1
|
Ahmad M, Liu M, Yang Z, Zhang H, Nabi G, Hao Y, Chen L. Perfluorooctane sulfonate causes DNA damage and apoptosis via oxidative stress in umbilical cord fibroblast cells of Yangtze finless porpoise. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 958:178030. [PMID: 39662399 DOI: 10.1016/j.scitotenv.2024.178030] [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: 09/12/2024] [Revised: 11/14/2024] [Accepted: 12/07/2024] [Indexed: 12/13/2024]
Abstract
Yangtze finless porpoise (YFP) is a critically endangered species in China. It has been found that YFP is constantly exposed to perfluorooctane sulfonate (PFOS) in aquatic environments, leading to significant bioaccumulation. However, the impacts of PFOS on YFP health and survival are still unknown. To circumvent the limitations in YFP research, this study used YFP umbilical cord fibroblast cell line and exposed the cells to PFOS for 48 h, with objectives to uncover the cytotoxicity and mechanisms of PFOS in YFP. A high-throughput proteomics assay showed that PFOS exposure at 50 μM for 48 h perturbed the proteome structure in YFP umbilical cord fibroblast cells. Functional annotation found the high relevance of oxidative stress, mitochondrial oxidative phosphorylation, and DNA damage to PFOS cytotoxic mechanisms. Concordantly, PFOS exposure significantly increased the deposition of reactive oxygen species (ROS) in YFP cells. The potential of mitochondria to produce ATP was also compromised by PFOS, which was accompanied by the higher permeability of mitochondrial membrane. In addition, exposure of YFP umbilical cord fibroblast cells to 50 μM PFOS damaged the DNA assembly as evidenced by the increase in the percentage of DNA fragmentation. Gene transcription and enzymatic activity of caspases were up-regulated by PFOS, subsequently favoring the occurrence of early and late apoptosis. It was notable that ROS scavenger could successfully mitigate the cytotoxicity of PFOS on oxidative stress and apoptosis, thus pinpointing ROS as the molecular initiating event in apoptosis endpoints. To our knowledge, this is the first study that investigates the detrimental effects of PFOS using YFP umbilical cord fibroblast cells. The data will support an accurate assessment of ecological risks imposed by environmental pollutants on the health and sustainability of YFP, which is especially important under the context of sharp decline in YFP population and national initiative in YFP conservation.
Collapse
Affiliation(s)
- Maaz Ahmad
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengyuan Liu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zixie Yang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haobo Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ghulam Nabi
- Department of Zoology, Institute of Molecular Biology and Biotechnology, University of Lahore, Lahore, Pakistan
| | - Yujiang Hao
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Lianguo Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| |
Collapse
|
2
|
Ahmad M, Hu C, Liu M, Zhang H, Shah SAUR, Nabi G, Hao Y, Chen L. Cytotoxicity and mechanisms of perfluorobutane sulfonate (PFBS) in umbilical cord fibroblast cells of Yangtze finless porpoise. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 276:107098. [PMID: 39298911 DOI: 10.1016/j.aquatox.2024.107098] [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: 07/27/2024] [Revised: 09/07/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Yangtze finless porpoises (YFP) accumulate high levels of per- and polyfluoroalkyl substances (PFASs). However, the health impacts of PFASs to YFP are still unknown because it is technically and ethically unfeasible to use the critically endangered YFP in toxicological exposures. To uncover the potential toxicities of PFASs to YFP, this study exposed a YFP umbilical cord fibroblast cell line to perfluorobutane sulfonate (PFBS), an emerging PFASs pollutant in the aquatic environments. After exposure, the cytotoxicity and mechanisms of PFBS were explored. Our preliminary experiments found that PFBS compromised the cell viability in a concentration and duration dependent manner. In an exposure of 48-h duration, the maximum no observed effect concentration (NOEC) of PFBS was determined to be 400 µM. High-throughput proteomics were then conducted to identify the differentially expressed proteins in YFP cells exposed to 400 µM PFBS for 48 h. The results found that PFBS exposure significantly perturbed the proteome fingerprints of YFP umbilical cord fibroblast cells. Functional annotation of differential proteins showed that PFBS had the potential to impair a variety of biological processes associated with the immunity, oxidative stress, metabolism, and proteolysis. Consistently, the intracellular levels of reactive oxygen species (ROS) and proinflammatory cytokine IL-1β were significantly increased by PFBS in YFP umbilical cord fibroblast cells. Overall, this study highlights the toxic effects of emerging PFASs on YFP and provides reference data to evaluate the health risks of aquatic pollution under the context of national YFP protection. To our knowledge, this is the first omics study using YFP umbilical cord fibroblast cells in ecotoxicology of PFASs, which is applicable to various cetacean species and pollutants.
Collapse
Affiliation(s)
- Maaz Ahmad
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chenyan Hu
- School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan 430072, China
| | - Mengyuan Liu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haobo Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Syed Ata Ur Rahman Shah
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ghulam Nabi
- Department of Zoology, Institute of Molecular Biology and Biotechnology, University of Lahore, Lahore, Pakistan
| | - Yujiang Hao
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Lianguo Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| |
Collapse
|
3
|
Wang J, Jia J, Wang D, Pan X, Xiong H, Li C, Jiang Y, Yan B. Zn 2+ loading as a critical contributor to the circ_0008553-mediated oxidative stress and inflammation in response to PM 2.5 exposures. J Environ Sci (China) 2023; 124:451-461. [PMID: 36182153 DOI: 10.1016/j.jes.2021.11.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/20/2021] [Accepted: 11/12/2021] [Indexed: 06/16/2023]
Abstract
Inflammation is a major adverse outcome induced by inhaled particulate matter with a diameter of ≤ 2.5 µm (PM2.5), and a critical trigger of most PM2.5 exposure-associated diseases. However, the key molecular events regulating the PM2.5-induced airway inflammation are yet to be elucidated. Considering the critical role of circular RNAs (circRNAs) in regulating inflammation, we predicted 11 circRNAs that may be involved in the PM2.5-induced airway inflammation using three previously reported miRNAs through the starBase website. A novel circRNA circ_0008553 was identified to be responsible for the PM2.5-activated inflammatory response in human bronchial epithelial cells (16HBE) via inducing oxidative stress. Using a combinatorial model PM2.5 library, we found that the synergistic effect of the insoluble core and loaded Zn2+ ions at environmentally relevant concentrations was the major contributor to the upregulation of circ_0008553 and subsequent induction of oxidative stress and inflammation in response to PM2.5 exposures. Our findings provided new insight into the intervention of PM2.5-induced adverse outcomes.
Collapse
Affiliation(s)
- Jingzhou Wang
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Jianbo Jia
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China.
| | - Dujia Wang
- School of Environmental Science and Engineering, Shandong University, Qingdao 266237, China
| | - Xiujiao Pan
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Haiyan Xiong
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Chengjun Li
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Yiguo Jiang
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China.
| | - Bing Yan
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China; School of Environmental Science and Engineering, Shandong University, Qingdao 266237, China.
| |
Collapse
|
4
|
Solan ME, Senthilkumar S, Aquino GV, Bruce ED, Lavado R. Comparative cytotoxicity of seven per- and polyfluoroalkyl substances (PFAS) in six human cell lines. Toxicology 2022; 477:153281. [PMID: 35933025 DOI: 10.1016/j.tox.2022.153281] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/17/2022] [Accepted: 08/01/2022] [Indexed: 01/09/2023]
Abstract
Human exposures to perfluoroalkyl and polyfluoroalkyl substances (PFAS) have been linked to several diseases associated with adverse health outcomes. Animal studies have been conducted, though these may not be sufficient due to the inherent differences in metabolic processes between humans and rodents. Acquiring relevant data on the health effects of short-chain PFAS can be achieved through methods supported by in vitro human cell-based models. Specifically, cytotoxicity assays are the crucial first step to providing meaningful information used for determining safety and providing baseline information for further testing. To this end, we exposed human cell lines representative of six different tissue types, including colon (CaCo-2), liver (HepaRG), kidney (HEK293), brain (HMC-3), lung (MRC-5), and muscle (RMS-13) to five short-chain PFAS and two legacy PFAS. The exposure of the individual PFAS was assessed using a range of concentrations starting from a low concentration (10-11 M) to a high concentration of (10-4 M). Our results indicated that CaCo-2 and HEK293 cells were the least sensitive to PFAS exposure, while HMC-3, HepaRG, MRC-5, and RMS-13 demonstrated significant decreases in viability in a relatively narrow range (EC50 ranging from 1 to 70 µM). The most sensitive cell line was the neural HMC-3 for all short- and long-chain PFAS (with EC50 ranging from 1.34 to 2.73 µM). Our data suggest that PFAS do not exert toxicity on all cell types equally, and the cytotoxicity estimates we obtained varied from previously reported values. Overall, this study is novel because it uses human cell lines that have not been widely used to understand human health outcomes associated with PFAS exposure.
Collapse
Affiliation(s)
- Megan E Solan
- Department of Environmental Science, Baylor University, Waco, TX 76798, USA
| | | | - Grace V Aquino
- Department of Environmental Science, Baylor University, Waco, TX 76798, USA
| | - Erica D Bruce
- Department of Environmental Science, Baylor University, Waco, TX 76798, USA
| | - Ramon Lavado
- Department of Environmental Science, Baylor University, Waco, TX 76798, USA.
| |
Collapse
|
5
|
Olugbami JO, Damoiseaux R, Odunola OA, Gimzewski JK. Mitigation of aflatoxin B1- and sodium arsenite-induced cytotoxicities in HUC-PC urinary bladder cells by curcumin and Khaya senegalensis. J Basic Clin Physiol Pharmacol 2020; 31:/j/jbcpp.ahead-of-print/jbcpp-2019-0309/jbcpp-2019-0309.xml. [PMID: 32324162 DOI: 10.1515/jbcpp-2019-0309] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/25/2020] [Indexed: 06/11/2023]
Abstract
Background Concomitant exposure to environmental/occupational toxicants such as aflatoxin B1 (AFB1) and arsenic in some regions of the world has been well reported. Therefore, this calls for the assessment of the efficacy of agents such as phytochemicals, which are already known for their ethno-medicinal uses in prophylaxis/remediation. We investigated the possible cytotoxic bio-interactions between AFB1 and sodium arsenite (SA) in urinary bladder cells. We also assessed the cytoprotective effects of curcumin and the ethanol stem bark extract of Khaya senegalensis (K2S). Methods The cells were exposed to graded levels of AFB1, SA, curcumin, and K2S for 24, 48, and 72 h. Subsequently, using optimum toxic concentrations of AFB1 and SA, respectively, the influence of non-toxic levels of curcumin and/or K2S was tested on exposure of the cells to AFB1 and/or SA. Hoechst 33342/propidium iodide staining technique was used to determine the end-points due to cytotoxicity with changes in adenosine triphosphate (ATP) levels determined using Promega's CellTiter-Glo luminescent assay. Results Co-treatment of the cells with AFB1 and SA resulted in synergy in cytotoxic effects. Cytotoxicity was reduced by 3.5- and 2.9-fold by pre-treatment of the cells with curcumin and K2S before treatment with AFB1, while post-treatment resulted in 1.1- and 2.6-fold reduction, respectively. Pre-exposure of the cells with curcumin and K2S before treatment with SA ameliorated cytotoxicity by 3.8- and 3.0-fold, but post-treatment caused a 1.2- and 1.3-fold reduction, respectively. Conclusions Pre-treatment of the cells with either curcumin or K2S exhibited cytoprotective effects by ameliorating AFB1- and SA-induced cytotoxicity with inferred tendencies to prevent carcinogenesis.
