collapse
Affiliation(s)
- Marco Peluso
- Cancer Risk Factor Branch, Cancer Prevention and Research Institute, Florence, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Wu A, Xu D, Lu D, Penning TM, Blair IA, Harvey RG. Synthesis of 13C4-labelled oxidized metabolites of the carcinogenic polycyclic aromatic hydrocarbon benzo[ a]pyrene. Tetrahedron 2012; 68:10.1016/j.tet.2012.05.130. [PMID: 24244053 PMCID: PMC3826453 DOI: 10.1016/j.tet.2012.05.130] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAHs), such as benzo[a]pyrene (BaP), are ubiquitous environmental contaminants that are implicated in causing lung cancer. BaP is a component of tobacco smoke that is transformed enzymatically to active forms that interact with DNA. We reported previously development of a sensitive stable isotope dilution LC/MS method for analysis of BaP metabolites. We now report efficient syntheses of 13C4-BaP and the complete set of its 13C4-labelled oxidized metabolites needed as internal standards They include the metabolites not involved in carcinogenesis (Group A) and the metabolites implicated in initiation of cancer (Group B). The synthetic approach is novel, entailing use of Pd-catalyzed Suzuki, Sonogashira, and Hartwig cross-coupling reactions combined with PtCl2-catalyzed cyclization of acetylenic compounds. This synthetic method requires fewer steps, employs milder conditions, and product isolation is simpler than conventional methods of PAH synthesis. The syntheses of 13C4-BaP and 13C4-BaP-8-ol each require only four steps, and the 13C-atoms are all introduced in a single step. 13C4-BaP-8-ol serves as the synthetic precursor of all the oxidized metabolites of 13C-BaP implicated in initiation of cancer. The isotopic purities of the synthetic 13C4-BaP metabolites were estimated to be ≥99.9%.
Collapse
Affiliation(s)
- Anhui Wu
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, United States
| | - Daiwang Xu
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, United States
| | - Ding Lu
- The Centers for Cancer Pharmacology and Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Trevor M. Penning
- The Centers for Cancer Pharmacology and Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Ian A. Blair
- The Centers for Cancer Pharmacology and Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Ronald G. Harvey
- The Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, United States
| |
Collapse
|
50
|
Jaiswal PK, Srivastava S, Gupta J, Thakur IS. Dibenzofuran induces oxidative stress, disruption of trans-mitochondrial membrane potential (ΔΨm) and G1 arrest in human hepatoma cell line. Toxicol Lett 2012; 214:137-44. [PMID: 22944260 DOI: 10.1016/j.toxlet.2012.08.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Revised: 08/17/2012] [Accepted: 08/17/2012] [Indexed: 01/14/2023]
Abstract
Dioxins are a class of extremely toxic environmentally persistent pollutant, comprised of halogenated dibenzo-p-dioxins, dibenzofurans and biphenyls. Despite significant human exposure via multiple routes, very little is known about toxicity induced by dibenzofuran (DF). Current study shed lights on the potential toxicity mechanism of DF using human hepatoma cell line (HepG2). It was observed that the exposure to DF potentiate oxidative stress, apoptosis and necrosis at 10μM within 8h in HepG2 cells. Interestingly, when we pre-incubated the cells with α-NF (1nM) for 12h, an aromatic hydrocarbon receptor antagonist, the IC(50) of DF increased by 14 folds indicating the cytoprotective ability of α-NF from DF induced toxicity. Furthermore, three additional metabolites were observed while studying the metabolic profile of DF in HepG2 cells with and without pre-incubation with α-NF using chromatography-mass spectroscopy (GC-MS). Of these, two metabolites were characterized as dihydroxylated derivative of DF and third metabolite was characterized as quinone derivative of DF. By flow cytometry and confocal laser microscopy analysis we followed the ROS formation after DF (10μM) exposure for 3h. Significantly low ROS was generated in cells which were pre-incubated with α-NF than cells which were not pre-incubated with α-NF underlining the importance of metabolism in DF toxicity. The same pattern of protection was consistent while measuring mitochondrial membrane potential (MMP), i.e., less MMP dip was observed in 'with α-NF pre-incubated and DF (10μM) exposed cells' than 'without α-NF pre-incubated but DF exposed cells'. In cell cycle studies, it was confirmed that cell population of HepG2 at G1 stage progressively increased in number (∼74%) within 24h. Thus, DF and its metabolites induce significantly higher cytotoxicity after metabolism in HepG2 cells than its parent compound (DF) by ROS formation, MMP dip and impaired cell cycle.
Collapse
|