Collapse
Affiliation(s)
- Jeremiah Olorunjuwon Olugbami
- Cancer Research and Molecular Biology (CRMB) Laboratories, Department of Biochemistry, University of Ibadan, Ibadan, OY 200005, Nigeria
- Nano and Pico Characterisation (NPC) Laboratories, California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Robert Damoiseaux
- Molecular Screening and Shared Resources (MSSR), California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
- Department of Molecular and Medicinal Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Oyeronke Adunni Odunola
- Cancer Research and Molecular Biology (CRMB) Laboratories, Department of Biochemistry, University of Ibadan, Ibadan, OY 200005, Nigeria
| | - James Kazimierz Gimzewski
- Nano and Pico Characterisation (NPC) Laboratories, California NanoSystems Institute (CNSI), University of California, Los Angeles, CA 90095, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
- International Center for Materials Nanoarchitectonics (MANA) Satellite, National Institute for Materials Science (NIMS), Tsukuba, Japan
| |
Collapse
|
6
|
Yuan W, Chen J, Huang H, Cai Z, Ling Q, Huang F, Huang Z. Low-Dose Arsenic Trioxide Modulates the Differentiation of Mouse Embryonic Stem Cells. Chem Res Toxicol 2018; 31:472-481. [PMID: 29767511 DOI: 10.1021/acs.chemrestox.8b00027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Arsenic (As) is a well-known environmental pollutant, while arsenic trioxide (ATO) has been proven to be an effective treatment for acute promyelocytic leukemia, however, the mechanism underlying its dual effects is not fully understood. Embryonic stem cells (ESCs) exhibit properties of stemness and serve as a popular model to investigate epigenetic modifiers including environmental pollutants. Herein, the effects of low-dose ATO on differentiation were evaluated in vitro using a mouse ESCs (mESCs) cell line, CGR8. Cells treated with 0.2-0.5 μM ATO for 3-4 days had slight inhibition of proliferation with elevation of apoptosis, but obvious alterations of differentiation by morphological checking and alkaline phosphatase (AP) staining. Moreover, ATO exposure significantly decreased the mRNA expression of the stemness maintenance genes including Oct4, Nanog, and Rex-1 ( P < 0.01), whereas obviously increased some tissue-specific differentiation marker genes such as Gata4, Gata-6, AFP, and IHH. These alterations were consistent with the differentiation phenotype induced by retinoic acid (RA) and the expression patterns of distinct pluripotency markers such as SSEA-1 and Oct4. Furthermore, low-dose ATO led to a quantitative increase in Caspase 3 (CASP3) activation and subsequent cleavage of Nanog around 27 kDa, which corresponded with the mouse Nanog cleaved by CASP3 in a tube cleavage assay. Taken together, we suggest that low-dose ATO exposure will induce differentiation, other than apoptosis, of ESCs, such effects might be tuned partially by ATO-induced CASP3 activation and Nanog cleavage coupling with other differentiation related genes involved. The present findings provide a preliminary action mechanism of arsenic on the cell fate determination.
Collapse
Affiliation(s)
- Wenlin Yuan
- Department of Biotechnology, School of Life Science and Technology , Jinan University , Guangzhou 510632 , Guangdong Province , China
| | - Jun Chen
- Department of Biotechnology, School of Life Science and Technology , Jinan University , Guangzhou 510632 , Guangdong Province , China
| | - Hongren Huang
- Department of Biotechnology, School of Life Science and Technology , Jinan University , Guangzhou 510632 , Guangdong Province , China
| | - Zhihui Cai
- Department of Biotechnology, School of Life Science and Technology , Jinan University , Guangzhou 510632 , Guangdong Province , China
| | - Qinjie Ling
- Department of Biotechnology, School of Life Science and Technology , Jinan University , Guangzhou 510632 , Guangdong Province , China
| | - Feng Huang
- Department of Rehabilitation Medicine, School of Medical Engineering , Foshan University , Foshan 528000 , Guangdong Province , China
| | - Zhi Huang
- Department of Biotechnology, School of Life Science and Technology , Jinan University , Guangzhou 510632 , Guangdong Province , China
| |
Collapse
|
7
|
Weatherly LM, Nelson AJ, Shim J, Riitano AM, Gerson ED, Hart AJ, de Juan-Sanz J, Ryan TA, Sher R, Hess ST, Gosse JA. Antimicrobial agent triclosan disrupts mitochondrial structure, revealed by super-resolution microscopy, and inhibits mast cell signaling via calcium modulation. Toxicol Appl Pharmacol 2018; 349:39-54. [PMID: 29630968 DOI: 10.1016/j.taap.2018.04.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/22/2018] [Accepted: 04/04/2018] [Indexed: 01/15/2023]
Abstract
The antimicrobial agent triclosan (TCS) is used in products such as toothpaste and surgical soaps and is readily absorbed into oral mucosa and human skin. These and many other tissues contain mast cells, which are involved in numerous physiologies and diseases. Mast cells release chemical mediators through a process termed degranulation, which is inhibited by TCS. Investigation into the underlying mechanisms led to the finding that TCS is a mitochondrial uncoupler at non-cytotoxic, low-micromolar doses in several cell types and live zebrafish. Our aim was to determine the mechanisms underlying TCS disruption of mitochondrial function and of mast cell signaling. We combined super-resolution (fluorescence photoactivation localization) microscopy and multiple fluorescence-based assays to detail triclosan's effects in living mast cells, fibroblasts, and primary human keratinocytes. TCS disrupts mitochondrial nanostructure, causing mitochondria to undergo fission and to form a toroidal, "donut" shape. TCS increases reactive oxygen species production, decreases mitochondrial membrane potential, and disrupts ER and mitochondrial Ca2+ levels, processes that cause mitochondrial fission. TCS is 60 × more potent than the banned uncoupler 2,4-dinitrophenol. TCS inhibits mast cell degranulation by decreasing mitochondrial membrane potential, disrupting microtubule polymerization, and inhibiting mitochondrial translocation, which reduces Ca2+ influx into the cell. Our findings provide mechanisms for both triclosan's inhibition of mast cell signaling and its universal disruption of mitochondria. These mechanisms provide partial explanations for triclosan's adverse effects on human reproduction, immunology, and development. This study is the first to utilize super-resolution microscopy in the field of toxicology.
Collapse
Affiliation(s)
- Lisa M Weatherly
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA; Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Andrew J Nelson
- Department of Physics and Astronomy, University of Maine, Orono, ME, USA
| | - Juyoung Shim
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Abigail M Riitano
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Erik D Gerson
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | - Andrew J Hart
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA
| | | | - Timothy A Ryan
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Roger Sher
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Samuel T Hess
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA; Department of Physics and Astronomy, University of Maine, Orono, ME, USA.
| | - Julie A Gosse
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA; Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, USA.
| |
Collapse
|
8
|
BonakdarYazdi B, Khodagholi F, Shaerzadeh F, Sharifzadeh A, Ahmadi R, Sanati M, Mehdizadeh H, Payandehmehr B, Vali L, Jahromi MM, Taghizadeh G, Sharifzadeh M. The effect of arsenite on spatial learning: Involvement of autophagy and apoptosis. Eur J Pharmacol 2017; 796:54-61. [PMID: 27993642 DOI: 10.1016/j.ejphar.2016.12.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 12/03/2016] [Accepted: 12/15/2016] [Indexed: 12/11/2022]
Abstract
Spatial learning plays a major role in one's information recording. Arsenic is one of ubiquitous environmental toxins with known neurological effects. However, studies investigating the effects of arsenic on spatial learning and related mechanisms are limited. This study was planned toexaminethe effects of bilateral intra-hippocampal infusion of different concentrations of sodium arsenite (5, 10 and 100nM, 5µl/side) on spatial learning in Wistar rats. Moreover, we evaluated levels of LC3-II, Atg7 and Atg12 as reliable biomarkers of autophagy and caspase-3 and Bax/Bcl-2 ratio as indicators of apoptosis in the hippocampus. Interestingly, low concentrations of sodium arsenite (5 and 10nM) significantly increased spatial acquisition but pre-training administration of sodium arsenite100nM did not significantly alter spatial learning. LC3-II levels were significantly increased in groups treated with sodium arsenite 5 and 10nM and decreased in the group receiving arsenite 100nM compared to the control group. Atg7 and Atg12 levels were obviously higher in all groups treated with sodium arsenite compared to control. However, caspase-3 cleavage and Bax/Bcl-2 ratio were notably greater in 100nM, and lesser in 5nM arsenite group in comparison with control animals. The results of this study showed that the low concentrations of sodium arsenite could facilitate spatial learning. This facilitation could be attributed to neuronal autophagy induced by low concentrations of sodium arsenite. These findings may help to clarify the regulatory pathways for apoptosis and autophagy balance due to sodium arsenite.
Collapse
Affiliation(s)
- Behnoosh BonakdarYazdi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Toxicology and poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Shaerzadeh
- Department of Physiology, faculty of medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Ramesh Ahmadi
- Department of Physiology, Azad University, Qom, Iran
| | - Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Toxicology and poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran
| | - Hajar Mehdizadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Toxicology and poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Borna Payandehmehr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Toxicology and poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Vali
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Kuwait University, Sulaibekhat, Kuwait
| | | | - Ghorban Taghizadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Toxicology and poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Occupational Therapy, Faculty of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Toxicology and poisoning Research Centre, Tehran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Bogen KT, Arnold LL, Chowdhury A, Pennington KL, Cohen SM. Low-dose dose-response for reduced cell viability after exposure of human keratinocyte (HEK001) cells to arsenite. Toxicol Rep 2016; 4:32-38. [PMID: 28959622 PMCID: PMC5615095 DOI: 10.1016/j.toxrep.2016.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 11/01/2016] [Accepted: 12/08/2016] [Indexed: 12/21/2022] Open
Abstract
The in vitro arsenite (AsIII) cytotoxicity dose-response (DR) of human keratinocytes (HEK001) was examined at greater statistical resolution than ever previously reported using the MTT assay to determine cell viability. Fifty-four 96-well plates were treated with AsIII concentrations of 0.25, 0.5, 1, 2, 3, 4, 5, 7, 10, 15, 20, 25, or 30 μM. Because of unexpected variation in viability response patterns, a two-stage DR analysis was used in which data on plate-specific viability (%), estimated as 100% times the ratio of measured viability in exposed to unexposed cells, were fit initially to a generalized lognormal response function positing that HEK001 cells studied consisted of: a proportion P of relatively highly sensitive (HS) cells, a proportion Po of relatively resistant cells, and a remaining (1-P-Po) fraction of typical-sensitivity (TS) cells exhibiting the intermediate level of AsIII sensitivity characteristic of most cells in each assay. The estimated fractions P and Po were used to adjust data from all 54 plates (and from the 28 plates yielding the best fits) to reflect the condition that P = Po = 0 to provide detailed DR analysis specifically for TS cells. Four DR models fit to the combined adjusted data were each very predictive (R2 > 0.97) overall but were inconsistent with at least one of the data set examined (p < 10-5). Adjusted mean responses at ≤3 μM were approximately equal (p > 0.30) and exceeded 100% significance (p ≤ 10-6). A low-dose hormetic model provided the best fit to the combined adjusted data for TS cells (R2 = 0.995). Marked variability in estimates of P (the proportion of apparent HS cells) was unexpected, not readily explained, and warrants further study using additional cell lines and assay methods, and in vivo.
Collapse
Affiliation(s)
| | - Lora L. Arnold
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Karen L. Pennington
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Samuel M. Cohen
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
- Havlik-Wall Professor of Oncology
| |
Collapse
|
10
|
Luijten M, Zwart EP, Dollé MET, de Pooter M, Cox JA, White PA, van Benthem J. Evaluation of the LacZ reporter assay in cryopreserved primary hepatocytes for In vitro genotoxicity testing. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2016; 57:643-655. [PMID: 27859631 DOI: 10.1002/em.22063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 10/24/2016] [Indexed: 06/06/2023]
Abstract
Assessment of genotoxic potential is an important step in the safety evaluation of chemical substances. Under most regulatory jurisdictions, the first tier of testing comprises a standard battery of in vitro genotoxicity tests in bacterial and mammalian cells. However, the mammalian cell tests commonly used exhibit a relatively high rate of misleading positive results, which may lead to unnecessary in vivo testing. We previously established a proof-of-concept for the LacZ reporter assay in proliferating primary hepatocytes as a promising alternative genotoxicity test. Here, cryopreserved instead of freshly isolated hepatocytes were used and the assay was evaluated in more detail. We examined the effect of cryopreservation on phenotype and metabolic capacity of the LacZ hepatocytes, and assessed the predictive performance of the assay by testing a set of substances comprising true positive, true negative, and misleading positive substances. Additionally, a historical negative control database was created and the type of mutations induced was analyzed for two of the substances tested. Our findings indicate that proliferating cryopreserved primary hepatocytes derived from LacZ plasmid mice retain their hepatocyte-specific characteristics and metabolic competence. Furthermore, we demonstrate that both gene mutations and genome rearrangements due to large deletions can be detected with the LacZ reporter assay. The assay seems to have a lower rate of misleading positive test results compared to the assays currently used. Together, our findings strongly support the use of the LacZ reporter assay in cryopreserved primary hepatocytes as follow-up to the standard in vitro test battery for genotoxicity testing. Environ. Mol. Mutagen. 57:643-655, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Edwin P Zwart
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Martijn E T Dollé
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Maaike de Pooter
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Julie A Cox
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Paul A White
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Jan van Benthem
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|
11
|
Gülden M, Kähler D, Seibert H. Incipient cytotoxicity: A time-independent measure of cytotoxic potency in vitro. Toxicology 2015; 335:35-45. [DOI: 10.1016/j.tox.2015.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/01/2015] [Accepted: 07/02/2015] [Indexed: 10/23/2022]
|
12
|
Silveira MAD, Ribeiro DL, Dos Santos TA, Vieira GM, Cechinato CN, Kazanovski M, Grégio d'Arce LP. Mutagenicity of two herbicides widely used on soybean crops by the Allium cepa test. Cytotechnology 2015; 68:1215-22. [PMID: 25947236 DOI: 10.1007/s10616-015-9881-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 04/28/2015] [Indexed: 01/01/2023] Open
Abstract
This study evaluated the mutagenic effects of two herbicides: Clorimurom Nortox(®) and Imazaquim Ultra Nortox(®) widely used on soybean crops in Brazil. As a test system, Allium cepa assay was used, which analyzes the frequency of micronuclei (MN), chromosomal aberrations (CA) and the mitotic index (MI). Four concentrations of each herbicide (50, 75, 100 and 125 %) were tested in triplicate using distilled water (negative control) and methyl methanesulfonate (positive control) as controls. Three experimental repetitions were realized. Clorimurom Nortox(®) showed a significantly lower MI than the negative control for the concentrations of 75, 100 and 125 %, but the CA was significantly increased at all concentrations. There was no recovery for CA or MI. The 125 % concentration of Imazaquim Ultra Nortox(®) was cytotoxic and also exerted an effect on the other parameters. The concentration of 100 % showed a statistically increased MN and there was no recovery, while the 75 % concentration significantly affected CA, with recovery observed. The two herbicides showed mutagenic damage in Allium cepa cells, which implies a careful handling of these products, to minimize the risk of human and environmental contamination.
Collapse
Affiliation(s)
- Maruhen Amir Datsch Silveira
- Laboratório de Mutagênese, CCBS, Centro de Ciências Biológicas e da Saúde, Colegiado de Ciências Biológicas, Universidade Estadual do Oeste do Paraná (UNIOESTE), Universitária Street, 2069 - Jardim Universitário, Postal 00711, Cascavel, PR, 85819-110, Brazil.
| | - Diego Luis Ribeiro
- Laboratório de Mutagênese, CCBS, Centro de Ciências Biológicas e da Saúde, Colegiado de Ciências Biológicas, Universidade Estadual do Oeste do Paraná (UNIOESTE), Universitária Street, 2069 - Jardim Universitário, Postal 00711, Cascavel, PR, 85819-110, Brazil
| | - Thayná Assakawa Dos Santos
- Laboratório de Mutagênese, CCBS, Centro de Ciências Biológicas e da Saúde, Colegiado de Ciências Biológicas, Universidade Estadual do Oeste do Paraná (UNIOESTE), Universitária Street, 2069 - Jardim Universitário, Postal 00711, Cascavel, PR, 85819-110, Brazil
| | - Gabriela Maciel Vieira
- Laboratório de Mutagênese, CCBS, Centro de Ciências Biológicas e da Saúde, Colegiado de Ciências Biológicas, Universidade Estadual do Oeste do Paraná (UNIOESTE), Universitária Street, 2069 - Jardim Universitário, Postal 00711, Cascavel, PR, 85819-110, Brazil
| | - Carlye Nicheli Cechinato
- Laboratório de Mutagênese, CCBS, Centro de Ciências Biológicas e da Saúde, Colegiado de Ciências Biológicas, Universidade Estadual do Oeste do Paraná (UNIOESTE), Universitária Street, 2069 - Jardim Universitário, Postal 00711, Cascavel, PR, 85819-110, Brazil
| | - Michele Kazanovski
- Laboratório de Mutagênese, CCBS, Centro de Ciências Biológicas e da Saúde, Colegiado de Ciências Biológicas, Universidade Estadual do Oeste do Paraná (UNIOESTE), Universitária Street, 2069 - Jardim Universitário, Postal 00711, Cascavel, PR, 85819-110, Brazil
| | - Luciana Paula Grégio d'Arce
- Laboratório de Mutagênese, CCBS, Centro de Ciências Biológicas e da Saúde, Colegiado de Ciências Biológicas, Universidade Estadual do Oeste do Paraná (UNIOESTE), Universitária Street, 2069 - Jardim Universitário, Postal 00711, Cascavel, PR, 85819-110, Brazil
| |
Collapse
|
13
|
Speit G, Vesely A, Schutz P, Linsenmeyer R, Bausinger J. The low molecular weight DNA diffusion assay as an indicator of cytotoxicity for the in vitro comet assay. Mutagenesis 2014; 29:267-77. [DOI: 10.1093/mutage/geu015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
14
|
Ireno IC, Baumann C, Stöber R, Hengstler JG, Wiesmüller L. Fluorescence-based recombination assay for sensitive and specific detection of genotoxic carcinogens in human cells. Arch Toxicol 2014; 88:1141-59. [PMID: 24671466 DOI: 10.1007/s00204-014-1229-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 03/18/2014] [Indexed: 10/25/2022]
Abstract
In vitro genotoxicity tests are known to suffer from several shortcomings, mammalian cell-based assays, in particular, from low specificities. Following a novel concept of genotoxicity detection, we developed a fluorescence-based method in living human cells. The assay quantifies DNA recombination events triggered by DNA double-strand breaks and damage-induced replication fork stalling predicted to detect a broad spectrum of genotoxic modes of action. To maximize sensitivities, we engineered a DNA substrate encompassing a chemoresponsive element from the human genome. Using this substrate, we screened various human tumor and non-transformed cell types differing in the DNA damage response, which revealed that detection of genotoxic carcinogens was independent of the p53 status but abrogated by apoptosis. Cell types enabling robust and sensitive genotoxicity detection were selected for the generation of reporter clones with chromosomally integrated DNA recombination substrate. Reporter cell lines were scrutinized with 21 compounds, stratified into five sets according to the established categories for identification of carcinogenic compounds: genotoxic carcinogens ("true positives"), non-genotoxic carcinogens, compounds without genotoxic or carcinogenic effect ("true negatives") and non-carcinogenic compounds, which have been reported to induce chromosomal aberrations or mutations in mammalian cell-based assays ("false positives"). Our results document detection of genotoxic carcinogens in independent cell clones and at levels of cellular toxicities <60 % with a sensitivity of >85 %, specificity of ≥90 % and detection of false-positive compounds <17 %. Importantly, through testing cyclophosphamide in combination with primary hepatocyte cultures, we additionally provide proof-of-concept for the identification of carcinogens requiring metabolic activation using this novel assay system.
Collapse
Affiliation(s)
- Ivanildce C Ireno
- Department of Obstetrics and Gynecology, University of Ulm, Prittwitzstrasse 43, 89075, Ulm, Germany
| | | | | | | | | |
Collapse
|
15
|
Qin Y, Ma Y, Zheng B, Zhang L, Zhao Y. Temporal and spatial variation of arsenic species in the Dahuofang reservoir in northeast China. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2014; 21:1054-1063. [PMID: 23872891 DOI: 10.1007/s11356-013-1969-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 06/27/2013] [Indexed: 06/02/2023]
Abstract
Overlying water, pore water, and sediment samples were collected from the Dahuofang reservoir in November 2011 and April 2012, respectively. Total arsenic and arsenic species including arsenite, arsenate, monomethylarsonic, and dimethylarsinic were analyzed by ICP-MS and HPLC-ICP-MS. The results indicated that the environments of the Dahuofang reservoir were in reduced conditions, arsenite was the predominant species in pore water and sediments in the reservoir. Arsenic concentrations in overlying water were very low in all the samples but showed different trend during the different time. In November, arsenic concentrations in the reservoir inlet were higher than that in the other sites, whereas arsenic showed accumulation from the upstream to downstream of the reservoir in samples collected in April. In pore water, arsenic concentrations were about 23 and 37 times higher than those in overlying water in November and April, respectively, and relatively high levels of arsenite were also detected in the pore water. In surface sediments, total arsenic and arsenic species content in the reservoir inlet showed the following decreasing order: R1 > R10 > R4. The results also showed that moderate ecological risks exist in pore water and sediments in the Dahuofang reservoir.
Collapse
Affiliation(s)
- Yanwen Qin
- National Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, China
| | | | | | | | | |
Collapse
|
16
|
Lee YS, Lee EK, Oh HH, Choi CS, Kim S, Jun HS. Sodium meta-arsenite ameliorates hyperglycemia in obese diabetic db/db mice by inhibition of hepatic gluconeogenesis. J Diabetes Res 2014; 2014:961732. [PMID: 25610880 PMCID: PMC4290036 DOI: 10.1155/2014/961732] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/01/2014] [Accepted: 12/03/2014] [Indexed: 12/29/2022] Open
Abstract
Sodium meta-arsenite (SA) is implicated in the regulation of hepatic gluconeogenesis-related genes in vitro; however, the effects in vivo have not been studied. We investigated whether SA has antidiabetic effects in a type 2 diabetic mouse model. Diabetic db/db mice were orally intubated with SA (10 mg kg(-1) body weight/day) for 8 weeks. We examined hemoglobin A1c (HbA1c), blood glucose levels, food intake, and body weight. We performed glucose, insulin, and pyruvate tolerance tests and analyzed glucose production and the expression of gluconeogenesis-related genes in hepatocytes. We analyzed energy metabolism using a comprehensive animal metabolic monitoring system. SA-treated diabetic db/db mice had reduced concentrations of HbA1c and blood glucose levels. Exogenous glucose was quickly cleared in glucose tolerance tests. The mRNA expressions of genes for gluconeogenesis-related enzymes, glucose 6-phosphatase (G6Pase), and phosphoenolpyruvate carboxykinase (PEPCK) were significantly reduced in the liver of SA-treated diabetic db/db mice. In primary hepatocytes, SA treatment decreased glucose production and the expression of G6Pase, PEPCK, and hepatocyte nuclear factor 4 alpha (HNF-4α) mRNA. Small heterodimer partner (SHP) mRNA expression was increased in hepatocytes dependent upon the SA concentration. The expression of Sirt1 mRNA and protein was reduced, and acetylated forkhead box protein O1 (FoxO1) was induced by SA treatment in hepatocytes. In addition, SA-treated diabetic db/db mice showed reduced energy expenditure. Oral intubation of SA ameliorates hyperglycemia in db/db mice by reducing hepatic gluconeogenesis through the decrease of Sirt1 expression and increase in acetylated FoxO1.
Collapse
MESH Headings
- Acetylation
- Animals
- Arsenites/pharmacology
- Biomarkers/blood
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- Body Weight/drug effects
- Cells, Cultured
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/genetics
- Disease Models, Animal
- Eating/drug effects
- Energy Metabolism/drug effects
- Forkhead Box Protein O1
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Gluconeogenesis/drug effects
- Glucose-6-Phosphatase/genetics
- Glucose-6-Phosphatase/metabolism
- Glycated Hemoglobin/metabolism
- Hepatocyte Nuclear Factor 4/genetics
- Hepatocyte Nuclear Factor 4/metabolism
- Hypoglycemic Agents/pharmacology
- Liver/drug effects
- Liver/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Obesity/complications
- Phosphoenolpyruvate Carboxykinase (ATP)/genetics
- Phosphoenolpyruvate Carboxykinase (ATP)/metabolism
- RNA, Messenger/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Signal Transduction/drug effects
- Sirtuin 1/genetics
- Sirtuin 1/metabolism
- Sodium Compounds/pharmacology
- Time Factors
Collapse
Affiliation(s)
- Young-Sun Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 7-45 Songdo-dong, Yeonsu-gu, Incheon 406-840, Republic of Korea
| | - Eun-Kyu Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 7-45 Songdo-dong, Yeonsu-gu, Incheon 406-840, Republic of Korea
| | - Hyun-Hee Oh
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 7-45 Songdo-dong, Yeonsu-gu, Incheon 406-840, Republic of Korea
| | - Cheol Soo Choi
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 7-45 Songdo-dong, Yeonsu-gu, Incheon 406-840, Republic of Korea
- Endocrinology, Internal Medicine, Gachon University Gil Medical Center, Namdong-gu, Guwol-dong 1198, Incheon 405-760, Republic of Korea
- Gachon Medical Research Institute, Gil Hospital, Incheon 405-760, Republic of Korea
| | - Sujong Kim
- Komipharm International Co. Ltd., 3188 Seongnam-dong, Jungwon-gu, Seongnam-si, Gyeonggi-do 462-827, Republic of Korea
| | - Hee-Sook Jun
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 7-45 Songdo-dong, Yeonsu-gu, Incheon 406-840, Republic of Korea
- Gachon Medical Research Institute, Gil Hospital, Incheon 405-760, Republic of Korea
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, 7-45 Songdo-dong, Yeonsu-gu, Incheon 406-840, Republic of Korea
- *Hee-Sook Jun:
| |
Collapse
|
17
|
Cohen SM, Arnold LL, Beck BD, Lewis AS, Eldan M. Evaluation of the carcinogenicity of inorganic arsenic. Crit Rev Toxicol 2013; 43:711-52. [DOI: 10.3109/10408444.2013.827152] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
18
|
Sobral O, Marin-Morales MA, Ribeiro R. Could contaminant induced mutations lead to a genetic diversity overestimation? ECOTOXICOLOGY (LONDON, ENGLAND) 2013; 22:838-846. [PMID: 23686739 DOI: 10.1007/s10646-013-1079-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/30/2013] [Indexed: 06/02/2023]
Abstract
Contaminant driven genetic erosion reported through the inspection of selectable traits can be underestimated using neutral markers. This divergence was previously reported in the aquatic system of an abandoned pyrite mine. The most sensitive genotypes of the microcrustacean cladoceran Daphnia longispina were found to be lacking in the impacted reservoir near the entrance of the metal rich acid mine drainage (AMD). Since that divergence could be, at least partially, accounted for by mutagenicity and genotoxicity of the AMD, the present study aimed at providing such a characterization. The Allium cepa chromosomal aberration assay, using root meristematic cells, was carried out, by exposing seeds to 100, 10, 1, and 0.1 % of the local AMD. Chromosomal aberrations, cell division phases and cell death were quantified after the AMD exposure and after 24 and 48 h recovery periods. The AMD revealed to be mutagenic and genotoxic, even after diluting it to 1 and 0.1 %. Dilutions within this range were previously found to be below the lethality threshold and to elicit sublethal effects on reproduction of locally collected D. longispina clonal lineages Significant mutagenic effects (micronuclei and chromosomal breaks) were also found at 0.1 % AMD, supporting that exposure may induce permanent genetic alterations. Recovery tests showed that AMD genotoxic effects persisted after the exposure.
Collapse
Affiliation(s)
- Olímpia Sobral
- Department of Life Sciences, IMAR-Instituto do Mar, University of Coimbra, Apartado 3046, 3001-401 Coimbra, Portugal
| | | | | |
Collapse
|
19
|
O'Donovan M. A critique of methods to measure cytotoxicity in mammalian cell genotoxicity assays. Mutagenesis 2012; 27:615-21. [PMID: 22952148 DOI: 10.1093/mutage/ges045] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Various methods have been used to estimate cytotoxicity in mammalian cell genotoxicity assays since their introduction more than four decades ago, and although there is no agreement on whether any single method is optimal, there is now a better appreciation of their limitations. Methods based on aspects of cellular function are inevitably inaccurate unless some estimate of cell number is included, and those using some measure of cell proliferation give different results depending on the mathematical model used. Although it would be desirable, it is not possible to provide a universal measure of cytotoxicity because the phenomenon is so complex. There is some flexibility in the limits of cytotoxicity proposed in regulatory guidelines, and it can be argued these could be even less precise. Also, to make valid comparisons of the performance of different test systems, novel or established, it would seem essential to use similar measures of cytotoxicity.
Collapse
Affiliation(s)
- Mike O'Donovan
- Safety Assessment, AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK.
| |
Collapse
|
20
|
Kim SC, Park SJ, Lee JR, Seo JC, Yang CH, Byun SH. Cytoprotective Activity of Glycyrrhizae radix Extract Against Arsenite-induced Cytotoxicity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 5:165-71. [PMID: 18604262 PMCID: PMC2396482 DOI: 10.1093/ecam/nem014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2006] [Accepted: 01/16/2007] [Indexed: 11/14/2022]
Abstract
Licorice, Glycyrrhizae radix, is one of the herbal medicines in East Asia that has been commonly used for treating various diseases, including stomach disorders. This study investigated the effect of licorice on arsenite (As)-induced cytotoxicity in H4IIE cells, a rat hepatocyte-derived cell line. Cell viability was significantly diminished in As-treated H4IIE cells in a time and concentration-dependent manner. Furthermore, results from flow cytometric assay and DNA laddering in H4IIE cells showed that As treatment induced apoptotic cell death by activating caspase-3. Licorice (0.1 and 1.0 mg ml(-1)) treatment significantly inhibited cell death and the activity of caspase-3 in response to As exposure. These results demonstrate that licorice induced a cytoprotective effect against As-induced cell death by inhibition of caspase-3.
Collapse
Affiliation(s)
- Sang Chan Kim
- College of Oriental Medicine and Research Center for Biomedical Resources of Oriental Medicine, Daegu Haany University, 165 Sang-dong, Suseong-gu, Daegu 706-060, Korea
| | | | | | | | | | | |
Collapse
|
21
|
Bailey KA, Hester SD, Knapp GW, Owen RD, Thai SF. Gene expression of normal human epidermal keratinocytes modulated by trivalent arsenicals. Mol Carcinog 2011; 49:981-98. [PMID: 20886546 DOI: 10.1002/mc.20677] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Chronic exposure to inorganic arsenic (iAs) is associated with the development of benign and malignant human skin lesions including nonmelanoma skin cancers. The precise arsenical form(s) responsible for this carcinogenic effect are unknown, although trivalent inorganic arsenic (iAs(III)) and two of its toxic metabolites, monomethylarsonous acid (MMA(III)) and methylarsinous acid (DMA(III)), are attractive candidates. In an effort to better understand and compare their toxic effects in the skin, we compared the global gene expression profiles of normal human epidermal keratinocytes (NHEKs) exposed to varying noncytotoxic/slightly cytotoxic concentrations of iAs(III), MMA(III), and DMA(III) for 24 h. Exposure to each arsenical treatment group exhibited a dose effect in the number of altered genes and the magnitude of expression change in NHEKs. The most significant gene expression changes associated with iAs(III) and MMA(III) exposure were consistent with several key events believed to be important to As-driven skin carcinogenesis, namely induction of oxidative stress, increased transcript levels of keratinocyte growth factors, and modulation of MAPK and NF-κB pathways. At both comparable arsenical concentrations and comparable NHEK toxicity, greater potential carcinogenic effects were observed in MMA(III)-exposed NHEKs than those exposed to iAs(III), including involvement of more proinflammatory signals and increased transcript levels of more growth factor genes. In contrast, none of these above-mentioned transcriptional trends were among the most significantly altered functions in the DMA(III) treatment group. This study suggests the relative capacity of each of the tested arsenicals to drive suspected key events in As-mediated skin carcinogenesis is MMA(III) > iAs(III) with little contribution from DMA(III).
Collapse
Affiliation(s)
- Kathryn A Bailey
- US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | | | | | | | | |
Collapse
|
22
|
Hutchinson LM, Trinh BM, Palmer RK, Preziosi CA, Pelletier JH, Nelson HM, Gosse JA. Inorganic arsenite inhibits IgE receptor-mediated degranulation of mast cells. J Appl Toxicol 2010; 31:231-41. [PMID: 20842677 DOI: 10.1002/jat.1585] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 07/23/2010] [Accepted: 07/27/2010] [Indexed: 11/10/2022]
Abstract
Millions of people worldwide are exposed to arsenic (As), a toxicant which increases the risk of various cancers, cardiovascular disease and several other health problems. Arsenic is a potent endocrine disruptor, including of the estrogen receptor. It was recently shown that environmental estrogen-receptor disruptors can affect the signaling of mast cells, which are important players in parasite defense, asthma and allergy. Antigen (Ag) or allergen crosslinking of IgE-bound receptors on mast cells leads to signaling, culminating in degranulation, the release of histamine and other mediators. Because As is an endocrine disruptor and because endocrine disruptors have been found to affect degranulation, here we have tested whether sodium arsenite affects degranulation. Using the rat basophilic leukemia (RBL) mast cell model, we have measured degranulation in a fluorescence assay. Arsenic alone had no effect on basal levels of degranulation. However, As strongly inhibited Ag-stimulated degranulation at environmentally relevant concentrations, in a manner that is very dependent on concentrations of both As and Ag. The concentrations of As effective at inhibiting degranulation were not cytotoxic. This inhibition may be a mechanism underlying the traditional Chinese medicinal use of As to treat asthma. These data indicate that As may inhibit the ability of humans to fight off parasitic disease.
Collapse
Affiliation(s)
- Lee M Hutchinson
- Department of Molecular & Biomedical Sciences, University of Maine, Orono, ME 04469, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Soria EA, Eynard AR, Bongiovanni GA. Cytoprotective effects of silymarin on epithelial cells against arsenic-induced apoptosis in contrast with quercetin cytotoxicity. Life Sci 2010; 87:309-15. [DOI: 10.1016/j.lfs.2010.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 06/18/2010] [Accepted: 07/02/2010] [Indexed: 10/19/2022]
|
24
|
McKim JM. Building a tiered approach to in vitro predictive toxicity screening: a focus on assays with in vivo relevance. Comb Chem High Throughput Screen 2010; 13:188-206. [PMID: 20053163 PMCID: PMC2908937 DOI: 10.2174/138620710790596736] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Accepted: 05/05/2009] [Indexed: 12/23/2022]
Abstract
One of the greatest challenges facing the pharmaceutical industry today is the failure of promising new drug candidates due to unanticipated adverse effects discovered during preclinical animal safety studies and clinical trials. Late stage attrition increases the time required to bring a new drug to market, inflates development costs, and represents a major source of inefficiency in the drug discovery/development process. It is generally recognized that early evaluation of new drug candidates is necessary to improve the process. Building in vitro data sets that can accurately predict adverse effects in vivo would allow compounds with high risk profiles to be deprioritized, while those that possess the requisite drug attributes and a lower risk profile are brought forward. In vitro cytotoxicity assays have been used for decades as a tool to understand hypotheses driven questions regarding mechanisms of toxicity. However, when used in a prospective manner, they have not been highly predictive of in vivo toxicity. Therefore, the issue may not be how to collect in vitro toxicity data, but rather how to translate in vitro toxicity data into meaningful in vivo effects. This review will focus on the development of an in vitro toxicity screening strategy that is based on a tiered approach to data collection combined with data interpretation.
Collapse
Affiliation(s)
- James M McKim
- CeeTox Inc., 4717 Campus Dr., Kalamazoo, MI 49008, USA.
| |
Collapse
|
25
|
Komissarova EV, Rossman TG. Arsenite induced poly(ADP-ribosyl)ation of tumor suppressor P53 in human skin keratinocytes as a possible mechanism for carcinogenesis associated with arsenic exposure. Toxicol Appl Pharmacol 2010; 243:399-404. [PMID: 20036271 PMCID: PMC2830301 DOI: 10.1016/j.taap.2009.12.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Revised: 12/09/2009] [Accepted: 12/12/2009] [Indexed: 11/17/2022]
Abstract
Arsenite is an environmental pollutant. Exposure to inorganic arsenic in drinking water is associated with elevated cancer risk, especially in skin. Arsenite alone does not cause skin cancer in animals, but arsenite can enhance the carcinogenicity of solar UV. Arsenite is not a significant mutagen at non-toxic concentrations, but it enhances the mutagenicity of other carcinogens. The tumor suppressor protein P53 and nuclear enzyme PARP-1 are both key players in DNA damage response. This laboratory demonstrated earlier that in cells treated with arsenite, the P53-dependent increase in p21(WAF1/CIP1) expression, normally a block to cell cycle progression after DNA damage, is deficient. Here we show that although long-term exposure of human keratinocytes (HaCaT) to a nontoxic concentration (0.1 microM) of arsenite decreases the level of global protein poly(ADP-ribosyl)ation, it increases poly(ADP-ribosyl)ation of P53 protein and PARP-1 protein abundance. We also demonstrate that exposure to 0.1 microM arsenite depresses the constitutive expression of p21 mRNA and P21 protein in HaCaT cells. Poly(ADP-ribosyl)ation of P53 is reported to block its activation, DNA binding and its functioning as a transcription factor. Our results suggest that arsenite's interference with activation of P53 via poly(ADP-ribosyl)ation may play a role in the comutagenic and cocarcinogenic effects of arsenite.
Collapse
Affiliation(s)
- Elena V. Komissarova
- The Nelson Institute of Environmental Medicine NYU Langone School of Medicine 57 Old Forge Road Tuxedo, NY 10987
| | - Toby G. Rossman
- The Nelson Institute of Environmental Medicine NYU Langone School of Medicine 57 Old Forge Road Tuxedo, NY 10987
| |
Collapse
|
26
|
Wang A, Kligerman AD, Holladay SD, Wolf DC, Robertson JL. Arsenate and dimethylarsinic acid in drinking water did not affect DNA damage repair in urinary bladder transitional cells or micronuclei in bone marrow. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2009; 50:760-770. [PMID: 19472316 DOI: 10.1002/em.20496] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Arsenic is a human skin, lung, and urinary bladder carcinogen, and may act as a cocarcinogen in the skin and urinary bladder. Possible modes of action of arsenic carcinogenesis/cocarcinogenesis include oxidative stress induction and inhibition of DNA damage repair. We investigated the effects of arsenic in drinking water on DNA damage repair in urinary bladder transitional cells and on micronucleus formation in bone marrow. F344 rats were given 100 ppm arsenate [As(V)] or dimethylarsinic acid [DMA(V)] in drinking water for 1 week. The in vivo repair of cyclophosphamide (CP)-induced DNA damage resulting from a single oral gavage of CP, and the in vitro repair of hydrogen peroxide (H(2)O(2))- or formaldehyde-induced DNA damage, resulting from adding H(2)O(2) or formaldehyde into cell medium, were measured by the Comet assay. DMA(V) effects were not observed on either CP-induced DNA damage induction or on DNA repair. Neither DMA(V) nor As(V) increased the H(2)O(2)- or formaldehyde-induced DNA damage, and neither inhibited the repair of H(2)O(2)-induced DNA damage. Neither DMA(V) nor As(V) increased the micronucleus frequency, nor did they elevate micronucleus frequency resulting from CP treatment above the level observed by the treatment with CP alone. These results suggest that arsenic carcinogenesis/cocarcinogenesis in the urinary bladder may not be via DNA damage repair inhibition. To our knowledge this is the first report of arsenic effects on DNA damage repair in the urinary bladder.
Collapse
Affiliation(s)
- Amy Wang
- Department of Biomedical Sciences and Pathobiology, Virginia Maryland Regional College of Veterinary Medicine, Blacksburg, Virginia 24061, USA.
| | | | | | | | | |
Collapse
|
27
|
Huang Z, Li J, Zhang S, Zhang X. Inorganic arsenic modulates the expression of selenoproteins in mouse embryonic stem cell. Toxicol Lett 2009; 187:69-76. [DOI: 10.1016/j.toxlet.2009.01.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 01/18/2009] [Accepted: 01/20/2009] [Indexed: 12/15/2022]
|
28
|
Rossman TG. Inappropriate cytotoxicity measurements. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2009; 50:81. [PMID: 19152380 DOI: 10.1002/em.20446] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
|
29
|
Liu Y, Templeton DM. Initiation of caspase-independent death in mouse mesangial cells by Cd2+: involvement of p38 kinase and CaMK-II. J Cell Physiol 2008; 217:307-18. [PMID: 18506790 DOI: 10.1002/jcp.21499] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cadmium (Cd) is a toxic metal with multiple effects on cell signaling and cell death. We studied the effects of Cd(2+) on quiescent mouse mesangial cells in serum-free conditions. Cadmium induces cell death over 6 h through annexin V+ states without or with causing uptake of propidium iodide, termed apoptotic and apoptosis-like death, respectively. Little or no necrosis is observed, and cell death is caspase-independent and associated with nuclear translocation of the apoptosis-inducing factor, AIF. We previously showed that Cd(2+) increased phosphorylation of Erk and CaMK-II, and CaMK-II activation increased cell death in an Erk-independent manner. Here we demonstrate that Cd(2+) increases Jnk and p38 kinase phosphorylation, and inhibition of p38-but not of Jnk-increases cell viability by suppressing apoptosis in preference to apoptosis-like death. Neither p38 kinase nor CaMK-II inhibition protects against a decrease in mitochondrial membrane potential, psi, indicating that kinase-mediated death is either independent of, or involves events downstream of a mitochondrial pathway. However, both the antioxidant N-acetyl cysteine (NAC) and the mitochondrial membrane-stabilizing agent cyclosporine A (CsA) partially preserve psi, suppress activation of p38 kinase, and partially protect the cells from Cd(2+)-induced death. Whereas the effect of CsA is on apoptosis, NAC acts on apoptosis-like death. Inhibition of glutathione synthesis exacerbates a Cd(2+)-dependent increase in cellular peroxides and favors apoptosis-like death over apoptosis. The caspase-independence of these modes of cell death is not due to an absence of this machinery in the mesangial cells: when they are exposed to Cd(2+) for longer periods in the presence of serum, procaspase-3 and PARP are cleaved and caspase inhibition is protective. We conclude that Cd(2+) can kill mesangial cells by multiple pathways, including caspase-dependent and -independent apoptotic and apoptosis-like death. Necrosis is not prominent. Activation of p38 kinase and of CaMK-II by Cd(2+) are associated with caspase-independent apoptosis that is not dependent on mitochondrial destabilization.
Collapse
Affiliation(s)
- Ying Liu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | | |
Collapse
|
30
|
Chanda D, Kim SJ, Lee IK, Shong M, Choi HS. Sodium arsenite induces orphan nuclear receptor SHP gene expression via AMP-activated protein kinase to inhibit gluconeogenic enzyme gene expression. Am J Physiol Endocrinol Metab 2008; 295:E368-79. [PMID: 18505831 DOI: 10.1152/ajpendo.00800.2007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sodium arsenite has been demonstrated to alter the expression of genes associated with glucose homeostasis in tissues involved in the pathogenesis of type 2 diabetes; however, the underlying molecular mechanism has not been fully elucidated yet. In this study, we report that the sodium arsenite-induced gene expression of the small heterodimer partner (SHP; NR0B2), an atypical orphan nuclear receptor, regulates the expression of hepatic gluconeogenic genes. Sodium arsenite augments hepatic SHP mRNA levels in an AMP-activated protein kinase (AMPK)-dependent manner. Sodium arsenite activated AMPK and was shown to perturb cellular ATP levels. The arsenite-induced SHP mRNA level was blocked by adenoviral overexpression of dominant negative AMPK (Ad-dnAMPKalpha) or by the AMPK inhibitor compound C in hepatic cell lines. We demonstrated the dose-dependent induction of SHP mRNA levels by sodium arsenite and repressed the forskolin/dexamethasone-induced gene expression of the key hepatic gluconeogenic genes phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase). Ad-dnAMPKalpha blocked the repressive effects of arsenite-induced SHP on PEPCK and G6Pase. Sodium arsenite inhibited the promoter activity of PEPCK and G6Pase, and this repression was abolished by small interfering (si)RNA SHP treatments. The knockdown of SHP expression by oligonucleotide siRNA SHP or adenoviral siRNA SHP released the sodium arsenite-mediated repression of forskolin/dexamethasone-stimulated PEPCK and G6Pase gene expression in a variety of hepatic cell lines. Results from our study suggest that sodium arsenite induces SHP via AMPK to inhibit the expression of hepatic gluconeogenic genes and also provide us with a novel molecular mechanism of arsenite-mediated regulation of hepatic glucose homeostasis.
Collapse
Affiliation(s)
- Dipanjan Chanda
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | | | | | | | | |
Collapse
|
31
|
Komissarova EV, Li P, Uddin AN, Chen X, Nadas A, Rossman TG. Gene expression levels in normal human lymphoblasts with variable sensitivities to arsenite: identification of GGT1 and NFKBIE expression levels as possible biomarkers of susceptibility. Toxicol Appl Pharmacol 2008; 226:199-205. [PMID: 17976673 PMCID: PMC2299492 DOI: 10.1016/j.taap.2007.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2007] [Revised: 08/30/2007] [Accepted: 09/05/2007] [Indexed: 12/21/2022]
Abstract
Drinking arsenic-contaminated water is associated with increased risk of neoplasias of the skin, lung, bladder and possibly other sites, as well as other diseases. Earlier, we showed that human lymphoblast lines from different normal unexposed donors showed variable sensitivities to the toxic effects of arsenite. In the present study, we used microarray analysis to compare the basal gene expression profiles between two arsenite-resistant (GM02707, GM00893) and two arsenite-sensitive lymphoblast lines (GM00546, GM00607). A number of genes were differentially expressed in arsenite-sensitive and arsenite-resistant cells. Among these, gamma-glutamyltranspeptidase 1 (GGT1) and NF kappa B inhibitor-epsilon (NFKBIE) showed higher expression levels in arsenite-resistant cells. RT-PCR analysis with gene-specific primers confirmed these results. Reduction of GGT1 expression level in arsenite-resistant lymphoblasts with GGT1-specific siRNA resulted in increased cell sensitivity to arsenite. In conclusion, we have demonstrated for the first time that expression levels of GGT1 and possibly NFKBIE might be useful as biomarkers of genetic susceptibility to arsenite. Expression microarrays can thus be exploited for identifying additional biomarkers of susceptibility to arsenite and to other toxicants.
Collapse
Affiliation(s)
- Elena V Komissarova
- The Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| | | | | | | | | | | |
Collapse
|
32
|
Sykora P, Snow ET. Modulation of DNA polymerase beta-dependent base excision repair in cultured human cells after low dose exposure to arsenite. Toxicol Appl Pharmacol 2007; 228:385-94. [PMID: 18252256 DOI: 10.1016/j.taap.2007.12.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 12/13/2007] [Accepted: 12/17/2007] [Indexed: 11/29/2022]
Abstract
Base excision repair (BER) is crucial for development and for the repair of endogenous DNA damage. However, unlike nucleotide excision repair, the regulation of BER is not well understood. Arsenic, a well-established human carcinogen, is known to produce oxidative DNA damage, which is repaired primarily by BER, whilst high doses of arsenic can also inhibit DNA repair. However, the mechanism of repair inhibition by arsenic and the steps inhibited are not well defined. To address this question we have investigated the regulation of DNA polymerase beta (Pol beta) and AP endonuclease (APE1), in response to low, physiologically relevant doses of arsenic. GM847 lung fibroblasts and HaCaT keratinocytes were exposed to sodium arsenite, As(III), and mRNA, protein levels and BER activity were assessed. Both Pol beta and APE1 mRNA exhibited significant dose-dependant down regulation at doses of As(III) above 1 microM. However, at lower doses Pol beta mRNA and protein levels, and consequently, BER activity were significantly increased. In contrast, APE1 protein levels were only marginally increased by low doses of As(III) and there was no correlation between APE1 and overall BER activity. Enzyme supplementation of nuclear extracts confirmed that Pol beta was rate limiting. These changes in BER correlated with overall protection against sunlight UV-induced toxicity at low doses of As(III) and produced synergistic toxicity at high doses. The results provide evidence that changes in BER due to low doses of arsenic could contribute to a non-linear, threshold dose response for arsenic carcinogenesis.
Collapse
Affiliation(s)
- Peter Sykora
- Centre for Cellular and Molecular Biology, School of Biological Sciences, Deakin University, Australia
| | | |
Collapse
|
33
|
Wang XJ, Sun Z, Chen W, Eblin KE, Gandolfi AJ, Zhang DD. Nrf2 protects human bladder urothelial cells from arsenite and monomethylarsonous acid toxicity. Toxicol Appl Pharmacol 2007; 225:206-13. [PMID: 17765279 PMCID: PMC2610476 DOI: 10.1016/j.taap.2007.07.016] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2007] [Revised: 07/25/2007] [Accepted: 07/26/2007] [Indexed: 11/28/2022]
Abstract
Arsenic is widely spread in our living environment and imposes a big challenge on human health worldwide. Arsenic damages biological systems through multiple mechanisms including the generation of reactive oxygen species. The transcription factor Nrf2 regulates the cellular antioxidant response that protects cells from various insults. In this study, the protective role of Nrf2 in arsenic toxicity was investigated in a human bladder urothelial cell line, UROtsa. Using a UROtsa cell line stably infected with Nrf2-siRNA, we clearly demonstrate that compromised Nrf2 expression sensitized the cells to As(III)- and MMA(III)-induced toxicity. On the other hand, the activation of the Nrf2 pathway by tert-butylhydroquinone (tBHQ) and sulforaphane (SF), the known Nrf2-inducers, rendered UROtsa cells more resistant to As(III) and MMA(III). Furthermore, the wild-type mouse embryo fibroblast (WT-MEF) cells were protected from As(III)- and MMA(III)-induced toxicity following Nrf2 activation by tBHQ or SF, whereas neither tBHQ nor SF conferred protection in the Nrf2(-/-)MEF cells, demonstrating that tBHQ- or SF-mediated protection against As(III)- and MMA(III)-induced toxicity depends on Nrf2 activation. These results, obtained by both loss of function and gain of function analyses, clearly demonstrate the protective role of Nrf2 in arsenic-induced toxicity. The current work lays the groundwork for using Nrf2 activators for therapeutic and dietary interventions against adverse effects of arsenic.
Collapse
Affiliation(s)
- Xiao-Jun Wang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | - Zheng Sun
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | - Weimin Chen
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | - Kylee E. Eblin
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | - A. Jay Gandolfi
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| | - Donna D. Zhang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721
| |
Collapse
|
34
|
Klein CB, Leszczynska J, Hickey C, Rossman TG. Further evidence against a direct genotoxic mode of action for arsenic-induced cancer. Toxicol Appl Pharmacol 2007; 222:289-97. [PMID: 17316729 PMCID: PMC1986829 DOI: 10.1016/j.taap.2006.12.033] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Revised: 12/26/2006] [Accepted: 12/29/2006] [Indexed: 11/18/2022]
Abstract
Arsenic in drinking water, a mixture of arsenite and arsenate, is associated with increased skin and other cancers in Asia and Latin America, but not the United States. Arsenite alone in drinking water does not cause skin cancers in experimental animals; therefore, it is not a complete carcinogen in skin. We recently showed that low concentrations of arsenite enhanced the tumorigenicity of solar UV irradiation in hairless mice, suggesting arsenic cocarcinogenesis with sunlight in skin cancer and perhaps with different carcinogenic partners for lung and bladder tumors. Cocarcinogenic mechanisms could include blocking DNA repair, stimulating angiogenesis, altering DNA methylation patterns, dysregulating cell cycle control, induction of aneuploidy and blocking apoptosis. Arsenicals are documented clastogens but not strong mutagens, with weak mutagenic activity reported at highly toxic concentrations of inorganic arsenic. Previously, we showed that arsenite, but not monomethylarsonous acid (MMA[III]), induced delayed mutagenesis in HOS cells. Here, we report new data on the mutagenicity of the trivalent methylated arsenic metabolites MMA(III) and dimethylarsinous acid [DMA(III)] at the gpt locus in Chinese hamster G12 cells. Both methylated arsenicals seemed mutagenic with apparent sublinear dose responses. However, significant mutagenesis occurred only at highly toxic concentrations of MMA(III). Most mutants induced by MMA(III) and DMA(III) exhibited transgene deletions. Some non-deletion mutants exhibited altered DNA methylation. A critical discussion of cell survival leads us to conclude that clastogenesis occurs primarily at highly cytotoxic arsenic concentrations, casting further doubt as to whether a genotoxic mode of action (MOA) for arsenicals is supportable.
Collapse
Affiliation(s)
- Catherine B. Klein
- The Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987
| | - Joanna Leszczynska
- The Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987
| | - Christina Hickey
- The Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987
| | - Toby G. Rossman
- The Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987
| |
Collapse
|
35
|
Fellows MD, O'Donovan MR. Cytotoxicity in cultured mammalian cells is a function of the method used to estimate it. Mutagenesis 2007; 22:275-80. [PMID: 17456508 DOI: 10.1093/mutage/gem013] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Up to prescribed limits, the maximum test compound concentrations used in mammalian cell genotoxicity assays in vitro are determined by cytotoxicity, unless limited by solubility in solvents or culture medium. However, 'cytotoxicity' is different in the various test systems, both in the methods used to estimate it and the levels of toxicity that must be achieved. For example, in cytogenetic assays, the acceptable level of toxicity is defined as a 'significant reduction (>50%)' in cell number, culture confluency or mitotic index (MI) (OECD 473, ICH S2A), whereas mutation tests require relative total growth or cloning efficiency (CE) to be reduced by 80-90% (OECD 476, ICH S2A). In this study using mouse lymphoma cells, it was shown that, for a variety of agents with differing modes of action, cytotoxicity varies considerably depending on the method used to estimate it. Specifically, trypan blue exclusion, MI and binucleate incidence all grossly underestimate cytotoxicity in comparison with cell growth or CE. If the performance of different test systems is to be compared, or if data from different assays are to be used for the meaningful assessment of a novel chemical entity, it is essential that similar methods to determine cytotoxicity are used for them all. The purpose of this paper is not to recommend a specific method to determine cytotoxicity, although it can be argued that any such method must quantify the proportion of cells capable of division following treatment, but rather to draw attention to the fact that apparent toxicity depends upon the method used to estimate it.
Collapse
Affiliation(s)
- Michael D Fellows
- Genetic Toxicology, Safety Assessment, AstraZeneca, R&D Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | | |
Collapse
|
36
|
A novel method for the evaluation of proximal tubule epithelial cellular necrosis in the intact rat kidney using ethidium homodimer. BMC PHYSIOLOGY 2007; 7:1. [PMID: 17319948 PMCID: PMC1810561 DOI: 10.1186/1472-6793-7-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Accepted: 02/23/2007] [Indexed: 01/08/2023]
Abstract
BACKGROUND Ethidium homodimer is a cell-membrane impermeant nuclear fluorochrome that has been widely used to identify necrotic cells in culture. Here, we describe a novel technique for evaluating necrosis of epithelial cells in the proximal tubule that involves perfusing ethidium homodimer through the intact rat kidney. As a positive control for inducing necrosis, rats were treated with 3.5, 1.75, 0.87 and 0.43 mg/kg mercuric chloride (Hg2+, intraperitoneal), treatments which have previously been shown to rapidly cause dose-dependent necrosis of the proximal tubule. Twenty-four h after the administration of Hg2+, ethidium homodimer (5 microM) was perfused through the intact left kidney while the animal was anesthetized. The kidney was then removed, placed in embedding medium, frozen and cryosectioned at a thickness of 5 microm. Sections were permeabilized with -20 degrees C methanol and then stained with 4',6-diamidino-2-phenylindole (DAPI) to label total nuclei. Total cell number was determined from the DAPI staining in random microscopic fields and the number of necrotic cells in the same field was determined by ethidium homodimer labeling. RESULTS The Hg2+-treated animals showed a dose-dependent increase in the number of ethidium labeled cells in the proximal tubule, but not in other segments of the nephron. Other results showed that a nephrotoxic dose of gentamicin also caused a significant increase in the number of ethidium labeled cells in the proximal tubule. CONCLUSION These results indicate that this simple and sensitive perfusion technique can be used to evaluate cellular necrosis in the proximal tubule with the three-dimensional cyto-architecture intact.
Collapse
|
37
|
Green RM, Graham M, O'Donovan MR, Chipman JK, Hodges NJ. Subcellular compartmentalization of glutathione: correlations with parameters of oxidative stress related to genotoxicity. Mutagenesis 2006; 21:383-90. [PMID: 17012304 DOI: 10.1093/mutage/gel043] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Glutathione (GSH) is a major component of the antioxidant defence system of mammalian cells and is found in subcellular pools within the cytoplasm, nucleus and mitochondria. To evaluate the relationships between these pools and parameters of oxidative stress related to genotoxicity, wild type (WT) and 8-oxo-2'-deoxyguanosine glycosylase 1 (OGG1)-null (mOGG1(-/-)) mouse embryonic fibroblasts (MEF) were treated with buthionine sulphoximine (BSO; 0-1000 microM, 24 h), an inhibitor of GSH biosynthesis. BSO treatment resulted in a concentration-dependent depletion of GSH from the cytoplasm, but depletion of mitochondrial and nuclear GSH occurred only at concentrations > or =100 microM. GSH levels were correlated with reactive oxygen species (ROS), lipid peroxidation (measured as the increase in the genotoxic end-product malondialdehyde (MDA)) and oxidative DNA modifications, measured as both frank DNA strand-breaks (FSB) and oxidized purine lesions (OxP) using the alkaline comet assay with formamidopyrimidine DNA glycosylase (FPG) modification; this system allowed for the identification of BSO-induced DNA modifications as primarily mutagenic 8-oxo-2'-deoxyguanosine lesions. A number of significant correlations were observed. First, negative linear correlations were observed between mitochondrial GSH and ROS (r = -0.985 and r = -0.961 for WT and mOGG1(-/-) MEF, respectively), and mitochondrial GSH and MDA (r = -0.967 and r = -0.963 for WT and mOGG1(-/-) MEF, respectively). Second, positive linear correlations were observed between ROS and MDA (r = 0.996 and r = 0.935 for WT and mOGG1(-/-) MEF, respectively), and ROS and OxP (r = 0.938 and r = 0.981 for WT and mOGG1(-/-) MEF, respectively). Finally, oxidative DNA modifications displayed a negative linear correlation with nuclear GSH (r = -0.963 and -0.951 between nuclear GSH and FSB and OxP, respectively, for WT MEF and r = -0.960 between nuclear GSH and OxP in mOGG1(-/-) MEF), thus, demonstrating the genotoxic potential of compounds that deplete GSH. The findings highlight the critical roles of the mitochondrial and nuclear GSH pools in protecting cellular components, particularly DNA, from oxidative modification.
Collapse
Affiliation(s)
- Richard M Green
- School of Biosciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | | | | | | |
Collapse
|
38
|
Tapio S, Grosche B. Arsenic in the aetiology of cancer. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2006; 612:215-246. [PMID: 16574468 DOI: 10.1016/j.mrrev.2006.02.001] [Citation(s) in RCA: 195] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Revised: 01/25/2006] [Accepted: 02/07/2006] [Indexed: 12/28/2022]
Abstract
Arsenic, one of the most significant hazards in the environment affecting millions of people around the world, is associated with several diseases including cancers of skin, lung, urinary bladder, kidney and liver. Groundwater contamination by arsenic is the main route of exposure. Inhalation of airborne arsenic or arsenic-contaminated dust is a common health problem in many ore mines. This review deals with the questions raised in the epidemiological studies such as the dose-response relationship, putative confounders and synergistic effects, and methods evaluating arsenic exposure. Furthermore, it describes the metabolic pathways of arsenic, and its biological modes of action. The role of arsenic in the development of cancer is elucidated in the context of combined epidemiological and biological studies. However, further analyses by means of molecular epidemiology are needed to improve the understanding of cancer aetiology induced by arsenic.
Collapse
Affiliation(s)
- Soile Tapio
- Federal Office for Radiation Protection, Department of Radiation Protection and Health, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany.
| | - Bernd Grosche
- Federal Office for Radiation Protection, Department of Radiation Protection and Health, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| |
Collapse
|
39
|
Gottschalg E, Moore NE, Ryan AK, Travis LC, Waller RC, Pratt S, Atmaca M, Kind CN, Fry JR. Phenotypic anchoring of arsenic and cadmium toxicity in three hepatic-related cell systems reveals compound- and cell-specific selective up-regulation of stress protein expression: implications for fingerprint profiling of cytotoxicity. Chem Biol Interact 2006; 161:251-61. [PMID: 16729991 DOI: 10.1016/j.cbi.2006.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Revised: 04/04/2006] [Accepted: 04/05/2006] [Indexed: 10/24/2022]
Abstract
Exposure of cells to toxic chemicals is known to up-regulate the expression of a number of stress proteins (SPs), including metallothionein (MT) and members of the heat shock protein (HSP) family, and this response may allow the development of a fingerprint profile to identify mechanisms of toxicity in an in vitro toxicology setting. To test this hypothesis, three hepatic-derived cell culture systems (rat hepatoma FGC4 cell line, rat hepatocytes, human hepatoma HepG2 cell line) were exposed to cadmium (as CdCl2) and arsenic (as NaAsO2), two compounds believed to exert their toxicity through an oxidative stress mechanism, under conditions of phenotypic anchoring defined as minimal and mild toxicity (approximately 5 and 25% reduction in neutral red uptake, respectively). The expression of six SPs--MT, HSP25/27, HSP40, HSP60, HSP70, and HSP90--was then determined by ELISA. Expression of four of these SPs--MT, HSP25/27, HSP40 and HSP70--was up-regulated in at least one experimental condition. However, the patterns of expression of these four SPs varied across the experimental conditions, according to differences in toxicant concentration and/or level of toxicity, cell-type and toxicant itself. This lack of uniformity in response of a focussed set of mechanistically defensible targets suggests that similar problems may emerge when using more global approaches based on genomics and proteomics, in which problems of redundancy in targets and uncertain mechanistic relevance will be greater.
Collapse
Affiliation(s)
- Elke Gottschalg
- School of Biomedical Sciences, Medical School (E Floor), Queen's Medical Centre, Nottingham NG7 2UH, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Brink A, Schulz B, Kobras K, Lutz WK, Stopper H. Time-dependent effects of sodium arsenite on DNA breakage and apoptosis observed in the comet assay. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2006; 603:121-8. [PMID: 16384727 DOI: 10.1016/j.mrgentox.2005.10.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Revised: 09/20/2005] [Accepted: 10/27/2005] [Indexed: 12/01/2022]
Abstract
To assess genotoxic effects of sodium arsenite (NaAsO2) the single-cell gel electrophoresis (comet assay) had been conducted in various studies indicating genotoxicity. However, DNA fragmentation due to NaAsO2-induced apoptosis may constitute a bias in the interpretation of the results. Apoptotic cells can show typically large and diffuse comets, which are usually excluded during genotoxicity analysis. It is controversial whether there is a time-window in which the apoptotic process generates comets that would falsely be interpreted to be the result of genotoxic DNA damage. Therefore, we evaluated frequency histograms for single-cell measures of tail DNA (% DNA in comet tail) in 30-min intervals after incubation of mouse lymphoma L5178Y cells with sodium arsenite (NaAsO2). In parallel, we evaluated apoptosis by measuring annexin V-positive cells with flow cytometry, and visualized apoptotic cells on slides by Hoechst bisbenzimide 33258 staining. The first observed effect at 30 min after treatment was an increase in annexin V-positive cells. At about 60 min the number of cells with moderate DNA migration increased in the comet-assay analysis. After 90 min, an increase in the number of cells with high levels of DNA migration was observed, which resulted in a bimodal distribution of cells with moderate and high levels of DNA migration. Hoechst-stained apoptotic cells could only be observed at later times (> or = 120 min). This means that the treatment would have been considered to be genotoxic if analysed at 120 min even if the cells with high levels of DNA migration would have been excluded. The occurrence of annexin V-positive cells preceded the appearance of cells with moderate levels of DNA migration. We hypothesize that these cells were early apoptotic cells and not indicative of genotoxic damage. We conclude that DNA-damaging effects of NaAsO2 cannot adequately be interpreted if the comet assay is not accompanied by separate analysis of early endpoints for induction of apoptosis.
Collapse
Affiliation(s)
- Andreas Brink
- Department of Toxicology, University of Würzburg, Versbacher Str. 9, D-97078 Würzburg, Germany
| | | | | | | | | |
Collapse
|
41
|
Abstract
Metals are essential for the normal functioning of living organisms. Their uses in biological systems are varied, but are frequently associated with sites of critical protein function, such as zinc finger motifs and electron or oxygen carriers. These functions only require essential metals in minute amounts, hence they are termed trace metals. Other metals are, however, less beneficial, owing to their ability to promote a wide variety of deleterious health effects, including cancer. Metals such as arsenic, for example, can produce a variety of diseases ranging from keratosis of the palms and feet to cancers in multiple target organs. The nature and type of metal-induced pathologies appear to be dependent on the concentration, speciation, and length of exposure. Unfortunately, human contact with metals is an inescapable consequence of human life, with exposures occurring from both occupational and environmental sources. A uniform mechanism of action for all harmful metals is unlikely, if not implausible, given the diverse chemical properties of each metal. In this chapter we will review the mechanisms of carcinogenesis of arsenic, cadmium, chromium, and nickel, the four known carcinogenic metals that are best understood. The key areas of speciation, bioavailability, and mechanisms of action are discussed with particular reference to the role of metals in alteration of gene expression and maintenance of genomic integrity.
Collapse
Affiliation(s)
- Troy R Durham
- Centre for Cellular and Molecular Biology, School of Biological and Chemical Sciences, Deakin University, 221 Burwood Highway, Burwood, Victoria, Australia 3125.
| | | |
Collapse
|
42
|
Sidhu JS, Ponce RA, Vredevoogd MA, Yu X, Gribble E, Hong SW, Schneider E, Faustman EM. Cell cycle inhibition by sodium arsenite in primary embryonic rat midbrain neuroepithelial cells. Toxicol Sci 2005; 89:475-84. [PMID: 16251481 DOI: 10.1093/toxsci/kfj032] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Arsenite (As3+) exposure during development has been associated with neural tube defects and other structural malformations, and with behavioral alterations including altered locomotor activity and operant learning. The molecular mechanisms underlying these effects are uncertain. Because arsenic can cross the placenta and accumulate in the developing neuroepithelium, we examined cell cycling effects of sodium arsenite (As3+ 0, 0.5, 1, 2, and 4 microM) on embryonic primary rat midbrain (gestational day [GD] 12) neuroepithelial cells over 48 h. There was a concentration- and time-dependent As3+-induced reduction in cell viability assessed by neutral red dye uptake assay but minimal apoptosis at concentrations below 4 microM. Morphologically, apoptosis was not apparent until 4 microM at 24 h, which was demonstrated by a marginal but statistically significant increase in cleaved caspase-3/7 activity. Cell cycling effects over several rounds of replication were determined by continuous 5-bromo-2'-deoxyuridine (BrdU) labeling and bivariate flow cytometric Hoechst-Propidium Iodide analysis. We observed a time- and concentration-dependent inhibition of cell cycle progression as early as 12 h after exposure (> or =0.5 microM). In addition, data demonstrated a concentration-dependent increase in cytostasis within all cell cycle phases, a decreased proportion of cells able to reach the second cell cycle, and a reduced cell cycle entry from gap 1 phase (G1). The proportion of affected cells and the severity of the cell cycle perturbation, which ranged from a decreased transition probability to complete cytostasis in all cell cycle phases, were also found to be concentration-dependent. Together, these data support a role for perturbed cell cycle progression in As3+ mediated neurodevelopmental toxicity.
Collapse
Affiliation(s)
- Jaspreet S Sidhu
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Kann S, Estes C, Reichard JF, Huang MY, Sartor MA, Schwemberger S, Chen Y, Dalton TP, Shertzer HG, Xia Y, Puga A. Butylhydroquinone protects cells genetically deficient in glutathione biosynthesis from arsenite-induced apoptosis without significantly changing their prooxidant status. Toxicol Sci 2005; 87:365-84. [PMID: 16014739 DOI: 10.1093/toxsci/kfi253] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Arsenic, first among the top environmentally hazardous substances, is associated with skin, lung, liver, kidney, prostate, and bladder cancer. Arsenic is also a cardiovascular and a central nervous system toxicant, and it has genotoxic and immunotoxic effects. Paradoxically, arsenic trioxide is used successfully in the treatment of acute promyelocytic leukemia and multiple myeloma. Arsenic induces oxidative stress, and its toxicity is decreased by free thiols and increased by glutathione depletion. To further characterize the role of glutathione and oxidative stress in the toxicity of arsenic, we have used fetal fibroblasts from Gclm(-/-) mice, which lack the modifier subunit of glutamate-cysteine ligase, the rate-limiting enzyme in glutathione biosynthesis. Gclm(-/-) mouse embryo fibroblasts (MEFs) are eight times more sensitive to arsenite-induced apoptotic death. Because of a dramatic decrease in glutathione levels, Gclm(-/-) MEFs have a high prooxidant status that is not significantly relieved by treatment with the phenolic antioxidant tBHQ; however, tBHQ blocks arsenite-induced apoptosis in both Gclm(+/+) and Gclm(-/-) cells, although it raises a significant antioxidant response only in Gclm(+/+) cells. Global gene expression profiles indicate that tBHQ is significantly effective in reversing arsenite-induced gene deregulation in Gclm(+/+) but not in Gclm(-/-) MEFs. This effect of tBHQ is evident in the expression of metalloproteases and chaperones, and in the expression of genes involved in DNA damage and repair, protein biosynthesis, cell growth and maintenance, apoptosis, and cell cycle regulation. These results suggest that regulation of glutathione levels by GCLM determines the sensitivity to arsenic-induced apoptosis by setting the overall ability of the cells to mount an effective antioxidant response.
Collapse
Affiliation(s)
- Simone Kann
- Center for Environmental Genetics and Department of Environmental Health, University of Cincinnati Medical Center, Ohio 45267-0056, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|